BINDING SITE IN TYPE 2 RYANODINE RECEPTOR

Information

  • Patent Application
  • 20230368862
  • Publication Number
    20230368862
  • Date Filed
    December 07, 2022
    a year ago
  • Date Published
    November 16, 2023
    5 months ago
Abstract
The present disclosure relates to methods and compositions useful for the identification of a ryanodine receptor modulator binding site in ryanodine receptor type 2 (RyR2). The present disclosure also provides compositions useful for the analysis of the ryanodine receptor modulator binding site in RyR2 via cryo-EM. The present disclosure further provides computational methods for identifying compounds that bind to RyR2.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in XML format and is hereby incorporated by reference in its entirety. Said XML copy, created on Dec. 7, 2022, is named 44010111US-PAT.xml and is 14,741 bytes in size.


BACKGROUND

The ryanodine receptor (RyR) is required for excitation-contraction coupling. Although RyR is tightly regulated, inherited mutations and stress-induced post-translational modifications can result in a Ca2+ leak in skeletal myopathies, heart failure, and exercise-induced sudden death. Compounds known as Rycals® repair the leaky RyR and are effective in preventing and treating disease symptoms and restoring normal RyR function.


INCORPORATION BY REFERENCE

All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.


SUMMARY OF THE INVENTION

In some embodiments, the present disclosure provides a composition comprising a complex suspended in a solid medium, the solid medium comprising vitreous ice, wherein the complex comprises a protein and a synthetic compound, wherein the protein is a ryanodine receptor 2 protein (RyR2) or a mutant thereof.


In some embodiments, the synthetic compound is




embedded image


or an ionized form thereof.


In some embodiments, the protein is a mutant RyR2, for example a RyR2 protein containing at least one mutation that is associated with Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT). In some embodiments, the RyR2 protein is post-translationally-modified RyR2 protein, for example a RyR2 protein containing at least one post-translational modification selected from phosphorylation, oxidation and nitrosylation. In some embodiments, the post-translationally modified RyR2 protein is associated with heart failure. In some embodiments, the RyR2 protein is a mutated and post-translationally modified RyR2 protein.


In some embodiments, the present disclosure provides a method for predicting a docked position of a target ligand in a binding site of a biomolecule, the method comprising:

    • receiving a template ligand-biomolecule structure, the template ligand-biomolecule structure comprising a template ligand docked in the binding site of the biomolecule;
    • comparing a pharmacophore model of the template ligand to a pharmacophore model of the target ligand;
    • overlapping the pharmacophore model of the target ligand with the pharmacophore model of the template ligand while the template ligand is in the binding site of the biomolecule; and
    • predicting the docked position of the target ligand in the binding site of the biomolecule based on a position of the pharmacophore model of the target ligand when overlapped with the pharmacophore model of the template ligand,
    • wherein the biomolecule is a RY1&2 domain of RyR2, wherein the template ligand-biomolecule structure is obtained by a process comprising subjecting a complex of the biomolecule and the template ligand to single-particle cryogenic electron microscopy analysis.


In some embodiments, the present disclosure provides a method of identifying a plurality of potential lead compounds, the method comprising the steps of:

    • (a) analyzing, using a computer system, an initial lead compound known to bind to a biomolecular target, the analyzing comprising partitioning, by providing a database of known reactions, the initial lead compound into atoms defining partitioned lead compound comprising a lead compound core and atoms defining a lead compound non-core, wherein the initial lead compound is partitioned using a computational retrosynthetic analysis of the initial lead compound;
    • (b) identifying, using the computer system, a plurality of alternative cores to replace the lead compound core in the initial lead compound, thereby generating a plurality of potential lead compounds each having a respective one of the plurality of alternative cores;
    • (c) calculating, using the computer system, a difference in binding free energy between the partitioned lead compound and each potential lead compound;
    • (d) predicting, using the computer system, whether each potential lead compound will bind to the biomolecular target and identifying a predicted active set of potential lead compounds based on the prediction;
    • (e) obtaining a synthesized set of at least some of the potential leads of the predicted active set to establish a first of potential lead compounds; and
    • (f) determining, empirically, an activity of each of the first set of synthesized potential lead compounds,
    • wherein the biomolecular target is a RY1&2 domain of RyR2, and the structure of the biomolecular target used in the predicting of (d) is obtained by a process comprising subjecting a complex of the biomolecular target and the initial lead compound to single-particle cryogenic electron microscopy analysis.


In some embodiments, the present disclosure provides a computer-implemented method of quantifying binding affinity between a ligand and a receptor molecule, the method comprising:

    • receiving by one or more computers, data representing a ligand molecule,
    • receiving by one or more computers, data representing a receptor molecule domain, using the data representing the ligand molecule and the data representing the receptor molecule domain in computer analysis to identify ring structure within the ligand, the ring structure being an entire ring or a fused ring;
    • using the data representative of the identified ligand ring structure to designate a first ring face and a second ring face opposite to the first ring face, and classifying the ring structure by:
    • a) determining proximity of receptor atoms to atoms on the first face of the ligand ring; and
    • b) determining proximity of receptor atoms to atoms on the second face of the ligand ring;
    • c) determining solvation of the first face of the ligand ring and solvation of the second face of the ligand ring;
    • classifying the identified ligand ring structure as buried, solvent exposed or having a single face exposed to solvent based on receptor atom proximity to and solvation of the first ring face and receptor atom proximity to and solvation of the second ring face;
    • quantifying the binding affinity between the ligand and the receptor molecule domain based at least in part on the classification of the ring structure; and
    • displaying, via computer, information related to the classification of the ring structure,
    • wherein the receptor molecule domain is a RY1&2 domain of RyR2, wherein the data representing a ligand molecule and the data representing a receptor molecule domain are obtained by a process comprising subjecting a complex comprising the ligand molecule and the receptor molecule domain to single-particle cryogenic electron microscopy analysis.


In some embodiments, the present disclosure provides a method comprising:

    • (a) determining an open probability (Po) of a first RyR2 protein, wherein the first RyR2 protein is treated with a test compound, and
    • (b) determining an open probability (Po) of a second RyR2 protein, wherein the second RyR2 protein is not treated with the test compound.


In some embodiments, the present disclosure provides a method comprising:

    • (a) contacting a first RyR2 protein with a test compound;
    • (b) providing a second RyR2 protein;
    • (c) subsequent to the contacting the first RyR2 protein with the test compound, measuring an open probability (Po) of the first RyR2 protein; and
    • (d) measuring an open probability (Po) of the second RyR2 protein.


In some embodiments, the present disclosure provides a method of identifying a compound having RyR2 modulatory activity, the method comprising:

    • (a) determining an open probability (Po) of a RyR2 protein;
    • (b) contacting the RyR2 protein with a test compound;
    • (c) determining an open probability (Po) of the RyR2 protein in the presence of the test compound; and
    • (d) determining a difference between the Po of the RyR2 protein in the presence and absence of the test compound;
    • wherein a reduction in the Po of the RyR2 protein in the presence of the test compound relative to the Po of the RyR2 protein in the absence of the test compound is indicative of the compound having RyR2 modulatory activity.


In some embodiments, the present disclosure provides a method for identifying a compound having RyR2 modulatory activity, comprising:

    • (a) contacting a RyR2 protein with a ligand having known RyR2 modulatory activity to create a mixture, wherein the RyR2 protein is a leaky RyR2, the leaky RyR2 comprising mutant RyR2 protein, post-translationally modified RyR2, or a combination thereof;
    • (b) contacting the mixture of step (a) with a test compound; and
    • (c) determining the ability of the test compound to displace the ligand from the RyR2 protein.


In some embodiments, the present disclosure provides a method for identifying a compound that preferentially binds to a mutated, post-translationally modified RyR2 or a combination thereof, comprising:

    • (a) determining binding affinity of a test compound to a first RyR2 protein, wherein the first RyR2 protein is a wild-type RyR2 protein;
    • (b) determining binding affinity of a test compound to a second RyR2 protein, wherein second first RyR2 protein is a mutant RyR2 protein, a post-translationally modified RyR2, or a combination thereof; and
    • (c) selecting a compound having a higher binding affinity to the second RyR2 protein relative to the first RyR2 protein.





BRIEF DESCRIPTION OF THE DRAWINGS

The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.



FIG. 1A depicts an immunoblot (left, top) and SDS-PAGE (left, middle) of human recombinant RyR2 expressed in HEK293 cells. Ratio of normalized intensities of the pS2808 and total RyR2 bands (right). FIG. 1B depicts an immunoblot (left, top) and SDS-PAGE (left, bottom) of dephosphorylated human recombinant RyR2 expressed in HEK293 cells.



FIGS. 2A-2J provide GSFSCs (top) and viewing angle distributions (middle) of the global nonuniform refinements performed in cryoSPARC before any local refinement for each structure, and FSC model-map performed in PHENIX (bottom) for DeP-RyR2-C (FIG. 2A), DeP-RyR2-O (FIG. 2B), P-RyR2-C (FIG. 2C), P-RyR2-O (FIG. 2D), P-RyR2+CaM-C (FIG. 2E), P-RyR2-R2474S-Pr, (FIG. 2F), P-RyR2-R2474S-O (FIG. 2G), P-RyR2-R2474S+Cpd1-C (FIG. 211), P-RyR2-R2474S+CaM-C (FIG. 2I), and P-RyR2-R2474S+CaM-O (FIG. 2J).



FIG. 3 depicts representative high-resolution details of cryo-EM maps showing the holes in proline and aromatic residues, precise side-chain conformations, and stabilized water molecules (blue arrows) for “primed” PKA RyR2-R2474S.



FIG. 4A and FIG. 4B depict local refinement cryo-EM maps colored by local resolution shown for “primed” PKA RyR2-R2474S.



FIGS. 5A-5K are flowcharts that summarize cryoSPARC processing of cryo-EM datasets to obtain final composite maps of DeP-RyR2 (FIG. 5A and FIG. 5B), P-RyR2 (FIG. 5C), P-RyR2+CaM (FIG. 5D and FIG. 5E), P-RyR2-R2474S (FIG. 5F and FIG. 5G), P-RyR2-R2474S+Cpd1 (FIG. 5H and FIG. 5I), and P-RyR2-R2474S+CaM (FIG. 5J and FIG. 5K).



FIGS. 6A-6C depict aligned atomic models of structures resolved by cryo-EM, focused on the pore and TM domains.



FIGS. 7A-7F show pore radii estimation calculated with HOLE for each structure resolved by cryo-EM.



FIG. 8A and FIG. 8B depict models of RyR2 with their respective cryo-EM maps centered on the ligand binding site of the closed (FIG. 8A) and open (FIG. 8B) state of representative RyR2 structures.



FIG. 9A shows overlapped models of open PKA-phosphorylated RyR2 (P-RyR2-0, PDB: 7U9R, yellow) and closed PKA-phosphorylated RyR2 (P-RyR2-C, PDB: 7U9Q, gray). FIG. 9B shows overlapped models of closed PKA-phosphorylated RyR2 (P-RyR2-C, PDB: 7U9Q, gray) and primed PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S-Pr, PDB: 7U9X, magenta). FIG. 9C shows overlapped models of primed PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S-Pr, PDB: 7U9X, magenta) and closed PKA-phosphorylated RyR2-R2474S+Compound 1 (P-RyR2-R2474S+Cpd1-C, PDB: 7UA1, cyan). FIG. 9D shows overlapped models of primed PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S-Pr, PDB: 7U9X, magenta) and closed PKA-phosphorylated RyR2-R2474S+CaM (P-RyR2-R2474S+CaM-C, PDB: 7UA3, cyan).



FIGS. 10A-10K depict pairwise comparisons of the cytosolic domains of all structures resolved by cryo-EM. Domains are labelled. Conformational changes are shown with arrows. The size of the arrows represents the amount of changes observed.



FIG. 11A depicts cryo-EM maps of closed PKA-phosphorylated RyR2 (gray) and primed PKA-phosphorylated RyR2-R2474S (magenta) from the side (left) and top (right) views. Conformation changes are shown with arrows. FIG. 11B depicts aligned models of closed PKA-phosphorylated RyR2 (PDB: 7U9Q, gray), open PKA-phosphorylated RyR2 (PDB: 7U9R, yellow), and primed PKA-phosphorylated RyR2-R2474S (PDB: 7U9X, magenta). FIG. 11C provides a close-up view of the region around residue 2474 of closed PKA-phosphorylated RyR2 (left) and primed PKA-phosphorylated RyR2-R2474S (right). Conformational changes are shown with arrows. Distances between closed PKA-phosphorylated RyR2 and primed PKA-phosphorylated RyR2-R2474S, and between closed and open PKA-phosphorylated RyR2 (in parentheses) are labeled.



FIG. 12A depicts cryo-EM maps of closed PKA-phosphorylated RyR2 (gray), closed PKA-phosphorylated RyR2-R2474S+Compound 1 (P-RyR2-R2474S+Cpd1-C, PDB: 7UA1, cyan), primed PKA-phosphorylated RyR2-R2474S (magenta) from the side (left) and top (right) views. Conformation changes are shown with arrows. FIG. 12B depicts aligned models of closed PKA-phosphorylated RyR2 (PDB: 7U9Q, gray), closed PKA-phosphorylated RyR2-R2474S+Compound 1 (P-RyR2-R2474S+Cpd1-C, PDB: 7UA1, cyan), open PKA-phosphorylated RyR2 (PDB: 7U9R, yellow), and primed PKA-phosphorylated RyR2-R2474S (PDB: 7U9X, magenta). FIG. 12C shows aligned models of closed PKA-phosphorylated RyR2 (P-RyR2-C, PDB: 7U9Q, gray), primed PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S-Pr, PDB: 7U9X, magenta), and closed PKA-phosphorylated RyR2-R2474S+Compound 1 (P-RyR2-R2474S+Cpd1-C, PDB: 7UA1, cyan). Conformational changes of the RYR1&2 and BSol domains are shown with arrows.



FIG. 13A depicts normalized differences in RMSD of primed PKA-phosphorylated RyR2-R2474S. FIG. 13B depicts normalized differences in RMSD of closed PKA-phosphorylated RyR2-R2474S+Cpd1.



FIG. 14A and FIG. 14B depict cryo-EM maps of local refinement cryoSPRAC jobs before 3D variability of “primed” PKA RyR2-R2474S (magenta), and closed PKA RyR2-R2474S+Compound 1 (cyan) from different views and map levels. FIG. 14C shows the closed PKA RyR2 model (PDB:7U9Q) with the aligned cryo-EM map centered on the RY1&2 domain from the top (top) and side (middle) views. FIG. 14D shows a close up of closed PKA R2474S RyR2+Compound 1 (P-RyR2-R2474S-C, PDB:7UA1) centered on the ryanodine receptor channel modulator binding site. FIG. 14E shows closed PKA RyR2-R2474S+Compound 1 (P-RyR2-R2474S-C, PDB:7UA1) centered on the BSol1-RY1&2 interface. Candidate residues involved in the BSol1-RY1&2 interaction are labeled. FIG. 14F depicts the same comparison provided in FIG. 14E but with distances between sidechains of candidate residues labeled in yellow.



FIG. 15, Panel A shows aligned cryo-EM maps of closed PKA-phosphorylated RyR2 (P-RyR2-C, gray) and closed PKA-phosphorylated RyR2+CaM (cyan). Panel B depicts JSol and CSol models of closed PKA-phosphorylated RyR2 (PDB: 7U9Q, gray) and open PKA-phosphorylated RyR2 (PDB: 7U9R, yellow). Panel C depicts JSol and CSol models of closed PKA-phosphorylated RyR2 (PDB: 7U9Q, gray) and closed PKA-phosphorylated RyR2+CaM (PDB: 7U9T, cyan). Panel D shows aligned cryo-EM maps of primed PKA-phosphorylated RyR2-R2474S (magenta) and closed PKA-phosphorylated RyR2-R2474S+CaM (cyan). Panel E depicts a model with cryo-EM map of closed PKA-phosphorylated RyR2-R2474S+CaM (PDB: 7UA3) centered on the BSol3 domain that is stabilized by CaM.



FIG. 16, Panel A illustrates a RMSD analysis of the closed PKA phosphorylated RyR2 (P-RyR2-C, PDB:7U9Q) vs. the closed (DeP-RyR2-C, PDB:7UA5) and open (DeP-RyR2-0, PDB:7UA9) states of dephosphorylated RyR2. Panel B depicts a 3D variability analysis of the RyR2 structures showing that the dynamic behavior of the RY3&4 domain is independent of the phosphorylation state, pore state, and mutation state. Panel C shows views of the “primed” PKA RyR2-R2474S model (P-RyR2-R2474S-Pr, PDB:7U9X) with the aligned cryo-EM map centered on the RY3&4 domain. Panel D shows the “primed” PKA RyR2-R2474S model (P-RyR2-R2474S-Pr, PDB:7U9X) centered on the interface between the RY3&4 and BSol1/SPRY3 domains. Panel E shows the atomic model of the RY3&4 domain of P-RyR2-R2474S-Pr centered on the poorly resolved phosphorylation loop. Panel F depicts cryo-EM maps of the particles in the closed state with destabilized RY3&4 domain (gray) and stabilized RY3&4 domain (magenta). Panel G depicts cryo-EM maps of the particles in the open state with destabilized RY3&4 domain (gray) and stabilized RY3&4 domain (magenta).



FIG. 17, Panel A and Panel B are cryo-EM maps of the closed particles with destabilized (gray) and stabilized (magenta) RY3&4 domain. A downward shift in surrounding domains was observed. Individual domains are labeled. Panel C and Panel D provide different points of view of the cryo-EM maps depicted in Panel A and Panel B, respectively.



FIG. 18 shows aligned models of the closed state of RyR2 with destabilized (gray) and stabilized (magenta) RY3&4 domain, and open state (yellow).



FIG. 19 is a chart illustrating an RMSD analysis of the closed state of RyR2 with stabilized RY3&4 domain.



FIG. 20A and FIG. 20B show a model with overlapped cryo-EM map of PKA-phosphorylated RyR2 (PDB: 7U9Q) highlighting intramembrane helices laterally positioned and encircling the TM domain from the side view (FIG. 20A) and bottom view (FIG. 20B).



FIG. 21A, shows sequence alignment of residues 4231-4320 between the secondary structure predicted by Jpred and the secondary structure from the cryo-EM-resolved model. FIG. 21B depicts the RyR2 model highlighting Sx helices. FIG. 21C depicts RyR2 model highlighting the interaction between Sx helices and the neighbor helical elements. FIG. 21D depicts RyR2 model highlighting a lysine rich linker. FIG. 21E depicts cryo-EM maps of closed PKA RyR2+CaM (gray), open PKA RyR2 (yellow), open PKA RyR2-R2474S (magenta), and open PKA RyR2-R2474S+CaM (cyan) centered on the Sx helices.



FIG. 22 shows single-channel current recordings traces from recombinant RyR2 at Ca2+150 nM, before (Panel A) and after (Panel B) the addition of xanthine 10 μM.



FIG. 23 depicts representative telemetric electrocardiogram (ECG) recordings of Ryr2R2474S/WT mice (n=4) during arrhythmia provocation stress testing by epinephrine injection (1 mg/kg epinephrine).



FIG. 24 shows SR Ca2+ leak measured in microsomes from Ryr2R2474S/WT mouse heart lysates. The Ca2+ leak was compared for hearts isolated from control Ryr2R2474S/WT mice (gray), Ryr2R2474S/WT mice treated with epinephrine (magenta), and Ryr2R2474S/WT mice treated with epinephrine and Compound 1 (cyan).



FIG. 25 illustrates sequence coverage of RyR2 provided in the mass spectroscopic analysis of hyperphosphorylated channels described in EXAMPLE 10.



FIG. 26, Panels A-C illustrate the proposed mechanism of CPVT-related RyR2 variants, other gain-of-function mutants, and heart failure. Panel A is a schematic representation of the normal function of RyR2. Panel B is a schematic representation of the CPVT-related Ca2+ leak during diastole under intense exercise or stress conditions. Panel C is a schematic representation of the heart failure-related primed state and Ca2+ leak.





DETAILED DESCRIPTION

Located on the sarco/endoplasmic reticulum (SR/ER) membrane, the ryanodine receptor (RyR) is the largest known ion channel, at over two megadaltons, and is the primary mediator of the Ca2+ release required for excitation-contraction coupling in cardiac and skeletal muscle. RyR is required for excitation-contraction coupling. RyR1 is the primary isoform in skeletal muscle while RyR2 is the predominant cardiac isoform. RyR1 and RyR2 are also found in neurons. RyR3 is present where RyR1 and RyR2 are each present, but with significantly lower expression levels. Beyond their expression pattern, RyR1 and RyR2 are unique in how each is activated. In skeletal muscle, RyR1 is activated by the direct, mechanical interaction with the dihydropyridine receptor (DHPR). RyR2 is instead activated by Ca2+ in the process termed calcium-induced calcium release (CICR) in which Ca2+ binding to RyR2 creates a cascade effect as the release of Ca2+ through the RyR creates a high local concentration of Ca2+, which can cause neighboring RyR channels to open. RyR, a tetramer, forms tetrads in muscle tissue and under normal conditions, undergoes cooperative activation through the process termed coupled gating.


The correct activation of RyR, and thus activation of the appropriate downstream Ca2+ signaling pathways, is regulated by multiple ligands and protein interactions. Aside from Ca2+, ATP, and caffeine, RyR also binds calmodulin (CaM). CaM is an inhibitor of ryanodine receptor type 2 (RyR2). CaM can act as either an activator of ryanodine receptor type 1 (RyR1) under low Ca2+ conditions (˜150 nM), such as those at rest, or an inhibitor of RyR1 under high Ca2+ conditions (>1 μM). High Ca2+ conditions occur locally following intracellular Ca2+ release. Calstabin, a second accessory protein, also binds the RyR. This interaction stabilizes the closed state of the channel. In disease states, RyR can be nitrosylated, oxidized and/or phosphorylated to cause calstabin to dissociate from the channel. This dissociation results in Ca2+ leaking into the cytosol and inappropriate triggering of downstream Ca2+ signaling pathways.


RyR comprises three major segments, each composed of several domains. The first, the cytosolic shell, consists of the N-terminal domain (NTD) with two segments (A & B) and an N-terminal solenoid, three SPRY domains, two RYR domains (RY1&2 and RY3&4), and the junctional and bridging solenoids (J-Sol and Br-Sol). The cytosolic shell also houses the calstabin binding site, which binds in a pocket formed by the Br-Sol and the SPRY domains, specifically SPRY1, and calmodulin, which binds on the other side of the Br-Sol from calstabin, with the N-terminal domain of CaM binding along the face of the Br-Sol while the C-terminal domain binds a peptide within a pocket of the Br-Sol.


Although RyR is tightly regulated, inherited mutations and stress-induced post-translational modifications (e.g., phosphorylation, nitrosylation and oxidation) can result in a Ca2+ leak. As a key player in Ca2+ signaling, leaky RyR channels are associated with a wide variety of disease states including skeletal muscle myopathies such as RyR-related myopathy (RYR-RM), dystrophies such as muscular dystrophy (e.g., Duchenne Muscular Dystrophy), cardiac diseases such as heart failure and catecholaminergic polymorphic ventricular tachycardia (CPVT), diabetes, and neurological disorders such as post-traumatic stress disorders (PTSD) and Alzheimer's disease.


Compounds known as ryanodine receptor modulators (also known as Rycals®) can repair leaky RyR and are effective in preventing and treating disease symptoms and restoring normal RyR function. Ryanodine receptor modulators can have efficacy in a host of diseases, both in vitro and in vivo using animal models. Ryanodine receptor modulators can repair the Ca2+ leak by preferentially binding to leaky RyR compared to normal RyR, and causing reassociation of calstabin, thus restabilizing the closed state of the channel. Mutations in RyR have been linked to rare genetic forms of cardiac and skeletal muscle disorders and ryanodine receptor modulators be effective in animal models in these disorders.


Given the structure of several ryanodine receptor modulator compounds, which contain aromatics and charged groups, ryanodine receptor modulators were initially hypothesized to bind near the caffeine binding site based on early cryo-electron microscopy (cryo-EM) structures with limited resolution. Advances in cryo-EM, and particularly direct detection cameras and novel processing methods including local refinement, have dramatically improved the resolution of cryo-EM maps, allowing unambiguous identification of ligand binding sites, including identification of a novel ATP binding site as described herein, and binding sites for Ca2+, and caffeine.


In some embodiments, the present disclosure utilizes cryo-EM techniques to generate a high resolution model of RyR2. In some embodiments, a high resolution model of RyR2 includes a ryanodine receptor modulator (e.g., Compound 1) bound to a ryanodine receptor modulator binding site in the RY1&2 domain of RyR2. In some embodiments, a ryanodine receptor modulator compound binds cooperatively with ATP and stabilizes the closed state of RyR2.


Ca2+, ATP, and xanthine are known to bind within the C-terminal domain (CTD) of RyR2. Disclosed herein is the identification of an additional ATP-binding site, in the periphery of the cytosolic shell of the RyR, in the RY1&2 domain that is comprised within the SPRY domain. In some embodiments, this region is also the ryanodine receptor modulator (Rycal) binding site. As demonstrated herein, ryanodine receptor modulator binding to RyR2 can increase in the presence of ATP. In some embodiments, Compound 1 binds in the RY1&2 domain cooperatively with ATP and stabilizes the closed state of the RyR2 channel despite the presence of activating ligands (Ca2+, ATP, and xanthine).


The present disclosure relates to methods and compositions useful for the identification of a binding site for ryanodine receptor modulators (Rycals) in ryanodine receptor type 2 (RyR2). The present disclosure also provides compositions useful for the analysis of the ryanodine receptor modulator binding site in RyR2 via cryo-EM. The present disclosure further provides methods (e.g., computational methods) for identifying compounds that bind to RyR2. The present disclosure further provides methods for screening for compounds that bind to RyR2 by utilizing a cryo-EM model of RyR2.


Methods of Structural Determination.

Cryogenic electron microscopy (cryo-EM) is a cryomicroscopy technique applied on samples cooled to cryogenic temperatures and embedded in an environment of vitreous water. An aqueous sample solution is applied to a grid-mesh and plunge-frozen in a cryogenic fluid such as liquid ethane or a mixture of liquid ethane and propane.


The structures of the disclosure can be determined using cryo-EM with a sample frozen at a temperature of from about −40° C. to about −280° C. In some embodiments, the cryo-EM used for structural determination uses a sample frozen at a temperature of from about −40° C. to about −100° C., from about −100° C., to about −150° C., from about −150° C. to about −175° C., from about −175° C. to about −200° C., from about −200° C. to about −225° C., from about −225° C. to about −250° C., or from about −250° C. to about −280° C. In some embodiments, the cryo-EM used for structural determination uses a sample frozen at a temperature of from about −40° C. to about −100° C. In some embodiments, the cryo-EM used for structural determination uses a sample frozen at a temperature of from about −150° C. to about −175° C. In some embodiments, the cryo-EM used for structural determination uses a sample frozen at a temperature of from about −175° C. to about −200° C. In some embodiments, the cryo-EM used for structural determination uses a sample frozen at a temperature of from about −250° C. to about −280° C. In some embodiments, the cryo-EM used for structural determination uses a sample frozen at a temperature of about −150° C., about −175° C., about −200° C., about −250° C., or about −280° C. In some embodiments, the cryo-EM used for structural determination uses a sample frozen at a temperature of about −175° C. In some embodiments, the cryo-EM used for structural determination uses a sample frozen at a temperature of about −200° C. In some embodiments, the cryo-EM used for structural determination uses a sample frozen in liquid nitrogen. In some embodiments, the cryo-EM used for structural determination uses a sample frozen in liquid helium. In some embodiments, the cryo-EM used for structural determination uses a sample frozen in liquid ethane. In some embodiments, the cryo-EM used for structural determination uses a sample frozen in liquid propane. In some embodiments, the cryo-EM used for structural determination uses a sample frozen in mixture of liquid nitrogen and liquid propane.


The structures of the disclosure can be determined using a protein concentration of from about 50 nM to about 5 μM. In some embodiments, a structure of the disclosure can be determined using a protein concentration of from about 50 nM to about 250 nM, from about 250 nM to about 500 nM, from about 500 nM to about 750 nM, from about 750 nM to about 1 μM, or from about 1 μM to about 5 μM. In some embodiments, a structure of the disclosure can be determined using a protein concentration of from about 50 nM to about 250 nM. In some embodiments, a structure of the disclosure can be determined using a protein concentration of from about 250 nM to about 500 nM. In some embodiments, a structure of the disclosure can be determined using a protein concentration of from about 500 nM to about 750 nM. In some embodiments, a structure of the disclosure can be determined using a protein concentration of from about 750 nM to about 1 μM. In some embodiments, a structure of the disclosure can be determined using a protein concentration of from about 1 μM to about 5 μM.


The structures of the disclosure can be determined using a sample solution with a pH of about 6.6, about 6.7, about 6.8, about 6.9, about 7.0, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, about 7.6, about 7.7, about 7.8, about 7.9, or about 8.0. In some embodiments, the sample solution has a pH of about 7.0. In some embodiments, the sample solution has a pH of about 7.1. In some embodiments, the sample solution has a pH of about 7.2. In some embodiments, the sample solution has a pH of about 7.3. In some embodiments, the sample solution has a pH of about 7.4. In some embodiments, the sample solution has a pH of about 7.5.


The structures of the disclosure (e.g., compositions comprising RyR2 and a ryanodine receptor modulator such as compound 1 bound to a ryanodine receptor modulator binding site on RyR2, and optionally an ATP molecule bound to an ATP binding site on the RyR2) can be determined at a resolution of from about 15 Å to about 2 Å. In some embodiments, the structures of the disclosure can be determined at a resolution of from about 15 Å to about 12 Å, from about 12 Å to about 9 Å, from about 9 Å to about 6 Å, from about 6 Å to about 5 Å, from about 5 Å to about 4 Å, from about 4 Å to about 3 Å, or from about 3 Å to about 2 Å. In some embodiments, the structures of the disclosure can be determined at a resolution of about 2.45 Å. In some embodiments, the structures of the disclosure is determined at a resolution of about 3.1 Å. In some embodiments, the structures of the disclosure is determined at a resolution from about 2 Å to about 3.5 Å, from about 2 Å to about 3.4 Å, from about 2 Å to about 3.3 Å, from about 2 Å to about 3.2 Å, from about 2 Å to about 3.1 Å, from about 2 Å to about 3 Å, from about 2 Å to about 2.9 Å, from about 2 Å to about 2.8 Å, from about 2 Å to about 2.7 Å, from about 2 Å to about 2.6 Å, from about 2 Å to about 2.5 Å, from about 2.1 Å to about 2.5 Å, from about 2.2 Å to about 2.5 Å, from about 2.3 Å to about 2.5 Å, or from about 2.4 Å to about 2.5 Å.


Compositions Containing Complexes of RyR2.

In some embodiments, the present disclosure provides compositions useful for the determination of the ryanodine receptor modulator binding site in RyR2 via methods such as cryo-EM. In some embodiments, the present disclosure provides a composition comprising a complex suspended in a solid medium, wherein the complex comprises a biomolecule (e.g., a protein) and a synthetic compound, wherein the protein is a ryanodine receptor 2 protein (RyR2) or a mutant thereof.


In some embodiments, the present disclosure provides a composition comprising a complex suspended in a solid medium, the solid medium is or comprises a cryo-electron microscopy grid, wherein the complex comprises a biomolecule (e.g., a protein) and a synthetic compound, wherein the protein is a ryanodine receptor 2 protein (RyR2) or a mutant thereof.


In some embodiments, the present disclosure provides a composition comprising a complex suspended in a non-biological solid medium, wherein the complex comprises a biomolecule (e.g., a protein) and a synthetic compound, wherein the protein is a ryanodine receptor 2 protein (RyR2) or a mutant thereof, optionally with one or more proteins associated with RyR2.


In some embodiments, the composition is prepared by a process, the process comprising vitrifying an aqueous solution that is applied to an electron microscopy grid, wherein the aqueous solution comprises the protein and the synthetic compound.


An electron microscopy grid is a support structure used to insert specimens, for example, for use in an electron microscope. The grid structures can be flat with various suitable materials (e.g., copper, gold, rhodium, nickel, molybdenum, ceramic, etc.) for the grids themselves. In some cases, the grid structure can have plating (e.g., rhodium), coating (e.g., carbon, gold, plastic, silicon nitride, etc.), a suitable thickness (e.g., from 20 to 50 micron), and a suitable diameter (e.g., 3 mm). The grid structures generally have crossing bars and spacings/holes between the bars (e.g., nanometer to micrometer scale holes). The bars can come in various suitable sizes or pitch, patterns (e.g., regular or irregular), and shapes (e.g., numbers or letters built into the grid bars).


In some embodiments, prior to the vitrifying, the aqueous solution is applied to the electron microscopy grid, and excess aqueous solution is removed from the electron microscopy grid by blotting the excess aqueous solution.


In some embodiments, the aqueous solution is dispensed onto the electron microscopy grid from a dispensing apparatus located on the side of the electron microscopy grid opposed to the side abutting blotting material. Once the liquid sample is dispensed onto the cryo-EM grid, the blotting material can pull excess solution through the electron microscopy grid to produce a thin liquid film of the aqueous solution on the electron microscopy grid.


In some embodiments, the vitrifying comprises plunge freezing the aqueous solution applied to the electron microscopy grid into liquid ethane chilled with liquid nitrogen.


In some embodiments, the aqueous solution further comprises a buffering agent. Suitable buffering agents can include, for example, zwitterionic amines, such as TAPS ([tris(hydroxymethyl)methylamino]propanesulfonic acid), Bicine (2-(bis(2-hydroxyethyl)amino)acetic acid), Tris (2-amino-2-(hydroxymethyl)propane-1,3-diol), and Tricine (N-[tris(hydroxymethyl)methyl]glycine), as well as zwitterionic sulfonic acids, such as TAPSO (3-[N-tris(hydroxymethyl)methylamino]-2-hydroxypropanesulfonic acid), HEPES (4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid), TES (2-[[1,3-dihydroxy-2-(hydroxymethyl)propan-2-yl]amino]ethanesulfonic acid), MOPS (3-(N-morpholino)propanesulfonic acid), PIPES (piperazine-N,N′-bis(2-ethanesulfonic acid)), and MES (2-(N-morpholino)ethanesulfonic acid). In some embodiments, the buffering agent is HEPES. In some embodiments, the buffering agent is EGTA.


In some embodiments, the aqueous solution further comprises a phospholipid. In some embodiments, the phospholipid is a phosphatidylcholine, such as, for example, 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1,2-dilinoleoyl-sn-glycero-3-phosphocholine (DLPC), 1,2-dihexanoyl-sn-glycero-3-phosphocholine (DHPC), 1,2-dilauroyl-sn-glycero-3-phosphocholine (DLPC), 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC), 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), or 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC). In some embodiments, the phospholipid is a phosphatidylserine, such as, for example, 1-palmitoyl-2-oleoyl-sn-glycero-3-phospho-L-serine (sodium salt) (POPS), or 1,2-dioleoyl-sn-glycero-3-phospho-L-serine (sodium salt) (DOPS). In some embodiments, the phospholipid is DOPS.


In some embodiments, the aqueous solution further comprises a surfactant. Surfactants can be used in a composition disclosed herein to increase the solubility of a protein (e.g. RyR2). In some embodiments, the surfactant is a zwitterionic surfactant, such as, for example, 3-[(3-cholamidopropyl)dimethylammonio]-1-propanesulfonate (CHAPS) or 3-([3-Cholamidopropyl]dimethylammonio)-2-hydroxy-1-propanesulfonate (CHAPSO). In some embodiments, the zwitterionic surfactant is CHAPS.


In some embodiments, the aqueous solution further comprises a disulfide-reducing agent, which can be, for example, tris (2-carboxyethyl) phosphine hydrochloride (TCEP), beta-mercaptoethanol (BME), tributylphosphine (TBP). or dithiothreitol (DTT). In some embodiments, the disulfide-reducing agent is TCEP.


In some embodiments, the aqueous solution further comprises a protease inhibitor. Suitable protease inhibitors can include, for example, 4-(2-aminoethyl)benzenesulfonyl fluoride (AEBSF), phenylmethylsulfonyl fluoride (PMSF), leupeptin, N-ethylmaleimide, antipain, pepstatin, alpha 2-macro-globulin, EDTA, bestatin, amastatin, and benzamidine. In some embodiments, the protease inhibitor is AEBSF. In some embodiments, the protease inhibitor is benzamidine hydrochloride.


In some embodiments, the aqueous solution further comprises xanthine. The concentration of xanthine in the aqueous solution can be, for example, about 1 mM to about 1000 μM, about 1 μM to about 900 μM, about 1 μM to about 800 μM, about 1 μM to about 700 μM, about 1 μM to about 600 μM, about 100 μM to about 1000 μM, about 100 μM to about 900 μM, about 100 μM to about 800 μM, about 100 μM to about 700 μM, about 100 μM to about 600 μM, about 200 μM to about 1000 μM, about 200 μM to about 900 μM, about 200 μM to about 800 μM, about 200 μM to about 700 μM, or about 200 μM to about 600 μM. In some embodiments, caffeine is present at a concentration of from about 200 μM to about 600 μM.


In some embodiments, xanthine is present at a concentration of about 100 μM, about 200 μM, about 300 μM, about 400 μM, about 500 μM, about 600 μM, about 700 μM, about 800 μM, about 900 μM, or about 1000 μM. In some embodiments, caffeine is present at a concentration of about 500 μM.


In some embodiments, the aqueous solution further comprises dissolved Ca2+. The concentration of dissolved Ca2+ in the aqueous solution can be, for example, about 1 μM to about 400 μM, about 1 μM to about 350 μM, about 1 μM to about 300 μM, about 1 μM to about 250 μM, about 1 μM to about 200 μM, about 50 μM to about 400 μM, about 50 μM to about 350 μM, about 50 μM to about 300 μM, about 50 μM to about 250 μM, or about 50 μM to about 200 μM. In some embodiments, dissolved Ca2+ is present at a concentration from about 50 μM to about 250 μM. In some embodiments, dissolved Ca2+ is present at a concentration from about 100 μM to about 200 μM.


In some embodiments, dissolved Ca2+ is present at a concentration of about 100 μM, about 110 μM, about 120 μM, about 130 μM, about 140 μM, about 150 μM, about 160 μM, about 170 μM, about 180 μM, about 190 μM, or about 200 μM. In some embodiments, dissolved Ca2+ is present at a concentration of about 150 μM.


The concentration of the protein in the aqueous solution can be, for example about 1 mg/mL to about 20 mg/mL, 2 mg/mL to about 20 mg/mL, 3 mg/mL to about 20 mg/mL, 4 mg/mL to about 20 mg/mL, 4 mg/mL to about 20 mg/mL, 1 mg/mL to about 15 mg/mL, 2 mg/mL to about 15 mg/mL, 3 mg/mL to about 15 mg/mL, 4 mg/mL to about 15 mg/mL, 1 mg/mL to about 10 mg/mL, 2 mg/mL to about 10 mg/mL, or 3 mg/mL to about 10 mg/mL. In some embodiments, the protein is present in the aqueous solution at a concentration from about 1 mg/mL to about 15 mg/mL. In some embodiments, the protein is present in the aqueous solution at a concentration from about 1 mg/mL to about 10 mg/mL. In some embodiments, the protein is present in the aqueous solution at a concentration from about 4 mg/mL to about 8 mg/mL.


In some embodiments, the protein is present in the aqueous solution at a concentration of about 1 mg/mL, about 2 mg/mL, about 3 mg/mL, about 4 mg/mL, about 5 mg/mL, about 6 mg/mL, about 7 mg/mL, about 8 mg/mL, about 9 mg/mL, about 10 mg/mL, about 11 mg/mL, about 12 mg/mL, about 13 mg/mL, about 14 mg/mL, about 15 mg/mL, about 16 mg/mL, about 17 mg/mL, about 18 mg/mL, about 19 mg/mL, or about 20 mg/mL.


In some embodiments, the aqueous solution further comprises sodium adenosine triphosphate (NaATP). The concentration of NaATP in the aqueous solution can be, for example, about 1 mM to about 15 mM, about 1 mM to about 50 mM, about 1 mM to about 30 mM, about 1 mM to about 30 mM, about 2 mM to about 30 mM, about 3 mM to about 30 mM, about 4 mM to about 30 mM, about 5 mM to about 30 mM, about 6 mM to about 30 mM, about 7 mM to about 30 mM, about 8 mM to about 30 mM, about 9 mM to about 30 mM, about 10 mM to about 30 mM, 1 mM to about 15 mM, about 2 mM to about 15 mM, about 3 mM to about 15 mM, about 4 mM to about 15 mM, or about 5 mM to about 15 mM. In some embodiments, NaATP is present in the aqueous solution at a concentration from about 3 mM to about 15 nM.


In some embodiments, NaATP is present in the aqueous solution at a concentration of about 5 mM, about 6 mM, about 7 mM, about 8 mM, about 9 mM, about 10 mM, about 11 mM, about 12 mM, about 13 mM, about 14 mM, about 15 mM, about 16 mM, about 17 mM, about 18 mM, about 19 mM, about 20 mM, about 25 mM, about 30 mM, about 35 mM, about 40 mM, about 45 mM, or about 50 mM. In some embodiments, the concentration of NaATP is about 10 mM.


In some embodiments, the aqueous solution further comprises cAMP (cyclic adenosine monophosphate). The concentration of cAMP in the aqueous solution can be, for example, about 1 μM to about 400 μM, about 1 μM to about 350 μM, about 1 μM to about 300 μM, about 1 μM to about 250 μM, about 1 μM to about 200 μM, about 50 μM to about 400 μM, about 50 μM to about 350 μM, about 50 μM to about 300 μM, or about 50 μM to about 250 μM. In some embodiments, cAMP is present the aqueous solution at a concentration from about 100 μM to about 300 μM. In some embodiments, cAMP is present at the aqueous solution a concentration from about 150 μM to about 250 μM.


In some embodiments, the aqueous solution is substantially free of cellular membrane. Prior to adding protein to the solution, the protein can be separated from cellular membranes by homogenization of cells containing the protein, and subjecting the resulting homogenate to chromatography.


In some embodiments, the aqueous solution further comprises calmodulin. In some embodiments, the calmodulin is human calmodulin. In some embodiments, the calmodulin has a sequence according to SEQ ID NO: 1.


The aqueous solution can be prepared or stored in a vessel. In some embodiments, the vessel is a vial, ampule, test tube, or microwell plate.


In some embodiments, the complex further comprises a nucleoside-containing molecule. In some embodiments, the nucleoside-containing molecule is a purine nucleoside-containing molecule. In some embodiments, the nucleoside-containing molecule is a nucleotide or nucleoside polyphosphate. In some embodiments, the nucleoside-containing molecule is an adenosine triphosphate (ATP) molecule.


In some embodiments, the nucleoside-containing molecule and the synthetic compound bind a RYR domain of the protein. In some embodiments, the RYR domain is a RY1&2 domain. In some embodiments, the RY1&2 domain has a three-dimensional structure according to TABLE 3. In some embodiments, the synthetic compound has a three-dimensional conformation according to TABLE 4. In some embodiments, the ATP molecule has a three-dimensional conformation according to TABLE 5. In some embodiments, the ATP molecule binds the protein and the synthetic compound. In some embodiments, the synthetic compound binds cooperatively with the ATP molecule in the RY 1&2 domain of RyR2. In some embodiments, the synthetic compound is a ryanodine receptor modulator, e.g., Compound 1.


In some embodiments, the complex further comprises a second ATP molecule, wherein both ATP molecules bind a common RYR domain of the protein.


In some embodiments, the complex further comprises a second binding site for a nucleoside-containing molecule. In some embodiments, the complex further comprises a second nucleoside-containing molecule. In some embodiments, the second nucleoside-containing molecule binds a C-terminal domain of the RyR2 protein. In some embodiments, the second nucleoside-containing molecule is a nucleotide or nucleoside polyphosphate. In some embodiments, the second nucleoside-containing molecule is a second ATP molecule.


In some embodiments, the complex further comprises calmodulin. In some embodiments, the calmodulin is human calmodulin.


In some embodiments, the complex further comprises calstabin (i.e., peptidyl-prolyl cis-trans isomerase FKBP1B). In some embodiments, the calstabin is human calstabin. In some embodiments, the calstabin has a sequence according to SEQ ID NO: 2.


In some embodiments, the RyR2 protein is in a resting (closed) state. In some embodiments, the RyR2 protein is in the primed state. In some embodiments, a primed state comprises a higher distribution of open probability (Po) as compared to a RyR2 in a resting (closed) state. In some embodiments, a primed state RyR2 comprises about 30% to about 60% of the RyR channel in an open state. In some embodiments, a primed state RyR2 comprises about 30%, about 35%, about 40%, about 45%, about 50%, about 55% or about 60% of the RyR channel in an open state.


In some embodiments, the complex further comprises a xanthine alkaloid molecule. In some embodiments, the complex further comprises a xanthine molecule, such as, for example, theobromine, theophylline, caffeine, or xanthine. In some embodiments, the complex further comprises a Ca2+ ion.


In some embodiments, the solid medium comprises vitreous ice. In some embodiments, the solid medium is substantially free of crystalline ice.


In some embodiments, the composition is substantially free of cellular membrane. In some embodiments, the RyR2 is a purified RyR2. In some embodiments, the RyR2 is a semi-purified RyR2 that is substantially free of cellular membrane.


In some embodiments, the composition further comprises additional complexes, wherein each of the additional complexes independently comprises the protein and the synthetic compound. In some embodiments, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or at least about 90% of the RyR2 protein in the additional complexes is in a closed state. In some embodiments, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or at least about 90% of the additional complexes is in an open state.


In some embodiments, the RyR2 protein is in a phosphorylated state, wherein the phosphorylated state is prepared by a process comprising contacting RyR2 protein with a phosphorylation reagent. In some embodiments, the phosphorylation reagent comprises protein kinase A. In some embodiments, the phosphorylation reagent further comprises ATP. In some embodiments, the phosphorylation reagent further comprises MgCl2.


In some embodiments, the protein is in a dephosphorylated state, wherein the dephosphorylated state is prepared by a process comprising contacting RyR2 protein with a dephosphorylation reagent. In some embodiments, the dephosphorylation reagent comprises phosphatase lambda. In some embodiments, the dephosphorylation reagent further comprises MnCl2.


In some embodiments, the synthetic compound binds a RYR domain of the protein. In some embodiments, the RYR domain is a RY1&2 domain.


In some embodiments, the protein is wild type RyR2. In some embodiments, the protein is mutant RyR2. In some embodiments, the mutant RyR2 is R2474S RyR2. In some embodiments, the protein is human RyR2. In some embodiments, the protein is a tetramer of RyR2 monomers, wherein each RyR2 monomer is SEQ ID NO: 3. In some embodiments, the protein is a tetramer of RyR2 monomers, wherein each RyR2 monomer is SEQ ID NO: 4. In some embodiments, the RyR2 protein is C4-symmetrical. In some embodiments, the protein comprises four RY1&2 domains, each with a three-dimensional conformation according to TABLE 3.


In some embodiments, the RyR2 protein is in a closed state. In some embodiments, the RyR2 protein is in an open state. In some embodiments, the RyR2 protein is in a primed state, wherein the RyR2 in the primed state has an open probability (Po) that is higher an open probability (Po) of the RyR2 in a closed state, and an open probability (Po) that is lower than an open probability (Po) of the RyR2 protein in an open state. In some embodiments, the protein is wild type RyR2. In some embodiments, the protein is a mutant RyR2. In some embodiments, the mutant RyR2 contains at least one mutation that is associated with Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT). In some embodiments, the mutation is R2474S. In some embodiments, the mutation is RyR2-R420Q. In some embodiments, the mutation is RyR2-R420W.


In some embodiments, the protein is a post-translationally modified RyR2 protein. In some embodiments, the post-translationally modified RyR2 protein is a phosphorylated RyR2 protein. In some embodiments, the post-translationally modified RyR2 is an oxidized RyR2 protein. In some embodiments, the post-translationally modified RyR2 is a nitrosylated RyR2. In some embodiments, the post-translationally modified RyR2 protein is associated with a cardiac disease. In some embodiments, the post-translationally modified RyR2 protein is associated with heart failure. In some embodiments, the post-translationally modified RyR2 protein is associated with a cardiac arrhythmia. In some embodiments, the RyR2 is a mutated and post-translationally modified RyR2


In some embodiments, the mutation destabilizes an interaction between NTD and BSol domains of the RyR2 protein. In some embodiments, the mutation destabilizes a cytosolic shell of the RyR2 protein, wherein the cytosolic shell comprises NTD, SPRY, JSol and BSol domains of the RyR2 proteins.


The relative difference in conformational states between a structure (e.g., a protein structure or protein domain structure) and a reference structure (e.g., a reference protein structure or a reference protein domain structure) can be quantified by calculating the average distance between atoms of the structure and the reference structure when the structure and reference structure are superimposed. A nonlimiting example of a measure of a difference in conformational states between a structure and a reference structure is root mean square deviation of atomic positions (RMSD), which can be defined as:






RMSD
=



1
N






i
=
1

N


δ
i
2








where δi is the distance between atom i and an analogous atom of a reference structure, or the mean position of the N equivalent atoms.


Root mean square deviation of atomic positions (RMSD) of a protein or protein domain structure relative to a reference protein or protein domain structure can be calculated computationally via software such as, for example, UCSF ChimeraX, SuperPose. LGA (Local-Global Alignment), or PDBeFold. In some embodiments, RMSD is calculated on the basis of Ca atomic coordinates of the structure and reference structure.


In some embodiments, the present disclosure provides a composition comprising a complex suspended in a solid medium, wherein the complex comprises a protein. In some embodiments, the present disclosure provides a composition comprising a complex suspended in vitreous ice, wherein the complex comprises a protein. In some embodiments, the protein is a ryanodine receptor protein or a mutant thereof. In some embodiments, the protein is a ryanodine receptor 2 protein (RyR2) or a mutant thereof. The composition can be, for example, a composition suitable for analysis via single-particle cryogenic electron microscopy, where the solid medium is an amorphous material such as vitreous ice.


In some embodiments, the composition is analyzed in a study, wherein the study comprises: (i) determining a structure of the protein (e.g., RyR2 or a mutant thereof) or a domain of the protein by subjecting the complex to single particle cryogenic electron microscopy analysis; and (ii) calculating RMSD of the protein or domain of the protein relative to a reference structure. In some embodiments, the protein is a RyR2 mutant (e.g, R2474S RyR2, RyR2-R420Q, or RyR2-R420W), and the reference structure is a structure of a wild type RyR2 protein. In some embodiments, the complex further comprises a synthetic compound (e.g., a ryanodine receptor channel modulator).


In some embodiments, the reference structure is obtained by a process comprising analyzing a reference composition via single particle cryogenic electron microscopy, wherein the reference composition comprises a reference complex suspended in a solid medium, wherein the reference complex comprises a reference protein. In some embodiments, the reference composition is prepared by a process comprising vitrifying a reference aqueous solution applied to an electron microscopy grid, wherein the reference aqueous solution comprises the reference complex. The reference aqueous solution can be identical to the aqueous solution used to prepare the composition, except that (a) the reference aqueous solution comprises the reference protein in place of the protein, and/or (b) the aqueous solution comprises a synthetic compound, the reference aqueous solution comprises calmodulin, the reference aqueous solution does not comprise the synthetic compound, and the aqueous solution does not comprise calmodulin. In some embodiments, the reference structure is a structure according to Protein Data Bank entry 7U9Q.


In some embodiments, the protein is R2474S RyR2, wherein if a study is conducted, the study comprising:

    • (i) determining a structure of a BSol2 domain of the protein in the complex by subjecting the complex to single particle cryogenic electron microscopy analysis, and
    • (ii) calculating root mean square deviation of atomic positions (RMSD) of the BSol2 domain of the protein relative to a BSol2 domain of a reference structure, wherein the reference structure is a structure of a wild type RyR2 protein in a closed state,
    • then the RMSD is no more than about 4.5, no more than about 4, no more than about 3.5, or no more than about 3.


In some embodiments, the protein is R2474S RyR2, wherein if a study is conducted, the study comprising:

    • (i) determining a structure of a BSol2 domain of the protein in the complex by subjecting the complex to single particle cryogenic electron microscopy analysis, and
    • (ii) calculating root mean square deviation of atomic positions (RMSD) of the BSol2 domain of the protein relative to a BSol2 domain of a reference structure, wherein the reference structure is a structure a wild type RyR2 protein in a closed state,
    • then the RMSD is from about 1 to about 4.5, about 1 to about 4, about 1 to about 3.5, about 1 to about 3, about 2 to 4.5, about 2 to about 4, about 2 to about 3.5, or about 2 to about 3.


In some embodiments, the protein is R2474S RyR2, wherein if a study is conducted, the study comprising:

    • (i) determining a structure of a BSol domain of the protein in the complex by subjecting the complex to single particle cryogenic electron microscopy analysis, and
    • (ii) calculating root mean square deviation of atomic positions (RMSD) of the BSol domain of the protein relative to a BSol domain of a reference structure, wherein the reference structure is a structure a wild type RyR2 protein in a closed state,
    • then the RMSD is no more than about 3, no more than about 2.5, no more than about 2, or no more than about 1.5.


In some embodiments, the protein is R2474S RyR2, wherein if a study is conducted, the study comprising:

    • (i) determining a structure of a BSol domain of the protein in the complex by subjecting the complex to single particle cryogenic electron microscopy analysis, and
    • (ii) calculating root mean square deviation of atomic positions (RMSD) of the BSol domain of the protein relative to a BSol domain of a reference structure, wherein the reference structure is a structure a wild type RyR2 protein in a closed state,
    • then the RMSD is from about 0.5 to about 3, about 0.5 to about 2.5, about 0.5 to about 2, about 0.5 to about 1.5, about 1 to 3, about 1 to about 2.5, about 1 to about 2, or about 1 to about 1.5.


In some embodiments, the protein is R2474S RyR2, wherein if a study is conducted, the study comprising:

    • (i) determining a structure of a NTD domain of the protein in the complex by subjecting the complex to single particle cryogenic electron microscopy analysis, and
    • (ii) calculating root mean square deviation of atomic positions (RMSD) of the NTD domain of the protein relative to a NTD domain of a reference structure, wherein the reference structure is a structure a wild type RyR2 protein in a closed state,
    • then the RMSD is no more than about 1.5, no more than about 1.4, no more than about 1.3, no more than about 1.2, no more than about 1.1, or no more than about 1.


In some embodiments, the protein is R2474S RyR2, wherein if a study is conducted, the study comprising:

    • (i) determining a structure of a NTD domain of the protein in the complex by subjecting the complex to single particle cryogenic electron microscopy analysis, and
    • (ii) calculating root mean square deviation of atomic positions (RMSD) of the NTD domain of the protein relative to a NTD domain of a reference structure, wherein the reference structure is a structure a wild type RyR2 protein in a closed state,
    • then the RMSD is from about 0.5 to about 1.6, about 0.5 to about 1.5, about 0.5 to about 1.4, about 0.5 to about 1.3, or about 0.5 to about 1.2.


In some embodiments, the protein is R2474S RyR2, wherein if a study is conducted, the study comprising:

    • (i) determining a structure of a SPRY domain of the protein in the complex by subjecting the complex to single particle cryogenic electron microscopy analysis, and
    • (ii) calculating root mean square deviation of atomic positions (RMSD) of the SPRY domain of the protein relative to a SPRY domain of a reference structure, wherein the reference structure is a structure a wild type RyR2 protein in a closed state,
    • then the RMSD is less than about 1.2, less than about 1.1, or less than about 1, less than about 0.9, less than about 0.8, or less than about 0.7.


In some embodiments, the protein is R2474S RyR2, wherein if a study is conducted, the study comprising:

    • (i) determining a structure of a SPRY domain of the protein in the complex by subjecting the complex to single particle cryogenic electron microscopy analysis, and
    • (ii) calculating root mean square deviation of atomic positions (RMSD) of the SPRY domain of the protein relative to a SPRY domain of a reference structure, wherein the reference structure is a structure a wild type RyR2 protein in a closed state,


      then the RMSD is from about 0.2 to about 1.3, about 0.2 to about 1.2, about 0.2 to about 1.1, about 0.2 to about 1, or about 0.2 to about 0.9.


In some embodiments, the reference protein is the wild type RyR2 protein. In some embodiments, the protein and the reference protein are each independently in a phosphorylated state. In some embodiments, the phosphorylated state of the protein or the reference protein is prepared by a process comprising contacting RyR2 protein or a mutant thereof with a phosphorylation reagent. In some embodiments, the phosphorylation reagent is protein kinase A. In some embodiments, the protein and the reference protein are each independently in a dephosphorylated state. In some embodiments, the dephosphorylated state of the protein or the reference protein is prepared by a process comprising contacting RyR2 protein or a mutant thereof with a dephosphorylation reagent. In some embodiments, the dephosphorylation reagent comprises phosphatase lambda.


In some embodiments, the protein and the reference protein are each independently in an oxidized state. In some embodiments, the protein and the reference protein are each independently in a nitrosylated state.


Compounds of the Disclosure.

The synthetic compound in the compositions described herein can be a ryanodine receptor modulator compound, such as a benzothiazepane derivative. Some benzothiazepine compounds are voltage-gated Ca2+ channel blockers, but ryanodine receptor modulator compounds can be free of any channel blocking activity. The inability of certain ryanodine receptor modulator compounds to block Ca2+ channels can be associated with the mechanism of stabilizing the closed state of the RyR without inhibiting the channel. In some embodiments, a ryanodine receptor modulator compounds are modulators of the RyR channel. In some embodiments, ryanodine receptor modulator compounds are allosteric modulators of the RyR channel.


Ryanodine receptor modulator compounds of the disclosure can be used as therapeutics because in some disease states, RyR leaks Ca2+ due to destabilization of the closed state of the channel after post-translational modifications such as nitrosylation, oxidation and phosphorylation. In other disease states, Ca2+ leak is present due to inherited mutations. The genetic mutations can predispose the RyR channel to post-translational modifications such as oxidation and nitrosylation, further exacerbating the leak. These mutations and post-translational modifications cause the stabilizing subunit, calstabin, to dissociate from the channel, increasing the open probability of the channel, resulting in Ca2+ leak. In disease models involving leaky RyR in cells, animals, and patients, treatment with a ryanodine receptor modulator compound can reverse the leak and restore calstabin binding.


In some embodiments, the synthetic compound comprises a benzazepane or benzothiazepane (e.g., 2,3,4,5-tetrahydro-1,4-benzothiazepine) moiety. In some embodiments, the synthetic compound comprises a benzothiazepane moiety. In some embodiments, the synthetic compound comprises a benzothiazepine moiety. In some embodiments, the synthetic compound comprises a 1,4-benzothiazepine moiety.


Chemical Groups.

The term “alkyl” as used herein refers to a linear or branched, saturated hydrocarbon having from 1 to 6 carbon atoms. Representative alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tert-butyl, pentyl, isopentyl, neopentyl, hexyl, isohexyl, and neohexyl. The term “C1-C4 alkyl” refers to a straight or branched chain alkane (hydrocarbon) radical containing from 1 to 4 carbon atoms, such as methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl, and isobutyl.


The term “alkenyl” as used herein refers to a linear or branched hydrocarbon having from 2 to 6 carbon atoms and having at least one carbon-carbon double bond. In one embodiment, the alkenyl has one or two double bonds. The alkenyl moiety may exist in the E or Z conformation and the compounds of the present invention include both conformations.


The term “alkynyl” as used herein refers to a linear or branched hydrocarbon having from 2 to 6 carbon atoms and having at least one carbon-carbon triple bond.


The term “aryl” as used herein refers to an aromatic group containing 1 to 3 aromatic rings, either fused or linked.


The term “cyclic group” as used herein includes a cycloalkyl group and a heterocyclic group.


The term “cycloalkyl” as used herein refers to a three- to seven-membered saturated or partially unsaturated carbon ring. Any suitable ring position of the cycloalkyl group may be covalently linked to the defined chemical structure. Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl.


The term “halogen” as used herein refers to fluorine, chlorine, bromine, and iodine.


The term “heterocyclic group” or “heterocyclic” or “heterocyclyl” or “heterocyclo” as used herein refers to fully saturated, or partially or fully unsaturated, including aromatic (i.e., “heteroaryl”) cyclic groups (for example, 4 to 7 membered monocyclic, 7 to 11 membered bicyclic, or 10 to 16 membered tricyclic ring systems) which have at least one heteroatom in at least one carbon atom-containing ring. Each ring of the heterocyclic group containing a heteroatom may have 1, 2, 3, or 4 heteroatoms selected from nitrogen atoms, oxygen atoms and/or sulfur atoms, where the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized. The heterocyclic group may be attached to the remainder of the molecule at any heteroatom or carbon atom of the ring or ring system. Examples of heterocyclic groups include, but are not limited to, azepanyl, azetidinyl, aziridinyl, dioxolanyl, furanyl, furazanyl, homo piperazinyl, imidazolidinyl, imidazolinyl, isothiazolyl, isoxazolyl, morpholinyl, oxadiazolyl, oxazolidinyl, oxazolyl, oxazolidinyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, piperazinyl, piperidinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazolyl, pyridoimidazolyl, pyridothiazolyl, pyridinyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, quinuclidinyl, tetrahydrofuranyl, thiadiazinyl, thiadiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiomorpholinyl, thiophenyl, triazinyl, and triazolyl. Examples of bicyclic heterocyclic groups include indolyl, isoindolyl, benzothiazolyl, benzoxazolyl, benzoxadiazolyl, benzothienyl, quinuclidinyl, quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuryl, benzofurazanyl, chromonyl, coumarinyl, benzopyranyl, cinnolinyl, quinoxalinyl, indazolyl, pyrrolopyridyl, furopyridinyl (such as furo[2,3-c]pyridinyl, furo[3,2-b]pyridinyl] or furo[2,3-b]pyridinyl), dihydroisoindolyl, dihydroquinazolinyl (such as 3,4-dihydro-4-oxo-quinazolinyl), triazinylazepinyl, tetrahydroquinolinyl and the like. Examples of tricyclic heterocyclic groups include carbazolyl, benzidolyl, phenanthrolinyl, acridinyl, phenanthridinyl, xanthenyl and the like.


The term “phenyl” as used herein refers to a substituted or unsubstituted phenyl group.


The aforementioned terms “alkyl,” “alkenyl,” “alkynyl,” “aryl,” “phenyl,” “cyclic group,” “cycloalkyl,” “heterocyclyl,” “heterocyclo,” and “heterocycle” can further be optionally substituted with one or more substituents. Examples of substituents include but are not limited to one or more of the following groups: hydrogen, halogen, CF3, OCF3, cyano, nitro, N3, oxo, cycloalkyl, alkenyl, alkynyl, heterocycle, aryl, alkylaryl, heteroaryl, ORa, SRa, S(═O)Re, S(═O)2Re, P(═O)2Re, S(═O)2ORa, P(═O)2ORa, NRbRc, NRbS(═O)2Re, NRbP(═O)2Re, S(═O)2NRbRc, P(═O)2NRbRc, C(═O)ORa, C(═O)Ra, C(═O)NRbRc, OC(═O)Ra, OC(═O)NRbRc, NR C(═O)ORa, NRdC(═O)NRbRc, NRdS(═O)2NRbRc, NRdP(═O)2NRbRc, NRbC(═O)Ra, or NRbP(═O)2Re, wherein Ra is hydrogen, alkyl, cycloalkyl, alkenyl, alkynyl, alkylaryl, heteroaryl, heterocycle, or aryl; Rb, Rc and Rd are independently hydrogen, alkyl, cycloalkyl, alkylaryl, heteroaryl, heterocycle, aryl, or said Rb and Rc, together with the N to which Rb and Rc are bonded optionally form a heterocycle; and Rc is alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, alkylaryl, heteroaryl, heterocycle, or aryl. In the aforementioned examples of substituents, groups such as alkyl, cycloalkyl, alkenyl, alkynyl, cycloalkenyl, alkylaryl; heteroaryl, heterocycle and aryl can themselves be optionally substituted.


Example substituents can further optionally include at least one labeling group, such as a fluorescent, a bioluminescent, a chemiluminescent, a colorimetric and a radioactive labeling group. A fluorescent labeling group can be selected from bodipy, dansyl, fluorescein, rhodamine, Texas red, cyanine dyes, pyrene, coumarins, Cascade Blue™, Pacific Blue, Marina Blue, Oregon Green, 4′,6-Diamidino-2-phenylindole (DAPI), indopyra dyes, lucifer yellow, propidium iodide, porphyrins, arginine, and variants and derivatives thereof. For example, ARM118 of the present invention contains a labeling group BODIPY, which is a family of fluorophores based on the 4,4-difluoro-4-bora-3a,4a-diaza-s-indacene moiety. For further information on fluorescent label moieties and fluorescence techniques, see, e.g., Handbook of Fluorescent Probes and Research Chemicals, by Richard P. Haughland, Sixth Edition, Molecular Probes, (1996), which is hereby incorporated by reference in its entirety. One of skill in the art can readily select a suitable labeling group, and conjugate such a labeling group to any of the compounds of the invention, without undue experimentation.


Pharmaceutically Acceptable Salts.

The disclosure provides the use of pharmaceutically-acceptable salts of any compound described herein. Pharmaceutically-acceptable salts include, for example, acid-addition salts and base-addition salts. The acid that is added to the compound to form an acid-addition salt can be an organic acid or an inorganic acid. A base that is added to the compound to form a base-addition salt can be an organic base or an inorganic base. In some embodiments, a pharmaceutically-acceptable salt is a metal salt. In some embodiments, a pharmaceutically-acceptable salt is an ammonium salt.


Metal salts can arise from the addition of an inorganic base to a compound of the disclosure. The inorganic base consists of a metal cation paired with a basic counterion, such as, for example, hydroxide, carbonate, bicarbonate, or phosphate. The metal can be an alkali metal, alkaline earth metal, transition metal, or main group metal. In some embodiments, the metal is lithium, sodium, potassium, cesium, cerium, magnesium, manganese, iron, calcium, strontium, cobalt, titanium, aluminum, copper, cadmium, or zinc.


In some embodiments, a metal salt is a lithium salt, a sodium salt, a potassium salt, a cesium salt, a cerium salt, a magnesium salt, a manganese salt, an iron salt, a calcium salt, a strontium salt, a cobalt salt, a titanium salt, an aluminum salt, a copper salt, a cadmium salt, or a zinc salt.


Ammonium salts can arise from the addition of ammonia or an organic amine to a compound of the present disclosure. In some embodiments, the organic amine is triethyl amine, diisopropyl amine, ethanol amine, diethanol amine, triethanol amine, morpholine, N-methylmorpholine, piperidine, N-methylpiperidine, N-ethylpiperidine, dibenzylamine, piperazine, pyridine, pyrazole, imidazole, or pyrazine.


In some embodiments, an ammonium salt is a triethyl amine salt, a trimethyl amine salt, a diisopropyl amine salt, an ethanol amine salt, a diethanol amine salt, a triethanol amine salt, a morpholine salt, an N-methylmorpholine salt, a piperidine salt, an N-methylpiperidine salt, an N-ethylpiperidine salt, a dibenzylamine salt, a piperazine salt, a pyridine salt, a pyrazole salt, a pyridazine salt, a pyrimidine salt, an imidazole salt, or a pyrazine salt.


Acid addition salts can arise from the addition of an acid to a compound of the present disclosure. In some embodiments, the acid is organic. In some embodiments, the acid is inorganic. In some embodiments, the acid is hydrochloric acid, hydrobromic acid, hydroiodic acid, nitric acid, nitrous acid, sulfuric acid, sulfurous acid, a phosphoric acid, isonicotinic acid, lactic acid, salicylic acid, tartaric acid, ascorbic acid, gentisic acid, gluconic acid, glucuronic acid, saccharic acid, formic acid, benzoic acid, glutamic acid, pantothenic acid, acetic acid, trifluoroacetic acid, mandelic acid, cinnamic acid, aspartic acid, stearic acid, palmitic acid, glycolic acid, propionic acid, butyric acid, fumaric acid, succinic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, oxalic acid, or maleic acid.


In some embodiments, the salt is a hydrochloride salt, a hydrobromide salt, a hydroiodide salt, a nitrate salt, a nitrite salt, a sulfate salt, a sulfite salt, a phosphate salt, isonicotinate salt, a lactate salt, a salicylate salt, a tartrate salt, an ascorbate salt, a gentisate salt, a gluconate salt, a glucuronate salt, a saccharate salt, a formate salt, a benzoate salt, a glutamate salt, a pantothenate salt, an acetate salt, a trifluoroacetate salt, a mandelate salt, a cinnamate salt, an aspartate salt, a stearate salt, a palmitate salt, a glycolate salt, a propionate salt, a butyrate salt, a fumarate salt, a hemifumarate salt, a succinate salt, a methanesulfonate salt, an ethanesulfonate salt, a benzenesulfonate salt, a p-toluenesulfonate salt, a citrate salt, an oxalate salt, or a maleate salt.


Compounds.

In some embodiments, a compound capable of binding RyR2 is a compound of Formula I:




embedded image


wherein,

    • n is 0, 1, or 2;
    • q is 0, 1, 2, 3, or 4;
    • each R is independently acyl, —O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, arylthio, arylamino, heteroarylthio, or heteroarylamino, each of which is independently substituted or unsubstituted; or halogen, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;
    • R1 is alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
    • R2 is alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H, —C(═O)R5, —C(═S)R6, —SO2R7, —P(═O)R8R9, or —(CH2)m—R10;
    • R3 is acyl, —O-acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or substituted; or H, —CO2Y, or —C(═O)NHY;
    • Y is alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
    • R4 is alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
    • each R5 is acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —NR15R16, —(CH2)tNR15R16, —NHNR15R16, —NHOH, —OR15, —C(═O)NHNR15R16, —CO2R15, —C(═O)NR15R16, or —CH2X;
    • each R6 is acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NHNR15R16, —NHOH, —NR15R16, or —CH2X;
    • each R7 is alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NR15R16, —NHNR15R16, —NHOH, or —CH2X;
    • each R8 and R9 are each independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or OH;
    • each R10 is —NR15R16, OH, —SO2R11, —NHSO2R11, C(═O)(R12), NHC═O(R12), —OC═O(R12), or —P(═O)R13R14;
    • each R11, R12, R13, and R14 is independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or H, OH, NH2, —NHNH2, or —NHOH;
    • each X is independently halogen, —CN, —CO2R15, —C(═O)NR15R16, —NR15R16, —OR15, —SO2R7, or —P(═O)R8R9; and
    • each R15 and R16 is independently acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted, or H; or R15 and R16 together with the N to which R15 and R16 are bonded form a heterocycle that is substituted or unsubstituted;
    • t is 1, 2, 3, 4, 5, or 6;
    • m is 1, 2, 3, or 4;


      or a pharmaceutically-acceptable salt thereof.


In some embodiments, R2 is unsubstituted alkyl.


In some embodiments, the present disclosure provides compounds of Formula I-a:




embedded image


wherein:

    • n is 0, 1, or 2;
    • q is 0, 1, 2, 3, or 4;
    • each R is independently acyl, —O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, arylthio, arylamino, heteroarylthio, or heteroarylamino, each of which is independently substituted or unsubstituted; or halogen, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;
    • R2 is alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H, —C(═O)R5, —C(═S)R6, —SO2R7, —P(═O)R8R9, or —(CH2)m—R10;
    • each R5 is acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —NR15R16, —NHNR15R16, —NHOH, —OR15, —C(═O)NHNR15R16, —CO2R15, —C(═O)NR15R16, —CH2X, or alkyl substituted by at least one labeling group, selected from a fluorescent group, a bioluminescent group, a chemiluminescent group, a colorimetric group, and a radioactive labeling group;
    • each R6 is acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NHNR15R16, —NHOH, —NR15R16, or —CH2X;
    • each R7 is alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NR15R16, N—NR15R16, —NHOH, or —CH2X;
    • each R8 and R9 are each independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or OH;
    • each R10 is —NR15R16, OH, —SO2R11, —NHSO2R11, C(═O)R12, NH(C═O)R12, —O(C═O)R12, or —P(═O)R13R14; m is 0, 1, 2, 3, or 4;
    • each R11, R12, R13, and R14 is independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or H, OH, NH2, —NHNH2, or —NHOH;
    • each X is halogen, —CN, —CO2R15, —C(═O)NR15R16, —NR15R16, —OR15, —SO2R7, or —P(═O)R8R9; and
    • each R15 and R16 is independently acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted, or H; or R15 and R16 together with the N to which R15 and R16 are bonded form a heterocycle that is substituted or unsubstituted;
    • or a pharmaceutically-acceptable salt thereof.


In some embodiments, the present disclosure provides a compound of formula I-a, wherein each R is independently halogen, —OH, OMe, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —S(═O)2C1-C4alkyl, —S(═O)C1-C4alkyl, —S—C1-C4alkyl, —OS(═O)2CF3, Ph, —NHCH2Ph, —C(═O)Me, —OC(═O)Me, morpholinyl, or propenyl; and n is 0, 1, or 2.


In some embodiments, the present disclosure provides a compound of formula I-a, wherein R2 is —C═O(R5), —C═S(R6), —SO2R7, —P(═O)R8R9, or —(CH2)m—R10.


In some embodiments, the present disclosure provides a compound of formula I-b:




embedded image


wherein

    • R′ and R″ are each independently acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, aryl, heteroaryl, arylthiol, heteroarylthio, arylamino, or heteroarylamino, each of which is independently substituted or substituted; or halogen, H, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;
    • R2 is alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H, —C(═O)R5, —C(═S)R6, —SO2R7, —P(═O)R8R9, or —(CH2)m—R10; and
    • n is 0, 1, or 2;


      or a pharmaceutically-acceptable salt thereof.


In some embodiments, the present disclosure provides a compound of formula I-b, wherein R′ and R″ are each independently H, halogen, —OH, OMe, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —S(═O)2C1-C4alkyl, —S(═O)C1-C4alkyl, —S—C1-C4alkyl, —OS(═O)2CF3, Ph, —NHCH2Ph, —C(═O)Me, —OC(═O)Me, morpholinyl, or propenyl; and n is 0, 1 or 2. In some embodiments, R′ is H or OMe, and R″ is H.


In some embodiments, the present disclosure provides a compound of formula I-b, wherein R2 is —C═O(R5), —C═S(R6), SO2R7, P(═O)R8R9, or —(CH2)m—R10.


In some embodiments, the present disclosure provides a compound formula of I-c:




embedded image




    • n is 0, 1, or 2;

    • q is 0, 1, 2, 3, or 4;

    • each R is independently acyl, —O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, arylthio, arylamino, heteroarylthio, or heteroarylamino, each of which is independently substituted or unsubstituted; or halogen, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;

    • each R7 is alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NR15R16, —NHNR15R16, —NHOH, or —CH2X;

    • or a pharmaceutically-acceptable salt thereof.





In some embodiments, the present disclosure provides a compound of formula I-c, wherein each R is independently halogen, —OH, OMe, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —S(═O)2C1-C4alkyl, —S(═O)C1-C4alkyl, —S—C1-C4alkyl, —OS(═O)2CF3, Ph, —NHCH2Ph, —C(═O)Me, —OC(═O)Me, morpholinyl, or propenyl; and n is 0, 1, or 2.


In some embodiments, the present disclosure provides a compound of formula I-c, wherein R7 is alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OH or —NR15R16.


In some embodiments, the present disclosure provides a compound of formula of I-d:




embedded image




    • n is 0, 1, or 2;

    • R′ and R″ are each independently acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, aryl, heteroaryl, arylthiol, heteroarylthio, arylamino, or heteroarylamino, each of which is independently substituted or substituted; or halogen, H, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;

    • each R7 is alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NR15R16, —NHNR15R16, —NHOH, or —CH2X,


      or a pharmaceutically-acceptable salt thereof.





In some embodiments, the present disclosure provides a compound of formula wherein R′ and R″ are each independently H, halogen, —OH, OMe, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —S(═O)2C1-C4alkyl, —S(═O)C1-C4alkyl, —S—C1-C4alkyl, —OS(═O)2CF3, Ph, —NHCH2Ph, —C(═O)Me, —OC(═O)Me, morpholinyl, or propenyl; and n is 0, 1 or 2. In some embodiments, R′ is H or OMe, and R″ is H.


In some embodiments, the present disclosure provides a compound of formula I-d, wherein R7 is alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OH, or —NR15R16.


In some embodiments, the present disclosure provides a compound of formula of I-e:




embedded image




    • n is 0, 1, or 2;

    • q is 0, 1, 2, 3, or 4;

    • each R is independently acyl, —O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, arylthio, arylamino, heteroarylthio, or heteroarylamino, each of which is independently substituted or unsubstituted; or halogen, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3; and

    • each R5 is acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —NR15R16, —NHNR15R16, —NHOH, —OR15, —C(═O)NHNR15R16, —CO2R15, —C(═O)NR15R16, —CH2X, or alkyl substituted by at least one labeling group, selected from a fluorescent group, a bioluminescent group, a chemiluminescent group, a colorimetric group, and a radioactive labeling group,


      or a pharmaceutically-acceptable salt thereof.





In some embodiments, the present disclosure provides a compound of formula I-e, wherein each R is independently halogen, —OH, OMe, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —S(═O)2C1-C4alkyl, —S(═O)C1-C4alkyl, —S—C1-C4alkyl, —OS(═O)2CF3, Ph, —NHCH2Ph, —C(═O)Me, —OC(═O)Me, morpholinyl, or propenyl; and n is 0, 1, or 2.


In some embodiments, the present disclosure provides a compound of formula I-e, wherein R5 is alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —NR15R16, —NHOH, —OR15, or —CH2X.


In some embodiments, the present disclosure provides a compound of formula of I-f:




embedded image




    • n is 0, 1, or 2;

    • R′ and R″ are each independently acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, aryl, heteroaryl, arylthiol, heteroarylthio, arylamino, or heteroarylamino, each of which is independently substituted or substituted; or halogen, H, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;

    • each R5 is acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —NR15R16, —NHNR15R16, —NHOH, —OR15, —C(═O)NHNR15R16, —CO2R15, —C(═O)NR15R16, —CH2X, or alkyl substituted by at least one labeling group, selected from a fluorescent group, a bioluminescent group, a chemiluminescent group, a colorimetric group, and a radioactive labeling group,


      or a pharmaceutically-acceptable salt thereof.





In some embodiments, the present disclosure provides a compound of formula I-f, wherein R′ and R″ are each independently H, halogen, —OH, OMe, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —S(═O)2C1-C4alkyl, —OS(═O)2CF3, Ph, —NHCH2Ph, —C(═O)Me, —OC(═O)Me, morpholinyl, or propenyl; and n is 0, 1 or 2. In some embodiments, R′ is H or OMe, and R″ is H.


In some embodiments, the present disclosure provides a compound of formula I-f, wherein R5 is alkyl, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —NR15R16, —NHOH, —OR15, or —CH2X.


In some embodiments, the present disclosure provides a compound of formula of I-g:




embedded image


wherein

    • n is 0, 1, or 2;
    • q is 0, 1, 2, 3, or 4;
    • W is S or O;
    • each R is independently acyl, —O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, arylthio, arylamino, heteroarylthio, or heteroarylamino, each of which is independently substituted or unsubstituted; or halogen, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;
    • each R15 and R16 is independently acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted, or H; or R15 and R16 together with the N to which R15 and R16 are bonded may form a heterocycle that is substituted or unsubstituted,


      or a pharmaceutically-acceptable salt thereof.


In some embodiments, the present disclosure provides a compound of formula I-g, wherein each R is independently selected from the group consisting of H, halogen, —OH, OMe, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —S(═O)2C1-C4alkyl, —S(═O)C1-C4alkyl, —S-C1-C4alkyl, —OS(═O)2CF3, Ph, —NHCH2Ph, —C(═O)Me, —OC(═O)Me, morpholinyl and propenyl; and n is 0, 1, or 2.


In some embodiments, the present disclosure provides a compound of formula I-g, wherein R15 and R16 are each independently alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl, each of which is independently substituted or unsubstituted; or H, OH, or NH2; or R15 and R16 together with the N to which they are bonded form a heterocycle that is substituted or unsubstituted.


In some embodiments, the present disclosure provides a compound of formula I-g, wherein W is O or S.


In some embodiments, the present disclosure provides a compound of formula of I-h:




embedded image




    • n is 0, 1, or 2;

    • W is S or O;

    • R′ and R″ are each independently acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, aryl, heteroaryl, arylthiol, heteroarylthio, arylamino, or heteroarylamino, each of which is independently substituted or substituted; or halogen, H, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3,


      or a pharmaceutically-acceptable salt thereof.





In some embodiments, the present disclosure provides a compound of formula wherein R′ and R″ are each independently H, halogen, —OH, OMe, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —S(═O)2C1-C4alkyl, —S(═O)C1-C4alkyl, —S—C1-C4alkyl, —OS(═O)2CF3, Ph, —NHCH2Ph, —C(═O)Me, —OC(═O)Me, morpholinyl, or propenyl; and n is 0, 1 or 2. In some embodiments, R′ is H or OMe, and R″ is H.


In some embodiments, the present disclosure provides a compound of formula I-h, wherein R15 and R16 are each independently alkyl, alkylamino, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or H, OH, NH2; or R15 and R16 together with the N to which R15 and R16 are bonded form a heterocycle that is substituted or unsubstituted.


In some embodiments, the present disclosure provides a compound of formula I-g, wherein W is O or S.


In some embodiments, the present disclosure provides a compound of formula of I-i:




embedded image


wherein

    • R17 is alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —NR15R16, —NHNR15R16, —NHOH, —OR15, or —CH2X;
    • n is 0, 1, or 2;
    • q is 0, 1, 2, 3, or 4; and
    • each R is independently acyl, —O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, arylthio, arylamino, heteroarylthio, or heteroarylamino, each of which is independently substituted or unsubstituted; or halogen, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3,


      or a pharmaceutically-acceptable salt thereof.


In some embodiments, the present disclosure provides a compound of formula I-i, wherein each R is independently halogen, —OH, OMe, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —S(═O)2C1-C4alkyl, —S(═O)C1-C4alkyl, —S—C1-C4alkyl, —OS(═O)2CF3, Ph, —NHCH2Ph, —C(═O)Me, —OC(═O)Me, morpholinyl, or propenyl; and n is 0, 1, or 2.


In some embodiments, the present disclosure provides a compound of formula I-i, wherein R17 is —NR15R16 or —OR15. In some embodiments, R17 is —OH, —OMe, —Net, —NHEt, —NHPh, —NH2, or —NHCH2pyridyl.


In some embodiments, the present disclosure provides a compound of formula of I-j:




embedded image




    • R′ and R″ are each independently acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, aryl, heteroaryl, arylthiol, heteroarylthio, arylamino, or heteroarylamino, each of which is independently substituted or substituted; or halogen, H, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;
      • R17 is selected from the group consisting of —NR15R16, —NHOH, —OR15, —CH2X, alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl; wherein each alkenyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl may be substituted or unsubstituted;
      • n is 0, 1, or 2,


        or a pharmaceutically-acceptable salt thereof.





In some embodiments, the present disclosure provides a compound of formula I-j, wherein R′ and R″ are each independently H, halogen, —OH, OMe, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —S(═O)2C1-C4alkyl, —S(═O)C1-C4alkyl, —S—C1-C4alkyl, —OS(═O)2CF3, Ph, —NHCH2Ph, —C(═O)Me, —OC(═O)Me, morpholinyl, or propenyl; and n is 0, 1 or 2. In some embodiments, R′ is H or OMe, and R″ is H.


In some embodiments, the present disclosure provides a compound of formula I-j, wherein R17 is NR15R16 or —OR15. In some embodiments, R17 is —OH, —OMe, —Net, —NHEt, —NHPh, —NH2, or —NHCH2pyridyl.


In some embodiments, the present disclosure provides a compound of formula I-k or I-k-1:




embedded image




    • each R is independently acyl, —O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, arylthio, arylamino, heteroarylthio, or heteroarylamino, each of which is independently substituted or unsubstituted; or halogen, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;

    • R′ and R″ are each independently acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, aryl, heteroaryl, arylthiol, heteroarylthio, arylamino, or heteroarylamino, each of which is independently substituted or substituted; or halogen, H, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;

    • R18 is alkyl, aryl, cycloalkyl, or heterocyclyl, each of which is independently substituted or unsubstituted; or —NR15R16, —C(═O)NR15R16, —(C═O)OR15, or —OR15;

    • q is 0, 1, 2, 3, or 4;

    • p is 1, 2, 3, 4, 5, 6, 7, 8 9, or 10; and

    • n is 0, 1, or 2,


      or a pharmaceutically-acceptable salt thereof.





In some embodiments, the present disclosure provides a compound of formula I-k, wherein each R is independently H, halogen, —OH, OMe, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —S(═O)2C1-C4alkyl, —S(═O)C1-C4alkyl, —S-C1-C4alkyl, —OS(═O)2CF3, Ph, —NHCH2Ph, —C(═O)Me, —OC(═O)Me, morpholinyl, or propenyl; and n is 0, 1 or 2. In some embodiments, R is OMe at position 7 of the benzothiazepine ring.


In some embodiments, the present disclosure provides a compound of formula I-k-1, wherein R′ and R″ are each independently H, halogen, —OH, OMe, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —S(═O)2C1-C4alkyl, —S(═O)C1-C4alkyl, —S-C1-C4alkyl, —OS(═O)2CF3, Ph, —NHCH2Ph, —C(═O)Me, —OC(═O)Me, morpholinyl, or propenyl; and n is 0, 1 or 2. In some embodiments, R′ is H or OMe, and R″ is H.


In some embodiments, the present disclosure provides a compound of formula I-k or I-k-1, wherein R18 is —NR15R16, —(C═O)OR15, —OR15, alkyl that is substituted or unsubstituted, or aryl that is substituted or unsubstituted. In some embodiments, m is 1, and R18 is Ph, —C(═O)OMe, C(═O)OH, aminoalkyl, NH2, NHOH, or NHCbz. In other embodiments, m is 0, and R18 is C1-C4 alkyl. In other embodiments, R18 is Me, Et, propyl, and butyl. In some embodiments, m is 2, and R18 is pyrrolidine, piperidine, piperazine, or morpholine. In some embodiments, m is 3, 4, 5, 5, 7, or 8, and R18 is a fluorescent labeling group selected from bodipy, dansyl, fluorescein, rhodamine, Texas red, cyanine dyes, pyrene, coumarins, Cascade Blue™, Pacific Blue, Marina Blue, Oregon Green, 4′,6-Diamidino-2-phenylindole (DAPI), indopyra dyes, lucifer yellow, propidium iodide, porphyrins, arginine, and variants and derivatives thereof.


In some embodiments, the present disclosure provides a compound of formula of I-1 or I-1-1:




embedded image


wherein

    • each R is independently acyl, —O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, arylthio, arylamino, heteroarylthio, or heteroarylamino, each of which is independently substituted or unsubstituted; or halogen, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;
    • R′ and R″ are each independently acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, aryl, heteroaryl, arylthiol, heteroarylthio, arylamino, or heteroarylamino, each of which is independently substituted or substituted; or halogen, H, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;
    • R6 is acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NHNR15R16, —NHOH, —NR15R16, or —CH2X;
    • q is 0, 1, 2, 3, or 4; and
    • n is 0, 1, or 2,


      or a pharmaceutically acceptable salt thereof.


In some embodiments, the present disclosure provides a compound of formula I-1, wherein each R is independently halogen, —OH, OMe, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —S(═O)2C1-C4alkyl, —S(═O)C1-C4alkyl, —S—C1-C4alkyl, —OS(═O)2CF3, Ph, —NHCH2Ph, —C(═O)Me, —OC(═O)Me, morpholinyl, or propenyl; and n is 0, 1 or 2. In some embodiments, R is OMe at position 7 of the benzothiazepine ring.


In some embodiments, the present disclosure provides a compound of formula I-1-1, wherein R′ and R″ are each independently H, halogen, —OH, OMe, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —S(═O)2C1-C4alkyl, —S(═O)C1-C4alkyl, —S—C1-C4alkyl, —OS(═O)2CF3, Ph, —NHCH2Ph, —C(═O)Me, —OC(═O)Me, morpholinyl, or propenyl; and n is 0, 1 or 2. In some embodiments, R′ is H or OMe, and R″ is H.


In some embodiments, the present disclosure provides a compound of formula I-1 or I-1-1, wherein R6 is acyl, alkenyl, alkyl, aryl, cycloalkyl, cycloalkylalkyl, heterocyclyl, and heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —NR15R16, —NHNR15R16, —OR15, —NHOH, or —CH2X. In some embodiments, R6 is —NR15R16. In some embodiments, R6 is —NHPh, pyrrolidine, piperidine, piperazine, morpholine. In some embodiments, R6 is alkoxyl. In some embodiments, R6 is —O-tBu.


In some embodiments, the present disclosure provides a compound of formula I-m or I-m-1:


wherein




embedded image




    • n is 0, 1, or 2;

    • q is 0, 1, 2, 3, or 4;

    • R′ and R″ are each independently acyl, alkyl, alkoxyl, alkylamino, alkylthio, cycloalkyl, aryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, aryl, heteroaryl, arylthiol, heteroarylthio, arylamino, or heteroarylamino, each of which is independently substituted or substituted; or halogen, H, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3; and

    • R8 and R9 are each independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or OH,


      or a pharmaceutically-acceptable salt thereof.





In some embodiments, the present disclosure provides a compound of formula I-m, wherein each R is independently halogen, —OH, OMe, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —S(═O)2C1-C4alkyl, —S(═O)C1-C4alkyl, —S—C1-C4alkyl, —OS(═O)2CF3, Ph, —NHCH2Ph, —C(═O)Me, —OC(═O)Me, morpholinyl, or propenyl; and n is 0, 1 or 2. In some embodiments, R is OMe at position 7 of the benzothiazepine ring.


In some embodiments, the present disclosure provides a compound of formula I-m-1, wherein R′ and R″ are each independently H, halogen, —OH, OMe, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —S(═O)2C1-C4alkyl, —S(═O)C1-C4alkyl, —S—C1-C4alkyl, —OS(═O)2CF3, Ph, —NHCH2Ph, —C(═O)Me, —OC(═O)Me, morpholinyl, or propenyl; and n is 0, 1 or 2. In some embodiments, R′ is H or OMe, and R″ is H.


In some embodiments, the present disclosure provides a compound of formula I-m or I-m-1, wherein R8 and R9 are each independently alkyl, aryl, OH, alkoxyl, or alkylamino. In some embodiments, R8 is C1-C4alkyl. In some embodiments, R8 is Me, Et, propyl or butyl. In some embodiments, R9 is aryl. In some embodiments, R9 is phenyl.


In some embodiments, the present disclosure provides a compound of formula I-n,




embedded image


wherein:

    • Rd is CH2, or NRa; and
    • Ra is H, alkoxy, (C1-C6 alkyl)-aryl, wherein the aryl is a disubstituted phenyl or a benzo[1,3]dioxo-5-yl group, or a Boc group.


      or a pharmaceutically-acceptable salt thereof.


In some embodiments, Ra is H.


Representative compounds of Formula I-n include without limitation S101, S102, S103, and S114.


In some embodiments, the present disclosure provides a compound of Formula I-o:




embedded image




    • wherein:

    • Re is (C1-C6 alkyl)-phenyl, (C1-C6 alkyl)-C(O)Rb, or substituted or unsubstituted C1-C6 alkyl; and

    • Rb is OH or —O—(C1-C6 alkyl),


      wherein the phenyl or the substituted alkyl is substituted with one or more of halogen, hydroxyl, C1-C6 alkyl, —O—(C1-C6 alkyl), —NH2, —NH(C1-C6 alkyl), —N(C1-C6 alkyl)2, cyano, or dioxolane,


      or a pharmaceutically-acceptable salt thereof.





Representative compounds of Formula I-o include without limitation S107, S110, S111, S120, and S121.


In some embodiments, the present disclosure provides a compound of Formula I-p:




embedded image


wherein:

    • Rc is —(C1-C6 alkyl)-NH2, —(C1-C6 alkyl)-ORf, wherein Rf is H or —C(O)—(C1-C6)alkyl, or —(C1-C6 alkyl)-NHRg, wherein Rg is carboxybenzyl.


In some embodiments, the present disclosure provides compounds of Formula II or Formula III:




embedded image


wherein:

    • n is 0, 1, or 2;
    • q is 0, 1, 2, 3, or 4;
    • each R is independently acyl, —O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, arylthio, arylamino, heteroarylthio, or heteroarylamino, each of which is independently substituted or unsubstituted; or halogen, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, ≤N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;
    • each R2 and R2a is independently alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H, —C(═O)R5, —C(═S)R6, —SO2R7, —P(═O)R8R9, or —(CH2)m—R10;
    • each R5 is acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —NR15R16, —NHNR15R16, —NHOH, —OR15, —C(═O)NHNR15R16, —CO2R15, —C(═O)NR15R16, —CH2X, or alkyl substituted by at least one labeling group, selected from a fluorescent group, a bioluminescent group, a chemiluminescent group, a colorimetric group, and a radioactive labeling group;
    • each R6 is acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NHNR15R16, —NHOH, —NR15R16, or —CH2X;
    • each R7 is alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NR15R16, —NHNR15R16, —NHOH, or —CH2X;
    • each R8 and R9 are each independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or OH;
    • each R10 is —NR15R16, OH, —SO2R11, —NHSO2R11, —C(═O)R12, NH(C═O)R12, —O(C═O)R12, or —P(═O)R13R14;
    • m is 0, 1, 2, 3, or 4;
    • each R11, R12, R13, and R14 is independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or H, OH, NH2, —NHNH2, or —NHOH;
    • each X is halogen, —CN, —CO2R15, —C(═O)NR15R16, —NR15R16, —OR15, —SO2R7, or —P(═O)R8R9; and
    • each R15 and R16 is independently acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted, or H; or R15 and R16 together with the N to which R15 and R16 are bonded form a heterocycle that is substituted or unsubstituted;


      or a pharmaceutically acceptable salt thereof.


In some embodiments, the compound of formula (I) is selected from:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


In some embodiments, the synthetic compound is a compound of Formula (I), (I-a), (I-b), (I-c), (I-d), (I-e), (I-f), (I-g), (I-h), (I-i), (I-j), (I-k), (I-k-1), (I-1), (I-1-1), (I-m), (I-m-1), (I-n), (I-o), (I-p), (II), or (III). In some embodiments, the synthetic compound is S1, S2, S3, S4, S5, S6, S7, S9, S1, S12, S13, S14, S19, S20, S22, S23, S24, S25, S26, S27, S36, S37, S38, S40, S43, S44, S45, S46, S47, S48, S49, S50, S51, S52, S53, S54, S55, S56, S57, S58, S59, S60, S61, S62, S63, S64, S66, S67, S68, S69, S70, S71, S72, S73, S74, S75, S76, S77, S78, S79, S80, S81, S82, S83, S84, S85, S86, S87, S88, S89, S90, S91, S92, S93, S94, S95, S96, S97, S98, S99, S100, S101, S102, S103, S104, S105, S107, S108, S109, S110, S111, S112, S113, S114, S115, S116, S117, S118, S119, S120, S121, S122, or S123, as herein defined.


In some embodiments, the synthetic compound is:




embedded image


or a pharmaceutically-acceptable salt thereof or an ionized form thereof.


In some embodiments, the synthetic compound is:




embedded image


or a pharmaceutically-acceptable salt or an ionized form thereof.


Compounds described herein may exist in their tautomeric form (for example, as an amide or imino ether). All such tautomeric forms are contemplated herein as part of the present disclosure.


All stereoisomers of the compounds of the present disclosure (for example, those which may exist due to asymmetric carbons on various substituents), including enantiomeric forms and diastereomeric forms, are contemplated within the scope of this invention. Individual stereoisomers of the compounds of the disclosure may, for example, be substantially free of other isomers (e.g., as a pure or substantially pure optical isomer having a specified activity), or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers. The chiral centers of the present invention may have the S or R configuration as defined by the IUPAC 1974 Recommendations. The racemic forms can be resolved by physical methods, such as, for example, fractional crystallization, separation or crystallization of diastereomeric derivatives or separation by chiral column chromatography. The individual optical isomers can be obtained from the racemates by any suitable method, including without limitation, conventional methods, such as, for example, salt formation with an optically active acid followed by crystallization.


Screening Methods.

The present disclosure provides methods for identifying a compound that binds to a biomolecular target (e.g. RyR2). In some embodiments, the methods described herein can include screening a library of three-dimensional compound structures to identify ligands that fit a binding pocket of the biomolecular target such as RyR2.


In some embodiments, provided is a method comprising:

    • (a) determining an open probability (Po) of a first RyR2 protein, wherein the first RyR2 protein is treated with a test compound, and
    • (b) determining an open probability (Po) of a second RyR2 protein, wherein the second RyR2 protein is not treated with the test compound.


In some embodiments, each of the determining the open probability (Po) of the first RyR2 protein and the determining the open probability (Po) of the second RyR2 protein comprises recording a single channel Ca2+ current. In some embodiments, the method further comprises determining a difference between the Po of the first RyR2 protein and Po of the second RyR2 protein. In some embodiments, the method further comprises identifying the test compound as a target for further analysis based on the difference between the Po of the first RyR2 protein and Po of the second RyR2 protein. In some embodiments, the Po of the first RyR2 protein is lower than the Po of the second RyR2 protein.


In some embodiments, the method further comprises performing an analogous assay wherein another compound is used in place of the test compound, wherein the analogous assay provides a difference between:

    • (a) an open probability (Po) of a third RyR2 protein, wherein the third RyR2 protein is treated with the other compound; and
    • (b) an open probability (Po) of a fourth RyR2 protein, wherein the fourth RyR2 protein is not treated with the other compound, wherein the test compound is prioritized over the other compound for the further analysis based on a comparison of:
    • (i) the difference between the Po of the first RyR2 protein and Po of the second RyR2 protein; with
    • (ii) a difference between the Po of the third RyR2 protein and Po of the fourth RyR2 protein.


In some embodiments, the difference of the Po in (b)(ii) is greater than the difference of the Po in (b)(i). In some embodiments, the RyR2 is a mutant RyR2. In some embodiments, the mutant RyR2 contains at least one mutation that is associated with Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT). In some embodiments, the mutation is R2474S. In some embodiments, the RyR2 protein is a post-translationally modified RyR2 protein. In some embodiments, the post-translationally modified RyR2 protein is a phosphorylated, oxidized or nitrosylated RyR2. In some embodiments, the RyR2 is a mutated and post-translationally modified RyR2.


In some embodiments, provided is a method comprising:

    • (a) contacting a first RyR2 protein with a test compound;
    • (b) providing a second RyR2 protein;
    • (c) subsequent to the contacting the first RyR2 protein with the test compound, measuring an open probability (Po) of the first RyR2 protein; and
    • (d) measuring an open probability (Po) of the second RyR2 protein.


In some embodiments, each of the determining the open probability (Po) of the first RyR2 protein and the determining the open probability (Po) of second RyR2 protein comprises recording a single channel Ca2+ current. In some embodiments, the method further comprises determining a difference between the Po of the first RyR2 protein and the Po of the second RyR2 protein. In some embodiments, the method further comprises identifying the test compound as a target for further analysis based on the difference between the Po of the first RyR2 protein and the Po of the second RyR2 protein. In some embodiments, the Po of the first RyR2 protein is lower than the Po of the second RyR2 protein.


In some embodiments, the method further comprises performing an analogous assay wherein another compound is used in place of the test compound, wherein the analogous assay provides a difference between:

    • (a) an open probability (Po) of a third RyR2 protein, wherein the third RyR2 protein is treated with the other compound; and
    • (b) an open probability (Po) of a fourth RyR2 protein, wherein the fourth RyR2 protein is not treated with the other compound,
    • wherein the test compound is prioritized over the other compound for the further analysis based on a comparison of:
    • (i) the difference between the Po of the first RyR2 protein and Po of the second RyR2 protein; with
    • (ii) a difference between the Po of the third RyR2 protein and Po of the fourth RyR2 protein.


In some embodiments, the difference of the Po in (b)(ii) is greater than the difference of the Po in (b)(i). In some embodiments, the RyR2 is a mutant RyR2. In some embodiments, the mutant RyR2 contains at least one mutation that is associated with Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT). In some embodiments, the mutation is R2474S. In some embodiments, the RyR2 protein is a post-translationally modified RyR2 protein. In some embodiments, the post-translationally modified RyR2 protein is a phosphorylated, oxidized or nitrosylated RyR2. In some embodiments, the RyR2 is a mutated and post-translationally modified RyR2. In some embodiments, the method further comprises: subsequent to the contacting the first RyR2 protein with the reagent and the test compound, fusing a first microsome containing the first RyR2 protein to a first planar lipid bilayer, and subsequent to the contacting the second RyR2 protein with the reagent, fusing a second microsome containing the second RyR2 protein to a second planar lipid bilayer.


In some embodiments, provided is a method of identifying a compound having RyR2 modulatory activity, the method comprising:

    • (a) determining an open probability (Po) of a RyR2 protein;
    • (b) contacting the RyR2 protein with a test compound;
    • (c) determining an open probability (Po) of the RyR2 protein in the presence of the test compound; and
    • (d) determining a difference between the Po of the RyR2 protein in the presence and absence of the test compound;
    • wherein a reduction in the Po of the RyR2 protein in the presence of the test compound relative to the Po of the RyR2 protein in the absence of the test compound is indicative of the compound having RyR2 modulatory activity.


In some embodiments, the RyR2 protein is a mutant RyR2 protein. In some embodiments, the mutant RyR2 contains at least one mutation that is associated with Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT). In some embodiments, the mutation is R2474S. In some embodiments, the RyR2 protein is a post-translationally modified RyR2 protein. In some embodiments, the post-translationally modified RyR2 protein is a phosphorylated, oxidized or nitrosylated RyR2. In some embodiments, the RyR2 protein is a mutated and post-translationally modified RyR2 protein. In some embodiments, the test compound preferentially binds to a mutant RyR2 relative to wild-type RyR2. In some embodiments, the test compound preferentially binds to post-translationally modified RyR2 relative to wild-type RyR2. In some embodiments, the test compound preferentially binds to a mutated and post-translationally modified RyR2 relative to a wild-type RyR2. In some embodiments, determining the open probability (Po) of the RyR2 protein comprises recording a single channel Ca2+ current.


In some embodiments, provided is a method for identifying a compound having RyR2 modulatory activity, comprising:

    • (a) contacting a RyR2 protein with a ligand having known RyR2 modulatory activity to create a mixture, wherein the RyR2 protein is a leaky RyR2, the leaky RyR2 comprising mutant RyR2 protein, post-translationally modified RyR2, or a combination thereof;
    • (b) contacting the mixture of step (a) with a test compound; and
    • (c) determining an ability of the test compound to displace the ligand from the RyR2 protein.


In some embodiments, the ligand is radiolabeled. In some embodiments, determining the ability of the test compound to displace the ligand from the RyR2 protein comprises determining a radioactive signal in the RyR2 protein. In some embodiments, the RyR2 protein is a mutant RyR2 protein. In some embodiments, the mutant RyR2 contains at least one mutation that is associated with Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT). In some embodiments, the mutation is R2474S. In some embodiments, the RyR2 protein is a post-translationally modified RyR2 protein. In some embodiments, the RyR2 protein is a mutated and post-translationally modified RyR2 protein. In some embodiments, the test compound preferentially binds to a mutant RyR2 relative to wild-type RyR2. In some embodiments, the test compound preferentially binds to post-translationally modified RyR2 relative to wild-type RyR2. In some embodiments, the test compound preferentially binds to a mutant and post-translationally modified RyR2 relative to a wild-type RyR2.


In some embodiments, provided is a method for identifying a compound that preferentially binds to a mutated, post-translationally modified RyR2 or a combination thereof, comprising:

    • (a) determining binding affinity of a test compound to a first RyR2 protein, wherein the first RyR2 protein is a wild-type RyR2 protein;
    • (b) determining binding affinity of a test compound to a second RyR2 protein, wherein second first RyR2 protein is a mutant RyR2 protein, a post-translationally modified RyR2, or a combination thereof, and
    • (c) selecting a compound having a higher binding affinity to the second RyR2 protein relative to the first RyR2 protein.


In some embodiments, the RyR2 protein is a mutant RyR2 protein. In some embodiments, the mutant RyR2 contains at least one mutation that is associated with Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT). In some embodiments, the mutation is R2474S. In some embodiments, the RyR2 protein is a post-translationally modified RyR2 protein. In some embodiments, the RyR2 protein is a mutated and post-translationally modified RyR2 protein. In some embodiments, the test compound preferentially binds to a mutant RyR2 relative to wild-type RyR2. In some embodiments, the test compound preferentially binds to post-translationally modified RyR2 relative to wild-type RyR2. In some embodiments, the test compound preferentially binds to a mutant and post-translationally modified RyR2 relative to a wild-type RyR2. In some embodiments, the test compound contains a benzothiazepane moiety.


In some embodiments, the test compound is a compound of Formula (I):




embedded image


wherein:

    • each R is independently acyl, —O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, arylthio, arylamino, heteroarylthio, or heteroarylamino, each of which is independently substituted or unsubstituted; or halogen, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;
    • R1 is alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
    • R2 is alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H, —C(═O)R5, —C(═S)R6, —SO2R7, —P(═O)R8R9, or —(CH2)m—R10;
    • R3 is acyl, —O-acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or substituted; or H, —CO2Y, or —C(═O)NHY;
    • Y is alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
    • R4 is alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
    • each R5 is acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —NR15R16, —(CH2)tNR15R16, —NHNR15R16, —NHOH, OR, —C(═O)NHNR15R16, —CO2R15, —C(═O)NR15R16, or —CH2X;
    • each R6 is acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NHNR15R16, —NHOH, —NR15R16, or —CH2X;
    • each R7 is alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NR15R16, —NHNR15R16, —NHOH, or —CH2X;
    • each R8 and R9 are each independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or OH;
    • each R10 is —NR15R16, OH, —SO2R11, —NHSO2R11, C(═O)(R12), NHC═O(R12), —OC═O(R12), or —P(═O)R13R14;
    • each R11, R12, R13, and R14 is independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or H, OH, —NH2, —NHNH2, or —NHOH;
    • each X is independently halogen, —CN, —CO2R15, —C(═O)NR15R16, —NR15R16, —OR15, —SO2R7, or —P(═O)R8R9;
    • each R15 and R16 is independently acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted, or H; or R15 and R16 together with the N to which R15 and R16 are bonded form a heterocycle that is substituted or unsubstituted;
    • n is 0, 1, or 2;
    • q is 0, 1, 2, 3, or 4;
    • t is 1, 2, 3, 4, 5, or 6; and
    • m is 1, 2, 3, or 4,


      or any other compound herein, or a pharmaceutically acceptable salt thereof.


In some embodiments, the test compound is a compound of Formula (I), (I-a), (I-b), (I-c), (I-d), (I-e), (I-f), (I-g), (I-h), (I-i), (I-j), (I-k), (I-k-1), (I-1), (I-1-1), (I-m), (I-m-1), (I-n), (I-o), (I-p), (II) or (III).


In some embodiments, the test compound is




embedded image


or an ionized form thereof.


In some embodiments, the test compound is




embedded image


or an ionized form thereof.


Computational Methods.

In some embodiments, a cryo-EM model disclosed herein can be used as a tool to screen for ryanodine receptor modulator compounds that bind RyR2. In some embodiments, a cryo-EM model disclosed herein can be used as a tool to screen for ryanodine receptor modulator compounds which preferentially bind leaky RyR2 (e.g., mutated RyR2, or post-translationally modified RyR2 (e.g., phosphorylated, dephosphorylated, oxidized and/or nitrosylated RyR2)) and stabilize the closed state of the RyR channel.


Structures of compounds (e.g., Compound 1) and biomolecular targets (e.g. RyR2) provided herein can be used in computational methods for identifying ligands that bind to a biomolecular target (e.g. RyR2). Such methods can include, for example, screening a library of three-dimensional compound structures to identify ligands that fit a binding pocket of the biomolecular target via a molecular docking system (e.g. Glide, DOCK, AutoDock, AutoDock Vina, FRED, and EnzyDock); de-novo generation of a structure of a ligand that binds the biomolecular target via a ligand structure prediction system (e.g., CHARMM, AMBER, or GROMACS); optimization of known ligands (e.g., Compound 1) by evaluating binding of proposed analogs within the binding cavity of the biomolecular target, and combinations of the preceding.


Structures of compounds (e.g., Compound 1) and biomolecular targets (e.g. RyR2) provided herein can be used in computational methods of predicting a docked position of a target ligand in a binding site of a biomolecule, such as the use of a computer to assist in predicting a docked position of a target ligand in a binding site of a biomolecule that is capable of undergoing an induced fit as disclosed in US20210193273A1, which is incorporated herein by reference in its entirety.


In some embodiments, the present disclosure provides a method for predicting a docked position of a target ligand in a binding site of a biomolecule, the method comprising:

    • receiving a template ligand-biomolecule structure, the template ligand-biomolecule structure comprising a template ligand docked in the binding site of the biomolecule;
    • comparing a pharmacophore model of the template ligand to a pharmacophore model of the target ligand;
    • overlapping the pharmacophore model of the target ligand with the pharmacophore model of the template ligand while the template ligand is in the binding site of the biomolecule; and
    • predicting the docked position of the target ligand in the binding site of the biomolecule based on a position of the pharmacophore model of the target ligand when overlapped with the pharmacophore model of the template ligand, wherein the biomolecule is a RY1&2 domain of RyR2, wherein the template ligand-biomolecule structure is obtained by a process comprising subjecting a complex of the biomolecule and the template ligand to single-particle cryogenic electron microscopy analysis.


In some embodiments, the RY1&2 domain comprises a structure according to TABLE 3. In some embodiments, the template ligand has a three-dimensional conformation according to TABLE 4. In some embodiments, the RY1&2 domain further comprises second binding site. In some embodiments, the second binding site is an ATP-binding site. In some embodiments, the RY1&2 domain further comprises a nucleoside-containing molecule. In some embodiments, the nucleoside-containing molecule is an ATP molecule. In some embodiments the target ligand cooperatively binds the RY1&2 domain with the ATP molecule. In some embodiments, the ATP molecule has a three-dimensional conformation according to TABLE 5. In some embodiments, the target ligand cooperatively binds the RY1&2 domain with the ATP molecule.


In some embodiments, the target ligand is a compound of Formula (I), (I-a), (I-b), (I-c), (I-d), (I-e), (I-f), (I-g), (I-h), (I-i), (I-j), (I-k), (I-k-1), (I-1), (I-1-1), (I-m), (I-m-1), (I-n), (I-o), (I-p), (II) or (III). In some embodiments, the target ligand and the template ligand are each independently a compound of Formula (I), (I-a), (I-b), (I-c), (I-d), (I-e), (I-f), (I-g), (I-h), (I-i), (I-j), (I-k), (I-k-1), (I-1), (I-1-1), (I-m), (I-m-1), (I-n), (I-o), (I-p), (II) or (III).


In some embodiments, the template ligand is




embedded image


In some embodiments, the template ligand is




embedded image


In some embodiments, the template ligand-biomolecule structure obtained by single-particle cryogenic electron microscopy analysis has a resolution from about 2 Å to about 3.5 Å, from about 2 Å to about 3.4 Å, from about 2 Å to about 3.3 Å, from about 2 Å to about 3.2 Å, from about 2 Å to about 3.1 Å, from about 2 Å to about 3 Å, from about 2 Å to about 2.9 Å, from about 2 Å to about 2.8 Å, from about 2 Å to about 2.7 Å, from about 2 Å to about 2.6 Å, from about 2 Å to about 2.5 Å, from about 2.1 Å to about 2.5 Å, from about 2.2 Å to about 2.5 Å, from about 2.3 Å to about 2.5 Å, or from about 2.4 Å to about 2.5 Å.


In some embodiments, the method further comprises selecting the target ligand from a plurality of ligand candidates, each of the ligand candidates being different from the template ligand, and wherein selecting the target ligand comprises comparing the pharmacophore model of the template ligand to a pharmacophore model of each respective one of the plurality of ligand candidates.


In some embodiments, the method further comprises receiving a plurality of template ligand-biomolecule structures, each template ligand-biomolecule structure having a different template ligand docked in the binding site of the biomolecule, and generating the pharmacophore model of the template ligand by combining information from each of the template ligands from the plurality of template ligand-biomolecule structures.


In some embodiments, the target ligand has more than one structural conformation in the unbound state, and the docked position of the target ligand in the binding site of the biomolecule is predicted by enumerating a set of potential target ligand conformations and overlapping a respective pharmacophore model of the target ligand for each of the potential target ligand conformations with the pharmacophore model of the template ligand while the template ligand is in the binding site of the biomolecule.


In some embodiments, predicting the docked position of the target ligand in the binding site of the biomolecule comprises ignoring at least one clash between the target ligand conformation's atomic coordinates and the biomolecule's atomic coordinates.


In some embodiments, the method further comprises, for each target ligand conformation, modifying atomic coordinates of the biomolecule to reduce clashes between the docked target ligand conformation's atomic coordinates and the biomolecule's atomic coordinates, thereby creating an altered ligand-biomolecule structure comprising the docked target ligand and an altered biomolecule.


In some embodiments, the method further comprises predicting a re-docked position of each target ligand conformation by predicting each target ligand conformation's position in the binding site of the altered biomolecule; and for each target ligand conformation, modifying atomic coordinates of the altered biomolecule to reduce clashes between the atomic coordinates of the target ligand conformation's re-docked position and the atomic coordinates of the altered biomolecule, thereby creating are-altered ligand-biomolecule structure comprising a re-docked target ligand and a re-altered biomolecule.


In some embodiments, the method further comprises ranking each altered and re-altered ligand-biomolecule structure using a scoring function.


In some embodiments, the method further comprises identifying a subset of high-ranking target ligands corresponding to target ligands having a threshold value for an empirical activity.


Structures of compounds (e.g., Compound 1) and biomolecular targets (e.g. RyR2) provided herein can be used in systems, devices, and methods that can generate lead compounds on the basis of known structure and activity of a lead compound (e.g., Compound 1) and the structure of a binding site for the lead compound, such as the systems, devices, and methods provided in US20210217500A1, which is incorporated herein by reference in its entirety.


In some embodiments, the present disclosure provides a method of identifying a plurality of potential lead compounds, the method comprising the steps of:

    • (a) analyzing, using a computer system, an initial lead compound known to bind to a biomolecular target, the analyzing comprising partitioning, by providing a database of known reactions, the initial lead compound into atoms defining partitioned lead compound comprising a lead compound core and atoms defining a lead compound non-core, wherein the initial lead compound is partitioned using a computational retrosynthetic analysis of the initial lead compound;
    • (b) identifying, using the computer system, a plurality of alternative cores to replace the lead compound core in the initial lead compound, thereby generating a plurality of potential lead compounds each having a respective one of the plurality of alternative cores;
    • (c) calculating, using the computer system, a difference in binding free energy between the partitioned lead compound and each potential lead compound;
    • (d) predicting, using the computer system, whether each potential lead compound binds to the biomolecular target and identifying a predicted active set of potential lead compounds based on the prediction;
    • (e) obtaining a synthesized set of at least some of the potential leads of the predicted active set to establish a first of potential lead compounds; and
    • (f) determining, empirically, an activity of each of the first set of synthesized potential lead compounds,
    • wherein the biomolecular target is a RY1&2 domain of RyR2, and the structure of the biomolecular target used in the predicting of (d) is obtained by a process comprising subjecting a complex of the biomolecular target and the initial lead compound to single-particle cryogenic electron microscopy analysis.


In some embodiments, the structure of the biomolecular target obtained by single-particle cryogenic electron microscopy analysis has a resolution from about 2 Å to about 3.5 Å, from about 2 Å to about 3.4 Å, from about 2 Å to about 3.3 Å, from about 2 Å to about 3.2 Å, from about 2 Å to about 3.1 Å, from about 2 Å to about 3 Å, from about 2 Å to about 2.9 Å, from about 2 Å to about 2.8 Å, from about 2 Å to about 2.7 Å, from about 2 Å to about 2.6 Å, from about 2 Å to about 2.5 Å, from about 2.1 Å to about 2.5 Å, from about 2.2 Å to about 2.5 Å, from about 2.3 Å to about 2.5 Å, or from about 2.4 Å to about 2.5 Å.


In some embodiments, the initial lead compound is a compound of Formula (I), (I-a), (I-b), (I-c), (I-d), (I-e), (I-f), (I-g), (I-h), (I-i), (I-j), (I-k), (I-k-1), (I-1), (I-1-1), (I-m), (I-m-1), (I-n), (I-o), (I-p), (II) or (III).


In some embodiments, the initial lead compound is




embedded image


In some embodiments, the initial lead compound is




embedded image


In some embodiments, the RY1&2 domain comprises a structure according to TABLE 3. In some embodiments, the RY1&2 domain contains an ATP molecule. In some embodiments, the ATP molecule has a three-dimensional conformation according to TABLE 5.


In some embodiments, the method further comprises obtaining a synthesized set of at least some of the potential lead compounds predicted to not bind with the biomolecular target to establish a second set of potential lead compounds and empirically determining an activity of each of the second set of synthesized potential lead compounds.


In some embodiments, the method further comprises comparing the empirically determined activity of each of the first set of synthesized potential lead compounds with a threshold activity level.


In some embodiments, the method further comprises comparing the empirically determined activity of each of the second set of synthesized potential lead compounds with a pre-determined activity level.


In some embodiments, the plurality of alternative cores are chosen from a database of synthetically feasible cores.


In some embodiments, the difference in binding free energy is calculated using a free energy perturbation technique.


In some embodiments, the generation of at least one potential lead compound comprises creating an additional covalent bond or annihilating an existing covalent bond, or both creating an additional first covalent bond and annihilating an existing second covalent bond different from the first covalent bond.


In some embodiments, the free energy perturbation technique uses a soft bond potential to calculate a bonded stretch interaction energy of existing covalent bonds for annihilation and additional covalent bonds for creation.


In some embodiments, the present disclosure provides a method for pharmaceutical drug discovery, comprising:

    • identifying an initial lead compound for binding to a biomolecular target;
    • using a method to identify a predicted active set of potential lead compounds for binding to the biomolecular target based on the initial lead compound, comprising:
      • (a) analyzing, using a computer system, an initial lead compound known to bind to a biomolecular target, the analyzing comprising partitioning, by providing a database of known reactions, the initial lead compound into atoms defining partitioned lead compound comprising a lead compound core and atoms defining a lead compound non-core, wherein the initial lead compound is partitioned using a computational retrosynthetic analysis of the initial lead compound;
      • (b) identifying, using the computer system, a plurality of alternative cores to replace the lead compound core in the initial lead compound, thereby generating a plurality of potential lead compounds each having a respective one of the plurality of alternative cores;
      • (c) calculating, using the computer system, a difference in binding free energy between the partitioned lead compound and each potential lead compound;
      • (d) predicting, using the computer system, whether each potential lead compound binds to the biomolecular target and identifying a predicted active set of potential lead compounds based on the prediction;
      • (e) obtaining a synthesized set of at least some of the potential leads of the predicted active set to establish a first of potential lead compounds; and
      • (f) determining, empirically, an activity of each of the first set of synthesized potential lead compounds,
      • (g) selecting one or more of the predicted active set of potential lead compounds for synthesis; and
      • (h) assaying the one or more synthesized selected compounds to assess each synthesized selected compounds suitability for in vivo use as a pharmaceutical compound,
    • wherein the biomolecular target is a RY1&2 domain of RyR2, and the structure of the biomolecular target used in the predicting of (d) is obtained by a process comprising subjecting a complex of the biomolecular target and the initial lead compound to single-particle cryogenic electron microscopy analysis.


In some embodiments, the structure of the biomolecular target obtained by single-particle cryogenic electron microscopy analysis has a resolution from about 2 Å to about 3.5 Å, from about 2 Å to about 3.4 Å, from about 2 Å to about 3.3 Å, from about 2 Å to about 3.2 Å, from about 2 Å to about 3.1 Å, from about 2 Å to about 3 Å, from about 2 Å to about 2.9 Å, from about 2 Å to about 2.8 Å, from about 2 Å to about 2.7 Å, from about 2 Å to about 2.6 Å, from about 2 Å to about 2.5 Å, from about 2.1 Å to about 2.5 Å, from about 2.2 Å to about 2.5 Å, from about 2.3 Å to about 2.5 Å, or from about 2.4 Å to about 2.5 Å.


In some embodiments, the initial lead compound is a compound of Formula (I), (I-a), (I-b), (I-c), (I-d), (I-e), (I-f), (I-g), (I-h), (I-i), (I-j), (I-k), (I-k-1), (I-1), (I-1-1), (I-m), (I-m-1), (I-n), (I-o), (I-p), (II) or (III).


In some embodiments, the initial lead compound is




embedded image


In some embodiments, the initial lead compound is




embedded image


In some embodiments, the RY1&2 domain comprises a structure according to TABLE 3. In some embodiments, the RY1&2 domain contains an ATP molecule. In some embodiments, the ATP molecule has a three-dimensional conformation according to TABLE 5.


Structures of compounds (e.g., Compound 1) and biomolecular targets (e.g. RyR2) provided herein can be used in methods that estimate binding affinity between a ligand and a receptor molecule, including the systems and methods disclosed in U.S. Pat. No. 8,160,820B2, which is incorporated by reference herein in its entirety.


In some embodiments, the present disclosure provides a computer-implemented method of quantifying binding affinity between a ligand and a receptor molecule domain, the method comprising:

    • receiving by one or more computers, data representing a ligand molecule,
    • receiving by one or more computers, data representing a receptor molecule domain,
    • using the data representing the ligand molecule and the data representing the receptor molecule domain in computer analysis to identify ring structure within the ligand, the ring structure being an entire ring or a fused ring;
    • using the data representative of the identified ligand ring structure to designate a first ring face and a second ring face opposite to the first ring face, and classifying the ring structure by:
    • a) determining proximity of receptor atoms to atoms on the first face of the ligand ring; and
    • b) determining proximity of receptor atoms to atoms on the second face of the ligand ring;
    • c) determining solvation of the first face of the ligand ring and solvation of the second face of the ligand ring;
    • classifying the identified ligand ring structure as buried, solvent exposed, or having a single face exposed to solvent based on receptor atom proximity to and solvation of the first ring face and receptor atom proximity to and solvation of the second ring face; quantifying the binding affinity between the ligand and the receptor molecule domain based at least in part on the classification of the ring structure; and
    • displaying, via computer, information related to the classification of the ring structure, wherein the receptor molecule domain is a RY1&2 domain of RyR2, wherein the data representing a ligand molecule and the data representing a receptor molecule domain are obtained by a process comprising subjecting a complex comprising the ligand molecule and the receptor molecule domain to single-particle cryogenic electron microscopy analysis.


In some embodiments, the structure of the receptor molecule domain obtained by single-particle cryogenic electron microscopy analysis has a resolution from about 2 Å to about 3.5 Å, from about 2 Å to about 3.4 Å, from about 2 Å to about 3.3 Å, from about 2 Å to about 3.2 Å, from about 2 Å to about 3.1 Å, from about 2 Å to about 3 Å, from about 2 Å to about 2.9 Å, from about 2 Å to about 2.8 Å, from about 2 Å to about 2.7 Å, from about 2 Å to about 2.6 Å, from about 2 Å to about 2.5 Å, from about 2.1 Å to about 2.5 Å, from about 2.2 Å to about 2.5 Å, from about 2.3 Å to about 2.5 Å, or from about 2.4 Å to about 2.5 Å.


In some embodiments, ligand molecule is a compound of Formula (I), (I-a), (I-b), (I-c), (I-d), (I-e), (I-f), (I-g), (I-h), (I-i), (I-j), (I-k), (I-k-1), (I-1), (I-1-1), (I-m), (I-m-1), (I-n), (I-o), (I-p), (II) or


In some embodiments, the ligand molecule is




embedded image


or a pharmaceutically-acceptable salt or ionized form thereof.


In some embodiments, the ligand molecule is




embedded image


or a pharmaceutically-acceptable salt or ionized form thereof.


In some embodiments, the complex further comprises a RyR2 protein, wherein the RY1&2 domain is a domain of the RyR2 protein.


In some embodiments, the data representing the receptor molecule domain represents a three-dimensional structure of the receptor molecule according to TABLE 3. In some embodiments, the data representing a ligand molecule represents a three-dimensional structure of the ligand molecule according to TABLE 4.


In some embodiments, the receptor molecule domain contains an ATP molecule. In some embodiments, the data representing the receptor molecule domain further comprises data representing a three-dimensional structure of the ATP molecule according to TABLE 5.


In some embodiments, quantifying the binding affinity includes a step that scores hydrophobic interactions between one or more ligand atoms and one or more receptor atoms by awarding a bonus for the presence of hydrophobic enclosure of one or more atoms of said ligand by the receptor molecule domain, said bonus being indicative of enhanced binding affinity between said ligand and said receptor molecule domain.


In some embodiments, the method further comprises calculating an initial binding affinity and then adjusting the initial binding affinity based on the classification of the ring structure as buried, solvent exposed, or solvent exposed on one face.


In some embodiments, the classification of a ring structure as buried, solvent exposed, or solvent exposed on one surface, includes using a parameter substantially correlated with the number of close contacts on both sides of the ring structure or part thereof with the receptor molecule domain.


In some embodiments, the number of close contacts at different distances between receptor atoms and the two ring faces are determined, an initial classification of the ring is made based on the numbers of these contacts, and this initial classification is then followed by calculation of a scoring function, said scoring function comprising identifying a first ring shell and a second ring shell, and calculating the number of water molecules in the first shell and in the second shell, or calculating the number of water molecules in the first and second shell combined.


In some embodiments, the scoring function for classification of the ring structure as buried, solvent exposed, or solvent exposed on one surface, includes using a parameter substantially correlated with the lipophilic-lipophilic pair score between the ring structure or part thereof and the receptor molecule domain.


In some embodiments, the scoring function used to classify a ring structure as buried, solvent exposed, or solvent exposed on one surface, includes calculating the degree of enclosure of each atom of the ring structure by atoms of the receptor.


In some embodiments, the scoring function used to classify a ring structure as buried, solvent exposed, or solvent exposed on one surface, includes using a parameter that is substantially correlated with the degree of enclosure of each atom of the ring structure by atoms of the receptor.


In some embodiments, the scoring function enabling classification of the ring structure as buried, solvent exposed, or solvent exposed on one surface, includes the use of a parameter corresponding to a hydrophobic interaction of the ring structure or part thereof with the receptor molecule domain.


In some embodiments, the information displayed by computer includes a depiction of at least one of the degree to which the ring structure is enclosed by atoms of the receptor molecule domain; water molecules surrounding the ring structure in a first shell or a second shell or both the first and the second shell of the ligand; a value of a lipophilic-lipophilic pair score of the ring structure; and a number of close contacts of a face of the ring structure with the receptor molecule domain.


In some embodiments, solvent exposed ring structures in the ligand, if any, are substantially ignored in quantifying the component of the binding affinity between the ligand and the receptor molecule domains, other than to recognize hydrogen bonds and other parameters that are independent of the classification of ring structure.


In some embodiments, hydrophobic contribution to binding affinity from ring structures classified as solvent exposed, if any, is substantially ignored in quantifying the component of the binding affinity.


In some embodiments, a ring structure is classified as buried, and the method further comprises identifying a quantity representative of a strain energy induced in the ligand-receptor complex by the buried ring structure, in which the quantification of the component of binding affinity is further based in part on strain energy.


In some embodiments, the method further comprises identifying a quantity representative of a strain energy induced in the ligand-receptor complex by the aggregate of the ring structures identified as buried; identifying a quantity representative of a total neutral-neutral hydrogen bond energy; and quantifying the component of binding affinity between the ligand and the receptor molecule domain based at least in part on the quantity representative of the strain energy induced in the receptor by the aggregate of the buried ring structures, and on the quantity representative of the total neutral-neutral hydrogen bond energy.


In some embodiments, quantifying the component of binding affinity further comprises identifying a hydrogen bond capping energy associated with the entire ligand, and the component of binding affinity is quantified based on a greater of the hydrogen bond capping energy and the quantity representative of the strain energy induced in the receptor by the aggregate of the identified structures.


In some embodiments, the method further comprises identifying a binding motif of the receptor molecule domain with respect to the ligand; identifying a reorganization energy of the receptor molecule domain based on the binding motif, and identifying a first ring structure as contributing to the reorganization energy, the quantity representative of strain energy being identified independently of the classification of the first ring structure.


In some embodiments, the component of binding affinity attributable to strain is quantified using at least one of: molecular dynamics, molecule mechanics, conformational searching and minimization.


In some embodiments, the information displayed by computer includes a depiction of solvent exposure, if any, of the ring structure.


In some embodiments, the information displayed by computer includes a depiction of burial, if any, of the ring structure.


In some embodiments, the information displayed by computer includes a depiction of at least one of: the degree to which the ring structure is enclosed by atoms of the receptor molecule domain; water molecules surrounding the ring structure in a first shell or a second shell or both the first and the second shell of the ligand; a value of a lipophilic-lipophilic pair score of the ring structure; and a number of close contacts of a face of the ring structure with the receptor molecule domain.


In some embodiments, the method further comprises performing a test on a physical sample that includes the ligand and the receptor molecule domain, test components being selected based at least in part on the binding affinity between the ligand or part thereof and the receptor molecule, or on the component of such binding affinity.


Embodiments of the subject matter and the functional operations described in this specification can be implemented in digital electronic circuitry, in tangibly-embodied computer software or firmware, in computer hardware, including the structures disclosed in this specification and their structural equivalents, or in combinations of one or more of them. Embodiments of the subject matter described in this specification can be implemented as one or more computer programs, i.e., one or more modules of computer program instructions encoded on a tangible non-transitory storage medium for execution by, or to control the operation of, data processing apparatus. The computer storage medium can be a machine-readable storage device, a machine-readable storage substrate, a random or serial access memory device, or a combination of one or more of them. Alternatively, or in addition, the program instructions can be encoded on an artificially generated propagated signal, e.g., a machine-generated electrical, optical, or electromagnetic signal, that is generated to encode information for transmission to suitable receiver apparatus for execution by a data processing apparatus.


The term “data processing apparatus” refers to data processing hardware and encompasses all kinds of apparatus, devices, and machines for processing data, including by way of example a programmable processor, a computer, or multiple processors or computers. The apparatus can also be, or further include, special purpose logic circuitry, e.g., an FPGA (field programmable gate array) or an ASIC (application specific integrated circuit). The apparatus can optionally include, in addition to hardware, code that creates an execution environment for computer programs, e.g., code that constitutes processor firmware, a protocol stack, a database management system, an operating system, or a combination of one or more of them.


A computer program, which is also referred to or described as a program, software, a software application, an app, a module, a software module, a script, or code, can be written in any form of programming language, including compiled or interpreted languages, or declarative or procedural languages; and can be deployed in any form, including as a stand-alone program or as a module, component, subroutine, or other unit suitable for use in a computing environment. A program can, but need not, correspond to a file in a file system. A program can be stored in a portion of a file that holds other programs or data, e.g., one or more scripts stored in a markup language document, in a single file dedicated to the program in question, or in multiple coordinated files, e.g., files that store one or more modules, sub programs, or portions of code. A computer program can be deployed to be executed on one computer or on multiple computers that are located at one site or distributed across multiple sites and interconnected by a data communication network.


The processes and logic flows described in this specification can be performed by one or more programmable computers executing one or more computer programs to perform functions by operating on input data and generating output. The processes and logic flows can also be performed by special purpose logic circuitry, e.g., an FPGA or an ASIC, or by a combination of special purpose logic circuitry and one or more programmed computers.


Computers suitable for the execution of a computer program can be based on general or special purpose microprocessors or both, or any other kind of central processing unit. Generally, a central processing unit receives instructions and data from a read only memory or a random access memory or both. The essential elements of a computer are a central processing unit for performing or executing instructions and one or more memory devices for storing instructions and data. The central processing unit and the memory can be supplemented by, or incorporated in, special purpose logic circuitry. Generally, a computer can also include, or be operatively coupled to receive data from or transfer data to, or both, one or more mass storage devices for storing data, e.g., magnetic, magneto optical disks, or optical disks. However, a computer need not have such devices. Moreover, a computer can be embedded in another device, e.g., a mobile telephone, a personal digital assistant (PDA), a mobile audio or video player, a game console, a Global Positioning System (GPS) receiver, or a portable storage device, e.g., a universal serial bus (USB) flash drive, to name just a few.


Computer readable media suitable for storing computer program instructions and data include all forms of non-volatile memory, media and memory devices, including by way of example semiconductor memory devices, e.g., EPROM, EEPROM, and flash memory devices; magnetic disks, e.g., internal hard disks or removable disks; magneto optical disks; and CD ROM and DVD-ROM disks.


To provide for interaction with a user, embodiments of the subject matter described in this specification can be implemented on a computer having a display device, e.g., a CRT (cathode ray tube) or LCD (liquid crystal display) monitor, for displaying information to the user and a keyboard and a pointing device, e.g., a mouse or a trackball, by which the user can provide input to the computer. Other kinds of devices can be used to provide for interaction with a user as well; for example, feedback provided to the user can be any form of sensory feedback, e.g., visual feedback, auditory feedback, or tactile feedback; and input from the user can be received in any form, including acoustic, speech, or tactile input. In addition, a computer can interact with a user by sending documents to and receiving documents from a device that is used by the user; for example, by sending web pages to a web browser on a user's device in response to requests received from the web browser. Also, a computer can interact with a user by sending text messages or other forms of message to a personal device, e.g., a smartphone that is running a messaging application, and receiving responsive messages from the user in return.


Embodiments of the subject matter described in this specification can be implemented in a computing system that includes a back end component, e.g., as a data server, or that includes a middleware component, e.g., an application server, or that includes a front end component, e.g., a client computer having a graphical user interface, a web browser, or an app through which a user can interact with an implementation of the subject matter described in this specification, or any combination of one or more such back end, middleware, or front end components. The components of the system can be interconnected by any form or medium of digital data communication, e.g., a communication network. Examples of communication networks include a local area network (LAN) and a wide area network (WAN), e.g., the Internet.


The computing system can include clients and servers. A client and server are generally remote from each other and typically interact through a communication network. The relationship of client and server arises by virtue of computer programs running on the respective computers and having a client-server relationship to each other. In some embodiments, a server transmits data, e.g., an HTML page, to a user device, e.g., for purposes of displaying data to and receiving user input from a user interacting with the device, which acts as a client. Data generated at the user device, e.g., a result of the user interaction, can be received at the server from the device.


EMBODIMENTS

The following non-limiting embodiments provide illustrative examples of the invention, but do not limit the scope of the invention.

    • Embodiment 1. A composition comprising a mixture of water and a protein, wherein the protein is a ryanodine receptor 2 protein (RyR2) or a mutant thereof.
    • Embodiment 2. The composition of embodiment 1, further comprising a synthetic compound.
    • Embodiment 3. The composition of embodiment 1 or embodiment 2, further comprising calmodulin.
    • Embodiment 4. The composition of embodiments 3, wherein the calmodulin is human calmodulin.
    • Embodiment 5. The composition of embodiment 3 or embodiment 4, wherein calmodulin is present in the mixture at a concentration from about 1 μM to about 200 μM.
    • Embodiment 6. The composition of embodiment 3 or embodiment 4, wherein calmodulin is present in the mixture at a concentration from about 1 μM to about 60 μM.
    • Embodiment 7. The composition of embodiment 3 or embodiment 4, wherein calmodulin is present in the mixture at a concentration of about 20 μM.
    • Embodiment 8. The composition of embodiment 3 or embodiment 4, wherein calmodulin is present in the mixture at a concentration of about 40 μM.
    • Embodiment 9. The composition of any one of embodiments 1-8, further comprising a buffering agent in the mixture.
    • Embodiment 10. The composition of embodiment 9, wherein the buffering agent is HEPES.
    • Embodiment 11. The composition of embodiment 9, wherein the buffering agent is EGTA.
    • Embodiment 12. The composition of any one of embodiments 1-11, further comprising a phospholipid in the mixture.
    • Embodiment 13. The composition of embodiment 12, wherein the phospholipid is dioleoylphosphatidylcholine (DOPC).
    • Embodiment 14. The composition of embodiment 12, wherein the phospholipid is 1,2-dioleoyl-sn-glycero-3-phospho-L-serine sodium salt (DOPS).
    • Embodiment 15. The composition of any one of embodiments 1-14, further comprising a zwitterionic surfactant in the mixture.
    • Embodiment 16. The composition of embodiment 15, wherein the zwitterionic surfactant is CHAPS.
    • Embodiment 17. The composition of any one of embodiments 1-16, further comprising a disulfide-reducing agent in the mixture.
    • Embodiment 18. The composition of embodiment 17, wherein the disulfide-reducing agent is TCEP.
    • Embodiment 19. The composition of embodiment 17, wherein the disulfide-reducing agent is dithiothreitol.
    • Embodiment 20. The composition of any one of embodiments 1-18, further comprising a protease inhibitor in the mixture.
    • Embodiment 21. The composition of embodiment 20, wherein the protease inhibitor is AEBSF.
    • Embodiment 22. The composition of embodiment 20, wherein the protease inhibitor is benzamidine or a salt thereof.
    • Embodiment 23. The composition of any one of embodiments 1-22, further comprising xanthine in the mixture.
    • Embodiment 24. The composition of embodiment 23, wherein xanthine is present in the mixture at a concentration from about 300 μM to about 700 μM.
    • Embodiment 25. The composition of embodiment 23, wherein xanthine is present in the mixture at a concentration of about 500 μM.
    • Embodiment 26. The composition of any one of embodiments 1-25, further comprising dissolved Ca2+ in the mixture.
    • Embodiment 27. The composition of embodiment 26, wherein dissolved Ca2+ is present in the mixture at a concentration from about 15 nM to about 500 nM.
    • Embodiment 28. The composition of embodiment 26, wherein dissolved Ca2+ is present in the mixture at a concentration from about 100 nM to about 300 nM.
    • Embodiment 29. The composition of embodiment 26, wherein dissolved Ca2+ is present in the mixture at a concentration of about 150 nM.
    • Embodiment 30. The composition of any one of embodiments 1-29, wherein the protein is present in the mixture at a concentration from about 0.1 mg/mL to about 50 mg/mL.
    • Embodiment 31. The composition of any one of embodiments 1-29, wherein the protein is present in the mixture at a concentration from about 0.1 mg/mL to about 20 mg/mL.
    • Embodiment 32. The composition of any one of embodiments 1-29, wherein the protein is present in the mixture at a concentration from about 1 mg/mL to about 10 mg/mL.
    • Embodiment 33. The composition of any one of embodiments 1-32, further comprising sodium adenosine triphosphate (NaATP) in the mixture.
    • Embodiment 34. The composition of embodiment 33, wherein the NaATP is present in the mixture at a concentration from about 3 mM to about 15 nM.
    • Embodiment 35. The composition of embodiment 33, wherein the NaATP is present in the mixture at a concentration from about 10 mM.
    • Embodiment 36. The composition of any one of embodiments 1-35, further comprising cyclic adenosine monophosphate (cAMP) in the mixture.
    • Embodiment 37. The composition of embodiment 36, wherein the cAMP is present in the mixture at a concentration from about 50 μM to about 500 μM.
    • Embodiment 38. The composition of embodiment 36, wherein the cAMP is present in the mixture at a concentration from about 200 μM.
    • Embodiment 39. The composition of any one of embodiments 1-38, wherein the protein is human RyR2.
    • Embodiment 40. The composition of any one of embodiments 1-39, wherein the protein is mutant RyR2.
    • Embodiment 41. The composition of embodiment 40, wherein the mutant RyR2 is R2474S-RyR2.
    • Embodiment 42. The composition of any one of embodiments 1-41, wherein the protein is in a phosphorylated state, wherein the phosphorylated state is prepared by a process comprising contacting the RyR2 protein with a phosphorylation reagent.
    • Embodiment 43. The composition of embodiment 42, wherein the phosphorylation reagent comprises protein kinase A.
    • Embodiment 44. The composition of embodiment 43, wherein the phosphorylation reagent further comprises ATP.
    • Embodiment 45. The composition of embodiment 43 or embodiment 44, wherein the phosphorylation reagent further comprises MgCl2.
    • Embodiment 46. The composition of any one of embodiments 1-41, wherein the protein is in a dephosphorylated state, wherein the dephosphorylated state is prepared by a process comprising contacting the RyR2 protein with a dephosphorylation reagent.
    • Embodiment 47. The composition of embodiment 46, wherein the dephosphorylation reagent comprises phosphatase lambda.
    • Embodiment 48. The composition of embodiment 47, wherein the dephosphorylation reagent further comprises MnCl2.
    • Embodiment 49. The composition of any one of embodiments 1-48, wherein the composition is substantially free of cellular membrane.
    • Embodiment 50. A composition comprising a complex suspended in a solid medium, wherein the complex comprises a protein, wherein the protein is a ryanodine receptor 2 protein (RyR2) or mutant thereof.
    • Embodiment 51. The composition of embodiment 50, wherein the composition is prepared by a process, the process comprising vitrifying an aqueous solution that is applied to an electron microscopy grid, wherein the aqueous solution comprises the complex.
    • Embodiment 52. The composition of embodiment 51, wherein, prior to the vitrifying, the aqueous solution is applied to the electron microscopy grid, and excess aqueous solution is removed from the electron microscopy grid by blotting the excess aqueous solution.
    • Embodiment 53. The composition of embodiment 51 or embodiment 52, wherein the vitrifying comprises plunge freezing the aqueous solution applied to the electron microscopy grid into liquid ethane chilled with liquid nitrogen.
    • Embodiment 54. The composition of any one of embodiments 51-53, wherein the aqueous solution further comprises a buffering agent.
    • Embodiment 55. The composition of embodiment 54, wherein the buffering agent is HEPES.
    • Embodiment 56. The composition of embodiment 54, wherein the buffering agent is EGTA.
    • Embodiment 57. The composition of any one of embodiments 51-56, wherein the aqueous solution further comprises a phospholipid.
    • Embodiment 58. The composition of embodiment 57, wherein the phospholipid is DOPS.
    • Embodiment 59. The composition of embodiment 57, wherein the phospholipid is DOPC.
    • Embodiment 60. The composition of any one of embodiments 51-59, wherein the aqueous solution further comprises a zwitterionic surfactant.
    • Embodiment 61. The composition of embodiment 60, wherein the zwitterionic surfactant is CHAPS.
    • Embodiment 62. The composition of any one of embodiments 51-61, wherein the aqueous solution further comprises a disulfide-reducing agent.
    • Embodiment 63. The composition of embodiment 62, wherein the disulfide-reducing agent is TCEP.
    • Embodiment 64. The composition of embodiment 62, wherein the disulfide-reducing agent is dithiothreitol.
    • Embodiment 65. The composition of any one of embodiments 51-64, wherein the aqueous solution further comprises a protease inhibitor.
    • Embodiment 66. The composition of embodiment 65, wherein the protease inhibitor is AEBSF.
    • Embodiment 67. The composition of embodiment 65, wherein the protease inhibitor is benzamidine or a salt thereof.
    • Embodiment 68. The composition of any one of embodiments 51-67, wherein the aqueous solution further comprises xanthine.
    • Embodiment 69. The composition of embodiment 68, wherein the xanthine is present in the aqueous solution at a concentration from about 300 μM to about 700 μM.
    • Embodiment 70. The composition of embodiment 68, wherein the xanthine is present in the aqueous solution at a concentration of about 500 μM.
    • Embodiment 71. The composition of any one of embodiments 51-70, wherein the aqueous solution further comprises dissolved Ca2+.
    • Embodiment 72. The composition of embodiment 71, wherein the dissolved Ca2+ is present in the aqueous solution at a concentration from about 15 μM to about 500 μM.
    • Embodiment 73. The composition of embodiment 71, wherein the dissolved Ca2+ is present in the aqueous solution at a concentration from about 100 nM to about 300 nM.
    • Embodiment 74. The composition of embodiment 71, wherein the dissolved Ca2+ is present in the aqueous solution at a concentration of about 150 nM.
    • Embodiment 75. The composition of any one of embodiments 50-74, wherein the protein is present in the aqueous solution at a concentration from about 0.1 mg/mL to about 50 mg/mL.
    • Embodiment 76. The composition of any one of embodiments 50-74, wherein the protein is present in the aqueous solution at a concentration from about 0.1 mg/mL to about 20 mg/mL.
    • Embodiment 77. The composition of any one of embodiments 50-74, wherein the protein is present in the aqueous solution at a concentration from about 0.1 mg/mL to about 10 mg/mL.
    • Embodiment 78. The composition of any one of embodiments 51-77, wherein the aqueous solution further comprises sodium adenosine triphosphate (NaATP).
    • Embodiment 79. The composition of embodiment 78, wherein the NaATP is present at a concentration from about 3 mM to about 15 nM.
    • Embodiment 80. The composition of embodiment 78, wherein the NaATP is present at a concentration of about 10 mM.
    • Embodiment 81. The composition of any one of embodiments 51-80, further comprising cyclic adenosine monophosphate (cAMP).
    • Embodiment 82. The composition of embodiment 81, wherein the cAMP is present in the aqueous solution at a concentration from about 50 μM to about 500 μM.
    • Embodiment 83. The composition of embodiment 81, wherein the cAMP is present in the aqueous solution at a concentration from about 200 μM.
    • Embodiment 84. The composition of any one of embodiments 51-83, wherein the aqueous solution further comprises calmodulin.
    • Embodiment 85. The composition of embodiment 84, wherein the calmodulin is human calmodulin.
    • Embodiment 86. The composition of embodiment 84 or embodiment 85, wherein calmodulin is present in the aqueous solution at a concentration from about 1 μM to about 200 μM.
    • Embodiment 87. The composition of embodiment 84 or embodiment 85, wherein calmodulin is present in the aqueous solution at a concentration from about 1 μM to about 60 μM.
    • Embodiment 88. The composition of embodiment 84 or embodiment 85, wherein calmodulin is present in the aqueous solution at a concentration of about 20 μM.
    • Embodiment 89. The composition of embodiment 84 or embodiment 85, wherein calmodulin is present in the aqueous solution at a concentration of about 40 μM.
    • Embodiment 90. The composition of any one of embodiments 50-77, wherein the complex further comprises calmodulin.
    • Embodiment 91. The composition of embodiment 90, wherein the calmodulin is human calmodulin.
    • Embodiment 92. The composition of any one of embodiments 50-91, wherein the complex further comprises calstabin.
    • Embodiment 93. The composition of embodiment 92, wherein the calstabin is calstabin-2.
    • Embodiment 94. The composition of embodiment 92, wherein the calstabin is human calstabin.
    • Embodiment 95. The composition of any one of embodiments 50-94, wherein the complex further comprises a xanthine molecule.
    • Embodiment 96. The composition of any one of embodiments 50-95, wherein the complex further comprises a Ca2+ ion.
    • Embodiment 97. The composition of any one of embodiments 50-96, wherein the RyR2 protein is in the closed state.
    • Embodiment 98. The composition of any one of embodiments 50-97, wherein the composition is substantially free of cellular membrane.
    • Embodiment 99. The composition of any one of embodiments 50-98, wherein the solid medium comprises vitreous ice.
    • Embodiment 100. The composition of embodiment 99, wherein the solid medium is substantially free of crystalline ice.
    • Embodiment 101. The composition of any one of embodiments 50-100, wherein the RyR2 protein is in a closed state.
    • Embodiment 102. The composition of any one of embodiments 50-100, wherein the RyR2 protein is in an open state.
    • Embodiment 103. The composition of any one of embodiments 50-100, wherein the RyR2 protein is in a primed state, wherein if the RyR2 protein is placed in a physiological medium, then the RyR2 protein in the primed state has an open probability (Po) that is higher than an open probability (Po) of the RyR2 in a closed state, and an open probability (Po) that is lower than an open probability (Po) of the RyR2 protein in an open state.
    • Embodiment 104. The composition of any one of embodiments 50-100, wherein the composition further comprises additional complexes, wherein each of the additional complexes independently comprises the protein.
    • Embodiment 105. The composition of embodiment 104, wherein at least about 50%, at least about 60%, at least about 70%, at least about 80%, or at least about 90% of the additional complexes are in the closed state.
    • Embodiment 106. The composition of any one of embodiments 50-100, wherein the protein is R2474S-RyR2, wherein if a study is conducted, the study comprising:
      • (i) determining a structure of a BSol2 domain of the protein in the complex by subjecting the complex to single particle cryogenic electron microscopy analysis, and
      • (ii) calculating root mean square deviation of atomic positions (RMSD) of the BSol2 domain of the protein relative to a BSol2 domain of a reference structure, wherein the reference structure is a structure of a wild type RyR2 protein in a closed state,
    • then the RMSD is no more than about 4.5, no more than about 4, no more than about 3.5, or no more than about 3.
    • Embodiment 107. The composition of any one of embodiments 50-100, wherein the protein is RyR2-R2474S, wherein if a study is conducted, the study comprising:
      • (i) determining a structure of a BSol2 domain of the protein in the complex by subjecting the complex to single particle cryogenic electron microscopy analysis, and
      • (ii) calculating root mean square deviation of atomic positions (RMSD) of the BSol2 domain of the protein relative to a BSol2 domain of a reference structure, wherein the reference structure is a structure of a wild type RyR2 protein in a closed state,
      • then the RMSD is from about 1 to about 4.5, about 1 to about 4, about 1 to about 3.5, about 1 to about 3, about 2 to 4.5, about 2 to about 4, about 2 to about 3.5, or about 2 to about 3. 103981 Embodiment 108. The composition of any one of embodiments 50-100, wherein the protein is RyR2-R2474S, wherein if a study is conducted, the study comprising:
      • (i) determining a structure of a BSol domain of the protein in the complex by subjecting the complex to single particle cryogenic electron microscopy analysis, and
      • (ii) calculating root mean square deviation of atomic positions (RMSD) of the BSol domain of the protein relative to a BSol domain of a reference structure, wherein the reference structure is a structure of a wild type RyR2 protein in a closed state,
      • then the RMSD is no more than about 3, no more than about 2.5, no more than about 2, or no more than about 1.5.
    • Embodiment 109. The composition of any one of embodiments 50-100, wherein the protein is RyR2-R2474S, wherein if a study is conducted, the study comprising:
      • (i) determining a structure of a BSol domain of the protein in the complex by subjecting the complex to single particle cryogenic electron microscopy analysis, and
      • (ii) calculating root mean square deviation of atomic positions (RMSD) of the BSol domain of the protein relative to a BSol domain of a reference structure, wherein the reference structure is a structure of a wild type RyR2 protein in a closed state,
      • then the RMSD is from about 0.5 to about 3, about 0.5 to about 2.5, about 0.5 to about 2, about 0.5 to about 1.5, about 1 to 3, about 1 to about 2.5, about 1 to about 2, or about 1 to about 1.5.
    • Embodiment 110. The composition of any one of embodiments 50-100, wherein the protein is RyR2-R2474S, wherein if a study is conducted, the study comprising:
      • (i) determining a structure of a NTD domain of the protein in the complex by subjecting the complex to single particle cryogenic electron microscopy analysis, and
      • (ii) calculating root mean square deviation of atomic positions (RMSD) of the NTD domain of the protein relative to a NTD domain of a reference structure, wherein the reference structure is a structure of a wild type RyR2 protein in a closed state,
      • then the RMSD is no more than about 1.5, no more than about 1.4, no more than about 1.3, no more than about 1.2, no more than about 1.1, or no more than about 1.
    • Embodiment 111. The composition of any one of embodiments 50-100, wherein the protein is RyR2-R2474S, wherein if a study is conducted, the study comprising:
      • (i) determining a structure of a NTD domain of the protein in the complex by subjecting the complex to single particle cryogenic electron microscopy analysis, and
      • (ii) calculating root mean square deviation of atomic positions (RMSD) of the NTD domain of the protein relative to a NTD domain of a reference structure, wherein the reference structure is a structure of a wild type RyR2 protein in a closed state,
      • then the RMSD is from about 0.5 to about 1.6, about 0.5 to about 1.5, about 0.5 to about 1.4, about 0.5 to about 1.3, or about 0.5 to about 1.2.
    • Embodiment 112. The composition of any one of embodiments 50-100, wherein the protein is RyR2-R2474S, wherein if a study is conducted, the study comprising:
      • (i) determining a structure of a SPRY domain of the protein in the complex by subjecting the complex to single particle cryogenic electron microscopy analysis, and
      • (ii) calculating root mean square deviation of atomic positions (RMSD) of the SPRY domain of the protein relative to a SPRY domain of a reference structure, wherein the reference structure is a structure of a wild type RyR2 protein in a closed state,
      • then the RMSD is less than about 1.2, less than about 1.1, or less than about 1, less than about 0.9, less than about 0.8, or less than about 0.7.
    • Embodiment 113. The composition of any one of embodiments 50-100, wherein the protein is RyR2-R2474S, wherein if a study is conducted, the study comprising:
      • (i) determining a structure of a SPRY domain of the protein in the complex by subjecting the complex to single particle cryogenic electron microscopy analysis, and
      • (ii) calculating root mean square deviation of atomic positions (RMSD) of the SPRY domain of the protein relative to a SPRY domain of a reference structure, wherein the reference structure is a structure of a wild type RyR2 protein in a closed state,
      • then the RMSD is from about 0.2 to about 1.3, about 0.2 to about 1.2, about 0.2 to about 1.1, about 0.2 to about 1, or about 0.2 to about 0.9.
    • Embodiment 114. The composition of any one of embodiments 106-113, wherein the reference structure is a structure according to Protein Data Bank entry 7U9Q.
    • Embodiment 115. The composition of any one of embodiments 106-113, wherein the RMSD is calculated via a process, the process comprising calculating a difference in atomic positions between a structure of the protein and a structure of the wild type RyR2 protein in the closed state, wherein the structure of the protein is determined via cryogenic electronic microscopy analysis of the complex.
    • Embodiment 116. The composition of any one of embodiments 106-115, wherein the wild type RyR2 protein in the closed state has a structure according to Protein Data Bank entry 7U9Q.
    • Embodiment 117. The composition of any one of embodiments 50-105, wherein the protein is wild type RyR2.
    • Embodiment 118. The composition of any one of embodiments 50-105, wherein the protein is a mutant RyR2.
    • Embodiment 119. The composition of embodiment 118, wherein the mutant RyR2 contains at least one mutation that is associated with Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT).
    • Embodiment 120. The composition of embodiment 119, wherein the mutation is RyR2-R2474S.
    • Embodiment 121. The composition of embodiment 119, wherein the mutation is RyR2-R420Q.
    • Embodiment 122. The composition of embodiment 119, wherein the mutation is RyR2-R420W.
    • Embodiment 123. The composition of any one of embodiments 119-122, wherein the mutation destabilizes an interaction between NTD and BSol domains of the RyR2 protein.
    • Embodiment 124. The composition of any one of embodiments 119-122, wherein the mutation destabilizes a cytosolic shell of the RyR2 protein, wherein the cytosolic shell comprises NTD, SPRY, JSol and BSol domains of the RyR2 proteins.
    • Embodiment 125. The composition of any one of embodiments 50-124, wherein the protein is a post-translationally modified RyR2 protein.
    • Embodiment 126. The composition of embodiment 125, wherein the post-translationally modified RyR2 protein is a phosphorylated, oxidized or nitrosylated RyR2.
    • Embodiment 127. The composition of embodiment 125 or 126, wherein the post-translationally modified RyR2 protein is a phosphorylated RyR2.
    • Embodiment 128. The composition of any one of embodiments 125-127, wherein the post-translationally modified RyR2 protein is associated with heart failure.
    • Embodiment 129. The composition of any one of embodiments 125-128, wherein the post-translational modification destabilizes an interaction between NTD and BSol domains of the RyR2 protein.
    • Embodiment 130. The composition of any one of embodiments 125-128, wherein the post-translational modification destabilizes a cytosolic shell of the RyR2 protein, wherein the cytosolic shell comprises NTD, SPRY, JSol and BSol domains of the RyR2 proteins.
    • Embodiment 131. The composition of any one of embodiments 105-130, wherein the RyR2 is a mutated and post-translationally modified RyR2.
    • Embodiment 132. The composition of any one of embodiments 50-131, wherein the protein is human RyR2.
    • Embodiment 133. The composition of any one of embodiments 50-132, wherein the protein is in a phosphorylated state, wherein the phosphorylated state is prepared by a process comprising contacting RyR2 protein with a phosphorylation reagent.
    • Embodiment 134. The composition of embodiment 133, wherein the phosphorylation reagent comprises protein kinase A.
    • Embodiment 135. The composition of embodiment 134, wherein the phosphorylation reagent further comprises ATP.
    • Embodiment 136. The composition of embodiment 133 or embodiment 134, wherein the phosphorylation reagent further comprises MgCl2.
    • Embodiment 137. The composition of any one of embodiments 50-132, wherein the protein is in a dephosphorylated state, wherein the dephosphorylated state is prepared by a process comprising contacting RyR2 protein with a dephosphorylation reagent.
    • Embodiment 138. The composition of embodiment 137, wherein the dephosphorylation reagent comprises phosphatase lambda.
    • Embodiment 139. The composition of embodiment 138, wherein the dephosphorylation reagent further comprises MnCl2.
    • Embodiment 140. The composition of any one of embodiments 1-105, wherein the protein is a tetramer of RyR2 monomers, wherein each RyR2 monomer is SEQ ID NO: 3.
    • Embodiment 141. The composition of any one of embodiments 1-105, wherein the protein is a tetramer of RyR2 monomers, wherein each RyR2 monomer is SEQ ID NO: 4.
    • Embodiment 142. The composition of any one of embodiments 50-141, wherein the complex further comprises a nucleoside-containing molecule.
    • Embodiment 143. The composition of embodiment 142, wherein the nucleoside-containing molecule is a purine nucleoside-containing molecule.
    • Embodiment 144. The composition of embodiment 142 or embodiment 143, wherein the nucleoside-containing molecule is a nucleotide or nucleoside polyphosphate.
    • Embodiment 145. The composition of any one of embodiments 142-144, wherein the nucleoside-containing molecule is an adenosine triphosphate (ATP) molecule.
    • Embodiment 146. The composition of embodiment 145, wherein the ATP molecule has a three-dimensional conformation according to TABLE 5.
    • Embodiment 147. The composition of embodiment 142 or embodiment 143, wherein the nucleoside-containing molecule is an adenosine diphosphate (ATP) molecule.
    • Embodiment 148. The composition of embodiment 147, wherein the complex further comprises a second ATP molecule, wherein both ATP molecules bind a common RYR domain of the protein.
    • Embodiment 149. The composition of embodiment 142, wherein the complex further comprises a second nucleoside-containing molecule.
    • Embodiment 150. The composition of embodiment 149, wherein the second nucleoside-containing molecule binds a C-terminal domain of the RyR2 protein.
    • Embodiment 151. The composition of embodiment 149 or embodiment 150, wherein the second nucleoside-containing molecule is a nucleotide or nucleoside polyphosphate.
    • Embodiment 152. The composition of any one of embodiments 149-151, wherein the second nucleoside-containing molecule is a second ATP molecule.
    • Embodiment 153. The composition of any one of embodiments 50-140, further comprising a synthetic compound.
    • Embodiment 154. The composition of embodiment 153, wherein the complex further comprises a nucleoside-containing molecule.
    • Embodiment 155. The composition of embodiment 154, wherein the nucleoside-containing molecule and the synthetic compound bind a RYR domain of the protein.
    • Embodiment 156. The composition of embodiment 155, wherein the RYR domain is a RY1&2 domain.
    • Embodiment 157. The composition of embodiment 156, wherein the RY1&2 domain has a three-dimensional structure according to TABLE 3.
    • Embodiment 158. The composition of any one of embodiments 154-157, wherein the nucleoside-containing molecule is a purine nucleoside-containing molecule.
    • Embodiment 159. The composition of any one of embodiments 154-158, wherein the nucleoside-containing molecule is a nucleotide or nucleoside polyphosphate.
    • Embodiment 160. The composition of any one of embodiments 154-159, wherein the nucleoside-containing molecule is an adenosine triphosphate (ATP) molecule.
    • Embodiment 161. The composition of embodiment 160, wherein the ATP molecule has a three-dimensional conformation according to TABLE 5.
    • Embodiment 162. The composition of embodiment 160 or embodiment 161, wherein the ATP molecule is cooperatively bound to the protein with the synthetic compound.
    • Embodiment 163. The composition of any one of embodiments 160-162, wherein the complex further comprises a second ATP molecule, wherein both ATP molecules bind a common RYR domain of the protein.
    • Embodiment 164. The composition of any one of embodiments 154-159, wherein the complex further comprises a second nucleoside-containing molecule.
    • Embodiment 165. The composition of embodiment 164, wherein the second nucleoside-containing molecule binds a C-terminal domain of the RyR2 protein.
    • Embodiment 166. The composition of embodiment 164, wherein the second nucleoside-containing molecule is a nucleotide or nucleoside polyphosphate.
    • Embodiment 167. The composition of any one of embodiments 164-166, wherein the second nucleoside-containing molecule is a second ATP molecule.
    • Embodiment 168. The composition of any one of embodiments 1-49 and 153, wherein the synthetic compound binds a RYR domain of the protein.
    • Embodiment 169. The composition of embodiment 168, wherein the RYR domain is a RY1&2 domain.
    • Embodiment 170. The composition of any one of embodiments 1-49 and 153-169, wherein the synthetic compound comprises a benzazepane, benzothiazepane, or benzodiazepane moiety.
    • Embodiment 171. The composition of any one of embodiments 1-49 and 153-169, wherein the synthetic compound comprises a benzothiazepane moiety.
    • Embodiment 172. The composition of any one of embodiments 1-49 and 153-169, wherein the synthetic compound is a compound of Formula (I):




embedded image




    • wherein:
      • each R is independently acyl, —O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, arylthio, arylamino, heteroarylthio, or heteroarylamino, each of which is independently substituted or unsubstituted; or halogen, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;
      • R1 is alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • R2 is alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H, —C(═O)R5, —C(═S)R6, —SO2R7, —P(═O)R8R9, or —(CH2)m—R10;
      • R3 is acyl, —O-acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or substituted; or H, —CO2Y, or —C(═O)NHY;
      • Y is alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • R4 is alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • each R5 is acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —NR15R16, —(CH2)tNR15R16, —NHNR15R16, —NHOH, —OR15, —C(═O)NHNR15R16, —CO2R15, —C(═O)NR15R16, or —CH2X;
      • each R6 is acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NHNR15R16, —NHOH, —NR15R16, or —CH2X;
      • each R7 is alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NR15R16, —NHNR15R16, —NHOH, or —CH2X;
      • each R8 and R9 are each independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or OH;
      • each R10 is —NR15R16, OH, —SO2R11, —NHSO2R11, C(═O)(R12), NHC═O(R12), —OC═O(R12), or —P(═O)R13R14;
      • each R11, R12, R13, and R14 is independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or H, OH, NH2, —NHNH2, or —NHOH;
      • each X is halogen, —CN, —CO2R15, —C(═O)NR15R16, —NR15R16, —OR15, —SO2R7, or —P(═O)R8R9;
      • each R15 and R16 is independently acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted, or H; or R15 and R16 together with the N to which R15 and R16 are bonded form a heterocycle that is substituted or unsubstituted;
      • n is 0, 1, or 2;
      • q is 0, 1, 2, 3, or 4;
      • t is 1, 2, 3, 4, 5, or 6; and
      • m is 1, 2, 3, or 4,

    • or a pharmaceutically-acceptable salt thereof.

    • Embodiment 173. The composition of any one of embodiments 1-49 and 153-172, wherein the synthetic compound is a compound of Formula (I-k):







embedded image




    • wherein:
      • each R is independently acyl, —O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, arylthio, arylamino, heteroarylthio, or heteroarylamino, each of which is independently substituted or unsubstituted; or halogen, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;
      • R18 is alkyl, aryl, cycloalkyl, or heterocyclyl, each of which is independently substituted or unsubstituted; or —NR15R16, —C(═O)NR15R16, —(C═O)OR15, or —OR15;
      • q is 0, 1, 2, 3, or 4;
      • p is 1, 2, 3, 4, 5, 6, 7, 8 9, or 10; and
      • n is 0, 1, or 2,

    • or a pharmaceutically-acceptable salt thereof.

    • Embodiment 174. The composition of embodiment 172 or embodiment 173, wherein each R is independently H, halogen, —OH, OMe, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —S(═O)2C1-C4alkyl, —S(═O)C1-C4alkyl, —S—C1-C4alkyl, —OS(═O)2CF3, Ph, —NHCH2Ph, —C(═O)Me, —OC(═O)Me, morpholinyl, or propenyl; and n is 0, 1 or 2.

    • Embodiment 175. The composition of any one of embodiments 172-174, wherein R18 is —NR15R16, —(C═O)OR15, —OR15, alkyl that is substituted or unsubstituted, or aryl that is substituted or unsubstituted

    • Embodiment 176. The composition of any one of embodiments 1-49 and 153-172, wherein the synthetic compound is a compound of Formula (I-0):







embedded image




    • wherein:
      • Re is (C1-C6 alkyl)-phenyl, (C1-C6 alkyl)-C(O)Rb, or substituted or unsubstituted —C1-C6 alkyl; and
      • Rb is OH or —O—(C1-C6 alkyl), wherein the phenyl or the substituted alkyl is substituted with one or more of halogen, hydroxyl, C1-C6 alkyl, —O—(C1-C6 alkyl), —NH2, —NH(C1-C6 alkyl), —N(C1-C6 alkyl)2, cyano, or dioxolane,

    • or a pharmaceutically-acceptable salt thereof.

    • Embodiment 177. The composition of any one of embodiments 1-49 and 153-176, wherein the synthetic compound is:







embedded image




    •  or an ionized form thereof.

    • Embodiment 178. The composition of any one of embodiments 1-49 and 153-176, wherein the synthetic compound is:







embedded image




    •  or an ionized form thereof.

    • Embodiment 179. The composition of embodiment 177, wherein the synthetic compound has a three-dimensional conformation according to TABLE 4.

    • Embodiment 180. A vessel containing the composition of any one of embodiments 1-49.

    • Embodiment 181. The vessel of embodiment 181, wherein the vessel is a vial, ampule, test tube, or microwell plate.

    • Embodiment 182. A method of determining a binding site of a synthetic compound in a protein, the method comprising subjecting a composition of any one of embodiments 153-179 to single-particle cryogenic electron microscopy analysis.

    • Embodiment 183. The method of embodiment 182, wherein the structure of the of protein obtained by single-particle cryogenic electron microscopy analysis has a resolution from about 2 Å to about 3.5 Å, from about 2 Å to about 3.4 Å, from about 2 Å to about 3.3 Å, from about 2 Å to about 3.2 Å, from about 2 Å to about 3.1 Å, from about 2 Å to about 3 Å, from about 2 Å to about 2.9 Å, from about 2 Å to about 2.8 Å, from about 2 Å to about 2.7 Å, from about 2 Å to about 2.6 Å, from about 2 Å to about 2.5 Å, from about 2.1 Å to about 2.5 Å, from about 2.2 Å to about 2.5 Å, from about 2.3 Å to about 2.5 Å, or from about 2.4 Å to about 2.5 Å.

    • Embodiment 184. A method for predicting a docked position of a target ligand in a binding site of a biomolecule, the method comprising:
      • receiving a template ligand-biomolecule structure, the template ligand-biomolecule structure comprising a template ligand docked in the binding site of the biomolecule;
      • comparing a pharmacophore model of the template ligand to a pharmacophore model of the target ligand;
      • overlapping the pharmacophore model of the target ligand with the pharmacophore model of the template ligand while the template ligand is in the binding site of the biomolecule; and
      • predicting the docked position of the target ligand in the binding site of the biomolecule based on a position of the pharmacophore model of the target ligand when overlapped with the pharmacophore model of the template ligand,
      • wherein the biomolecule is a RY1&2 domain of RyR2, wherein the template ligand-biomolecule structure is obtained by a process comprising subjecting a complex of the biomolecule and the template ligand to single-particle cryogenic electron microscopy analysis.

    • Embodiment 185. The method of embodiment 184, wherein the RY1&2 domain comprises a structure according to TABLE 3.

    • Embodiment 186. The method of embodiment 184, wherein the template ligand has a three-dimensional conformation according to TABLE 4.

    • Embodiment 187. The method of embodiment 184, wherein the RY1&2 domain further comprises a nucleoside-containing molecule.

    • Embodiment 188. The method of embodiment 187, wherein the nucleoside-containing molecule is an ATP molecule.

    • Embodiment 189. The method of embodiment 188, wherein the ATP molecule has a three-dimensional conformation according to TABLE 5.

    • Embodiment 190. The method of embodiment 187 or embodiment 188, wherein the target ligand cooperatively binds the RY1&2 domain with the ATP molecule.

    • Embodiment 191. The method of embodiment 184, wherein the target ligand and the template ligand are each independently a compound of Formula (I):







embedded image




    • wherein:
      • each R is independently acyl, —O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, arylthio, arylamino, heteroarylthio, or heteroarylamino, each of which is independently substituted or unsubstituted; or halogen, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;
      • R1 is alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • R2 is alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H, —C(═O)R5, —C(═S)R6, —SO2R7, —P(═O)R8R9, or —(CH2)m—R10;
      • R3 is acyl, —O-acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or substituted; or H, —CO2Y, or —C(═O)NHY;
      • Y is alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • R4 is alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • each R5 is acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —NR15R16, —(CH2)tNR15R16, —NHNR15R16, —NHOH, OR, —C(═O)NHNR15R16, —CO2R15, —C(═O)NR15R16, or —CH2X;
      • each R6 is acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NHNR15R16, —NHOH, —NR15R16, or —CH2X;
      • each R7 is alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NR15R16, —NHNR15R16, —NHOH, or —CH2X;
      • each R8 and R9 are each independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or OH;
      • each R10 is —NR15R16, OH, —SO2R11, —NHSO2R11, C(═O)(R12), NHC═O(R12), —OC═O(R12), or —P(═O)R13R14;
      • each R11, R12, R13, and R14 is independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or H, OH, NH2, —NHNH2, or —NHOH;
      • each X is independently halogen, —CN, —CO2R15, —C(═O)NR15R16, —NR15R16, —OR15, —SO2R7, or —P(═O)R8R9;
      • each R15 and R16 is independently acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted, or H; or R15 and R16 together with the N to which R15 and R16 are bonded form a heterocycle that is substituted or unsubstituted;
      • n is 0, 1, or 2;
      • q is 0, 1, 2, 3, or 4;
      • t is 1, 2, 3, 4, 5, or 6; and
      • m is 1, 2, 3, or 4.

    • Embodiment 192. The method of any one of embodiments 184-192, wherein the template ligand is







embedded image




    •  or an ionized form thereof.

    • Embodiment 193. The method of any one of embodiments 184-192, wherein the template ligand is







embedded image




    •  or an ionized form thereof.

    • Embodiment 194. The method of embodiment 184, further comprising selecting the target ligand from a plurality of ligand candidates, each of the ligand candidates being different from the template ligand, and wherein selecting the target ligand comprises comparing the pharmacophore model of the template ligand to a pharmacophore model of each respective one of the plurality of ligand candidates.

    • Embodiment 195. The method of embodiment 184, further comprising receiving a plurality of template ligand-biomolecule structures, each template ligand-biomolecule structure having a different template ligand docked in the binding site of the biomolecule, and generating the pharmacophore model of the template ligand by combining information from each of the template ligands from the plurality of template ligand-biomolecule structures.

    • Embodiment 196. The method of embodiment 184, wherein the target ligand has more than one structural conformation in an unbound state, and the docked position of the target ligand in the binding site of the biomolecule is predicted by enumerating a set of potential target ligand conformations and overlapping a respective pharmacophore model of the target ligand for each of the potential target ligand conformations with the pharmacophore model of the template ligand while the template ligand is in the binding site of the biomolecule.

    • Embodiment 197. The method of embodiment 196, wherein predicting the docked position of the target ligand in the binding site of the biomolecule comprises ignoring at least one clash between the target ligand conformation's atomic coordinates and the biomolecule's atomic coordinates.

    • Embodiment 198. The method of embodiment 197, further comprising, for each target ligand conformation, modifying atomic coordinates of the biomolecule to reduce clashes between the docked target ligand conformation's atomic coordinates and the biomolecule's atomic coordinates, thereby creating an altered ligand-biomolecule structure comprising the docked target ligand and an altered biomolecule.

    • Embodiment 199. The method of embodiment 198, further comprising, predicting a re-docked position of each target ligand conformation by predicting each target ligand conformation's position in the binding site of the altered biomolecule; and
      • for each target ligand conformation, modifying atomic coordinates of the altered biomolecule to reduce clashes between the atomic coordinates of the target ligand conformation's re-docked position and the atomic coordinates of the altered biomolecule,
      • thereby creating a re-altered ligand-biomolecule structure comprising a re-docked target ligand and a re-altered biomolecule.

    • Embodiment 200. The method of embodiment 199, further comprising ranking each altered and re-altered ligand-biomolecule structure using a scoring function.

    • Embodiment 201. The method of embodiment 200, further comprising identifying a subset of high-ranking target ligands corresponding to target ligands having a threshold value for an empirical activity.

    • Embodiment 202. A method of identifying a plurality of potential lead compounds, the method comprising the steps of:
      • (a) analyzing, using a computer system, an initial lead compound known to bind to a biomolecular target, the analyzing comprising partitioning, by providing a database of known reactions, the initial lead compound into atoms defining partitioned lead compound comprising a lead compound core and atoms defining a lead compound non-core, wherein the initial lead compound is partitioned using a computational retrosynthetic analysis of the initial lead compound;
      • (b) identifying, using the computer system, a plurality of alternative cores to replace the lead compound core in the initial lead compound, thereby generating a plurality of potential lead compounds each having a respective one of the plurality of alternative cores;
      • (c) calculating, using the computer system, a difference in binding free energy between the partitioned lead compound and each potential lead compound;
      • (d) predicting, using the computer system, whether each potential lead compound will bind to the biomolecular target and identifying a predicted active set of potential lead compounds based on the prediction;
      • (e) obtaining a synthesized set of at least some of the potential leads of the predicted active set to establish a first of potential lead compounds; and
      • (f) determining, empirically, an activity of each of the first set of synthesized potential lead compounds,
      • wherein the biomolecular target is a RY1&2 domain of RyR2, and the structure of the biomolecular target used in the predicting of (d) is obtained by a process comprising subjecting a complex of the biomolecular target and the initial lead compound to single-particle cryogenic electron microscopy analysis.

    • Embodiment 203. The method of embodiment 202, wherein the structure of the of the biomolecular target obtained by single-particle cryogenic electron microscopy analysis has a resolution from about 2 Å to about 3.5 Å, from about 2 Å to about 3.4 Å, from about 2 Å to about 3.3 Å, from about 2 Å to about 3.2 Å, from about 2 Å to about 3.1 Å, from about 2 Å to about 3 Å, from about 2 Å to about 2.9 Å, from about 2 Å to about 2.8 Å, from about 2 Å to about 2.7 Å, from about 2 Å to about 2.6 Å, from about 2 Å to about 2.5 Å, from about 2.1 Å to about 2.5 Å, from about 2.2 Å to about 2.5 Å, from about 2.3 Å to about 2.5 Å, or from about 2.4 Å to about 2.5 Å.

    • Embodiment 204. The method of embodiment 202, wherein the initial lead compound is a compound of Formula (I):







embedded image




    • wherein.
      • each R is independently acyl, —O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, arylthio, arylamino, heteroarylthio, or heteroarylamino, each of which is independently substituted or unsubstituted; or halogen, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;
      • R1 is alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • R2 is alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H, —C(═O)R5, —C(═S)R6, —SO2R7, —P(═O)R8R9, —(CH2)m—R10;
      • R3 is acyl, —O-acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or substituted; or H, —CO2Y, or —C(═O)NHY;
      • Y is alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • R4 is alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • each R5 is acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —NR15R16, —(CH2)tNR15R16, —NHNR15R16, —NHOH, OR, —C(═O)NHNR15R16, —CO2R15, —C(═O)NR15R16, or —CH2X;
      • each R6 is acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NHNR15R16, —NHOH, —NR15R16, or —CH2X;
      • each R7 is alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NR15R16, —NHNR15R16, —NHOH, or —CH2X;
      • each R8 and R9 are each independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or OH;
      • each R10 is —NR15R16, OH, —SO2R11, —NHSO2R11, C(═O)(R12), NHC═O(R12), —OC═O(R12), or —P(═O)R13R14;
      • each R11, R12, R13, and R14 is independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or H, OH, NH2, —NHNH2, or —NHOH;
      • each X is independently halogen, —CN, —CO2R15, —C(═O)NR15R16, —NR15R16, —OR15, —SO2R7, or —P(═O)R8R9;
      • each R15 and R16 is independently acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted, or H; or R15 and R16 together with the N to which R15 and R16 are bonded form a heterocycle that is substituted or unsubstituted;
      • n is 0, 1, or 2;
      • q is 0, 1, 2, 3, or 4;
      • t is 1, 2, 3, 4, 5, or 6; and
      • m is 1, 2, 3, or 4.

    • Embodiment 205. The method of embodiment 202, wherein the initial lead compound is







embedded image




    •  or an ionized form thereof.

    • Embodiment 206. The method of embodiment 202, wherein the initial lead compound is







embedded image




    •  or an ionized form thereof.

    • Embodiment 207. The method of embodiment 202, wherein the RY1&2 domain comprises a structure according to TABLE 3.

    • Embodiment 208. The method of embodiment 202, wherein the RY1&2 domain contains an ATP molecule.

    • Embodiment 209. The method of embodiment 208, wherein the ATP molecule has a three-dimensional conformation according to TABLE 5.

    • Embodiment 210. The method of embodiment 202, further comprising obtaining a synthesized set of at least some of the potential lead compounds predicted to not bind with the biomolecular target to establish a second set of potential lead compounds and empirically determining an activity of each of the second set of synthesized potential lead compounds.

    • Embodiment 211. The method of embodiment 202, further comprising comparing the empirically determined activity of each of the first set of synthesized potential lead compounds with a threshold activity level.

    • Embodiment 212. The method of embodiment 203, further comprising comparing the empirically determined activity of each of the second set of synthesized potential lead compounds with a pre-determined activity level.

    • Embodiment 213. The method of embodiment 202, wherein the plurality of alternative cores are chosen from a database of synthetically feasible cores.

    • Embodiment 214. The method of embodiment 202, wherein the difference in binding free energy is calculated using a free energy perturbation technique.

    • Embodiment 215. The method of embodiment 210, wherein the generation of at least one potential lead compound comprises creating an additional covalent bond or annihilating an existing covalent bond, or both creating an additional first covalent bond and annihilating an existing second covalent bond different from the first covalent bond.

    • Embodiment 216. The method of embodiment 211, wherein the free energy perturbation technique uses a soft bond potential to calculate a bonded stretch interaction energy of existing covalent bonds for annihilation and additional covalent bonds for creation.

    • Embodiment 217. A method for pharmaceutical drug discovery, comprising:
      • identifying an initial lead compound for binding to a biomolecular target; using the method of embodiment 202 to identify a predicted active set of potential lead compounds for binding to the biomolecular target based on the initial lead compound; selecting one or more of the predicted active set of potential lead compounds for synthesis; and
      • assaying the one or more synthesized selected compounds to assess each synthesized selected compounds suitability for in vivo use as a pharmaceutical compound,
      • wherein the biomolecular target is a RY1&2 domain of RyR2, and the structure of the biomolecular target used in the predicting of (d) is obtained by a process comprising subjecting a complex of the biomolecular target and the initial lead compound to single-particle cryogenic electron microscopy analysis.

    • Embodiment 218. The method of embodiment 217, wherein the initial lead compound is compound of Formula (I):







embedded image




    • wherein:
      • each R is independently acyl, —O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, arylthio, arylamino, heteroarylthio, or heteroarylamino, each of which is independently substituted or unsubstituted; or halogen, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;
      • R1 is alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • R2 is alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H, —C(═O)R5, —C(═S)R6, —SO2R7, —P(═O)R8R9, —(CH2)m—R10;
      • R3 is acyl, —O-acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or substituted; or H, —CO2Y, or —C(═O)NHY;
      • Y is alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • R4 is alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • each R5 is acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —NR15R16, —(CH2)tNR15R16, —NHNR15R16, —NHOH, OR, —C(═O)NHNR15R16, —CO2R15, —C(═O)NR15R16, or —CH2X;
      • each R6 is acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NHNR15R16, —NHOH, —NR15R16, or —CH2X;
      • each R7 is alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NR15R16, —NHNR15R16, —NHOH, or —CH2X;
      • each R8 and R9 are each independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or OH;
      • each R10 is —NR15R16, OH, —SO2R11, —NHSO2R11, C(═O)(R12), NHC═O(R12), —OC═O(R12), or —P(═O)R13R14;
      • each R11, R12, R13, and R14 is independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or H, OH, NH2, —NHNH2, or —NHOH;
      • each X is independently halogen, —CN, —CO2R15, —C(═O)NR15R16, —NR15R16, —OR15, —SO2R7, or —P(═O)R8R9;
      • each R15 and R16 is independently acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted, or H; or R15 and R16 together with the N to which R15 and R16 are bonded form a heterocycle that is substituted or unsubstituted;
      • n is 0, 1, or 2;
      • q is 0, 1, 2, 3, or 4;
      • t is 1, 2, 3, 4, 5, or 6; and
      • m is 1, 2, 3, or 4.

    • Embodiment 219. The method of embodiment 217, wherein the initial lead compound is







embedded image




    •  or an ionized form thereof.

    • Embodiment 220. The method of embodiment 217, wherein the initial lead compound is







embedded image




    •  or an ionized form thereof.

    • Embodiment 221. The method of embodiment 217, wherein the RY1&2 domain comprises a structure according to TABLE 3.

    • Embodiment 222. The method of embodiment 217, wherein the RY1&2 domain contains an ATP molecule.

    • Embodiment 223. The method of embodiment 221, wherein the ATP molecule has a three-dimensional conformation according to TABLE 5.

    • Embodiment 224. A computer-implemented method of quantifying binding affinity between a ligand and a receptor molecule, the method comprising:
      • receiving by one or more computers, data representing a ligand molecule,
      • receiving by one or more computers, data representing a receptor molecule domain,
      • using the data representing the ligand molecule and the data representing the receptor molecule domain in computer analysis to identify ring structure within the ligand, the ring structure being an entire ring or a fused ring;
      • using the data representative of the identified ligand ring structure to designate a first ring face and a second ring face opposite to the first ring face, and classifying the ring structure by:
      • a) determining proximity of receptor atoms to atoms on the first face of the ligand ring; and
      • b) determining proximity of receptor atoms to atoms on the second face of the ligand ring;
      • c) determining solvation of the first face of the ligand ring and solvation of the second face of the ligand ring;
      • classifying the identified ligand ring structure as buried, solvent exposed or having a single face exposed to solvent based on receptor atom proximity to and solvation of the first ring face and receptor atom proximity to and solvation of the second ring face;
      • quantifying the binding affinity between the ligand and the receptor molecule domain based at least in part on the classification of the ring structure; and
      • displaying, via computer, information related to the classification of the ring structure, wherein the receptor molecule domain is a RY1&2 domain of RyR2, wherein the data representing a ligand molecule and the data representing a receptor molecule domain are obtained by a process comprising subjecting a complex comprising the ligand molecule and the receptor molecule domain to single-particle cryogenic electron microscopy analysis.

    • Embodiment 225. The method of embodiment 224, wherein the initial lead compound is compound of Formula (I):







embedded image




    • wherein:
      • each R is independently acyl, —O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, arylthio, arylamino, heteroarylthio, or heteroarylamino, each of which is independently substituted or unsubstituted; or halogen, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;
      • R1 is alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • R2 is alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H, —C(═O)R5, —C(═S)R6, —SO2R7, —P(═O)R8R9, or —(CH2)m—R10;
      • R3 is acyl, —O-acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or substituted; or H, —CO2Y, or —C(═O)NHY;
      • Y is alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • R4 is alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • each R5 is acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —NR15R16, —(CH2)tNR15R16, —NHNR15R16, —NHOH, —OR15, —C(═O)NHNR15R16, —CO2R15, —C(═O)NR15R16, or —CH2X;
      • each R6 is acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NHNR15R16, —NHOH, —NR15R16, or —CH2X;
      • each R7 is alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NR15R16, —NHNR15R16, —NHOH, or —CH2X;
      • each R8 and R9 are each independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or OH;
      • each R10 is —NR15R16, OH, —SO2R11, —NHSO2R11, C(═O)(R12), NHC═O(R12), —OC═O(R12), or —P(═O)R13R14;
      • each R11, R12, R13, and R14 is independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or H, OH, —NH2, —NHNH2, or —NHOH;
      • each X is independently halogen, —CN, —CO2R15, —C(═O)NR15R16, —NR15R16, —OR15, —SO2R7, or —P(═O)R8R9;
      • each R15 and R16 is independently acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted, or H; or R15 and R16 together with the N to which R15 and R16 are bonded form a heterocycle that is substituted or unsubstituted;
      • n is 0, 1, or 2;
      • q is 0, 1, 2, 3, or 4;
      • t is 1, 2, 3, 4, 5, or 6; and
      • m is 1, 2, 3, or 4.

    • Embodiment 226. The method of embodiment 224, wherein the ligand molecule is







embedded image




    •  or an ionized form thereof.

    • Embodiment 227. The method of embodiment 224, wherein the ligand molecule is







embedded image




    •  or an ionized form thereof.

    • Embodiment 228. The method of embodiment 224, wherein the complex further comprises a RyR2 protein, wherein the RY1&2 domain is a domain of the RyR2 protein.

    • Embodiment 229. The method of embodiment 224, wherein the data representing the receptor molecule domain represents a three-dimensional structure of the receptor molecule according to TABLE 3.

    • Embodiment 230. The method of embodiment 224, wherein the data representing a ligand molecule represents a three-dimensional structure of the ligand molecule according to TABLE 4.

    • Embodiment 231. The method of embodiment 224, wherein the receptor molecule domain contains an ATP molecule.

    • Embodiment 232. The method of embodiment 231, wherein the data representing the receptor molecule domain further comprises data representing a three-dimensional structure of the ATP molecule according to TABLE 5.

    • Embodiment 233. The method of embodiment 224, wherein quantifying the binding affinity includes a step that scores hydrophobic interactions between one or more ligand atoms and one or more receptor atoms by awarding a bonus for the presence of hydrophobic enclosure of one or more atoms of said ligand by the receptor molecule domain, said bonus being indicative of enhanced binding affinity between said ligand and said receptor molecule domain.

    • Embodiment 234. The method of embodiment 224, further comprising calculating an initial binding affinity and then adjusting the initial binding affinity based on the classification of the ring structure as buried, solvent exposed or solvent exposed on one face.

    • Embodiment 235. The method of embodiment 224, wherein the classification of a ring structure as buried, solvent exposed, or solvent exposed on one surface, includes using a parameter substantially correlated with the number of close contacts on both sides of the ring structure or part thereof with the receptor molecule domain.

    • Embodiment 236. The method of embodiment 224, wherein the number of close contacts at different distances between receptor atoms and the two ring faces are determined, an initial classification of the ring is made based on the numbers of these contacts, and this initial classification is then followed by calculation of a scoring function, said scoring function comprising identifying a first ring shell and a second ring shell, and calculating the number of water molecules in the first shell and in the second shell, or calculating the number of water molecules in the first and second shell combined.

    • Embodiment 237. The method of embodiment 236, wherein the scoring function allowing classification of the ring structure as buried, solvent exposed, or solvent exposed on one surface, includes using a parameter substantially correlated with the lipophilic-lipophilic pair score between the ring structure or part thereof and the receptor molecule domain.

    • Embodiment 238. The method of embodiment 236, wherein the scoring function used to classify a ring structure as buried, solvent exposed, or solvent exposed on one surface, includes calculating the degree of enclosure of each atom of the ring structure by atoms of the receptor.

    • Embodiment 239. The method of embodiment 236, wherein the scoring function used to classify a ring structure as buried, solvent exposed, or solvent exposed on one surface, includes using a parameter that is substantially correlated with the degree of enclosure of each atom of the ring structure by atoms of the receptor.

    • Embodiment 240. The method of embodiment 224 or embodiment 236, wherein the scoring function allowing classification of the ring structure as buried, solvent exposed, or solvent exposed on one surface, includes the use of a parameter corresponding to a hydrophobic interaction of the ring structure or part thereof with the receptor molecule domain.

    • Embodiment 241. The method of embodiment 240, wherein the information displayed by computer includes a depiction of at least one of
      • the degree to which the ring structure is enclosed by atoms of the receptor molecule domain;
      • water molecules surrounding the ring structure in a first shell or a second shell or both the first and the second shell of the ligand;
      • a value of a lipophilic-lipophilic pair score of the ring structure; and
      • a number of close contacts of a face of the ring structure with the receptor molecule domain.

    • Embodiment 242. The method of embodiment 224, wherein solvent exposed ring structures in the ligand, if any, are substantially ignored in quantifying the component of the binding affinity between the ligand and the receptor molecule domains, other than to recognize hydrogen bonds and other parameters that are independent of the classification of ring structure.

    • Embodiment 243. The method of embodiment 224, wherein hydrophobic contribution to binding affinity from ring structures classified as solvent exposed, if any, is substantially ignored in quantifying the component of the binding affinity.

    • Embodiment 244. The method of embodiment 224, wherein a ring structure is classified as buried, and the method further comprises:
      • identifying a quantity representative of a strain energy induced in the ligand-receptor complex by the buried ring structure, in which the quantification of the component of binding affinity is further based in part on strain energy.

    • Embodiment 245. The method of embodiment 244, further comprising
      • identifying a quantity representative of a strain energy induced in the ligand-receptor complex by the aggregate of the ring structures identified as buried;
      • identifying a quantity representative of a total neutral-neutral hydrogen bond energy; and
      • quantifying the component of binding affinity between the ligand and the receptor molecule domain based at least in part on the quantity representative of the strain energy induced in the receptor by the aggregate of the buried ring structures, and on the quantity representative of the total neutral-neutral hydrogen bond energy.

    • Embodiment 246. The method of embodiment 245, wherein
      • quantifying the component of binding affinity further comprises identifying a hydrogen bond capping energy associated with the entire ligand, and
      • the component of binding affinity is quantified based on a greater of the hydrogen bond capping energy and the quantity representative of the strain energy induced in the receptor by the aggregate of the identified structures.

    • Embodiment 247. The method of embodiment 245, further comprising:
      • identifying a binding motif of the receptor molecule domain with respect to the ligand;
      • identifying a reorganization energy of the receptor molecule domain based on the binding motif; and
      • identifying a first ring structure as contributing to the reorganization energy,
      • the quantity representative of strain energy being identified independently of the classification of the first ring structure.

    • Embodiment 248. The method of embodiment 244, wherein the component of binding affinity attributable to strain is quantified using at least one of: molecular dynamics, molecule mechanics, conformational searching and minimization.

    • Embodiment 249. The method of embodiment 224, wherein the information displayed by computer includes a depiction of solvent exposure, if any, of the ring structure.

    • Embodiment 250. The method of embodiment 224, wherein the information displayed by computer includes a depiction of burial, if any, of the ring structure.

    • Embodiment 251. The method of embodiment 224, wherein the information displayed by computer includes a depiction of at least one of
      • the degree to which the ring structure is enclosed by atoms of the receptor molecule domain;
      • water molecules surrounding the ring structure in a first shell or a second shell or both the first and the second shell of the ligand;
      • a value of a lipophilic-lipophilic pair score of the ring structure; and
      • a number of close contacts of a face of the ring structure with the receptor molecule domain.

    • Embodiment 252. The method of embodiment 224, further comprising,
      • performing a test on a physical sample that includes the ligand and the receptor molecule domain, test components being selected based at least in part on the binding affinity between the ligand or part thereof and the receptor molecule, or on the component of such binding affinity.

    • Embodiment 253. A method comprising:
      • (a) determining an open probability (Po) of a first RyR2 protein, wherein the first RyR2 protein is treated with a test compound, and
      • (b) determining an open probability (Po) of a second RyR2 protein, wherein the second RyR2 protein is not treated with the test compound.

    • Embodiment 254. The method of embodiment 253, wherein each of the determining the open probability (Po) of the first RyR2 protein and the determining the open probability (Po) of the second RyR2 protein comprises recording a single channel Ca2+ current.

    • Embodiment 255. The method of embodiments 253-254, further comprising determining a difference between the Po of the first RyR2 protein and Po of the second RyR2 protein.

    • Embodiment 256. The method of embodiment 255, further comprising identifying the test compound as a target for further analysis based on the difference between the Po of the first RyR2 protein and Po of the second RyR2 protein.

    • Embodiment 257. The method of embodiment 256, wherein the Po of the first RyR2 protein is lower than the Po of the second RyR2 protein.

    • Embodiment 258. The method of embodiment 255, further comprising performing an analogous assay wherein another compound is used in place of the test compound, wherein the analogous assay provides a difference between:
      • (a) an open probability (Po) of a third RyR2 protein, wherein the third RyR2 protein is treated with the other compound; and
      • (b) an open probability (Po) of a fourth RyR2 protein, wherein the fourth RyR2 protein is not treated with the other compound,
      • wherein the test compound is prioritized over the other compound for the further analysis based on a comparison of:
      • (i) the difference between the Po of the first RyR2 protein and Po of the second RyR2 protein; with
      • (ii) a difference between the Po of the third RyR2 protein and Po of the fourth RyR2 protein.

    • Embodiment 259. The method of any one of embodiments 255-258, wherein the difference of the Po in (b)(ii) is greater than the difference of the Po in (b)(i).

    • Embodiment 260. The methods of any one of embodiments 255-259, wherein the RyR2 is a mutant RyR2.

    • Embodiment 261. The method of embodiment 260, wherein the mutant RyR2 contains at least one mutation that is associated with Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT).

    • Embodiment 262. The composition of embodiment 261, wherein the mutation is RyR2-R2474S.

    • Embodiment 263. The composition of embodiment 261, wherein the mutation is RyR2-R420Q.

    • Embodiment 264. The composition of embodiment 261, wherein the mutation is RyR2-R420W.

    • Embodiment 265. The composition of any one of embodiments 260-264, wherein the mutation destabilizes an interaction between NTD and BSol domains of the RyR2 protein.

    • Embodiment 266. The composition of any one of embodiments 260-265, wherein the mutation destabilizes a cytosolic shell of the RyR2 protein, wherein the cytosolic shell comprises NTD, SPRY, JSol and BSol domains of the RyR2 proteins.

    • Embodiment 267. The method of any one of embodiments 255-266, wherein the RyR2 protein is a post-translationally modified RyR2 protein.

    • Embodiment 268. The method of embodiment 268, wherein the post-translationally modified RyR2 protein is a phosphorylated, oxidized or nitrosylated RyR2.

    • Embodiment 269. The composition of embodiment 267 or embodiment 268, wherein the post-translationally modified RyR2 protein is a phosphorylated RyR2.

    • Embodiment 270. The composition of any one of embodiments 267-269, wherein the post-translationally modified RyR2 protein is associated with heart failure.

    • Embodiment 271. The composition of any one of embodiments 267-270, wherein the post-translational modification destabilizes an interaction between NTD and BSol domains of the RyR2 protein.

    • Embodiment 272. The composition of any one of embodiments 267-271, wherein the post-translational modification destabilizes a cytosolic shell of the RyR2 protein, wherein the cytosolic shell comprises NTD, SPRY, JSol and BSol domains of the RyR2 proteins.

    • Embodiment 273. The method of any one of embodiments 255-272, wherein the RyR2 is a mutated and post-translationally modified RyR2.

    • Embodiment 274. A method comprising:
      • (a) contacting a first RyR2 protein with a test compound;
      • (b) providing a second RyR2 protein;
      • (c) subsequent to the contacting the first RyR2 protein with the test compound, measuring an open probability (Po) of the first RyR2 protein; and
      • (d) measuring an open probability (Po) of the second RyR2 protein.

    • Embodiment 275. The method of embodiment 274, wherein each of the determining the open probability (Po) of the first RyR2 protein and the determining the open probability (Po) of second RyR2 protein comprises recording a single channel Ca2+ current.

    • Embodiment 276. The method of any one of embodiments 274-275, further comprising determining a difference between the Po of the first RyR2 protein and the Po of the second RyR2 protein.

    • Embodiment 277. The method of embodiment 276, further comprising identifying the test compound as a target for further analysis based on the difference between the Po of the first RyR2 protein and the Po of the second RyR2 protein.

    • Embodiment 278. The method of embodiment 277, wherein the Po of the first RyR2 protein is lower than the Po of the second RyR2 protein.

    • Embodiment 279. The method of embodiment 276, further comprising performing an analogous assay wherein another compound is used in place of the test compound, wherein the analogous assay provides a difference between:
      • (a) an open probability (Po) of a third RyR2 protein, wherein the third RyR2 protein is treated with the other compound; and
      • (b) an open probability (Po) of a fourth RyR2 protein, wherein the fourth RyR2 protein is not treated with the other compound,
      • wherein the test compound is prioritized over the other compound for the further analysis based on a comparison of:
      • (i) the difference between the Po of the first RyR2 protein and Po of the second RyR2 protein; with
      • (ii) a difference between the Po of the third RyR2 protein and Po of the fourth RyR2 protein.

    • Embodiment 280. The method of any one of embodiments 276-279, wherein the difference of the Po in (b)(ii) is greater than the difference of the Po in (b)(i).

    • Embodiment 281. The methods of any one of embodiments 274-280, wherein the RyR2 is a mutant RyR2.

    • Embodiment 282. The method of embodiment 281, wherein the mutant RyR2 contains at least one mutation that is associated with Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT).

    • Embodiment 283. The composition of embodiment 282, wherein the mutation is RyR2-R2474S.

    • Embodiment 284. The composition of embodiment 282, wherein the mutation is RyR2-R420Q.

    • Embodiment 285. The composition of embodiment 282, wherein the mutation is RyR2-R420W.

    • Embodiment 286. The composition of any one of embodiments 282-285, wherein the mutation destabilizes an interaction between NTD and BSol domains of the RyR2 protein.

    • Embodiment 287. The composition of any one of embodiments 282-286, wherein the mutation destabilizes a cytosolic shell of the RyR2 protein, wherein the cytosolic shell comprises NTD, SPRY, JSol and BSol domains of the RyR2 proteins.

    • Embodiment 288. The method of any one of embodiments 274-287, wherein the RyR2 protein is a post-translationally modified RyR2 protein.

    • Embodiment 289. The method of embodiment 288, wherein the post-translationally modified RyR2 protein is a phosphorylated, oxidized or nitrosylated RyR2.

    • Embodiment 290. The method of any one of embodiments 274-289, wherein the RyR2 is a mutated and post-translationally modified RyR2.

    • Embodiment 291. The method of any one of embodiments 253-290, further comprising: subsequent to the contacting the first RyR2 protein with the reagent and the test compound, fusing a first microsome containing the first RyR2 protein to a first planar lipid bilayer, and subsequent to the contacting the second RyR2 protein with the reagent, fusing a second microsome containing the second RyR2 protein to a second planar lipid bilayer.

    • Embodiment 292. A method of identifying a compound having RyR2 modulatory activity, the method comprising:
      • (a) determining an open probability (Po) of a RyR2 protein;
      • (b) contacting the RyR2 protein with a test compound;
      • (c) determining an open probability (Po) of the RyR2 protein in the presence of the test compound; and
      • (d) determining a difference between the Po of the RyR2 protein in the presence and absence of the test compound;
      • wherein a reduction in the Po of the RyR2 protein in the presence of the test compound relative to the Po of the RyR2 protein in the absence of the test compound is indicative of the compound having RyR2 modulatory activity.

    • Embodiment 293. The method of embodiment 292, wherein the RyR2 protein is a mutant RyR2 protein.

    • Embodiment 294. The method of embodiment 293, wherein the mutant RyR2 contains at least one mutation that is associated with Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT).

    • Embodiment 295. The composition of embodiment 294, wherein the mutation is RyR2-R2474S.

    • Embodiment 296. The composition of embodiment 294, wherein the mutation is RyR2-R420Q.

    • Embodiment 297. The composition of embodiment 294, wherein the mutation is RyR2-R420W.

    • Embodiment 298. The composition of any one of embodiments 294-297, wherein the mutation destabilizes an interaction between NTD and BSol domains of the RyR2 protein.

    • Embodiment 299. The composition of any one of embodiments 294-297, wherein the mutation destabilizes a cytosolic shell of the RyR2 protein, wherein the cytosolic shell comprises NTD, SPRY, JSol and BSol domains of the RyR2 proteins.

    • Embodiment 300. The method of any one of embodiments 292-299, wherein the RyR2 protein is a post-translationally modified RyR2 protein.

    • Embodiment 301. The method of embodiment 300, wherein the post-translationally modified RyR2 protein is a phosphorylated, oxidized or nitrosylated RyR2.

    • Embodiment 302. The method of any one of embodiments 292-301, wherein the RyR2 protein is a mutated and post-translationally modified RyR2 protein.

    • Embodiment 303. The method of any one of embodiments 292-302, wherein the test compound preferentially binds to a mutant RyR2 relative to wild-type RyR2.

    • Embodiment 304. The method of any one of embodiments 292-303, wherein the test compound preferentially binds to post-translationally modified RyR2 relative to wild-type RyR2.

    • Embodiment 305. The method of any one of embodiments 292-305, wherein the test compound preferentially binds to a mutated and post-translationally modified RyR2 relative to a wild-type RyR2.

    • Embodiment 306. The method of any one of embodiments 292-305, wherein determining the open probability (Po) of the RyR2 protein comprises recording a single channel Ca2+ current.

    • Embodiment 307. A method for identifying a compound having RyR2 modulatory activity, comprising:
      • (a) contacting a RyR2 protein with a ligand having known RyR2 modulatory activity to create a mixture, wherein the RyR2 protein is a leaky RyR2, the leaky RyR2 comprising mutant RyR2 protein, post-translationally modified RyR2, or a combination thereof;
      • (b) contacting the mixture of step (a) with a test compound; and
      • (c) determining an ability of the test compound to displace the ligand from the RyR2 protein.

    • Embodiment 308. The method of embodiment 307, wherein the ligand is radiolabeled.

    • Embodiment 309. The method of embodiment 307 or embodiment 308, wherein determining the ability of the test compound to displace the ligand from the RyR2 protein comprises determining a radioactive signal in the RyR2 protein.

    • Embodiment 310. The method of any one of embodiments 307-309, wherein the RyR2 protein is a mutant RyR2 protein.

    • Embodiment 311. The method of embodiment 309 or 310, wherein the mutant RyR2 contains at least one mutation that is associated with Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT).

    • Embodiment 312. The composition of embodiment 311, wherein the mutation is RyR2-R2474S.

    • Embodiment 313. The composition of embodiment 311, wherein the mutation is RyR2-R420Q.

    • Embodiment 314. The composition of embodiment 311, wherein the mutation is RyR2-R420W.

    • Embodiment 315. The composition of any one of embodiments 311-314, wherein the mutation destabilizes an interaction between NTD and BSol domains of the RyR2 protein.

    • Embodiment 316. The composition of any one of embodiments 311-315, wherein the mutation destabilizes a cytosolic shell of the RyR2 protein, wherein the cytosolic shell comprises NTD, SPRY, JSol and BSol domains of the RyR2 proteins.

    • Embodiment 317. The method of any one of embodiments 307-316, wherein the RyR2 protein is a post-translationally modified RyR2 protein.

    • Embodiment 318. The method of any one of embodiments 307-316, wherein the RyR2 protein is a mutated and post-translationally modified RyR2 protein.

    • Embodiment 319. The method of any one of embodiments 307-318, wherein the test compound preferentially binds to a mutant RyR2 relative to wild-type RyR2.

    • Embodiment 320. The method of any one of embodiments 307-318, wherein the test compound preferentially binds to post-translationally modified RyR2 relative to wild-type RyR2.

    • Embodiment 321. The method of any one of embodiments 307-318, wherein the test compound preferentially binds to a mutant and post-translationally modified RyR2 relative to a wild-type RyR2.

    • Embodiment 322. A method for identifying a compound that preferentially binds to a mutated, post-translationally modified RyR2 or a combination thereof, comprising:
      • (a) determining binding affinity of a test compound to a first RyR2 protein, wherein the first RyR2 protein is a wild-type RyR2 protein;
      • (b) determining binding affinity of a test compound to a second RyR2 protein, wherein second first RyR2 protein is a mutant RyR2 protein, a post-translationally modified RyR2, or a combination thereof, and
      • (c) selecting a compound having a higher binding affinity to the second RyR2 protein relative to the first RyR2 protein.

    • Embodiment 323. The method of embodiment 322, wherein the RyR2 protein is a mutant RyR2 protein.

    • Embodiment 324. The method of embodiment 322 or 323, wherein the mutant RyR2 contains at least one mutation that is associated with Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT).

    • Embodiment 325. The composition of embodiment 324, wherein the mutation is RyR2-R2474S.

    • Embodiment 326. The composition of embodiment 324, wherein the mutation is RyR2-R420Q.

    • Embodiment 327. The composition of embodiment 324, wherein the mutation is RyR2-R420W.

    • Embodiment 328. The composition of any one of embodiments 324-327, wherein the mutation destabilizes an interaction between NTD and BSol domains of the RyR2 protein.

    • Embodiment 329. The composition of any one of embodiments 324-328, wherein the mutation destabilizes a cytosolic shell of the RyR2 protein, wherein the cytosolic shell comprises NTD, SPRY, JSol and BSol domains of the RyR2 proteins.

    • Embodiment 330. The method of any one of embodiments 322-329, wherein the RyR2 protein is a post-translationally modified RyR2 protein.

    • Embodiment 331. The method of any one of embodiments 322-329, wherein the RyR2 protein is a mutated and post-translationally modified RyR2 protein.

    • Embodiment 332. The method of any one of embodiments 253-331, wherein the test compound preferentially binds to a mutant RyR2 relative to wild-type RyR2.

    • Embodiment 333. The method of any one of embodiments 253-331, wherein the test compound preferentially binds to post-translationally modified RyR2 relative to wild-type RyR2.

    • Embodiment 334. The method of any one of embodiments 253-331, wherein the test compound preferentially binds to a mutant and post-translationally modified RyR2 relative to a wild-type RyR2.

    • Embodiment 335. The method of any one of embodiments 253-334, wherein the test compound contains a benzothiazepane moiety.

    • Embodiment 336. The method of any one of embodiments 253-335, wherein the test compound is a compound of Formula (I):







embedded image




    • wherein:
      • each R is independently acyl, —O-acyl, alkyl, alkoxyl, alkylamino, alkylarylamino, alkylthio, cycloalkyl, alkylaryl, aryl, heteroaryl, heterocyclyl, heterocyclylalkyl, alkenyl, alkynyl, arylthio, arylamino, heteroarylthio, or heteroarylamino, each of which is independently substituted or unsubstituted; or halogen, —OH, —NH2, —NO2, —CN, —CF3, —OCF3, —N3, —SO3H, —S(═O)2alkyl, —S(═O)alkyl, or —OS(═O)2CF3;
      • R1 is alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • R2 is alkyl, aryl, alkylaryl, heteroaryl, cycloalkyl, cycloalkylalkyl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H, —C(═O)R5, —C(═S)R6, —SO2R7, —P(═O)R8R9, or —(CH2)m—R10;
      • R3 is acyl, —O-acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or substituted; or H, —CO2Y, or —C(═O)NHY;
      • Y is alkyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • R4 is alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, heteroaryl, or heterocyclyl, each of which is independently substituted or unsubstituted; or H;
      • each R5 is acyl, alkyl, alkenyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —NR15R16, —(CH2)tNR15R16, —NHNR15R16, —NHOH, OR, —C(═O)NHNR15R16, —CO2R15, —C(═O)NR15R16, or —CH2X;
      • each R6 is acyl, alkenyl, alkyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NHNR15R16, —NHOH, —NR15R16, or —CH2X;
      • each R7 is alkyl, alkenyl, alkynyl, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or —OR15, —NR15R16, —NHNR15R16, —NHOH, or —CH2X;
      • each R8 and R9 are each independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or OH;
      • each R10 is —NR15R16, OH, —SO2R11, —NHSO2R11, C(═O)(R12), NHC═O(R12), —OC═O(R12), or —P(═O)R13R14;
      • each R11, R12, R13, and R14 is independently acyl, alkenyl, alkoxyl, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted; or H, OH, NH2, —NHNH2, or —NHOH;
      • each X is independently halogen, —CN, —CO2R15, —C(═O)NR15R16, —NR15R16, —OR15, —SO2R7, or —P(═O)R8R9;
      • each R15 and R16 is independently acyl, alkenyl, alkoxyl, OH, NH2, alkyl, alkylamino, aryl, alkylaryl, cycloalkyl, cycloalkylalkyl, heteroaryl, heterocyclyl, or heterocyclylalkyl, each of which is independently substituted or unsubstituted, or H; or R15 and R16 together with the N to which R15 and R16 are bonded form a heterocycle that is substituted or unsubstituted;
      • n is 0, 1, or 2;
      • q is 0, 1, 2, 3, or 4;
      • t is 1, 2, 3, 4, 5, or 6; and
      • m is 1, 2, 3, or 4,


        or any other compound herein, or a pharmaceutically acceptable salt thereof.





EXAMPLES
Example 1: Generation of Stable Cell Lines

Complementary DNA (cDNA) for human RyR2 was subcloned into the A1.2 vector in two steps, using the Nhe I-Xho I fragment and then the Not I-Nhe I fragment of human RYR2. Human RYR2 was inserted 3′ of a cytomegalovirus (CMV) promoter and followed in 5′ by internal ribosomal entry site (IRES)-green fluorescent protein (GFP). The sequence of the construct was confirmed by Sanger sequencing and restriction digestion with Afl III enzyme. The resulting vector (G418-resistant) was cotransfected into HEK293T cells with a plasmid carrying a puromycin resistance gene using calcium chloride. Cells were maintained in a G418- and puromycin-containing medium for approximately 3 weeks and then underwent two cycles of clonal selection, where the top 0.1% of the most highly fluorescent cells were propagated.


Example 2: Generation of Human RYR2-R2474S DNA and HEK293 Transfection

Constructs expressing RyR2-R2474S were formed by introducing the respective mutation into fragments of human RYR2 using the QuikChange II XL Site-Directed Mutagenesis Kit (Agilent). Three nucleotide changes were introduced (WT sequence: AGGGTCTAT to R2474S mutant: AGCGTATAC). The first was the mutation R2474S, and the other two were silent mutations that introduced a BstZ171 restriction site (GTATAC) to facilitate screening for mutant clones. Each fragment was subcloned into a full-length human RYR2 construct in pCMV5 vector, confirmed by sequencing, and expressed in 293T/17 cells using Lipofectamine 2000 (Thermo Fisher Scientific). For final expression, HEK293 cells grown in 150-mm dishes with Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% (v/v) fetal bovine serum (Invitrogen), penicillin (100 U/ml), streptomycin (100 μg/ml), and 2 mM 1-glutamine were cotransfected with 25 mg per dish of human RYR2-R2474S cDNA using PEI MAX at a 1:5 ratio (Polysciences). Cells were collected 48 hours after transfection.


Example 3: Purification of Recombinant GST-Calstabin-2

Recombinant human GST-calstabin-2 was expressed in BL21 (DE3) Escherichia co/i cells with a thrombin protease cleavage site between GST and calstabin-2. Protein expression was induced with 0.8 mM isopropyl-o-d-thiogalactopyranoside (IPTG) added to E. coli at an OD600 (optical density at 600 nm) of 0.8 with overnight incubation at 18° C. before centrifugation for 10 min at 6500 g. The pellets were resuspended in buffer A (phosphate-buffered saline+0.5 mM AEBSF) and lysed using an emulsiflex (Avestin EmulsiFlex-C3). The lysate was pelleted by centrifugation for 10 min at 100,000 g. The supernatant was then loaded into a 5-ml GSTrap HP column (Cytiva) and washed with 5 column volume (CV) of buffer A to remove contaminants before elution with buffer B [tris (pH 8), 2 mM DTT, and 20 mM glutathione]. Fractions containing GST-calstabin-2 were pooled, concentrated, and dialyzed overnight at 4° C. into buffer A. final concentration was determined by spectroscopy using NanoDrop 1000 (Thermo Fisher Scientific) with absorbance at 280 nm and an extinction coefficient of 46,200 M−1 cm−1. GST-calstabin-2 was stored at −80° C.


Example 4: Purification of Recombinant Calmodulin (CaM) and TEV Protease

Recombinant human calmodulin (CaM) was expressed in BL21 (DE3) E. coli cells with an N-terminal 6-histidine tag and a tobacco etch virus (TEV) protease cleavage site. Protein expression was induced with 0.8 mM IPTG added to E. coli at an OD600 of 0.8 with overnight incubation at 18° C. before centrifugation for 10 min at 6500 g and storage at −80° C. CaM was purified using a two-step 5-ml HisTrap HP column (Cytiva) purification. In brief, the pellets were resuspended in buffer A [20 mM Hepes (pH 7.5), 150 mM NaCl, 20 mM imidazole, 5 mM 2-Mercaptoethanol, and 0.5 mM AEBSF] and lysed using an emulsiflex (Avestin EmulsiFlex-C3). The lysate was pelleted by centrifugation for 10 min at 100,000 g. The supernatant was then loaded over a HisTrap column and washed with 5 CV of buffer A to remove contaminants before elution using a linear gradient from buffer A to buffer B (buffer A containing 500 mM imidazole). Fractions containing CaM were pooled, 1 to 2 mg of purified TEV protease was added, and the mixture was dialyzed overnight at 4° C. into buffer C (buffer A with no imidazole). CaM was then loaded onto a HisTrap column with the flowthrough collected and the wash fractionated to retain fractions containing CaM before elution of TEV and any remaining contaminants with a linear gradient from buffer C to buffer B. The flowthrough and any fractions containing CaM were pooled, concentrated to >2 mM, and determined by spectroscopy using NanoDrop 1000 (Thermo Fisher Scientific) with absorbance at 280 nm and the extinction coefficient of CaM (3000 M−1 cm−1). CaM was stored at −20° C. TEV protease was purified in the same manner except for using an uncleavable his-tag and thus ending after the first HisTrap column wherein the purified protease was stored at −80° C. in buffer C with 10% glycerol.


Example 5: Purification and Treatment of Recombinant Human RyR2

To obtain high-resolution structures of WT and mutant human RyR2, the purification of channels expressed recombinantly in HEK293 cells was optimized. All purification steps were performed on ice unless otherwise stated. HEK293 cells (25 to 50 dishes) expressing human RyR2 or RyR2-R2474S (prepared according to EXAMPLE 1 and EXAMPLE 2) were harvested by centrifugation for 10 min at 1500 g. The pellet fraction was resuspended in tris malate buffer [10 mM tris malate (pH 6.8), 1 mM EGTA, 1 mM dithiothreitol (DTT), 1 mM benzamidine, 0.5 mM 4-benzenesulfonyl fluoride hydrochloride (AEBSF), and protease inhibitor cocktail] and was sonicated with six pulses of 20 s at 35% amplitude. The membrane fraction was precipitated by centrifugation at 100,000 g for 30 min and was resuspended with a glass homogenizer in CHAPS buffer [10 mM Hepes (pH 7.4), 1 M NaCl, 1.5% CHAPS, 0.5% phosphatidylcholine (PC), 1 mM EGTA, 2 mM DTT, 0.5 mM AEBSF, 1 mM benzamidine, and protease inhibitor cocktail]. The sample was diluted 1:4 with the same buffer without NaCl. To achieve stabilization of peripherical domains, 100 nmol of GST-calstabin-2 was added to both tris malate buffer and CHAPS buffer. The remaining insoluble material was separated with a second centrifugation at 100,000 g for 30 min.


The supernatant-containing detergent-solubilized human RyR2—was filtered and loaded into a 5-ml HiTrap Q HP column (Cytiva) previously equilibrated with buffer A [10 mM Hepes (pH 7.4), 0.4% CHAPS, 1 mM EGTA, 0.001% dioleoylphosphatidylcholine (DOPC), 250 mM NaCl, and 0.5 mM TCEP (tris(2-carboxyethyl)phosphine)]. The HiTrap Q HP column was eluted with a linear gradient between 300 and 600 mM NaCl. The fractions containing human RyR2 (300 to 350 mM NaCl) were pooled, and 100 nmol of GST-calstabin-2 (prepared according to EXAMPLE 3) was added. The pooled fractions were loaded into a 1-ml GSTrap HP column (Cytiva), which was left recirculating overnight. The GSTrap HP column was washed with buffer A and eluted with glutathione buffer {10 mM Hepes (pH 8), 0.4% CHAPS, 1 mM EGTA, 0.001% DOPC, 200 mM NaCl, 10 mM GSH [glutathione (reduced form)], and 1 mM DTT}. Immediately after, a second 1-ml HiTrap Q HP column (Cytiva) binding/elution step was applied to separate human RyR2 from the excess of unbound GST-calstabin-2 and GSH.


Six preparations of RyR2 and RyR2-R2474S were prepared:

    • (1) Dephosphorylated RyR2 (DeP-RyR2);
    • (2) PKA phosphorylated RyR2 (P-RyR2);
    • (3) PKA phosphorylated RyR2+20 μM CaM (P-RyR2+CaM);
    • (4) PKA phosphorylated RyR2-R2474S (P-RyR2-R2474S);
    • (5) PKA phosphorylated RyR2-R2474S+20M Compound 1 (P-RyR2-R2474S+Cpd1); and
    • (6) PKA phosphorylated RyR2-R2474S+40 μM CaM (P-RyR2-R2474S+CaM).


For phosphorylated samples (P-RyR2, P-RyR2+CaM, P-RyR2-R2474S, P-RyR2-R2474S+Cpd1, and P-RyR2-R2474S+CaM), simultaneous cleavage of GST tag and PKA phosphorylation was performed by addition of 50 U of thrombin and 100 U of PKA (+10 mM EGTA, 8 mM MgCl2, and 100 μM ATP for activity), respectively, for 30 min on ice. For samples requiring dephosphorylation treatment (DeP-RyR2), PKA was replaced by 2000 U of phosphatase lambda (PX from NEB, +1×Protein MetalloPhosphatases (PMP) buffer, and 1 mM MnCl2). Each sample was concentrated to 0.5 ml, and a gel filtration step was run with TSKgel G4SWXL (TOSOH Biosciences) with buffer A. RyR2 fractions were pooled and concentrated to a concentration of 4 to 8 mg/ml (with centrifugal filters of 100-kDa cutoff) and were filtered (with centrifugal filters of 0.22-μm cutoff) to eliminate aggregates.


To resemble exercise diastolic conditions, 10 mM NaATP, 500 μM xanthine, 150 nM Ca2+ free (650 μM total Ca2+), and 200 μM cAMP were added to all samples. 20 μM CaM (prepared according to EXAMPLE 4) was added to PKA-phosphorylated RyR2 (P-RyR2+CaM), 40 μM CaM was added to PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S+CaM), and 500 μM Compound 1 was added to PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S+Cpd1). MaxChelator webserver was used to calculate total/free Ca2+ concentrations.


Quality control was assessed by SDS-polyacrylamide gel electrophoresis (SDS-PAGE) and immunoblots using anti-RyR-5029 and anti-RyR2-pS2809 antibodies for total and phosphorylated RyR2, respectively. FIG. 1A depicts an immunoblot (left, top) and SDS-PAGE (left, middle) of purification of human recombinant RyR2 expressed in HEK293 cells. Ratio of normalized intensities of the pS2808 and total RyR2 bands (right). After PKA phosphorylation, the intensity increases ˜3-fold, reaching a saturation point. RyR2 expressed in HEK293 cells show a basal phosphorylation of S2808 as confirmed by mass spectrometry. FIG. 1B depicts an immunoblot (left, top) and SDS-PAGE (left, bottom) of purification of dephosphorylated (with phosphatase lambda) human recombinant RyR2 expressed in HEK293 cells.


Example 6: Cryo-EM Analysis
Methods.
Sample Preparation and Data Collection.

Each of the final samples (3 μl each) prepared in EXAMPLE 5 was applied to UltrAuFoil holey gold grids (Quantifoil R 0.6/1.0, Au 300) previously cleaned with easiGlow (PELCO). Grids were blotted with ashless filter paper (Whatman) using blot force 10 and blot time 8 s before vitrification by plunge-freezing into liquid ethane chilled with liquid nitrogen using Vitrobot Mark IV (Thermo Fisher Scientific) operated at 4° C. with 100% relative humidity.


Prepared grids were screened in-house on a Glacios Cryo-TEM (Thermo Fisher Scientific) microscope with a 200-kV x-FEG source and a Falcon 3EC direct electron detector (Thermo Fisher Scientific). Microscope operations and data collection were carried out using EPU software (Thermo Fisher Scientific). High-resolution data collection was performed at Columbia University on a Titan Krios 300-kV (Thermo Fisher Scientific) microscope equipped with an energy filter (slit width 20 eV) and a K3 direct electron detector (Gatan). Data were collected using Leginon and at a nominal magnification of ×105,000 in electron counting mode, corresponding to a pixel size of 0.83 Å. The electron dose rate was set to 16 e/pixel per second with 2.5-s exposures for a total dose of 50 to 60 e/A2.


Data Processing and Model Building.

Cryo-EM data processing was performed in cryoSPARC with image stacks aligned using Patch motion, defocus value estimation by Patch CTF estimation. Particle picking was performed using the template picker with templates created from preexisting cryo-EM maps. Particles were subjected to 2D classification in cryoSPARC with 100 classes. Particles from the highest-resolution classes were pooled for ab initio 3D reconstruction with a single class followed by homogeneous refinement with C4 symmetry imposed. 3D variability and further clustering were performed using a mask comprising the TM domain to separate those particles in the closed and open states followed by heterogeneous refinement with four classes to further select the best particles. C4 symmetry expansion was performed before local refinements. The masks used were TaF+TM+CTD domains (residues 4131 to 4967), calstabin-2+NTD+SPRY domains (residues 1 to 1646), JSol+CSol domains (residues 1700 to 2476, 3590 to 4130), and BSol domain (residues 2400 to 3344). Only the TaF+TM+CTD mask used C4 symmetry. Smaller masks, for a second round of 3D variability analysis, clustering, and local refinement, were RY1&2 (residues 862 to 1076), RY3&4 (residues 2685 to 2909), and BSol2 (residues 3042 to 3344). Local refinements used a dynamical mask with a far distance of 10, 50, and 150 Å for the initial masks, small masks, and expanded RY3&4 mask for comparing the effect of RY3&4 stabilization, respectively. The resulting maps were combined in ChimeraX to generate a composite map before calibration of the pixel size using correlation coefficients with a map generated from the crystal structure of the NTD of RyR2 (PDB ID: 4JKQ). The pixel size was altered by 0.001 Å per step, up to 20 steps in each direction.


An initial model of RYR2 was generated with Phenix tool sculptor from a 2.45-Å structure of RyR1 (PDB ID: 7TZC). Truncated residues were manually corrected to obtain the full side chains. Only domains RY1&2 and RY3&4, which show weak cryo-EM density and confidence in structure, were based on crystallographic structures. The initial model of RyR2 RY1&2 domain was generated with Phenix tool sculptor from the RY1&2 domain of RyR3 with ATP (PDB ID: 6UHIH). The model of RyR2 RY3&4 domain was obtained as is (PDB ID: 4ETV). Calstabin-2 and CaM models were obtained as is (PDB ID: 6JI8). Model building was performed in Coot and refined with Phenix tool RealSpaceRefine. Figures of the final structure were created using ChimeraX. cryo-EM statistics are summarized in TABLE 1A and TABLE 1B.











TABLE 1A









Sample











Dephosphorylated
PKA phosphorylated
PKA phosphorylated



hRyR2
hRyR2
hRyR2 + CaM














In vitro treatment
Pλ dephosphorylation
PKA phosphorylation
PKA phosphorylation












State
closed
open
closed
open
closed


PDB ID
7UA5
7UA9
7U9Q
7U9R
7U9T


EMDB ID
EMD-26415
EMD-26416
EMD-26405
EMD-26407
EMD-26408


Data collection


Microscope
FEI Titan
FEI Titan
FEI Titan
FEI Titan
FEI Titan



Krios
Krios
Krios
Krios
Krios


Detector
Gatan K3
Gatan K3
Gatan K3
Gatan K3
Gatan K3


Voltage (kV)
300
300
300
300
300


Magnification
105,000
105,000
105,000
105,000
105,000


Exposure (e/Å2)
58
58
50.54
58
58


Defocus range (μm)
−0.4 to −1.2
−0.4 to −1.2
−0.4 to −1.2
−0.4 to −1.2
−0.4 to −1.2


Pixel size (Å)
0.83
0.83
0.844
0.83
0.83


Processing


Software
cryoSPARC
cryoSPARC
cryoSPARC
cryoSPARC
cryoSPARC


Symmetry
C4
C4
C4
C4
C4


Initial particles (N)
148,746
148,746
94,476
148,830
148,830


Final particles (N)
90,375
36,491
94,476
20,156
93,881


Global map res. (Å)*
2.83
3.59
3.04
3.54
2.68


Local maps res. range
2.52-4.06
2.92-5.45
2.76-4.13
3.13-3.71
2.41-3.49


(Å)


Model Composition


Peptide chains
8
8
8
8
12


Nonhydrogen
138,608
138,656
138,608
138,656
143,236


Protein residues
17,324
17,324
17,324
17,324
17,912


Ligands
12
20
12
20
12


Mean B factors (Å2)


Protein
107.18
135.07
120.33
111.46
109.79


Ligands
149.67
187.73
203.75
116.42
176.02


R.m.s. deviations


Bond length (Å)
0.003
0.010
0.003
0.003
0.004


Bond angles (°)
0.779
0.877
0.848
0.848
0.81


Ramachandran


Favored (%)
96.84
96.83
96.55
97.48
96.4


Allowed (%)
3.07
3.10
3.36
2.43
3.53


Disallowed (%)
0.09
0.07
0.09
0.09
0.07


Validation


MolProbity score
1.86
2.06
1.82
2.02
1.99


Clashscore
10.84
14.72
12.18
12.81
10.53


Rotamer outliers (%)
1.41
1.82
0.66
2.45
1.89


FSC model 0.5 (Å)#
2.88
3.38
3.20
3.88
2.92





*Map resolution of the non-uniform refinement determined by CryoSPARC before local masks.



Map resolution range represents the range determined by local refinements in cryoSPARC using the masks described in Methods.




#Value obtained from Phenix validation tool.
















TABLE 1B









Model











PKA phosphorylated
PKA phosphorylated
PKA phosphorylated



hRyR2-R2474S
hRyR2-R2474S + Cpd1
hRyR2-R2474S + CaM














In vitro treatment
PKA phosphorylation
PKA phosphorylation
PKA phosphorylation












State
primed
open
closed
closed
open


PDB ID
7U9X
7U9Z
7UA1
7UA3
7UA4


EMDB ID
EMD-26409
EMD-26410
EMD-26412
EMD-26413
EMD-26414


Data collection


Microscope
FEI Titan
FEI Titan
FEI Titan
FEI Titan
FEI Titan



Krios
Krios
Krios
Krios
Krios


Detector
Gatan K3
Gatan K3
Gatan K3
Gatan K3
Gatan K3


Voltage (kV)
300
300
300
300
300


Magnification
105,000
105,000
105,000
105,000
105,000


Exposure (e/Å2)
58.2
58.2
58.2
58.2
58.2


Defocus range (μm)
−0.4 to −1.2
−0.4 to −1.2
−0.4 to −1.2
−0.4 to −1.2
−0.4 to −1.2


Pixel size (Å)
0.83
0.83
0.83
0.83
0.83


Processing


Software
cryoSPARC
cryoSPARC
cryoSPARC
cryoSPARC
cryoSPARC


Symmetry
C4
C4
C4
C4
C4


Initial particles (N)
271,481
271,481 +
133,548
205,393
205,393




133,548


Final particles (N)
212,141
42,295
113,172
73,052
102,257


Global map res. (Å)*
2.58
3.23
2.93
2.87
2.93


Local maps res. range
2.25-2.97
2.79-3.76
2.53-3.56
2.52-3.46
2.46-3.32


(Å)


Model Composition


Peptide chains
8
8
8
12
12


Nonhydrogen
138,588
138,688
138,680
150,252
147,856


Protein residues
17,324
17,332
17,324
18,768
18,472


Ligands
12
20
16
12
20


Mean B factors (Å2)


Protein
85.11
112.14
96.72
101.58
105.11


Ligands
110.48
180.94
135.79
136.45
199.47


R.m.s. deviations


Bond length (Å)
0.003
0.003
0.003
0.003
0.006


Bond angles (°)
0.614
0.752
0.618
0.687
0.729


Ramachandran


Favored (%)
97.7
95.26
96.56
95.93
96.36


Allowed (%)
2.28
4.6
3.32
3.92
3.55


Disallowed (%)
0.02
0.14
0.12
0.15
0.09


Validation


MolProbity score
1.55
1.96
1.69
1.78
1.84


Clashscore
9.1
11.55
8.72
9.46
10.59


Rotamer outliers (%)
0.11
1.19
0.29
0.52
1.19


FSC model 0.5 (Å)#
2.65
3.16
3.03
2.97
2.90





*Map resolution of the non-uniform refinement determined by CryoSPARC before local masks.



Map resolution range represents the range determined by local refinements in cryoSPARC using the masks described in Methods.



#Value obtained from Phenix validation tool.






Normalized Difference in RMSD Analyses: Pairwise 1-Domain Comparison.

This analysis determined the conformation of a certain query domain from a model X relative to reference closed and open states of RyR2. To this end, the RMSD of Ca of the query domain was measured between model X and the closed state (RMSDX-closed), and between model X and the open state (RMSDX-open). The normalized difference in RMSD was calculated as RMSDX-closed/(RMSDX-closed+RMSDX-open). A value of 0 means that the conformation of the query domain is identical to the same domain in the closed state. A value of 1 means that the conformation of the query domain is identical to the same domain in the open state. A value of 0.5 means that the conformation of the query domain is equally distant to the closed and open states. The error, considered as the intrinsic variability between atomic models, was calculated as the RMSD between aligned domains of the respective models. Propagation of error was calculated using the webserver uncertaintycalculator.com. Bar graphs were made with GraphPad Prism software. RMSD was measured in ChimeraX. When needed, previous alignment of the atomic models centered on the control domains was performed.


Results.
CPVT-Linked Mutation RyR2-R2474S Induction of Primed State, and Compound I and CaM Induction of Closed State.

For cryo-EM experiments, conditions that resemble those found during the resting phase of the cardiac cycle known as diastole (150 nM free Ca12 and 10 mM ATP) were used. Under these conditions, wild-type RyR2 channels are tightly closed, but CPVT RyR2 variants are leaky. Wild-type RyR2 and RyR2-R2474S were pretreated with PKA to induce hyperphosphorylation and to mimic exercise-induced β-adrenergic signaling, which can be a trigger for fatal cardiac arrhythmias in patients with CPVT (FIG. 1A). Xanthine, a physiologically relevant analog of the RyR2 agonist caffeine, was added (500 μM). Xanthine activates RyRs and increases to micromolar range during exercise and can be endogenous activator of RyR2 during exercise.


Across the six preparations of RyR1 subjected to cryo-EM analysis, 10 structures of human RyR2 were obtained:


Dephosphorylated RyR2 (DeP-RyR2)





    • (i) Open state (DeP-RyR2-0)

    • (ii) Closed state (DeP-RyR2-C)





PKA Phosphorylated RyR2 (P-RyR2)





    • (iii) Open state (P-RyR2-0)

    • (iv) Closed state (P-RyR2-C)





PKA Phosphorylated RyR2+20 μM CaM (P-RyR2+CaM)





    • (v) Closed state (P-RyR2+CaM-C)





PKA Phosphorylated RyR2-R2474S (P-RyR2-R2474S)





    • (vi) Open state (P-RyR2-R2474S-O)

    • (vii) Primed state (P-RyR2-R2474S-Pr)





PKA Phosphorylated RyR2-R2474S+20M Compound I (P-RyR2-R2474S+Cpd1)





    • (viii) Closed state (P-RyR2-R2474S+Cpd1-C). Compound 1 shifts the equilibrium of the RyR2-R2474S channel pore from a primed state to a closed state. In this case, the cytosolic shell is in a primed state, but less open in the presence than the absence of Compound 1. In other words, in the presence of Compound 1, more RyR2-R2474S particles are in a closed state than in the absence of the compound.





PKA Phosphorylated RyR2-R2474S+40 μM CaM (P-RyR2-R2474S+CaM)





    • (ix) Open state (P-RyR2-R2474S+CaM-O)

    • (x) Closed state (P-RyR2-R2474S+CaM-C)





˜3-Å resolution was obtained for most structures. FIGS. 2A-2J provide GSFSCs (top) and viewing angle distributions (middle) of the global nonuniform refinements performed in cryoSPARC before any local refinement for each structure, and FSC model-map performed in PHENIX (bottom) for DeP-RyR2-C (FIG. 2A), DeP-RyR2-0 (FIG. 2B), P-RyR2-C (FIG. 2C), P-RyR2-0 (FIG. 2D), P-RyR2+CaM-C (FIG. 2E), P-RyR2-R2474S-Pr, (FIG. 2F), P-RyR2-R2474S-0 (FIG. 2G), P-RyR2-R2474S+Cpd1-C (FIG. 211), P-RyR2-R2474S+CaM-C (FIG. 2I), and P-RyR2-R2474S+CaM-O (FIG. 2J).


The highest-resolution cryo-EM coulombic potential maps (cryo-EM maps) show well-defined densities of most domains, including the NTD, SPRY1-3, JSol, BSol1, and activation core and channel domains, where near-atomic details including “holes” in aromatic residues were observed. FIG. 3 depicts representative high-resolution details of cryo-EM maps showing the holes in proline and aromatic residues, precise side-chain conformations, and stabilized water molecules (blue arrows). Respective regions of the JSol (left), CSol (center), and TaF+CTD domains (right) show that these details are distributed over the structure. FIG. 4A and FIG. 4B depict local refinement cryo-EM maps colored by local resolution shown for the “primed” PKA RyR2-R2474S, which showed the highest resolution and was used for building the initial atomic model.


For the remaining dynamic domains usually missing in previously published structures (RY1&2, RY3&4, and BSol2), the local resolution was reduced but sufficient to follow the backbone and detect bulky side chains. The cryo-EM map quality was sufficient to build atomic models with a high level of confidence for the previously less well-resolved domains of RyR2 and the accessory proteins calstabin-2 (known as PPIase FKBP1B or, alternatively, as FKBP12.6) and CaM. Using this high-resolution model of the human RyR2 isoform, the residue span and nomenclature of domains and subdomains for human RyR2 were updated as shown in TABLE 2.









TABLE 2







Nomenclature for Human RyR2









Identification
Symbol
Residue Span





Cytosolic shell

  (1-3633)


N-terminal domain
NTD
 (1-639)


N-terminal domain A
NTD-A
 (1-219)


N-terminal domain B
NTD-B
(220-408)


N-terminal solenoid
NSol
(409-639)


SPRY domain
SPRY
 (640-1646)


SP1a/ryanodine receptor domain 1
SPRY1
(640-861, 1463-1483,




1595-1646)


SP1a/ryanodine receptor domain 2
SPRY2
(1076-1255)


SP1a/ryanodine receptor domain 3
SPRY3
(1256-1462, 1484-1594)


RYR repeats 1 & 2
RY1&2
 (862-1076)


Junctional solenoid
JSol
(1647-2108)


Bridging solenoid
BSol
(2109-3564)


Bridging solenoid 1
BSo11
(2109-2681, 2916-3042)


Bridging solenoid 2
BSo12
(3042-3344)


Bridging solenoid 3
BSo13
(3345-3564)


RYR repeats 3 & 4
RY3&4
(2682-2915)


Shell-core linker peptide
SCLP
(3565-3633)


Activation core and channel

(3634-4967)


Core solenoid
CSol
(3634-4130)


EF-hand pair
EF1&2
(4016-4090)


Thumb and forefingers domain
TaF
(4131-4209)


Transmembrane domain
TM
(4237-4886)


Auxiliary intramembrane helices
Sx
(4237-4310)


Pseudo voltage sensor domain
pVSD
(4480-4750)


Channel pore domain
Pore
(4751-4886)


C-terminal domain
CTD
(4887-4967)


Zn-finger domain
ZnF
(4887-4914)









To separate the particles in the closed state from those in the open states in the same dataset, local three-dimensional (3D) variability, clustering, and heterogeneous refinement were performed, which rendered better results than 3D classification or heterogeneous refinement alone. This improvement was attributed to the dynamic cytosolic shell of RyR2, which, being almost 90% of the mass of the protein, can predominate and result in mixed populations.



FIGS. 5A-5K are flowcharts that summarize the entire cryoSPARC processing of the cryo-EM datasets to obtain final composite maps of DeP-RyR2 (FIG. 5A and FIG. 5B), P-RyR2 (FIG. 5C), P-RyR2+CaM (FIG. 5D and FIG. 5E), P-RyR2-R2474S (FIG. 5F and FIG. 5G), P-RyR2-R2474S+Cpd1 (FIG. 5H and FIG. 5I), and P-RyR2-R2474S+CaM (FIG. 5J and FIG. 5K). Number of particles and resolution achieved for each refinement are shown for each step. Masks used are shown only in (FIG. 5B) but are identical in all processing procedures depicted in FIGS. 5A-5K. For better clarity, cryo-EM maps of the 3D variability of Compound 1 are shown in FIG. 5I.


The three-dimensional atomic coordinates as determined by cryo-EM for the RY1&2 domain in PKA phosphorylated RyR2-R2474S treated with 20M Compound 1 (P-RyR2-R2474S+Cpd1-C, residues 862-1076), are provided in TABLE 3. The three-dimensional atomic coordinates for Compound 1 and ATP bound to the RY1&2 domain of the P-RyR2-R2474S+Cpd1-C structure are provided in TABLE 4 and TABLE 5, respectively.









TABLE 3







Three-dimensional atomic coordinates of RY1&2 domain.















Id1
type_symbol2
label_atom_id3
label_comp_id4
label_seq_id5
Cartn_x6
Cartn_y7
Cartn_z8
B_iso_or_equiv9


















6450
N
N
PHE
862
74.748
182.682
181.942
121.08


6451
C
CA
PHE
862
74.218
181.323
181.964
121.08


6452
C
C
PHE
862
74.061
180.829
180.533
121.08


6453
O
O
PHE
862
73.314
181.419
179.745
121.08


6454
C
CB
PHE
862
72.885
181.267
182.706
121.08


6455
C
CG
PHE
862
72.197
179.935
182.614
121.08


6456
C
CD1
PHE
862
72.898
178.761
182.827
121.08


6457
C
CD2
PHE
862
70.849
179.858
182.308
121.08


6458
C
CE1
PHE
862
72.268
177.535
182.739
121.08


6459
C
CE2
PHE
862
70.213
178.635
182.22
121.08


6460
C
CZ
PHE
862
70.924
177.472
182.435
121.08


6461
N
N
THR
863
74.763
179.752
180.205
117.62


6462
C
CA
THR
863
74.615
179.081
178.918
117.62


6463
C
C
THR
863
74.3
177.614
179.172
117.62


6464
O
O
THR
863
75.19
176.857
179.603
117.62


6465
C
CB
THR
863
75.876
179.226
178.067
117.62


6466
O
OG1
THR
863
76.027
180.592
177.665
117.62


6467
C
CG2
THR
863
75.783
178.351
176.825
117.62


6468
N
N
PRO
864
73.066
177.169
178.935
121.81


6469
C
CA
PRO
864
72.74
175.752
179.141
121.81


6470
C
C
PRO
864
73.41
174.899
178.075
121.81


6471
O
O
PRO
864
73.23
175.123
176.875
121.81


6472
C
CB
PRO
864
71.213
175.715
179.022
121.81


6473
C
CG
PRO
864
70.86
176.897
178.186
121.81


6474
C
CD
PRO
864
71.96
177.919
178.316
121.81


6475
N
N
ILE
865
74.184
173.918
178.52
129.57


6476
C
CA
ILE
865
75.048
173.139
177.643
129.57


6477
C
C
ILE
865
74.427
171.76
177.436
129.57


6478
O
O
ILE
865
74.069
171.074
178.398
129.57


6479
C
CB
ILE
865
76.483
173.049
178.198
129.57


6480
C
CG1
ILE
865
77.44
172.506
177.134
129.57


6481
C
CG2
ILE
865
76.55
172.252
179.502
129.57


6482
C
CD1
ILE
865
77.617
173.431
175.947
129.57


6483
N
N
PRO
866
74.226
171.348
176.189
138.28


6484
C
CA
PRO
866
73.634
170.033
175.925
138.28


6485
C
C
PRO
866
74.641
168.911
176.121
138.28


6486
O
O
PRO
866
75.85
169.13
176.223
138.28


6487
C
CB
PRO
866
73.204
170.125
174.454
138.28


6488
C
CG
PRO
866
73.318
171.584
174.092
138.28


6489
C
CD
PRO
866
74.408
172.118
174.951
138.28


6490
N
N
VAL
867
74.118
167.687
176.171
145.84


6491
C
CA
VAL
867
74.967
166.508
176.085
145.84


6492
C
C
VAL
867
75.276
166.226
174.622
145.84


6493
O
O
VAL
867
74.513
166.599
173.72
145.84


6494
C
CB
VAL
867
74.297
165.296
176.756
145.84


6495
C
CG1
VAL
867
74.14
165.542
178.245
145.84


6496
C
CG2
VAL
867
72.949
165.01
176.111
145.84


6497
N
N
ASP
868
76.409
165.574
174.378
164.98


6498
C
CA
ASP
868
76.797
165.25
173.014
164.98


6499
C
C
ASP
868
75.807
164.266
172.403
164.98


6500
O
O
ASP
868
75.265
163.397
173.091
164.98


6501
C
CB
ASP
868
78.21
164.668
172.987
164.98


6502
C
CG
ASP
868
78.729
164.459
171.578
164.98


6503
O
OD1
ASP
868
78.217
165.119
170.649
164.98


6504
O
OD2
ASP
868
79.652
163.636
171.398
164.98


6505
N
N
THR
869
75.558
164.419
171.102
166.07


6506
C
CA
THR
869
74.63
163.557
170.388
166.07


6507
C
C
THR
869
75.235
162.888
169.162
166.07


6508
O
O
THR
869
74.484
162.373
168.328
166.07


6509
C
CB
THR
869
73.379
164.343
169.964
166.07


6510
O
OG1
THR
869
73.77
165.526
169.257
166.07


6511
C
CG2
THR
869
72.554
164.729
171.182
166.07


6512
N
N
SER
870
76.559
162.91
169.006
171.46


6513
C
CA
SER
870
77.182
162.13
167.942
171.46


6514
C
C
SER
870
77.225
160.65
168.301
171.46


6515
O
O
SER
870
77.075
159.787
167.429
171.46


6516
C
CB
SER
870
78.589
162.657
167.66
171.46


6517
O
OG
SER
870
79.429
162.501
168.791
171.46


6518
N
N
GLN
871
77.43
160.342
169.584
173.01


6519
C
CA
GLN
871
77.625
158.961
170.01
173.01


6520
C
C
GLN
871
76.309
158.212
170.179
173.01


6521
O
O
GLN
871
76.319
156.989
170.357
173.01


6522
C
CB
GLN
871
78.401
158.933
171.326
173.01


6523
C
CG
GLN
871
77.523
159.153
172.551
173.01


6524
C
CD
GLN
871
77.232
160.62
172.811
173.01


6525
O
OE1
GLN
871
76.725
161.327
171.941
173.01


6526
N
NE2
GLN
871
77.546
161.081
174.016
173.01


6527
N
N
ILE
872
75.178
158.913
170.133
166.17


6528
C
CA
ILE
872
73.904
158.318
170.522
166.17


6529
C
C
ILE
872
73.5
157.244
169.521
166.17


6530
O
O
ILE
872
73.535
157.443
168.301
166.17


6531
C
CB
ILE
872
72.813
159.398
170.663
166.17


6532
C
CG1
ILE
872
72.644
160.176
169.358
166.17


6533
C
CG2
ILE
872
73.145
160.344
171.807
166.17


6534
C
CD1
ILE
872
71.528
161.195
169.389
166.17


6535
N
N
VAL
873
73.134
156.079
170.05
173.29


6536
C
CA
VAL
873
72.581
154.981
169.27
173.29


6537
C
C
VAL
873
71.38
154.43
170.023
173.29


6538
O
O
VAL
873
71.28
154.577
171.246
173.29


6539
C
CB
VAL
873
73.618
153.866
169.011
173.29


6540
C
CG1
VAL
873
74.755
154.379
168.139
173.29


6541
C
CG2
VAL
873
74.151
153.317
170.326
173.29


6542
N
N
LEU
874
70.47
153.807
169.294
176.69


6543
C
CA
LEU
874
69.303
153.216
169.932
176.69


6544
C
C
LEU
874
69.679
151.859
170.523
176.69


6545
O
O
LEU
874
70.272
151.029
169.827
176.69


6546
C
CB
LEU
874
68.155
153.09
168.926
176.69


6547
C
CG
LEU
874
66.831
152.423
169.31
176.69


6548
C
CD1
LEU
874
65.703
153.034
168.499
176.69


6549
C
CD2
LEU
874
66.875
150.932
169.056
176.69


6550
N
N
PRO
875
69.365
151.609
171.797
174.54


6551
C
CA
PRO
875
69.841
150.384
172.441
174.54


6552
C
C
PRO
875
69.26
149.149
171.781
174.54


6553
O
O
PRO
875
68.117
149.165
171.292
174.54


6554
C
CB
PRO
875
69.354
150.538
173.893
174.54


6555
C
CG
PRO
875
68.181
151.452
173.793
174.54


6556
C
CD
PRO
875
68.491
152.402
172.677
174.54


6557
N
N
PRO
876
70.001
148.036
171.769
176.91


6558
C
CA
PRO
876
69.699
146.963
170.805
176.91


6559
C
C
PRO
876
68.48
146.119
171.14
176.91


6560
O
O
PRO
876
68.011
145.386
170.262
176.91


6561
C
CB
PRO
876
70.977
146.109
170.803
176.91


6562
C
CG
PRO
876
71.805
146.567
171.957
176.91


6563
C
CD
PRO
876
71.15
147.725
172.635
176.91


6564
N
N
HIS
877
67.974
146.144
172.375
173.58


6565
C
CA
HIS
877
66.801
145.324
172.669
173.58


6566
C
C
HIS
877
65.531
146.15
172.838
173.58


6567
O
O
HIS
877
64.49
145.594
173.202
173.58


6568
C
CB
HIS
877
67.043
144.455
173.907
173.58


6569
C
CG
HIS
877
67.253
145.227
175.173
173.58


6570
N
ND1
HIS
877
68.464
145.79
175.511
173.58


6571
C
CD2
HIS
877
66.411
145.5
176.198
173.58


6572
C
CE1
HIS
877
68.356
146.389
176.684
173.58


6573
N
NE2
HIS
877
67.12
146.229
177.122
173.58


6574
N
N
LEU
878
65.588
147.461
172.592
174.63


6575
C
CA
LEU
878
64.427
148.312
172.832
174.63


6576
C
C
LEU
878
63.662
148.689
171.567
174.63


6577
O
O
LEU
878
62.798
149.571
171.63
174.63


6578
C
CB
LEU
878
64.836
149.59
173.571
174.63


6579
C
CG
LEU
878
65.149
149.484
175.067
174.63


6580
C
CD1
LEU
878
66.489
148.834
175.323
174.63


6581
C
CD2
LEU
878
65.088
150.86
175.715
174.63


6582
N
N
GLU
879
63.95
148.061
170.424
183.39


6583
C
CA
GLU
879
63.233
148.416
169.2
183.39


6584
C
C
GLU
879
61.745
148.104
169.313
183.39


6585
O
O
GLU
879
60.902
148.997
169.168
183.39


6586
C
CB
GLU
879
63.83
147.686
167.996
183.39


6587
C
CG
GLU
879
65.239
148.107
167.639
183.39


6588
C
CD
GLU
879
66.27
147.395
168.481
183.39


6589
O
OE1
GLU
879
65.864
146.537
169.29
183.39


6590
O
OE2
GLU
879
67.476
147.69
168.336
183.39


6591
N
N
ARG
880
61.403
146.839
169.572
185.93


6592
C
CA
ARG
880
60.001
146.44
169.617
185.93


6593
C
C
ARG
880
59.271
147.022
170.821
185.93


6594
O
O
ARG
880
58.036
147.056
170.826
185.93


6595
C
CB
ARG
880
59.89
144.915
169.63
185.93


6596
C
CG
ARG
880
60.425
144.267
170.896
185.93


6597
C
CD
ARG
880
60.086
142.786
170.95
185.93


6598
N
NE
ARG
880
60.823
142.019
169.953
185.93


6599
C
CZ
ARG
880
61.944
141.356
170.2
185.93


6600
N
NH1
ARG
880
62.487
141.341
171.407
185.93


6601
N
NH2
ARG
880
62.536
140.691
169.212
185.93


6602
N
N
ILE
881
60.002
147.48
171.837
184.38


6603
C
CA
ILE
881
59.395
147.982
173.064
184.38


6604
C
C
ILE
881
59.237
149.498
173.041
184.38


6605
O
O
ILE
881
58.747
150.081
174.02
184.38


6606
C
CB
ILE
881
60.207
147.537
174.294
184.38


6607
C
CG1
ILE
881
59.359
147.587
175.56
184.38


6608
C
CG2
ILE
881
61.411
148.429
174.489
184.38


6609
C
CD1
ILE
881
60.04
146.974
176.745
184.38


6610
N
N
ARG
882
59.629
150.154
171.945
191.08


6611
C
CA
ARG
882
59.509
151.606
171.873
191.08


6612
C
C
ARG
882
58.056
152.046
171.982
191.08


6613
O
O
ARG
882
57.749
153.027
172.668
191.08


6614
C
CB
ARG
882
60.133
152.127
170.579
191.08


6615
C
CG
ARG
882
60.023
153.634
170.422
191.08


6616
C
CD
ARG
882
60.829
154.367
171.481
191.08


6617
N
NE
ARG
882
60.748
155.813
171.318
191.08


6618
C
CZ
ARG
882
59.785
156.57
171.826
191.08


6619
N
NH1
ARG
882
58.799
156.048
172.537
191.08


6620
N
NH2
ARG
882
59.811
157.883
171.615
191.08


6621
N
N
GLU
883
57.142
151.322
171.332
195.18


6622
C
CA
GLU
883
55.729
151.666
171.451
195.18


6623
C
C
GLU
883
55.254
151.546
172.893
195.18


6624
O
O
GLU
883
54.762
152.525
173.47
195.18


6625
C
CB
GLU
883
54.888
150.778
170.533
195.18


6626
C
CG
GLU
883
54.872
151.22
169.079
195.18


6627
C
CD
GLU
883
54.298
152.615
168.899
195.18


6628
O
OE1
GLU
883
53.367
152.981
169.649
195.18


6629
O
OE2
GLU
883
54.776
153.346
168.005
195.18


6630
N
N
LYS
884
55.476
150.385
173.517
193.06


6631
C
CA
LYS
884
54.968
150.179
174.869
193.06


6632
C
C
LYS
884
55.541
151.221
175.814
193.06


6633
O
O
LYS
884
54.833
151.728
176.689
193.06


6634
C
CB
LYS
884
55.293
148.769
175.368
193.06


6635
C
CG
LYS
884
54.702
148.458
176.751
193.06


6636
C
CD
LYS
884
55.659
148.736
177.908
193.06


6637
C
CE
LYS
884
56.787
147.732
177.97
193.06


6638
N
NZ
LYS
884
57.707
148.022
179.106
193.06


6639
N
N
LEU
885
56.815
151.576
175.634
189.96


6640
C
CA
LEU
885
57.387
152.659
176.423
189.96


6641
C
C
LEU
885
56.725
153.993
176.103
189.96


6642
O
O
LEU
885
56.601
154.844
176.986
189.96


6643
C
CB
LEU
885
58.895
152.737
176.194
189.96


6644
C
CG
LEU
885
59.693
151.539
176.713
189.96


6645
C
CD1
LEU
885
61.157
151.651
176.323
189.96


6646
C
CD2
LEU
885
59.541
151.4
178.219
189.96


6647
N
N
ALA
886
56.28
154.191
174.86
196.35


6648
C
CA
ALA
886
55.6
155.435
174.507
196.35


6649
C
C
ALA
886
54.281
155.585
175.261
196.35


6650
O
O
ALA
886
54.041
156.604
175.923
196.35


6651
C
CB
ALA
886
55.366
155.489
172.997
196.35


6652
N
N
GLU
887
53.413
154.57
175.183
200.88


6653
C
CA
GLU
887
52.174
154.671
175.962
200.88


6654
C
C
GLU
887
52.436
154.604
177.462
200.88


6655
O
O
GLU
887
51.659
155.163
178.245
200.88


6656
C
CB
GLU
887
51.114
153.628
175.573
200.88


6657
C
CG
GLU
887
50.495
153.769
174.174
200.88


6658
C
CD
GLU
887
51.31
153.163
173.067
200.88


6659
O
OE1
GLU
887
52.344
152.561
173.373
200.88


6660
O
OE2
GLU
887
50.911
153.286
171.89
200.88


6661
N
N
ASN
888
53.529
153.969
177.888
197.83


6662
C
CA
ASN
888
53.86
153.965
179.307
197.83


6663
C
C
ASN
888
54.207
155.371
179.781
197.83


6664
O
O
ASN
888
53.744
155.813
180.837
197.83


6665
C
CB
ASN
888
55.02
153.003
179.564
197.83


6666
C
CG
ASN
888
55.287
152.792
181.038
197.83


6667
O
OD1
ASN
888
54.481
153.169
181.885
197.83


6668
N
ND2
ASN
888
56.43
152.195
181.353
197.83


6669
N
N
ILE
889
55.005
156.097
178.996
199.93


6670
C
CA
ILE
889
55.34
157.476
179.333
199.93


6671
C
C
ILE
889
54.099
158.353
179.274
199.93


6672
O
O
ILE
889
53.953
159.294
180.059
199.93


6673
C
CB
ILE
889
56.457
157.999
178.412
199.93


6674
C
CG1
ILE
889
57.747
157.213
178.647
199.93


6675
C
CG2
ILE
889
56.689
159.485
178.641
199.93


6676
C
CD1
ILE
889
58.293
157.342
180.052
199.93


6677
N
N
HIS
890
53.182
158.06
178.349
207.04


6678
C
CA
HIS
890
51.899
158.759
178.347
207.04


6679
C
C
HIS
890
51.161
158.537
179.665
207.04


6680
O
O
HIS
890
50.592
159.474
180.245
207.04


6681
C
CB
HIS
890
51.058
158.29
177.155
207.04


6682
C
CG
HIS
890
49.668
158.852
177.119
207.04


6683
N
ND1
HIS
890
48.712
158.552
178.066
207.04


6684
C
CD2
HIS
890
49.073
159.694
176.241
207.04


6685
C
CE1
HIS
890
47.591
159.187
177.776
207.04


6686
N
NE2
HIS
890
47.783
159.887
176.673
207.04


6687
N
N
GLU
891
51.167
157.294
180.153
208.01


6688
C
CA
GLU
891
50.507
156.981
181.416
208.01


6689
C
C
GLU
891
51.155
157.721
182.581
208.01


6690
O
O
GLU
891
50.458
158.301
183.422
208.01


6691
C
CB
GLU
891
50.533
155.472
181.655
208.01


6692
C
CG
GLU
891
49.534
154.695
180.815
208.01


6693
C
CD
GLU
891
50.094
153.377
180.318
208.01


6694
O
OE1
GLU
891
50.853
152.731
181.071
208.01


6695
O
OE2
GLU
891
49.776
152.987
179.175
208.01


6696
N
N
LEU
892
52.49
157.707
182.651
203.06


6697
C
CA
LEU
892
53.17
158.47
183.698
203.06


6698
C
C
LEU
892
52.871
159.956
183.592
203.06


6699
O
O
LEU
892
52.673
160.623
184.61
203.06


6700
C
CB
LEU
892
54.684
158.24
183.688
203.06


6701
C
CG
LEU
892
55.304
156.969
184.278
203.06


6702
C
CD1
LEU
892
55.059
155.738
183.465
203.06


6703
C
CD2
LEU
892
56.799
157.182
184.472
203.06


6704
N
N
TRP
893
52.837
160.497
182.375
209.79


6705
C
CA
TRP
893
52.554
161.917
182.208
209.79


6706
C
C
TRP
893
51.174
162.266
182.748
209.79


6707
O
O
TRP
893
51.022
163.219
183.523
209.79


6708
C
CB
TRP
893
52.683
162.297
180.732
209.79


6709
C
CG
TRP
893
52.582
163.767
180.461
209.79


6710
C
CD1
TRP
893
53.571
164.695
180.607
209.79


6711
C
CD2
TRP
893
51.441
164.473
179.958
209.79


6712
N
NE1
TRP
893
53.111
165.939
180.246
209.79


6713
C
CE2
TRP
893
51.807
165.829
179.842
209.79


6714
C
CE3
TRP
893
50.143
164.092
179.603
209.79


6715
C
CZ2
TRP
893
50.922
166.805
179.386
209.79


6716
C
CZ3
TRP
893
49.266
165.062
179.151
209.79


6717
C
CH2
TRP
893
49.66
166.402
179.047
209.79


6718
N
N
VAL
894
50.159
161.477
182.386
212.83


6719
C
CA
VAL
894
48.805
161.806
182.824
212.83


6720
C
C
VAL
894
48.652
161.6
184.331
212.83


6721
O
O
VAL
894
48.038
162.427
185.019
212.83


6722
C
CB
VAL
894
47.755
161.019
182.013
212.83


6723
C
CG1
VAL
894
47.885
161.341
180.532
212.83


6724
C
CG2
VAL
894
47.874
159.522
182.24
212.83


6725
N
N
MET
895
49.229
160.522
184.881
215.43


6726
C
CA
MET
895
49.084
160.301
186.318
215.43


6727
C
C
MET
895
49.857
161.345
187.115
215.43


6728
O
O
MET
895
49.384
161.811
188.155
215.43


6729
C
CB
MET
895
49.523
158.887
186.714
215.43


6730
C
CG
MET
895
50.991
158.561
186.505
215.43


6731
S
SD
MET
895
51.393
156.846
186.907
215.43


6732
C
CE
MET
895
50.666
155.964
185.529
215.43


6733
N
N
ASN
896
51.043
161.731
186.646
214.43


6734
C
CA
ASN
896
51.798
162.779
187.314
214.43


6735
C
C
ASN
896
51.031
164.092
187.283
214.43


6736
O
O
ASN
896
50.93
164.792
188.297
214.43


6737
C
CB
ASN
896
53.164
162.927
186.645
214.43


6738
C
CG
ASN
896
54.117
163.786
187.441
214.43


6739
O
OD1
ASN
896
53.779
164.292
188.511
214.43


6740
N
ND2
ASN
896
55.322
163.958
186.919
214.43


6741
N
N
LYS
897
50.449
164.425
186.128
216.99


6742
C
CA
LYS
897
49.693
165.665
186.016
216.99


6743
C
C
LYS
897
48.476
165.661
186.933
216.99


6744
O
O
LYS
897
48.125
166.702
187.5
216.99


6745
C
CB
LYS
897
49.271
165.886
184.562
216.99


6746
C
CG
LYS
897
48.513
167.181
184.316
216.99


6747
C
CD
LYS
897
49.39
168.391
184.616
216.99


6748
C
CE
LYS
897
50.505
168.529
183.584
216.99


6749
N
NZ
LYS
897
51.357
169.737
183.8
216.99


6750
N
N
ILE
898
47.825
164.508
187.1
222.49


6751
C
CA
ILE
898
46.595
164.478
187.887
222.49


6752
C
C
ILE
898
46.879
164.37
189.387
222.49


6753
O
O
ILE
898
46.108
164.888
190.202
222.49


6754
C
CB
ILE
898
45.667
163.35
187.395
222.49


6755
C
CG1
ILE
898
44.258
163.532
187.964
222.49


6756
C
CG2
ILE
898
46.215
161.989
187.762
222.49


6757
C
CD1
ILE
898
43.499
164.69
187.355
222.49


6758
N
N
GLU
899
47.971
163.716
189.789
222.71


6759
C
CA
GLU
899
48.315
163.636
191.206
222.71


6760
C
C
GLU
899
49.36
164.657
191.643
222.71


6761
O
O
GLU
899
49.868
164.552
192.764
222.71


6762
C
CB
GLU
899
48.784
162.225
191.596
222.71


6763
C
CG
GLU
899
47.652
161.236
191.908
222.71


6764
C
CD
GLU
899
47.263
160.34
190.75
222.71


6765
O
OE1
GLU
899
47.985
160.316
189.738
222.71


6766
O
OE2
GLU
899
46.225
159.653
190.855
222.71


6767
N
N
LEU
900
49.699
165.635
190.801
223.05


6768
C
CA
LEU
900
50.414
166.802
191.306
223.05


6769
C
C
LEU
900
49.497
167.783
192.023
223.05


6770
O
O
LEU
900
49.971
168.829
192.48
223.05


6771
C
CB
LEU
900
51.148
167.529
190.175
223.05


6772
C
CG
LEU
900
52.518
166.988
189.763
223.05


6773
C
CD1
LEU
900
53.037
167.72
188.535
223.05


6774
C
CD2
LEU
900
53.506
167.097
190.914
223.05


6775
N
N
GLY
901
48.206
167.473
192.133
227.61


6776
C
CA
GLY
901
47.242
168.346
192.765
227.61


6777
C
C
GLY
901
46.22
168.948
191.827
227.61


6778
O
O
GLY
901
45.286
169.607
192.299
227.61


6779
N
N
TRP
902
46.362
168.743
190.522
230.04


6780
C
CA
TRP
902
45.421
169.285
189.556
230.04


6781
C
C
TRP
902
44.301
168.285
189.281
230.04


6782
O
O
TRP
902
44.419
167.087
189.552
230.04


6783
C
CB
TRP
902
46.132
169.656
188.254
230.04


6784
C
CG
TRP
902
47.177
170.725
188.413
230.04


6785
C
CD1
TRP
902
48.254
170.704
189.252
230.04


6786
C
CD2
TRP
902
47.233
171.978
187.721
230.04


6787
N
NE1
TRP
902
48.98
171.863
189.12
230.04


6788
C
CE2
TRP
902
48.374
172.662
188.187
230.04


6789
C
CE3
TRP
902
46.432
172.587
186.75
230.04


6790
C
CZ2
TRP
902
48.732
173.923
187.716
230.04


6791
C
CZ3
TRP
902
46.789
173.839
186.285
230.04


6792
C
CH2
TRP
902
47.929
174.493
186.767
230.04


6793
N
N
GLN
903
43.201
168.796
188.732
235.31


6794
C
CA
GLN
903
42.032
167.975
188.457
235.31


6795
C
C
GLN
903
41.333
168.498
187.211
235.31


6796
O
O
GLN
903
41.534
169.641
186.792
235.31


6797
C
CB
GLN
903
41.072
167.95
189.652
235.31


6798
C
CG
GLN
903
40.422
169.29
189.954
235.31


6799
C
CD
GLN
903
41.228
170.119
190.936
235.31


6800
O
OE1
GLN
903
42.405
169.848
191.175
235.31


6801
N
NE2
GLN
903
40.597
171.136
191.51
235.31


6802
N
N
TYR
904
40.508
167.636
186.62
235.11


6803
C
CA
TYR
904
39.791
167.988
185.401
235.11


6804
C
C
TYR
904
38.795
169.111
185.666
235.11


6805
O
O
TYR
904
38.073
169.1
186.667
235.11


6806
C
CB
TYR
904
39.07
166.757
184.85
235.11


6807
C
CG
TYR
904
38.151
167.042
183.686
235.11


6808
C
CD1
TYR
904
38.642
167.571
182.5
235.11


6809
C
CD2
TYR
904
36.79
166.778
183.772
235.11


6810
C
CE1
TYR
904
37.804
167.83
181.433
235.11


6811
C
CE2
TYR
904
35.944
167.034
182.711
235.11


6812
C
CZ
TYR
904
36.456
167.56
181.544
235.11


6813
O
OH
TYR
904
35.616
167.817
180.484
235.11


6814
N
N
GLY
905
38.751
170.08
184.753
235.87


6815
C
CA
GLY
905
37.875
171.219
184.896
235.87


6816
C
C
GLY
905
37.31
171.703
183.576
235.87


6817
O
O
GLY
905
37.768
171.317
182.497
235.87


6818
N
N
PRO
906
36.276
172.548
183.641
235.54


6819
C
CA
PRO
906
35.668
173.063
182.404
235.54


6820
C
C
PRO
906
36.564
174.016
181.628
235.54


6821
O
O
PRO
906
36.775
173.828
180.425
235.54


6822
C
CB
PRO
906
34.403
173.775
182.907
235.54


6823
C
CG
PRO
906
34.17
173.238
184.291
235.54


6824
C
CD
PRO
906
35.533
172.97
184.837
235.54


6825
N
N
VAL
907
37.1
175.038
182.295
233.89


6826
C
CA
VAL
907
37.833
176.112
181.637
233.89


6827
C
C
VAL
907
39.171
176.297
182.344
233.89


6828
O
O
VAL
907
39.31
176.005
183.535
233.89


6829
C
CB
VAL
907
37.018
177.428
181.626
233.89


6830
C
CG1
VAL
907
36.736
177.904
183.046
233.89


6831
C
CG2
VAL
907
37.704
178.511
180.796
233.89


6832
N
N
ARG
908
40.16
176.782
181.595
230.2


6833
C
CA
ARG
908
41.526
176.928
182.088
230.2


6834
C
C
ARG
908
41.692
178.304
182.723
230.2


6835
O
O
ARG
908
41.503
179.329
182.061
230.2


6836
C
CB
ARG
908
42.526
176.719
180.951
230.2


6837
C
CG
ARG
908
43.994
176.7
181.369
230.2


6838
C
CD
ARG
908
44.632
178.082
181.28
230.2


6839
N
NE
ARG
908
46.068
178.042
181.531
230.2


6840
C
CZ
ARG
908
46.831
179.115
181.687
230.2


6841
N
NH1
ARG
908
46.327
180.337
181.622
230.2


6842
N
NH2
ARG
908
48.132
178.959
181.913
230.2


6843
N
N
ASP
909
42.053
178.32
184.005
233.34


6844
C
CA
ASP
909
42.372
179.543
184.725
233.34


6845
C
C
ASF
909
43.584
179.297
185.61
233.34


6846
O
O
ASF
909
43.808
178.182
186.088
233.34


6847
C
CB
ASP
909
41.189
180.032
185.574
233.34


6848
C
CG
ASP
909
40.093
180.666
184.741
233.34


6849
O
OD1
ASF
909
40.415
181.309
183.719
233.34


6850
O
OD2
ASP
909
38.907
180.524
185.108
233.34


6851
N
N
ASP
910
44.371
180.355
185.823
233.95


6852
C
CA
ASP
910
45.572
180.224
186.641
233.95


6853
C
C
ASP
910
45.226
180.019
188.111
233.95


6854
O
O
ASP
910
45.833
179.181
188.789
233.95


6855
C
CB
ASP
910
46.467
181.453
186.461
233.95


6856
C
CG
ASP
910
45.744
182.754
186.76
233.95


6857
O
OD1
ASP
910
44.517
182.72
186.991
233.95


6858
O
OD2
ASP
910
46.406
183.813
186.764
233.95


6859
N
N
ASN
911
44.252
180.774
188.623
234.55


6860
C
CA
ASN
911
43.873
180.647
190.027
234.55


6861
C
C
ASN
911
43.117
179.353
190.293
234.55


6862
O
O
ASN
911
43.117
178.858
191.426
234.55


6863
C
CB
ASN
911
43.028
181.847
190.454
234.55


6864
C
CG
ASN
911
41.801
182.034
189.584
234.55


6865
O
OD1
ASN
911
40.746
181.458
189.846
234.55


6866
N
ND2
ASN
911
41.935
182.842
188.539
234.55


6867
N
N
LYS
912
42.467
178.795
189.269
233.92


6868
C
CA
LYS
912
41.677
177.584
189.463
233.92


6869
C
C
LYS
912
42.553
176.378
189.775
233.92


6870
O
O
LYS
912
42.105
175.455
190.465
233.92


6871
C
CB
LYS
912
40.823
177.314
188.225
233.92


6872
C
CG
LYS
912
39.411
177.867
188.312
233.92


6873
C
CD
LYS
912
38.652
177.644
187.014
233.92


6874
C
CE
LYS
912
38.12
176.224
186.927
233.92


6875
N
NZ
LYS
912
37.586
175.908
185.574
233.92


6876
N
N
ARG
913
43.794
176.369
189.281
232.94


6877
C
CA
ARG
913
44.713
175.243
189.459
232.94


6878
C
C
ARG
913
44.093
173.941
188.958
232.94


6879
O
O
ARG
913
44.263
172.878
189.558
232.94


6880
C
CB
ARG
913
45.153
175.108
190.919
232.94


6881
C
CG
ARG
913
45.959
176.286
191.447
232.94


6882
C
CD
ARG
913
47.363
176.318
190.864
232.94


6883
N
NE
ARG
913
47.455
177.187
189.697
232.94


6884
C
CZ
ARG
913
48.551
177.349
188.968
232.94


6885
N
NH1
ARG
913
49.674
176.713
189.258
232.94


6886
N
NH2
ARG
913
48.52
178.171
187.923
232.94


6887
N
N
GLN
914
43.367
174.026
187.846
235.52


6888
C
CA
GLN
914
42.646
172.897
187.279
235.52


6889
C
C
GLN
914
43.173
172.618
185.879
235.52


6890
O
O
GLN
914
43.654
173.527
185.195
235.52


6891
C
CB
GLN
914
41.14
173.176
187.24
235.52


6892
C
CG
GLN
914
40.447
172.967
188.579
235.52


6893
C
CD
GLN
914
38.99
172.579
188.436
235.52


6894
O
OE1
GLN
914
38.671
171.47
188.013
235.52


6895
N
NE2
GLN
914
38.096
173.493
188.794
235.52


6896
N
N
HIS
915
43.088
171.359
185.459
235.61


6897
C
CA
HIS
915
43.644
170.927
184.178
235.61


6898
C
C
HIS
915
42.519
170.492
183.25
235.61


6899
O
O
HIS
915
42.106
169.318
183.286
235.61


6900
C
CB
HIS
915
44.641
169.788
184.388
235.61


6901
C
CG
HIS
915
45.301
169.318
183.13
235.61


6902
N
ND1
HIS
915
46.278
170.047
182.486
235.61


6903
C
CD2
HIS
915
45.134
168.189
182.402
235.61


6904
C
CE1
HIS
915
46.68
169.39
181.413
235.61


6905
N
NE2
HIS
915
46.002
168.258
181.34
235.61


6906
N
N
PRO
916
41.991
171.387
182.412
234.91


6907
C
CA
PRO
916
40.875
171.003
181.533
234.91


6908
C
C
PRO
916
41.28
170.113
180.373
234.91


6909
O
O
PRO
916
40.4
169.579
179.687
234.91


6910
C
CB
PRO
916
40.342
172.353
181.028
234.91


6911
C
CG
PRO
916
40.909
173.378
181.952
234.91


6912
C
CD
PRO
916
42.22
172.839
182.416
234.91


6913
N
N
CYS
917
42.577
169.931
180.129
233.71


6914
C
CA
CYS
917
43.041
169.258
178.922
233.71


6915
C
C
CYS
917
42.78
167.756
178.927
233.71


6916
O
O
CYS
917
43.107
167.089
177.939
233.71


6917
C
CB
CYS
917
44.533
169.524
178.713
233.71


6918
S
SG
CYS
917
45.001
171.266
178.842
233.71


6919
N
N
LEU
918
42.206
167.205
179.997
230.57


6920
C
CA
LEU
918
41.789
165.803
180.008
230.57


6921
C
C
LEU
918
40.485
165.67
179.22
230.57


6922
O
O
LEU
918
39.4
165.453
179.762
230.57


6923
C
CB
LEU
918
41.641
165.292
181.435
230.57


6924
C
CG
LEU
918
42.922
164.777
182.094
230.57


6925
C
CD1
LEU
918
42.815
164.841
183.609
230.57


6926
C
CD2
LEU
918
43.231
163.36
181.631
230.57


6927
N
N
VAL
919
40.616
165.806
177.902
231.33


6928
C
CA
VAL
919
39.484
165.813
176.985
231.33


6929
C
C
VAL
919
39.901
165.04
175.738
231.33


6930
O
O
VAL
919
41.029
164.542
175.655
231.33


6931
C
CB
VAL
919
39.045
167.255
176.651
231.33


6932
C
CG1
VAL
919
40.035
167.92
175.698
231.33


6933
C
CG2
VAL
919
37.606
167.307
176.126
231.33


6934
N
N
GLU
920
38.993
164.921
174.772
229.76


6935
C
CA
GLU
920
39.283
164.206
173.538
229.76


6936
C
C
GLU
920
40.49
164.816
172.827
229.76


6937
O
O
GLU
920
40.868
165.969
173.055
229.76


6938
C
CB
GLU
920
38.064
164.217
172.616
229.76


6939
C
CG
GLU
920
37.089
163.078
172.865
229.76


6940
C
CD
GLU
920
36.013
163.441
173.871
229.76


6941
O
OE1
GLU
920
35.931
164.626
174.254
229.76


6942
O
OE2
GLU
920
35.25
162.54
174.277
229.76


6943
N
N
PHE
921
41.092
164.019
171.941
228.6


6944
C
CA
PHE
921
42.341
164.415
171.298
228.6


6945
C
C
PHE
921
42.148
165.524
170.271
228.6


6946
O
O
PHE
921
43.121
165.937
169.63
228.6


6947
C
CB
PHE
921
42.996
163.196
170.642
228.6


6948
C
CG
PHE
921
42.176
162.584
169.538
228.6


6949
C
CD1
PHE
921
41.252
161.589
169.814
228.6


6950
C
CD2
PHE
921
42.34
162.991
168.223
228.6


6951
C
CE1
PHE
921
40.499
161.021
168.803
228.6


6952
C
CE2
PHE
921
41.59
162.427
167.208
228.6


6953
C
CZ
PHE
921
40.669
161.44
167.498
228.6


6954
N
N
SER
922
40.916
166.005
170.087
230.33


6955
C
CA
SER
922
40.666
167.048
169.096
230.33


6956
C
C
SER
922
41.44
168.32
169.42
230.33


6957
O
O
SER
922
42.179
168.842
168.576
230.33


6958
C
CB
SER
922
39.167
167.339
169.012
230.33


6959
O
OG
SER
922
38.704
167.963
170.197
230.33


6960
N
N
LYS
923
41.287
168.834
170.64
226.95


6961
C
CA
LYS
923
41.992
170.041
171.052
226.95


6962
C
C
LYS
923
43.325
169.735
171.724
226.95


6963
O
O
LYS
923
44.217
170.591
171.713
226.95


6964
C
CB
LYS
923
41.105
170.872
171.993
226.95


6965
C
CG
LYS
923
41.692
172.216
172.444
226.95


6966
C
CD
LYS
923
42.425
172.136
173.78
226.95


6967
C
CE
LYS
923
41.471
171.932
174.942
226.95


6968
N
NZ
LYS
923
42.204
171.87
176.237
226.95


6969
N
N
LEU
924
43.482
168.535
172.282
220.74


6970
C
CA
LEU
924
44.664
168.213
173.066
220.74


6971
C
C
LEU
924
45.923
168.412
172.224
220.74


6972
O
O
LEU
924
46.016
167.902
171.103
220.74


6973
C
CB
LEU
924
44.589
166.775
173.578
220.74


6974
C
CG
LEU
924
45.496
166.367
174.744
220.74


6975
C
CD1
LEU
924
44.856
165.23
175.527
220.74


6976
C
CD2
LEU
924
46.888
165.968
174.276
220.74


6977
N
N
PRO
925
46.898
169.163
172.735
213.7


6978
C
CA
PRO
925
48.038
169.569
171.902
213.7


6979
C
C
PRO
925
48.844
168.385
171.388
213.7


6980
O
O
PRO
925
49.502
167.668
172.152
213.7


6981
C
CB
PRO
925
48.866
170.444
172.848
213.7


6982
C
CG
PRO
925
47.902
170.94
173.865
213.7


6983
C
CD
PRO
925
46.867
169.866
174.029
213.7


6984
N
N
GLU
926
48.8
168.192
170.068
209.72


6985
C
CA
GLU
926
49.663
167.2
169.441
209.72


6986
C
C
GLU
926
51.127
167.583
169.595
209.72


6987
O
O
GLU
926
51.997
166.71
169.684
209.72


6988
C
CB
GLU
926
49.298
167.038
167.966
209.72


6989
C
CG
GLU
926
47.983
166.311
167.73
209.72


6990
C
CD
GLU
926
48.009
164.874
168.216
209.72


6991
O
OE1
GLU
926
49.089
164.247
168.171
209.72


6992
O
OE2
GLU
926
46.95
164.372
168.645
209.72


6993
N
N
GLN
927
51.419
168.886
169.64
205.13


6994
C
CA
GLN
927
52.781
169.315
169.93
205.13


6995
C
C
GLN
927
53.181
168.933
171.35
205.13


6996
O
O
GLN
927
54.337
168.583
171.599
205.13


6997
C
CB
GLN
927
52.931
170.822
169.7
205.13


6998
C
CG
GLN
927
52.268
171.714
170.738
205.13


6999
C
CD
GLN
927
50.794
171.939
170.466
205.13


7000
O
OE1
GLN
927
50.169
171.198
169.708
205.13


7001
N
NE2
GLN
927
50.23
172.968
171.087
205.13


7002
N
N
GLU
928
52.234
168.968
172.293
201.87


7003
C
CA
GLU
928
52.537
168.507
173.645
201.87


7004
C
C
GLU
928
52.755
167
173.68
201.87


7005
O
O
GLU
928
53.627
166.512
174.407
201.87


7006
C
CB
GLU
928
51.426
168.91
174.614
201.87


7007
C
CG
GLU
928
51.602
168.359
176.023
201.87


7008
C
CD
GLU
928
52.903
168.792
176.676
201.87


7009
O
OE1
GLU
928
53.428
169.87
176.325
201.87


7010
O
OE2
GLU
928
53.409
168.042
177.537
201.87


7011
N
N
ARG
929
51.975
166.245
172.902
202.57


7012
C
CA
ARG
929
52.229
164.809
172.799
202.57


7013
C
C
ARG
929
53.623
164.541
172.242
202.57


7014
O
O
ARG
929
54.364
163.695
172.764
202.57


7015
C
CB
ARG
929
51.173
164.147
171.913
202.57


7016
C
CG
ARG
929
49.765
164.139
172.48
202.57


7017
C
CD
ARG
929
48.78
163.639
171.433
202.57


7018
N
NE
ARG
929
47.394
163.733
171.875
202.57


7019
C
CZ
ARG
929
46.793
162.842
172.652
202.57


7020
N
NH1
ARG
929
47.43
161.771
173.096
202.57


7021
N
NH2
ARG
929
45.521
163.03
172.99
202.57


7022
N
N
ASN
930
53.997
165.264
171.185
198.21


7023
C
CA
ASN
930
55.319
165.094
170.593
198.21


7024
C
C
ASN
930
56.41
165.481
171.58
198.21


7025
O
O
ASN
930
57.456
164.829
171.647
198.21


7026
C
CB
ASN
930
55.429
165.918
169.311
198.21


7027
C
CG
ASN
930
54.574
165.366
168.188
198.21


7028
O
OD1
ASN
930
54.508
164.154
167.981
198.21


7029
N
ND2
ASN
930
53.913
166.254
167.455
198.21


7030
N
N
TYR
931
56.183
166.543
172.357
193.04


7031
C
CA
TYR
931
57.146
166.944
173.377
193.04


7032
C
C
TYR
931
57.293
165.87
174.445
193.04


7033
O
O
TYR
931
58.405
165.593
174.907
193.04


7034
C
CB
TYR
931
56.727
168.273
174.007
193.04


7035
C
CG
TYR
931
56.819
169.458
173.072
193.04


7036
C
CD1
TYR
931
57.525
169.375
171.878
193.04


7037
C
CD2
TYR
931
56.197
170.66
173.382
193.04


7038
C
CE1
TYR
931
57.609
170.457
171.021
193.04


7039
C
CE2
TYR
931
56.276
171.746
172.532
193.04


7040
C
CZ
TYR
931
56.983
171.639
171.353
193.04


7041
O
OH
TYR
931
57.064
172.719
170.504
193.04


7042
N
N
ASN
932
56.181
165.254
174.848
191.16


7043
C
CA
ASN
932
56.241
164.183
175.835
191.16


7044
C
C
ASN
932
57.069
163.015
175.314
191.16


7045
O
O
ASN
932
57.976
162.519
176
191.16


7046
C
CB
ASN
932
54.823
163.72
176.175
191.16


7047
C
CG
ASN
932
54.041
164.757
176.957
191.16


7048
O
OD
ASN
932
54.558
165.377
177.882
191.16


7049
N
ND2
ASN
932
52.783
164.954
176.581
191.16


7050
N
N
LEU
933
56.787
162.585
174.082
180.56


7051
C
CA
LEU
933
57.53
161.471
173.498
180.56


7052
C
C
LEU
933
59.006
161.808
173.33
180.56


7053
O
O
LEU
933
59.882
160.981
173.623
180.56


7054
C
CB
LEU
933
56.913
161.067
172.158
180.56


7055
C
CG
LEU
933
55.706
160.126
172.18
180.56


7056
C
CD1
LEU
933
56.168
158.816
172.759
180.56


7057
C
CD2
LEU
933
54.519
160.65
172.968
180.56


7058
N
N
GLN
934
59.308
163.024
172.868
179.21


7059
C
CA
GLN
934
60.7
163.37
172.635
179.21


7060
C
C
GLN
934
61.462
163.544
173.937
179.21


7061
O
O
GLN
934
62.645
163.205
173.989
179.21


7062
C
CB
GLN
934
60.825
164.629
171.767
179.21


7063
C
CG
GLN
934
60.288
165.909
172.371
179.21


7064
C
CD
GLN
934
61.29
166.602
173.274
179.21


7065

OE1
GLN
934
62.501
166.494
173.076
179.21


7066
N
NE2
GLN
934
60.79
167.313
174.278
179.21


7067
N
N
MET
935
60.82
164.031
175.004
175.62


7068
C
CA
MET
935
61.551
164.126
176.261
175.62


7069
C
C
MET
935
61.738
162.749
176.876
175.62


7070
O
O
MET
935
62.745
162.505
177.541
175.62


7071
C
CB
MET
935
60.859
165.051
177.264
175.62


7072
C
CG
MET
935
59.511
164.591
177.775
175.62


7073
S
SD
MET
935
58.81
165.808
178.905
175.62


7074
C
CE
MET
935
57.258
165.028
179.327
175.62


7075
N
N
SER
936
60.8
161.825
176.646
166.44


7076
C
CA
SER
936
61.014
160.454
177.104
166.44


7077
C
C
SER
936
62.206
159.82
176.395
166.44


7078
O
O
SER
936
63.08
159.212
177.034
166.44


7079
C
CB
SER
936
59.755
159.622
176.876
166.44


7080
O
OG
SER
936
59.358
159.662
175.517
166.44


7081
N
N
LEU
937
62.261
159.954
175.067
164.45


7082
C
CA
LEU
937
63.375
159.361
174.334
164.45


7083
C
C
LEU
937
64.684
160.072
174.656
164.45


7084
O
O
LEU
937
65.739
159.433
174.727
164.45


7085
C
CB
LEU
937
63.094
159.362
172.828
164.45


7086
C
CG
LEU
937
62.884
160.648
172.027
164.45


7087
C
CD1
LEU
937
64.201
161.279
171.593
164.45


7088
C
CD2
LEU
937
61.997
160.372
170.821
164.45


7089
N
N
GLU
938
64.636
161.386
174.883
162.4


7090
C
CA
GLU
938
65.834
162.11
175.29
162.4


7091
C
C
GLU
938
66.281
161.678
176.677
162.4


7092
O
O
GLU
938
67.481
161.586
176.948
162.4


7093
C
CB
GLU
938
65.576
163.616
175.252
162.4


7094
C
CG
GLU
938
66.817
164.471
175.456
162.4


7095
C
CD
GLU
938
67.719
164.486
174.238
162.4


7096
O
OE1
GLU
938
67.209
164.268
173.119
162.4


7097
O
OE2
GLU
938
68.935
164.724
174.397
162.4


7098
N
N
THR
939
65.33
161.387
177.564
153.44


7099
C
CA
THR
939
65.675
160.889
178.887
153.44


7100
C
C
THR
939
66.402
159.556
178.789
153.44


7101
O
O
THR
939
67.472
159.375
179.382
153.44


7102
C
CB
THR
939
64.41
160.753
179.735
153.44


7103
O
OG1
THR
939
63.783
162.035
179.874
153.44


7104
C
CG2
THR
939
64.747
160.203
181.114
153.44


7105
N
N
LEU
940
65.847
158.617
178.017
151.97


7106
C
CA
LEU
940
66.495
157.311
177.896
151.97


7107
C
C
LEU
940
67.848
157.431
177.199
151.97


7108
O
O
LEU
940
68.812
156.741
177.565
151.97


7109
C
CB
LEU
940
65.574
156.321
177.178
151.97


7110
C
CG
LEU
940
65.088
156.531
175.741
151.97


7111
C
CD1
LEU
940
66.103
156.041
174.709
151.97


7112
C
CD2
LEU
940
63.741
155.854
175.534
151.97


7113
N
N
LYS
941
67.949
158.325
176.213
150.11


7114
C
CA
LYS
941
69.225
158.537
175.544
150.11


7115
C
C
LYS
941
70.244
159.137
176.505
150.11


7116
O
O
LYS
941
71.435
158.822
176.438
150.11


7117
C
CB
LYS
941
69.024
159.427
174.318
150.11


7118
C
CG
LYS
941
68.431
158.67
173.133
150.11


7119
C
CD
LYS
941
68.604
159.391
171.805
150.11


7120
C
CE
LYS
941
68.539
158.397
170.655
150.11


7121
N
NZ
LYS
941
69.379
158.785
169.493
150.11


7122
N
N
THR
942
69.789
159.986
177.427
148.53


7123
C
CA
THR
942
70.706
160.557
178.405
148.53


7124
C
C
THR
942
71.146
159.52
179.432
148.53


7125
O
O
THR
942
72.307
159.529
179.855
148.53


7126
C
CB
THR
942
70.07
161.764
179.092
148.53


7127
O
OG1
THR
942
68.736
161.441
179.496
148.53


7128
C
CG2
THR
942
70.044
162.955
178.148
148.53


7129
N
N
LEU
943
70.248
158.617
179.852
146.13


7130
C
CA
LEU
943
70.712
157.55
180.74
146.13


7131
C
C
LEU
943
71.731
156.66
180.04
146.13


7132
O
O
LEU
943
72.731
156.268
180.649
146.13


7133
C
CB
LEU
943
69.586
156.683
181.33
146.13


7134
C
CG
LEU
943
68.343
157.119
182.127
146.13


7135
C
CD1
LEU
943
67.074
157.251
181.308
146.13


7136
C
CD2
LEU
943
68.108
156.174
183.3
146.13


7137
N
N
LEU
944
71.506
156.326
178.764
149.98


7138
C
CA
LEU
944
72.514
155.516
178.079
149.98


7139
C
C
LEU
944
73.812
156.298
177.9
149.98


7140
O
O
LEU
944
74.902
155.716
177.929
149.98


7141
C
CB
LEU
944
72.002
154.996
176.731
149.98


7142
C
CG
LEU
944
71.582
155.887
175.561
149.98


7143
C
CD1
LEU
944
72.779
156.319
174.722
149.98


7144
C
CD2
LEU
944
70.564
155.176
174.689
149.98


7145
N
N
ALA
945
73.716
157.619
177.718
144.56


7146
C
CA
ALA
945
74.915
158.444
177.611
144.56


7147
C
C
ALA
945
75.675
158.502
178.928
144.56


7148
O
O
ALA
945
76.904
158.635
178.93
144.56


7149
C
CB
ALA
945
74.546
159.853
177.149
144.56


7150
N
N
LEU
946
74.967
158.412
180.055
142.66


7151
C
CA
LEU
946
75.631
158.369
181.353
142.66


7152
C
C
LEU
946
76.406
157.076
181.558
142.66


7153
O
O
LEU
946
77.169
156.971
182.525
142.66


7154
C
CB
LEU
946
74.611
158.544
182.48
142.66


7155
C
CG
LEU
946
73.894
159.891
182.583
142.66


7156
C
CD1
LEU
946
72.822
159.846
183.662
142.66


7157
C
CD2
LEU
946
74.888
161.008
182.859
142.66


7158
N
N
GLY
947
76.225
156.095
180.68
145.9


7159
C
CA
GLY
947
76.848
154.798
180.823
145.9


7160
C
C
GLY
947
75.933
153.707
181.324
145.9


7161
O
O
GLY
947
76.407
152.595
181.581
145.9


7162
N
N
CYS
948
74.639
153.986
181.467
150.29


7163
C
CA
CYS
948
73.682
153.02
181.997
150.29


7164
C
C
CYS
948
73.35
152.002
180.914
150.29


7165
O
O
CYS
948
72.482
152.229
180.068
150.29


7166
C
CB
CYS
948
72.418
153.726
182.477
150.29


7167
S
SG
CYS
948
72.668
154.946
183.782
150.29


7168
N
N
HIS
949
74.045
150.865
180.933
150.01


7169
C
CA
HIS
949
73.691
149.758
180.044
150.01


7170
C
C
HIS
949
72.556
148.957
180.689
150.01


7171
O
O
HIS
949
72.731
147.88
181.262
150.01


7172
C
CB
HIS
949
74.923
148.925
179.699
150.01


7173
C
CG
HIS
949
75.55
148.217
180.861
150.01


7174
N
ND1
HIS
949
76.4
148.847
181.744
150.01


7175
C
CD2
HIS
949
75.501
146.92
181.249
150.01


7176
C
CE1
HIS
949
76.82
147.978
182.645
150.01


7177
N
NE2
HIS
949
76.29
146.8
182.367
150.01


7178
N
N
VAL
950
71.357
149.539
180.582
154.38


7179
C
CA
VAL
950
70.18
149.02
181.267
154.38


7180
C
C
VAL
950
69.908
147.582
180.844
154.38


7181
O
O
VAL
950
70.033
147.218
179.668
154.38


7182
C
CB
VAL
950
68.968
149.921
180.973
154.38


7183
C
CG1
VAL
950
67.704
149.365
181.613
154.38


7184
C
CG2
VAL
950
69.238
151.342
181.438
154.38


7185
N
N
GLY
951
69.528
146.753
181.816
156.18


7186
C
CA
GLY
951
69.304
145.346
181.563
156.18


7187
C
C
GLY
951
67.87
144.943
181.847
156.18


7188
O
O
GLY
951
67.223
145.451
182.767
156.18


7189
N
N
ILE
952
67.381
144.014
181.033
165.46


7190
C
CA
ILE
952
66.071
143.408
181.244
165.46


7191
C
C
ILE
952
66.207
142.302
182.284
165.46


7192
O
O
ILE
952
67.027
141.388
182.137
165.46


7193
C
CB
ILE
952
65.486
142.89
179.916
165.46


7194
C
CG1
ILE
952
64.035
142.435
180.096
165.46


7195
C
CG2
ILE
952
66.367
141.812
179.275
165.46


7196
C
CD1
ILE
952
63.223
142.473
178.818
165.46


7197
N
N
SER
953
65.44
142.41
183.37
173.51


7198
C
CA
SER
953
65.581
141.436
184.447
173.51


7199
C
C
SER
953
64.59
140.286
184.299
173.51


7200
O
O
SER
953
64.984
139.149
184.026
173.51


7201
C
CB
SER
953
65.404
142.129
185.8
173.51


7202
O
OG
SER
953
64.165
142.815
185.862
173.51


7203
N
N
ASP
954
63.295
140.585
184.403
186.74


7204
C
CA
ASF
954
62.238
139.576
184.433
186.74


7205
C
C
ASP
954
60.941
140.191
183.933
186.74


7206
O
O
ASP
954
60.655
141.36
184.209
186.74


7207
C
CB
ASP
954
62.028
139.009
185.848
186.74


7208
C
CG
ASF
954
63.162
138.106
186.31
186.74


7209
O
OD1
ASP
954
63.788
137.436
185.466
186.74


7210
O
OD2
ASP
954
63.429
138.07
187.529
186.74


7211
N
N
GLU
955
60.156
139.396
183.202
201.71


7212
C
CA
GLU
955
58.81
139.803
182.815
201.71


7213
C
C
GLU
955
57.856
139.846
184.004
201.71


7214
C
O
GLU
955
56.788
140.462
183.91
201.71


7215
C
CB
GLU
955
58.27
138.853
181.741
201.71


7216
C
CG
GLU
955
57.007
139.326
181.032
201.71


7217
C
CD
GLU
955
55.742
138.769
181.659
201.71


7218
O
OE1
GLU
955
55.756
137.597
182.089
201.71


7219
C
OE2
GLU
955
54.735
139.505
181.725
201.71


7220
N
N
HIS
956
58.231
139.231
185.128
209.87


7221
C
CA
HIS
956
57.313
139.029
186.243
209.87


7222
C
C
HIS
956
56.89
140.322
186.932
209.87


7223
O
O
HIS
956
56.158
140.255
187.923
209.87


7224
C
CB
HIS
956
57.943
138.089
187.272
209.87


7225
C
CG
HIS
956
58.347
136.762
186.71
209.87


7226
N
ND1
HIS
956
57.435
135.778
186.397
209.87


7227
C
CD2
HIS
956
59.565
136.258
186.402
209.87


7228
C
CE1
HIS
956
58.074
134.723
185.922
209.87


7229
N
NE2
HIS
956
59.367
134.989
185.914
209.87


7230
N
N
ALA
957
57.339
141.486
186.457
211.98


7231
C
CA
ALA
957
56.913
142.74
187.07
211.98


7232
C
C
ALA
957
55.409
142.944
186.925
211.98


7233
O
O
ALA
957
54.739
143.391
187.865
211.98


7234
C
CB
ALA
957
57.676
143.913
186.455
211.98


7235
N
N
GLU
958
54.862
142.622
185.749
217.11


7236
C
CA
GLU
958
53.426
142.759
185.528
217.11


7237
C
C
GLU
958
52.638
141.812
186.424
217.11


7238
O
O
GLU
958
51.535
142.146
186.873
217.11


7239
C
CB
GLU
958
53.101
142.506
184.055
217.11


7240
C
CG
GLU
958
51.635
142.683
183.694
217.11


7241
C
CD
GLU
958
51.446
143.286
182.315
217.11


7242
O
OE1
GLU
958
52.428
143.331
181.544
217.11


7243
O
OE2
GLU
958
50.315
143.713
182.001
217.11


7244
N
N
ASP
959
53.18
140.622
186.685
218.73


7245
C
CA
ASP
959
52.516
139.695
187.595
218.73


7246
C
C
ASP
959
52.599
140.18
189.038
218.73


7247
O
O
ASP
959
51.626
140.071
189.793
218.73


7248
C
CB
ASP
959
53.131
138.302
187.462
218.73


7249
C
CG
ASP
959
52.888
137.686
186.098
218.73


7250
O
OD1
ASP
959
51.857
138.01
185.472
218.73


7251
O
OD2
ASP
959
53.73
136.878
185.651
218.73


7252
N
N
LYS
960
53.754
140.717
189.438
217.59


7253
C
CA
LYS
960
53.946
141.118
190.828
217.59


7254
C
C
LYS
960
53.134
142.36
191.172
217.59


7255
O
O
LYS
960
52.626
142.483
192.293
217.59


7256
C
CB
LYS
960
55.43
141.352
191.118
217.59


7257
C
CG
LYS
960
56.283
140.094
191.095
217.59


7258
C
CD
LYS
960
57.681
140.371
191.627
217.59


7259
C
CE
LYS
960
58.478
141.257
190.684
217.59


7260
N
NZ
LYS
960
58.781
140.575
189.398
217.59


7261
N
N
VAL
961
53.008
143.3
190.232
217.24


7262
C
CA
VAL
961
52.268
144.525
190.512
217.24


7263
C
C
VAL
961
50.8
144.188
190.746
217.24


7264
O
O
VAL
961
50.181
143.433
189.983
217.24


7265
C
CB
VAL
961
52.452
145.555
189.38
217.24


7266
C
CG1
VAL
961
51.896
145.044
188.06
217.24


7267
C
CG2
VAL
961
51.801
146.88
189.759
217.24


7268
N
N
LYS
962
50.247
144.722
191.832
214.27


7269
C
CA
LYS
962
48.89
144.414
192.257
214.27


7270
C
C
LYS
962
48.207
145.697
192.706
214.27


7271
O
O
LYS
962
48.843
146.583
193.283
214.27


7272
C
CB
LYS
962
48.893
143.364
193.384
214.27


7273
C
CG
LYS
962
47.517
142.912
193.857
214.27


7274
C
CD
LYS
962
47.104
143.619
195.138
214.27


7275
C
CE
LYS
962
47.949
143.157
196.315
214.27


7276
N
NZ
LYS
962
47.568
143.842
197.581
214.27


7277
N
N
LYS
963
46.911
145.794
192.425
211.36


7278
C
CA
LYS
963
46.162
147.006
192.725
211.36


7279
C
C
LYS
963
45.867
147.116
194.216
211.36


7280
O
O
LYS
963
45.227
146.24
194.806
211.36


7281
C
CB
LYS
963
44.855
147.023
191.932
211.36


7282
C
CG
LYS
963
44.017
148.274
192.13
211.36


7283
C
CD
LYS
963
42.794
148.259
191.227
211.36


7284
C
CE
LYS
963
41.87
149.428
191.522
211.36


7285
N
NZ
LYS
963
42.504
150.733
191.184
211.36


7286
N
N
MET
964
46.334
148.203
194.827
208.65


7287
C
CA
MET
964
45.974
148.501
196.205
208.65


7288
C
C
MET
964
44.638
149.24
196.259
208.65


7289
O
O
MET
964
43.922
149.37
195.261
208.65


7290
C
CB
MET
964
47.064
149.323
196.894
208.65


7291
C
CG
MET
964
48.295
148.532
197.318
208.65


7292
S
SD
MET
964
49.371
148.037
195.963
208.65


7293
C
CE
MET
964
50.142
149.603
195.565
208.65


7294
N
N
LYS
965
44.306
149.735
197.449
210.93


7295
C
CA
LYS
965
43.057
150.458
197.655
210.93


7296
C
C
LYS
965
43.199
151.882
197.133
210.93


7297
O
O
LYS
965
44.052
152.647
197.591
210.93


7298
C
CB
LYS
965
42.675
150.465
199.135
210.93


7299
C
CG
LYS
965
42.043
149.171
199.633
210.93


7300
C
CD
LYS
965
43.075
148.07
199.825
210.93


7301
C
CE
LYS
965
42.448
146.817
200.41
210.93


7302
N
NZ
LYS
965
43.418
145.689
200.456
210.93


7303
N
N
LEU
966
42.359
152.235
196.163
219.26


7304
C
CA
LEU
966
42.447
153.523
195.505
219.26


7305
C
C
LEU
966
41.093
154.222
195.535
219.26


7306
O
O
LEU
966
40.082
153.63
195.132
219.26


7307
C
CB
LEU
966
42.939
153.337
194.057
219.26


7308
C
CG
LEU
966
43.291
154.533
193.174
219.26


7309
C
CD1
LEU
966
44.312
154.116
192.136
219.26


7310
C
CD2
LEU
966
42.062
155.04
192.478
219.26


7311
N
N
PRO
967
41.032
155.467
196.004
227.55


7312
C
CA
PRO
967
39.737
156.141
196.162
227.55


7313
C
C
PRO
967
39.095
156.486
194.825
227.55


7314
O
O
PRO
967
39.755
156.607
193.79
227.55


7315
C
CB
PRO
967
40.088
157.41
196.95
227.55


7316
C
CG
PRO
967
41.438
157.14
197.551
227.55


7317
C
CD
PRO
967
42.137
156.262
196.562
227.55


7318
N
N
LYS
968
37.771
156.662
194.871
230.22


7319
C
CA
LYS
968
37.012
156.97
193.663
230.22


7320
C
C
LYS
968
37.441
158.295
193.044
230.22


7321
O
O
LYS
968
37.536
158.4
191.816
230.22


7322
C
CB
LYS
968
35.514
156.998
193.975
230.22


7323
C
CG
LYS
968
34.939
155.688
194.51
230.22


7324
C
CD
LYS
968
34.847
154.611
193.435
230.22


7325
C
CE
LYS
968
35.971
153.591
193.556
230.22


7326
N
NZ
LYS
968
35.825
152.489
192.566
230.22


7327
N
N
ASN
969
37.695
159.314
193.869
230.88


7328
C
CA
ASN
969
38.126
160.603
193.335
230.88


7329
C
C
ASN
969
39.492
160.498
192.665
230.88


7330
O
O
ASA
969
39.717
161.09
191.603
230.88


7331
C
CB
ASN
969
38.139
161.657
194.445
230.88


7332
C
CG
ASN
969
39.046
161.282
195.606
230.88


7333
O
OD1
ASN
969
39.761
160.282
195.559
230.88


7334
N
ND2
ASN
969
39.017
162.09
196.659
230.88


7335
N
N
TYR
970
40.416
159.747
193.268
230.04


7336
C
CA
TYR
970
41.729
159.555
192.666
230.04


7337
C
C
TYR
970
41.674
158.663
191.433
230.04


7338
O
O
TYR
970
42.592
158.708
190.607
230.04


7339
C
CB
TYR
970
42.706
158.972
193.69
230.04


7340
C
CG
TYR
970
43.122
159.948
194.769
230.04


7341
C
CD1
TYR
970
42.917
161.314
194.615
230.04


7342
C
CD2
TYR
970
43.728
159.506
195.938
230.04


7343
C
CE1
TYR
970
43.298
162.21
195.596
230.04


7344
C
CE2
TYR
970
44.113
160.395
196.925
230.04


7345
C
CZ
TYR
970
43.895
161.745
196.748
230.04


7346
O
OH
TYR
970
44.276
162.634
197.727
230.04


7347
N
N
GLN
971
40.63
157.849
191.294
228.26


7348
C
CA
GLN
971
40.477
157.03
190.098
228.26


7349
C
C
GLN
971
40.222
157.925
188.892
228.26


7350
O
O
GLN
971
39.365
158.813
188.934
228.26


7351
C
CB
GLN
971
39.336
156.028
190.279
228.26


7352
C
CG
GLN
971
39.244
154.976
189.183
228.26


7353
C
CD
GLN
971
38.318
155.378
188.053
228.26


7354
O
OE1
GLN
971
37.386
156.158
188.246
228.26


7355
N
NE2
GLN
971
38.57
154.842
186.864
228.26


7356
N
N
LEU
972
40.969
157.69
187.817
223.62


7357
C
CA
LEU
972
40.879
158.547
186.644
223.62


7358
C
C
LEU
972
39.544
158.361
185.936
223.62


7359
O
O
LEU
972
39.109
157.235
185.676
223.62


7360
C
CB
LEU
972
42.026
158.255
185.677
223.62


7361
C
CG
LEU
972
43.412
158.07
186.296
223.62


7362
C
CD1
LEU
972
44.407
157.577
185.257
223.62


7363
C
CD2
LEU
972
43.888
159.365
186.929
223.62


7364
N
N
THR
973
38.894
159.482
185.622
222.81


7365
C
CA
THR
973
37.655
159.455
184.857
222.81


7366
C
C
THR
973
37.898
159.301
183.362
222.81


7367
O
O
THR
973
36.933
159.136
182.607
222.81


7368
C
CB
THR
973
36.845
160.727
185.117
222.81


7369
O
OG1
THR
973
37.682
161.876
184.93
222.81


7370
C
CG2
THR
973
36.302
160.73
186.539
222.81


7371
N
N
SER
974
39.158
159.355
182.921
220.23


7372
C
CA
SER
974
39.467
159.213
181.502
220.23


7373
C
C
SER
974
39.129
157.825
180.975
220.23


7374
O
O
SER
974
38.943
157.657
179.765
220.23


7375
C
CB
SER
974
40.945
159.521
181.257
220.23


7376
O
OG
SER
974
41.745
158.367
181.45
220.23


7377
N
N
GLY
975
39.048
156.826
181.853
215.94


7378
C
CA
GLY
975
38.723
155.466
181.476
215.94


7379
C
C
GLY
975
39.871
154.49
181.608
215.94


7380
O
O
GLY
975
39.628
153.282
181.723
215.94


7381
N
N
TYR
976
41.111
154.971
181.596
211.6


7382
C
CA
TYR
976
42.252
154.091
181.8
211.6


7383
C
C
TYR
976
42.359
153.705
183.27
211.6


7384
O
O
TYR
976
41.993
154.476
184.162
211.6


7385
C
CB
TYR
976
43.541
154.764
181.33
211.6


7386
C
CG
TYR
976
44.739
153.84
181.306
211.6


7387
C
CD1
TYR
976
44.948
152.971
180.243
211.6


7388
C
CD2
TYR
976
45.662
153.839
182.343
211.6


7389
C
CE1
TYR
976
46.04
152.124
180.215
211.6


7390
C
CE2
TYR
976
46.758
152.995
182.324
211.6


7391
C
CZ
TYR
976
46.941
152.141
181.258
211.6


7392
O
OH
TYR
976
48.03
151.299
181.235
211.6


7393
N
N
LYS
977
42.867
152.501
183.52
208.77


7394
C
CA
LYS
977
42.982
151.986
184.878
208.77


7395
C
C
LYS
977
44.318
152.408
185.472
208.77


7396
O
O
LYS
977
45.373
152.026
184.942
208.77


7397
C
CB
LYS
977
42.85
150.463
184.892
208.77


7398
C
CG
LYS
977
42.848
149.858
186.286
208.77


7399
C
CD
LYS
977
42.627
148.356
186.236
208.77


7400
C
CE
LYS
977
42.63
147.754
187.631
208.77


7401
N
NZ
LYS
977
42.413
146.282
187.605
208.77


7402
N
N
PRO
978
44.331
153.185
186.553
207.52


7403
C
CA
PRO
978
45.603
153.584
187.162
207.52


7404
C
C
PRO
978
46.368
152.376
187.678
207.52


7405
O
O
PRO
978
45.788
151.367
188.086
207.52


7406
C
CB
PRO
978
45.176
154.5
188.316
207.52


7407
C
CG
PRO
978
43.789
154.934
187.971
207.52


7408
C
CD
PRO
978
43.177
153.777
187.248
207.52


7409
N
N
ALA
979
47.694
152.492
187.657
194.87


7410
C
CA
ALA
979
48.582
151.445
188.161
194.87


7411
C
C
ALA
979
49.579
152.054
189.137
194.87


7412
O
O
ALA
979
50.778
152.15
188.846
194.87


7413
C
CB
ALA
979
49.3
150.736
187.013
194.87


7414
N
N
PRO
980
49.117
152.478
190.318
189.41


7415
C
CA
PRO
980
50.058
152.931
191.35
189.41


7416
C
C
PRO
980
50.625
151.741
192.108
189.41


7417
O
O
PRO
980
49.886
150.976
192.733
189.41


7418
C
CB
PRO
980
49.188
153.822
192.243
189.41


7419
C
CG
PRO
980
47.83
153.205
192.15
189.41


7420
C
CD
PRO
980
47.718
152.568
190.778
189.41


7421
N
N
MET
981
51.94
151.564
192.031
176.42


7422
C
CA
MET
981
52.582
150.398
192.616
176.42


7423
C
C
MET
981
53.214
150.74
193.965
176.42


7424
O
O
MET
981
53.339
151.906
194.35
176.42


7425
C
CB
MET
981
53.604
149.818
191.634
176.42


7426
C
CG
MET
981
54.752
150.748
191.238
176.42


7427
S
SD
MET
981
56.084
150.887
192.442
176.42


7428
C
CE
MET
981
56.822
149.264
192.28
176.42


7429
N
N
ASP
982
53.615
149.694
194.687
167.59


7430
C
CA
ASF
982
54.168
149.83
196.032
167.59


7431
C
C
ASP
982
55.602
150.342
195.937
167.59


7432
O
O
ASF
982
56.566
149.575
195.98
167.59


7433
C
CB
ASP
982
54.103
148.495
196.771
167.59


7434
C
CG
ASP
982
54.458
148.62
198.239
167.59


7435
O
OD1
ASP
982
53.912
149.524
198.907
167.59


7436
O
OD2
ASP
982
55.28
147.816
198.727
167.59


7437
N
N
LEU
983
55.745
151.659
195.807
163.5


7438
C
CA
LEU
983
57.04
152.282
195.565
163.5


7439
C
C
LEU
983
57.842
152.527
196.838
163.5


7440
O
O
LEU
983
58.963
153.04
196.756
163.5


7441
C
CB
LEU
983
56.858
153.601
194.805
163.5


7442
C
CG
LEU
983
56.294
154.833
195.523
163.5


7443
C
CD1
LEU
983
56.575
156.078
194.696
163.5


7444
C
CD2
LEU
983
54.801
154.708
195.79
163.5


7445
N
N
SER
984
57.302
152.18
198.008
162.16


7446
C
CA
SER
984
58.019
152.451
199.25
162.16


7447
C
C
SER
984
59.183
151.487
199.453
162.16


7448
O
O
SER
984
60.276
151.907
199.853
162.16


7449
C
CB
SER
984
57.06
152.386
200.439
162.16


7450
O
OG
SER
984
56.57
151.071
200.633
162.16


7451
N
N
PHE
985
58.977
150.195
199.182
157


7452
C
CA
PHE
985
59.991
149.21
199.543
157


7453
C
C
PHE
985
61.216
149.268
198.639
157


7454
O
O
PHE
985
62.27
148.747
199.019
157


7455
C
CB
PHE
985
59.401
147.796
199.539
157


7456
C
CG
PHE
985
59.283
147.178
198.174
157


7457
C
CD1
PHE
985
58.194
147.449
197.366
157


7458
C
CD2
PHE
985
60.257
146.309
197.706
157


7459
C
CE1
PHE
985
58.081
146.872
196.114
157


7460
C
CE2
PHE
985
60.152
145.734
196.455
157


7461
C
CZ
PHE
985
59.063
146.017
195.657
157


7462
N
N
ILE
986
61.108
149.879
197.459
150.75


7463
C
CA
ILE
986
62.274
150.017
196.595
150.75


7464
C
C
ILE
986
63.249
150.994
197.236
150.75


7465
O
O
ILE
986
62.926
152.171
197.445
150.75


7466
C
CB
ILE
986
61.872
150.485
195.189
150.75


7467
C
CG1
ILE
986
61.014
149.436
194.481
150.75


7468
C
CG2
ILE
986
63.113
150.786
194.364
150.75


7469
C
CD1
ILE
986
59.531
149.65
194.652
150.75


7470
N
N
LYS
987
64.448
150.514
197.547
139.36


7471
C
CA
LYS
987
65.479
151.334
198.167
139.36


7472
C
C
LYS
987
66.317
151.978
197.07
139.36


7473
O
O
LYS
987
66.966
151.277
196.285
139.36


7474
C
CB
LYS
987
66.343
150.493
199.11
139.36


7475
C
CG
LYS
987
67.57
151.197
199.685
139.36


7476
C
CD
LYS
987
68.845
150.864
198.921
139.36


7477
C
CE
LYS
987
70.03
151.649
199.464
139.36


7478
N
NZ
LYS
987
71.245
151.497
198.618
139.36


7479
N
N
LEU
988
66.286
153.307
197.009
132.98


7480
C
CA
LEU
988
67.048
154.047
196.013
132.98


7481
C
C
LEU
988
68.505
154.122
196.443
132.98


7482
O
O
LEU
988
68.847
154.834
197.394
132.98


7483
C
CB
LEU
988
66.471
155.448
195.826
132.98


7484
C
CG
LEU
988
66.932
156.213
194.585
132.98


7485
C
CD1
LEU
988
66.519
155.49
193.316
132.98


7486
C
CD2
LEU
988
66.371
157.621
194.611
132.98


7487
N
N
THR
989
69.356
153.381
195.747
130.98


7488
C
CA
THR
989
70.777
153.406
196.044
130.98


7489
C
C
THR
989
71.346
154.789
195.709
130.98


7490
O
O
THR
989
70.915
155.43
194.735
130.98


7491
C
CB
THR
989
71.481
152.286
195.262
130.98


7492
O
OG1
THR
989
70.831
151.04
195.542
130.98


7493
C
CG2
THR
989
72.95
152.146
195.634
130.98


7494
N
N
PRO
990
72.278
155.304
196.521
133.56


7495
C
CA
PRO
990
72.781
156.667
196.276
133.56


7496
C
C
PRO
990
73.356
156.872
194.887
133.56


7497
O
O
PRO
990
73.39
158.012
194.408
133.56


7498
C
CB
PRO
990
73.845
156.865
197.367
133.56


7499
C
CG
PRO
990
74.045
155.548
198.014
133.56


7500
C
CD
PRO
990
72.852
154.704
197.738
133.56


7501
N
N
SER
991
73.807
155.812
194.214
133.78


7502
C
CA
SER
991
74.237
155.969
192.829
133.78


7503
C
C
SER
991
73.086
156.443
191.947
133.78


7504
O
O
SER
991
73.229
157.422
191.202
133.78


7505
C
CB
SER
991
74.817
154.656
192.303
133.78


7506
O
OG
SER
991
73.823
153.65
192.222
133.78


7507
N
N
GLN
992
71.932
155.77
192.019
133.67


7508
C
CA
GLN
992
70.776
156.245
191.263
133.67


7509
C
C
GLN
992
70.304
157.598
191.763
133.67


7510
O
O
GLN
992
69.742
158.377
190.993
133.67


7511
C
CB
GLN
992
69.624
155.239
191.284
133.67


7512
C
CG
GLN
992
69.915
153.948
190.554
133.67


7513
C
CD
GLN
992
70.697
153.003
191.404
133.67


7514
O
OE1
GLN
992
70.748
153.17
192.612
133.67


7515
N
NE2
GLN
992
71.317
152.007
190.787
133.67


7516
N
N
GLU
993
70.517
157.908
193.043
138.41


7517
C
CA
GLU
993
70.239
159.279
193.474
138.41


7518
C
C
GLU
993
71.119
160.276
192.719
138.41


7519
O
O
GLU
993
70.661
161.355
192.32
138.41


7520
C
CB
GLU
993
70.427
159.418
194.984
138.41


7521
C
CG
GLU
993
70.186
160.834
195.491
138.41


7522
C
CD
GLU
993
68.723
161.242
195.442
138.41


7523
O
OE1
GLU
993
67.849
160.355
195.533
138.41


7524
O
OE2
GLU
993
68.451
162.453
195.298
138.41


7525
N
N
ALA
994
72.388
159.924
192.508
138.7


7526
C
CA
ALA
994
73.288
160.795
191.755
138.7


7527
C
C
ALA
994
72.854
160.921
190.297
138.7


7528
O
O
ALA
994
72.868
162.022
189.727
138.7


7529
C
CB
ALA
994
74.722
160.275
191.845
138.7


7530
N
N
MET
995
72.482
159.803
189.666
144.61


7531
C
CA
MET
995
71.959
159.904
188.304
144.61


7532
C
C
MET
995
70.653
160.689
188.263
144.61


7533
O
O
MET
995
70.356
161.343
187.263
144.61


7534
C
CB
MET
995
71.773
158.535
187.635
144.61


7535
C
CG
MET
995
73.056
157.862
187.123
144.61


7536
S
SD
MET
995
74.226
157.138
188.28
144.61


7537
C
CE
MET
995
73.322
155.66
188.735
144.61


7538
N
N
VAL
996
69.865
160.643
189.336
139.77


7539
C
CA
VAL
996
68.663
161.468
189.425
139.77


7540
C
C
VAL
996
69.033
162.947
189.459
139.77


7541
O
O
VAL
996
68.381
163.784
188.82
139.77


7542
C
CB
VAL
996
67.832
161.045
190.652
139.77


7543
C
CG1
VAL
996
66.859
162.12
191.044
139.77


7544
C
CG2
VAL
996
67.07
159.762
190.351
139.77


7545
N
N
ASP
997
70.088
163.289
190.201
140.79


7546
C
CA
ASP
997
70.562
164.67
190.214
140.79


7547
C
C
ASP
997
71.003
165.109
188.823
140.79


7548
O
O
ASP
997
70.666
166.212
188.368
140.79


7549
C
CB
ASP
997
71.717
164.821
191.204
140.79


7550
C
CG
ASP
997
71.276
164.671
192.646
140.79


7551
O
OD1
ASP
997
70.111
164.996
192.951
140.79


7552
O
OD2
ASP
997
72.1
164.226
193.474
140.79


7553
N
N
LYS
998
71.757
164.253
188.131
141.72


7554
C
CA
LYS
998
72.156
164.566
186.761
141.72


7555
C
C
LYS
998
70.957
164.673
185.829
141.72


7556
O
O
LYS
998
70.949
165.515
184.927
141.72


7557
C
CB
LYS
998
73.159
163.539
186.238
141.72


7558
C
CG
LYS
998
74.445
163.467
187.036
141.72


7559
C
CD
LYS
998
75.195
164.788
186.88
141.72


7560
C
CE
LYS
998
76.578
164.755
187.506
141.72


7561
N
NZ
LYS
998
76.527
164.685
188.989
141.72


7562
N
N
LEU
999
69.943
163.826
186.012
143.25


7563
C
CA
LEU
999
68.747
163.922
185.179
143.25


7564
C
C
LEU
999
68.036
165.25
185.395
143.25


7565
O
O
LEU
999
67.581
165.878
184.434
143.25


7566
C
CB
LEU
999
67.8
162.755
185.465
143.25


7567
C
CG
LEU
999
68.212
161.349
185.013
143.25


7568
C
CD1
LEU
999
66.988
160.463
184.812
143.25


7569
C
CD2
LEU
999
69.069
161.387
183.754
143.25


7570
N
N
ALA
1000
67.929
165.69
186.65
142.71


7571
C
CA
ALA
1000
67.324
166.989
186.923
142.71


7572
C
C
ALA
1000
68.119
168.11
186.271
142.71


7573
O
O
ALA
1000
67.549
168.977
185.594
142.71


7574
C
CB
ALA
1000
67.217
167.213
188.431
142.71


7575
N
N
GLU
1001
69.442
168.094
186.45
143.2


7576
C
CA
GLU
1001
70.289
169.123
185.857
143.2


7577
C
C
GLU
1001
70.133
169.147
184.341
143.2


7578
O
O
GLU
1001
69.97
170.212
183.735
143.2


7579
C
CB
GLU
1001
71.749
168.876
186.242
143.2


7580
C
CG
GLU
1001
72.736
169.871
185.653
143.2


7581
C
CD
GLU
1001
72.739
171.195
186.388
143.2


7582
O
OE1
GLU
1001
72.108
171.276
187.462
143.2


7583
O
OE2
GLU
1001
73.368
172.154
185.89
143.2


7584
N
N
ASN
1002
70.152
167.969
183.716
141.05


7585
C
CA
ASN
1002
70.122
167.884
182.262
141.05


7586
C
C
ASN
1002
68.763
168.287
181.707
141.05


7587
O
O
ASN
1002
68.688
168.97
180.681
141.05


7588
C
CB
ASN
1002
70.488
166.467
181.825
141.05


7589
C
CG
ASN
1002
70.875
166.385
180.365
141.05


7590
O
OD1
ASN
1002
70.718
167.343
179.608
141.05


7591
N
ND2
ASN
1002
71.396
165.235
179.962
141.05


7592
N
N
ALA
1003
67.676
167.872
182.362
137.21


7593
C
CA
ALA
1003
66.351
168.281
181.914
137.21


7594
C
C
ALA
1003
66.177
169.787
182.034
137.21


7595
O
O
ALA
1003
65.616
170.43
181.137
137.21


7596
C
CB
ALA
1003
65.275
167.547
182.714
137.21


7597
N
N
HIS
1004
66.661
170.372
183.134
139.95


7598
C
CA
HIS
1004
66.576
171.819
183.285
139.95


7599
C
C
HIS
1004
67.396
172.528
182.214
139.95


7600
O
O
HIS
1004
66.961
173.543
181.658
139.95


7601
C
CB
HIS
1004
67.034
172.219
184.688
139.95


7602
C
CG
HIS
1004
67.221
173.693
184.87
139.95


7603
N
ND1
HIS
1004
66.267
174.491
185.463
139.95


7604
C
CD2
HIS
1004
68.255
174.51
184.561
139.95


7605
C
CE1
HIS
1004
66.699
175.738
185.499
139.95


7606
N
NEZ
HIS
1004
67.903
175.777
184.958
139.95


7607
N
N
ASN
1005
68.586
172.002
181.907
138.12


7608
C
CA
ASN
1005
69.408
172.594
180.856
138.12


7609
C
C
ASN
1005
68.729
172.498
179.495
138.12


7610
O
O
ASN
1005
68.789
173.439
178.698
138.12


7611
C
CB
ASN
1005
70.781
171.923
180.817
138.12


7612
C
CG
ASN
1005
71.556
172.107
182.106
138.12


7613
O
OD1
ASN
1005
71.243
172.983
182.912
138.12


7614
N
ND2
ASN
1005
72.574
171.278
182.308
138.12


7615
N
N
VAL
1006
68.094
171.361
179.204
131.81


7616
C
CA
VAL
1006
67.394
171.2
177.931
131.81


7617
C
C
VAL
1006
66.238
172.186
177.832
131.81


7618
O
O
VAL
1006
66.02
172.81
176.785
131.81


7619
C
CB
VAL
1006
66.918
169.745
177.762
131.81


7620
C
CG1
VAL
1006
65.945
169.634
176.6
131.81


7621
C
CG2
VAL
1006
68.107
168.823
177.546
131.81


7622
N
N
TRP
1007
65.481
172.342
178.92
133.96


7623
C
CA
TRP
1007
64.398
173.32
178.935
133.96


7624
C
C
TRP
1007
64.921
174.734
178.71
133.96


7625
O
O
TRP
1007
64.338
175.508
177.938
133.96


7626
C
CB
TRP
1007
63.643
173.233
180.259
133.96


7627
C
CG
TRP
1007
62.64
174.317
180.435
133.96


7628
C
CD1
TRP
1007
61.395
174.385
179.885
133.96


7629
C
CD2
TRP
1007
62.804
175.506
181.211
133.96


7630
N
NE1
TRP
1007
60.769
175.544
180.278
133.96


7631
C
CE2
TRP
1007
61.616
176.25
181.092
133.96


7632
C
CE3
TRP
1007
63.843
176.014
181.997
133.96


7633
C
CZ2
TRP
1007
61.436
177.473
181.732
133.96


7634
C
CZ3
TRP
1007
63.662
177.227
182.631
133.96


7635
C
CH2
TRP
1007
62.469
177.942
182.496
133.96


7636
N
N
ALA
1008
66.018
175.091
179.382
132.48


7637
C
CA
ALA
1008
66.588
176.423
179.214
132.48


7638
C
C
ALA
1008
67.065
176.641
177.784
132.48


7639
O
O
ALA
1008
66.869
177.72
177.214
132.48


7640
C
CB
ALA
1008
67.734
176.633
180.203
132.48


7641
N
N
ARG
1009
67.695
175.626
177.189
130.16


7642
C
CA
ARG
1009
68.138
175.735
175.804
130.16


7643
C
C
ARG
1009
66.958
175.922
174.862
130.16


7644
O
O
ARG
1009
67.022
176.729
173.927
130.16


7645
C
CB
ARG
1009
68.928
174.492
175.404
130.16


7646
C
CG
ARG
1009
69.536
174.579
174.018
130.16


7647
C
CD
ARG
1009
70.331
173.333
173.706
130.16


7648
N
NE
ARG
1009
69.474
172.156
173.625
130.16


7649
C
CZ
ARG
1009
69.499
171.147
174.485
130.16


7650
N
NH1
ARG
1009
70.309
171.149
175.531
130.16


7651
N
NH2
ARG
1009
68.684
170.113
174.296
130.16


7652
N
N
ASP
1010
65.88
175.168
175.082
128.45


7653
C
CA
ASP
1010
64.7
175.309
174.238
128.45


7654
C
C
ASP
1010
64.112
176.707
174.355
128.45


7655
O
O
ASP
1010
63.74
177.321
173.348
128.45


7656
C
CB
ASP
1010
63.662
174.252
174.611
128.45


7657
C
CG
ASP
1010
64.069
172.858
174.175
128.45


7658
O
OD1
ASP
1010
64.822
172.74
173.186
128.45


7659
O
OD2
ASP
1010
63.637
171.881
174.822
128.45


7660
N
N
ARG
1011
64.042
177.239
175.576
125.59


7661
C
CA
ARG
1011
63.508
178.585
175.755
125.59


7662
C
C
ARG
1011
64.41
179.635
175.116
125.59


7663
O
O
ARG
1011
63.921
180.627
174.565
125.59


7664
C
CB
ARG
1011
63.294
178.873
177.241
125.59


7665
C
CG
ARG
1011
61.857
178.656
177.699
125.59


7666
C
CD
ARG
1011
61.398
177.232
177.43
125.59


7667
N
NE
ARG
1011
59.987
177.039
177.744
125.59


7668
C
CZ
ARG
1011
59.278
175.983
177.373
125.59


7669
N
NH1
ARG
1011
59.818
175
176.671
125.59


7670
N
NH2
ARG
1011
57.994
175.911
177.712
125.59


7671
N
N
ILE
1012
65.729
179.439
175.177
120.25


7672
C
CA
ILE
1012
66.642
180.403
174.57
120.25


7673
C
C
ILE
1012
66.534
180.365
173.048
120.25


7674
O
O
ILE
1012
66.558
181.409
172.386
120.25


7675
C
CB
ILE
1012
68.081
180.158
175.057
120.25


7676
C
CG1
ILE
1012
68.202
180.539
176.534
120.25


7677
C
CG2
ILE
1012
69.077
180.949
174.226
120.25


7678
C
CD1
ILE
1012
69.582
180.353
177.106
120.25


7679
N
N
ARG
1013
66.41
179.168
172.469
121.71


7680
C
CA
ARG
1013
66.123
179.068
171.039
121.71


7681
C
C
ARG
1013
64.802
179.737
170.689
121.71


7682
O
O
ARG
1013
64.679
180.364
169.63
121.71


7683
C
CB
ARG
1013
66.128
177.605
170.598
121.71


7684
C
CG
ARG
1013
67.474
176.938
170.761
121.71


7685
C
CD
ARG
1013
68.425
177.453
169.669
121.71


7686
N
NE
ARG
1013
69.417
178.465
170.042
121.71


7687
C
CZ
ARG
1013
70.072
178.564
171.194
121.71


7688
N
NH1
ARG
1013
69.957
177.654
172.148
121.71


7689
N
NH2
ARG
1013
70.896
179.591
171.381
121.71


7690
N
N
GLN
1014
63.801
179.613
171.559
118.04


7691
C
CA
GLN
1014
62.566
180.356
171.347
118.04


7692
C
C
GLN
1014
62.798
181.857
171.446
118.04


7693
O
O
GLN
1014
62.059
182.639
170.839
118.04


7694
C
CB
GLN
1014
61.502
179.913
172.35
118.04


7695
C
CG
GLN
1014
61.012
178.495
172.124
118.04


7696
C
CD
GLN
1014
60.363
178.318
170.768
118.04


7697
O
OE1
GLN
1014
60.898
177.632
169.898
118.04


7698
N
NE2
GLN
1014
59.205
178.939
170.579
118.04


7699
N
N
GLY
1015
63.815
182.275
172.196
116.62


7700
C
CA
GLY
1015
64.142
183.676
172.355
116.62


7701
C
C
GLY
1015
63.968
184.217
173.757
116.62


7702
O
O
GLY
1015
64.124
185.426
173.96
116.62


7703
N
N
TRP
1016
63.647
183.365
174.727
118.72


7704
C
CA
TRP
1016
63.439
183.816
176.095
118.72


7705
C
C
TRP
1016
64.741
184.317
176.707
118.72


7706
O
O
TRP
1016
65.834
183.859
176.364
118.72


7707
C
CB
TRP
1016
62.873
182.683
176.949
118.72


7708
C
CG
TRP
1016
61.473
182.307
176.602
118.72


7709
C
CD1
TRP
1016
61.069
181.52
175.565
118.72


7710
C
CD2
TRP
1016
60.286
182.694
177.3
118.72


7711
N
NE1
TRP
1016
59.701
181.397
175.57
118.72


7712
C
CE2
TRP
1016
59.196
182.11
176.626
118.72


7713
C
CE3
TRP
1016
60.038
183.484
178.427
118.72


7714
C
CZ2
TRP
1016
57.88
182.287
177.042
118.72


7715
C
CZ3
TRP
1016
58.731
183.658
178.839
118.72


7716
C
CH2
TRP
1016
57.669
183.063
178.148
118.72


7717
N
N
THR
1017
64.617
185.274
177.625
106.56


7718
C
CA
THR
1017
65.77
185.76
178.368
106.56


7719
C
C
THR
1017
65.387
185.924
179.831
106.56


7720
O
O
THR
1017
64.214
186.098
180.167
106.56


7721
C
CB
THR
1017
66.305
187.077
177.792
106.56


7722
O
OG1
THR
1017
67.52
187.437
178.461
106.56


7723
C
CG2
THR
1017
65.287
188.181
177.951
106.56


7724
N
N
TYR
1018
66.39
185.852
180.701
107.48


7725
C
CA
TYR
1018
66.136
185.845
182.134
107.48


7726
C
C
TYR
1018
65.584
187.187
182.602
107.48


7727
O
O
TYR
1018
65.921
188.248
182.07
107.48


7728
C
CB
TYR
1018
67.413
185.513
182.905
107.48


7729
C
CG
TYR
1018
67.343
185.864
184.373
107.48


7730
C
CD1
TYR
1018
66.607
185.087
185.256
107.48


7731
C
CD2
TYR
1018
68.004
186.977
184.875
107.48


7732
C
CE1
TYR
1018
66.535
185.404
186.597
107.48


7733
C
CE2
TYR
1018
67.937
187.302
186.216
107.48


7734
C
CZ
TYR
1018
67.201
186.512
187.072
107.48


7735
O
OH
TYR
1018
67.131
186.83
188.408
107.48


7736
N
N
GLY
1019
64.719
187.125
183.614
109.48


7737
C
CA
GLY
1019
64.16
188.312
184.228
109.48


7738
C
C
GLY
1019
63.784
188.02
185.664
109.48


7739
O
O
GLY
1019
63.891
186.888
186.138
109.48


7740
N
N
ILE
1020
63.355
189.068
186.367
107.17


7741
C
CA
ILE
1020
62.934
188.895
187.754
107.17


7742
C
C
ILE
1020
61.583
188.193
187.827
107.17


7743
O
O
ILE
1020
61.332
187.396
188.738
107.17


7744
C
CB
ILE
1020
62.914
190.248
188.487
107.17


7745
C
CG1
ILE
1020
62.046
191.26
187.734
107.17


7746
C
CG2
ILE
1020
64.328
190.766
188.682
107.17


7747
C
CD1
ILE
1020
61.726
192.502
188.534
107.17


7748
N
N
GLN
1021
60.694
188.474
186.877
109.49


7749
C
CA
GLN
1021
59.344
187.934
186.912
109.49


7750
C
C
GLN
1021
58.862
187.709
185.486
109.49


7751
O
O
GLN
1021
59.43
188.236
184.526
109.49


7752
C
CB
GLN
1021
58.402
188.86
187.695
109.49


7753
C
CG
GLN
1021
58.222
190.248
187.096
109.49


7754
C
CD
GLN
1021
57.14
190.295
186.039
109.49


7755
O
OE1
GLN
1021
56.171
189.541
186.096
109.49


7756
N
NE2
GLN
1021
57.303
191.181
185.063
109.49


7757
N
N
GLN
1022
57.805
186.911
185.36
114.61


7758
C
CA
GLN
1022
57.328
186.491
184.048
114.61


7759
C
C
GLN
1022
56.737
187.664
183.277
114.61


7760
O
O
GLN
1022
55.763
188.284
183.712
114.61


7761
C
CB
GLN
1022
56.29
185.379
184.199
114.61


7762
C
CG
GLN
1022
55.584
184.971
182.905
114.61


7763
C
CD
GLN
1022
56.505
184.348
181.861
114.61


7764
O
OE1
GLN
1022
57.724
184.292
182.028
114.61


7765
N
NE2
GLN
1022
55.913
183.876
180.771
114.61


7766
N
N
ASP
1023
57.322
187.953
182.116
110.03


7767
C
CA
ASP
1023
56.827
188.995
181.22
110.03


7768
C
C
ASP
1023
56.693
188.379
179.835
110.03


7769
O
O
ASP
1023
57.7
188.127
179.168
110.03


7770
C
CB
ASP
1023
57.761
190.203
181.195
110.03


7771
C
CG
ASP
1023
57.191
191.366
180.405
110.03


7772
O
OD1
ASP
1023
57.076
191.251
179.166
110.03


7773
O
OD2
ASP
1023
56.856
192.398
181.024
110.03


7774
N
N
VAL
1024
55.452
188.129
179.413
108.87


7775
C
CA
VAL
1024
55.221
187.506
178.115
108.87


7776
C
C
VAL
1024
55.564
188.465
176.981
108.87


7777
O
O
VAL
1024
56.048
188.041
175.924
108.87


7778
C
CB
VAL
1024
53.77
187
178.018
108.87


7779
C
CG1
VAL
1024
53.549
185.852
178.989
108.87


7780
C
CG2
VAL
1024
52.789
188.127
178.302
108.87


7781
N
N
LYS
1025
55.316
189.762
177.172
107.89


7782
C
CA
LYS
1025
55.556
190.73
176.106
107.89


7783
C
C
LYS
1025
57.037
190.817
175.761
107.89


7784
O
O
LYS
1025
57.419
190.76
174.586
107.89


7785
C
CB
LYS
1025
55.016
192.102
176.513
107.89


7786
C
CG
LYS
1025
53.56
192.336
176.144
107.89


7787
C
CD
LYS
1025
52.622
191.602
177.088
107.89


7788
C
CE
LYS
1025
52.681
192.184
178.49
107.89


7789
N
NZ
LYS
1025
51.724
191.508
179.409
107.89


7790
N
N
ASN
1026
57.888
190.951
176.774
108.61


7791
C
CA
ASN
1026
59.327
191.044
176.57
108.61


7792
C
C
ASN
1026
60.017
189.688
176.619
108.61


7793
O
O
ASN
1026
61.242
189.629
176.477
108.61


7794
C
CB
ASN
1026
59.945
191.979
177.612
108.61


7795
C
CG
ASN
1026
59.228
193.31
177.694
108.61


7796
O
OD1
ASN
1026
58.455
193.667
176.805
108.61


7797
N
ND2
ASN
1026
59.48
194.054
178.765
108.61


7798
N
N
ARG
1027
59.258
188.608
176.812
113.82


7799
C
CA
ARG
1027
59.795
187.249
176.875
113.82


7800
C
C
ARG
1027
60.886
187.14
177.941
113.82


7801
O
O
ARG
1027
62.001
186.667
177.689
113.82


7802
C
CB
ARG
1027
60.298
186.795
175.504
113.82


7803
C
CG
ARG
1027
60.013
185.334
175.209
113.82


7804
C
CD
ARG
1027
60.71
184.874
173.947
113.82


7805
N
NE
ARG
1027
60.113
185.457
172.751
113.82


7806
C
CZ
ARG
1027
59.362
184.787
171.889
113.82


7807
N
NH1
ARG
1027
59.094
183.502
172.058
113.82


7808
N
NH2
ARG
1027
58.867
185.421
170.83
113.82


7809
N
N
ARG
1028
60.548
187.602
179.142
107.72


7810
C
CA
ARG
1028
61.411
187.517
180.311
107.72


7811
C
C
ARG
1028
60.894
186.407
181.215
107.72


7812
O
O
ARG
1028
59.725
186.418
181.611
107.72


7813
C
CB
ARG
1028
61.447
188.842
181.074
107.72


7814
C
CG
ARG
1028
61.663
190.07
180.207
107.72


7815
C
CD
ARG
1028
63.109
190.197
179.768
107.72


7816
N
NE
ARG
1028
64.036
190.259
180.891
107.72


7817
C
CZ
ARG
1028
64.334
191.364
181.559
107.72


7818
N
NH1
ARG
1028
63.789
192.528
181.247
107.72


7819
N
NH2
ARG
1028
65.203
191.301
182.564
107.72


7820
N
N
ASN
1029
61.768
185.465
181.543
119.07


7821
C
CA
ASN
1029
61.424
184.306
182.345
119.07


7822
C
C
ASN
1029
62.165
184.365
183.67
119.07


7823
O
O
ASN
1029
63.387
184.559
183.678
119.07


7824
C
CB
ASN
1029
61.781
183.018
181.594
119.07


7825
C
CG
ASN
1029
61.129
181.789
182.191
119.07


7826
O
OD1
ASN
1029
60.456
181.859
183.217
119.07


7827
N
ND2
ASN
1029
61.311
180.654
181.532
119.07


7828
N
N
PRO
1030
61.467
184.279
184.805
121.93


7829
C
CA
PRO
1030
62.177
184.312
186.092
121.93


7830
C
C
PRO
1030
62.994
183.064
186.366
121.93


7831
O
O
PRO
1030
63.999
183.142
187.082
121.93


7832
C
CB
PRO
1030
61.046
184.482
187.117
121.93


7833
C
CG
PRO
1030
59.817
184.015
186.413
121.93


7834
C
CD
PRO
1030
60.009
184.375
184.976
121.93


7835
N
N
ARG
1031
62.599
181.918
185.817
136.32


7836
C
CA
ARG
1031
63.222
180.648
186.163
136.32


7837
C
C
ARG
1031
64.466
180.356
185.328
136.32


7838
O
O
ARG
1031
65.175
179.385
185.612
136.32


7839
C
CB
ARG
1031
62.185
179.522
186.02
136.32


7840
C
CG
ARG
1031
62.634
178.14
186.48
136.32


7841
C
CD
ARG
1031
61.514
177.118
186.363
136.32


7842
N
NE
ARG
1031
61.92
175.808
186.859
136.32


7843
C
CZ
ARG
1031
62.487
174.865
186.12
136.32


7844
N
NH1
ARG
1031
62.726
175.047
184.832
136.32


7845
N
NH2
ARG
1031
62.821
173.709
186.687
136.32


7846
N
N
LEU
1032
64.779
181.194
184.34
125.78


7847
C
CA
LEU
1032
65.906
180.934
183.449
125.78


7848
C
C
LEU
1032
67.229
181.186
184.164
125.78


7849
O
O
LEU
1032
67.94
182.153
183.876
125.78


7850
C
CB
LEU
1032
65.805
181.795
182.191
125.78


7851
C
CG
LEU
1032
65.036
181.185
181.019
125.78


7852
C
CD1
LEU
1032
64.975
182.162
179.86
125.78


7853
C
CD2
LEU
1032
65.673
179.875
180.587
125.78


7854
N
N
VAL
1033
67.559
180.304
185.101
131


7855
C
CA
VAL
1033
68.823
180.354
185.834
131


7856
C
C
VAL
1033
69.402
178.948
185.877
131


7857
O
O
VAL
1033
68.685
177.961
185.656
131


7858
C
CB
VAL
1033
68.636
180.913
187.264
131


7859
C
CG1
VAL
1033
68.173
182.358
187.221
131


7860
C
CG2
VAL
1033
67.657
180.055
188.046
131


7861
N
N
PRO
1034
70.706
178.822
186.133
131.72


7862
C
CA
PRO
1034
71.29
177.484
186.294
131.72


7863
C
C
PRO
1034
70.633
176.727
187.44
131.72


7864
O
O
PRO
1034
70.253
177.306
188.46
131.72


7865
C
CB
PRO
1034
72.77
177.769
186.581
131.72


7866
C
CG
PRO
1034
72.844
179.23
186.911
131.72


7867
C
CD
PRO
1034
71.735
179.873
186.152
131.72


7868
N
N
TYR
1035
70.502
175.41
187.253
143.13


7869
C
CA
TYR
1035
69.766
174.586
188.209
143.13


7870
C
C
TYR
1035
70.405
174.618
189.591
143.13


7871
O
O
TYR
1035
69.702
174.519
190.604
143.13


7872
C
CB
TYR
1035
69.664
173.151
187.683
143.13


7873
C
CG
TYR
1035
68.826
172.204
188.525
143.13


7874
C
CD1
TYR
1035
67.55
171.834
188.12
143.13


7875
C
CD2
TYR
1035
69.332
171.634
189.692
143.13


7876
C
CE1
TYR
1035
66.782
170.962
188.871
143.13


7877
C
CE2
TYR
1035
68.571
170.761
190.45
143.13


7878
C
CZ
TYR
1035
67.298
170.428
190.033
143.13


7879
O
OH
TYR
1035
66.539
169.557
190.779
143.13


7880
N
N
THR
1036
71.73
174.76
189.655
137.31


7881
C
CA
THR
1036
72.403
174.811
190.948
137.31


7882
C
C
THR
1036
71.888
175.967
191.795
137.31


7883
O
O
THR
1036
71.798
175.852
193.023
137.31


7884
C
CB
THR
1036
73.914
174.929
190.749
137.31


7885
O
OG1
THR
1036
74.21
176.148
190.057
137.31


7886
C
CG2
THR
1036
74.436
173.754
189.936
137.31


7887
N
N
LEU
1037
71.538
177.082
191.16
138.34


7888
C
CA
LEU
1037
71.005
178.248
191.847
138.34


7889
C
C
LEU
1037
69.486
178.348
191.754
138.34


7890
O
O
LEU
1037
68.924
179.385
192.119
138.34


7891
C
CB
LEU
1037
71.641
179.522
191.285
138.34


7892
C
CG
LEU
1037
73.141
179.691
191.529
138.34


7893
C
CD1
LEU
1037
73.665
180.92
190.801
138.34


7894
C
CD2
LEU
1037
73.444
179.77
193.018
138.34


7895
N
N
LEU
1038
68.815
177.304
191.276
141.13


7896
C
CA
LEU
1038
67.38
177.362
191.042
141.13


7897
C
C
LEU
1038
66.613
177.456
192.361
141.13


7898
O
O
LEU
1038
67.114
177.102
193.431
141.13


7899
C
CB
LEU
1038
66.925
176.135
190.249
141.13


7900
C
CG
LEU
1038
65.52
176.143
189.646
141.13


7901
C
CD1
LEU
1038
65.376
177.292
188.667
141.13


7902
C
CD2
LEU
1038
65.22
174.82
188.963
141.13


7903
N
N
ASP
1039
65.383
177.959
192.272
148.25


7904
C
CA
ASP
1039
64.518
178.049
193.438
148.25


7905
C
C
ASP
1039
64.113
176.658
193.918
148.25


7906
O
O
ASP
1039
64.089
175.691
193.153
148.25


7907
C
CB
ASP
1039
63.276
178.888
193.127
148.25


7908
C
CG
ASP
1039
62.489
178.357
191.943
148.25


7909
O
OD1
ASP
1039
62.968
177.42
191.272
148.25


7910
O
OD2
ASP
1039
61.386
178.881
191.683
148.25


7911
N
N
ASP
1040
63.798
176.569
195.211
149.98


7912
C
CA
ASP
1040
63.552
175.269
195.828
149.98


7913
C
C
ASP
1040
62.278
174.614
195.302
149.98


7914
O
O
ASP
1040
62.27
173.411
195.02
149.98


7915
C
CB
ASP
1040
63.495
175.417
197.349
149.98


7916
C
CG
ASP
1040
62.426
176.396
197.806
149.98


7917
O
OD1
ASP
1040
61.798
177.046
196.943
149.98


7918
O
OD2
ASP
1040
62.215
176.515
199.031
149.98


7919
N
N
ARG
1041
61.193
175.381
195.166
148.91


7920
C
CA
ARG
1041
59.891
174.778
194.891
148.91


7921
C
C
ARG
1041
59.863
174.104
193.523
148.91


7922
O
O
ARG
1041
59.492
172.93
193.404
148.91


7923
C
CB
ARG
1041
58.791
175.836
194.987
148.91


7924
C
CG
ARG
1041
57.382
175.264
195.109
148.91


7925
C
CD
ARG
1041
56.702
175.106
193.757
148.91


7926
N
NE
ARG
1041
56.524
176.377
193.068
148.91


7927
C
CZ
ARG
1041
56.233
176.492
191.779
148.91


7928
N
NH1
ARG
1041
56.082
175.428
191.007
148.91


7929
N
NH2
ARG
1041
56.092
177.705
191.251
148.91


7930
N
N
THR
1042
60.247
174.837
192.475
151.28


7931
C
CA
THR
1042
60.167
174.289
191.124
151.28


7932
C
C
THR
1042
61.148
173.141
190.931
151.28


7933
O
O
THR
1042
60.819
172.135
190.288
151.28


7934
C
CB
THR
1042
60.421
175.386
190.092
151.28


7935
O
OG1
THR
1042
61.776
175.84
190.2
151.28


7936
C
CG2
THR
1042
59.479
176.557
190.321
151.28


7937
N
N
LYS
1043
62.366
173.281
191.463
149.39


7938
C
CA
LYS
1043
63.32
172.185
191.364
149.39


7939
C
C
LYS
1043
62.829
170.969
192.132
149.39


7940
O
O
LYS
1043
63.004
169.84
191.669
149.39


7941
C
CB
LYS
1043
64.705
172.626
191.846
149.39


7942
C
CG
LYS
1043
64.911
172.648
193.352
149.39


7943
C
CD
LYS
1043
66.391
172.648
193.695
149.39


7944
C
CE
LYS
1043
67.058
173.935
193.242
149.39


7945
N
NZ
LYS
1043
68.474
174.03
193.693
149.39


7946
N
N
LYS
1044
62.173
171.177
193.277
155.01


7947
C
CA
LYS
1044
61.587
170.058
194.007
155.01


7948
C
C
LYS
1044
60.498
169.378
193.186
155.01


7949
O
O
LYS
1044
60.416
168.145
193.15
155.01


7950
C
CB
LYS
1044
61.029
170.54
195.346
155.01


7951
C
CG
LYS
1044
60.456
169.441
196.228
155.01


7952
C
CD
LYS
1044
61.551
168.533
196.763
155.01


7953
C
CE
LYS
1044
60.999
167.531
197.764
155.01


7954
N
NZ
LYS
1044
60.112
166.525
197.117
155.01


7955
N
N
SER
1045
59.656
170.167
192.515
154.74


7956
C
CA
SER
1045
58.574
169.597
191.716
154.74


7957
C
C
SER
1045
59.117
168.766
190.558
154.74


7958
O
O
SER
1045
58.723
167.606
190.367
154.74


7959
C
CB
SER
1045
57.663
170.712
191.202
154.74


7960
O
OG
SER
1045
58.177
171.278
190.009
154.74


7961
N
N
ASN
1046
60.03
169.344
189.774
152.74


7962
C
CA
ASN
1046
60.611
168.598
188.661
152.74


7963
C
C
ASN
1046
61.378
167.383
189.165
152.74


7964
O
O
ASN
1046
61.333
166.306
188.555
152.74


7965
C
CB
ASN
1046
61.519
169.505
187.83
152.74


7966
C
CG
ASN
1046
60.875
170.839
187.508
152.74


7967
O
OD1
ASN
1046
61.518
171.886
187.585
152.74


7968
N
ND2
ASN
1046
59.598
170.808
187.149
152.74


7969
N
N
LYS
1047
62.072
167.537
190.294
152.3


7970
C
CA
LYS
1047
62.866
166.46
190.859
152.3


7971
C
C
LYS
1047
61.987
165.293
191.284
152.3


7972
O
O
LYS
1047
62.34
164.131
191.058
152.3


7973
C
CB
LYS
1047
63.664
167.011
192.04
152.3


7974
C
CG
LYS
1047
64.678
166.078
192.646
152.3


7975
C
CD
LYS
1047
65.891
165.961
191.749
152.3


7976
C
CE
LYS
1047
67.036
165.308
192.489
152.3


7977
N
NZ
LYS
1047
66.551
164.279
193.452
152.3


7978
N
N
ASP
1048
60.833
165.578
191.896
158.86


7979
C
CA
ASP
1048
59.935
164.495
192.282
158.86


7980
C
C
ASP
1048
59.287
163.853
191.066
158.86


7981
O
O
ASP
1048
59.114
162.628
191.029
158.86


7982
C
CB
ASP
1048
58.877
164.981
193.285
158.86


7983
C
CG
ASP
1048
57.881
165.965
192.692
158.86


7984
O
OD1
ASP
1048
57.201
165.636
191.697
158.86


7985
O
OD2
ASP
1048
57.75
167.067
193.262
158.86


7986
N
N
SER
1049
58.94
164.652
190.054
158.47


7987
C
CA
SER
1049
58.393
164.069
188.834
158.47


7988
C
C
SER
1049
59.377
163.079
188.224
158.47


7989
O
O
SER
1049
59.005
161.962
187.837
158.47


7990
C
CB
SER
1049
58.056
165.172
187.832
158.47


7991
O
OG
SER
1049
59.174
166.006
187.597
158.47


7992
N
N
LEU
1050
60.652
163.457
188.165
155.57


7993
C
CA
LEU
1050
61.626
162.591
187.515
155.57


7994
C
C
LEU
1050
62.051
161.428
188.412
155.57


7995
O
O
LEU
1050
62.377
160.346
187.905
155.57


7996
C
CB
LEU
1050
62.811
163.424
187.032
155.57


7997
C
CG
LEU
1050
63.674
164.207
188.014
155.57


7998
C
CD1
LEU
1050
64.737
163.345
188.649
155.57


7999
C
CD2
LEU
1050
64.279
165.382
187.286
155.57


8000
N
N
ARG
1051
62.041
161.599
189.744
156.42


8001
C
CA
ARG
1051
62.264
160.413
190.567
156.42


8002
C
C
ARG
1051
61.116
159.431
190.406
156.42


8003
O
O
ARG
1051
61.336
158.218
190.428
156.42


8004
C
CB
ARG
1051
62.459
160.695
192.067
156.42


8005
C
CG
ARG
1051
61.289
161.273
192.864
156.42


8006
C
CD
ARG
1051
61.686
161.429
194.354
156.42


8007
N
NE
ARG
1051
62.755
162.365
194.683
156.42


8008
C
CZ
ARG
1051
62.581
163.655
194.935
156.42


8009
N
NH1
ARG
1051
61.374
164.184
195.025
156.42


8010
N
NH2
ARG
1051
63.644
164.417
195.173
156.42


8011
N
N
GLU
1052
59.888
159.932
190.246
160.07


8012
C
CA
GLU
1052
58.764
159.048
189.957
160.07


8013
C
C
GLU
1052
58.962
158.323
188.634
160.07


8014
O
O
GLU
1052
58.665
157.13
188.521
160.07


8015
C
CB
GLU
1052
57.457
159.84
189.944
160.07


8016
C
CG
GLU
1052
56.213
158.964
189.922
160.07


8017
C
CD
GLU
1052
55.971
158.25
191.237
160.07


8018
O
OE1
GLU
1052
56.411
158.764
192.286
160.07


8019
O
OE2
GLU
1052
55.341
157.171
191.222
160.07


8020
N
N
ALA
1053
59.452
159.032
187.616
158.31


8021
C
CA
ALA
1053
59.713
158.379
186.334
158.31


8022
C
C
ALA
1053
60.752
157.267
186.474
158.31


8023
O
O
ALA
1053
60.546
156.141
185.994
158.31


8024
C
CB
ALA
1053
60.167
159.412
185.303
158.31


8025
N
N
VAL
1054
61.868
157.56
187.146
154.17


8026
C
CA
VAL
1054
62.92
156.56
187.313
154.17


8027
C
C
VAL
1054
62.417
155.39
188.152
154.17


8028
O
O
VAL
1054
62.732
154.225
187.876
154.17


8029
C
CB
VAL
1054
64.177
157.204
187.927
154.17


8030
C
CG1
VAL
1054
65.245
156.153
188.184
154.17


8031
C
CG2
VAL
1054
64.713
158.294
187.011
154.17


8032
N
N
ARG
1055
61.62
155.678
189.182
155.46


8033
C
CA
ARG
1055
61.1
154.619
190.035
155.46


8034
C
C
ARG
1055
60.08
153.759
189.304
155.46


8035
O
O
ARG
1055
59.987
152.56
189.569
155.46


8036
C
CB
ARG
1055
60.492
155.218
191.303
155.46


8037
C
CG
ARG
1055
60.155
154.189
192.363
155.46


8038
C
CD
ARG
1055
61.421
153.657
193.014
155.46


8039
N
NE
ARG
1055
62.064
154.643
193.874
155.46


8040
C
CZ
ARG
1055
61.718
154.88
195.132
155.46


8041
N
NH1
ARG
1055
60.733
154.216
195.715
155.46


8042
N
NH2
ARG
1055
62.378
155.805
195.823
155.46


8043
N
N
THR
1056
59.315
154.339
188.378
160.35


8044
C
CA
THR
1056
58.412
153.528
187.568
160.35


8045
C
C
THR
1056
59.191
152.643
186.603
160.35


8046
O
O
THR
1056
58.804
151.494
186.35
160.35


8047
C
CB
THR
1056
57.436
154.425
186.809
160.35


8048
O
OG1
THR
1056
56.6
155.12
187.743
160.35


8049
C
CG2
THR
1056
56.558
153.595
185.887
160.35


8050
N
N
LEU
1057
60.295
153.159
186.059
154.78


8051
C
CA
LEU
1057
61.16
152.309
185.245
154.78


8052
C
C
LEU
1057
61.704
151.146
186.071
154.78


8053
O
O
LEU
1057
61.708
149.993
185.616
154.78


8054
C
CB
LEU
1057
62.29
153.148
184.643
154.78


8055
C
CG
LEU
1057
63.241
152.54
183.606
154.78


8056
C
CD1
LEU
1057
63.798
153.638
182.713
154.78


8057
C
CD2
LEU
1057
64.379
151.784
184.258
154.78


8058
N
N
LEU
1058
62.138
151.426
187.302
155.36


8059
C
CA
LEU
1058
62.588
150.361
188.195
155.36


8060
C
C
LEU
1058
61.456
149.401
188.544
155.36


8061
O
O
LEU
1058
61.692
148.205
188.746
155.36


8062
C
CB
LEU
1058
63.184
150.958
189.47
155.36


8063
C
CG
LEU
1058
64.472
151.767
189.324
155.36


8064
C
CD1
LEU
1058
64.853
152.405
190.649
155.36


8065
C
CD2
LEU
1058
65.589
150.869
188.826
155.36


8066
N
N
GLY
1059
60.228
149.91
188.647
159.8


8067
C
CA
GLY
1059
59.096
149.045
188.931
159.8


8068
C
C
GLY
1059
58.812
148.079
187.799
159.8


8069
O
O
GLY
1059
58.535
146.899
188.03
159.8


8070
N
N
TYR
1060
58.872
148.567
186.558
162.62


8071
C
CA
TYR
1060
58.85
147.65
185.423
162.62


8072
C
C
TYR
1060
60.082
146.757
185.39
162.62


8073
O
O
TYR
1060
60.064
145.719
184.718
162.62


8074
C
CB
TYR
1060
58.699
148.411
184.106
162.62


8075
C
CG
TYR
1060
57.255
148.638
183.717
162.62


8076
C
CD1
TYR
1060
56.231
148.431
184.632
162.62


8077
C
CD2
TYR
1060
56.912
149.022
182.428
162.62


8078
C
CE1
TYR
1060
54.908
148.623
184.282
162.62


8079
C
CE2
TYR
1060
55.591
149.215
182.068
162.62


8080
C
CZ
TYR
1060
54.594
149.015
182.998
162.62


8081
O
OH
TYR
1060
53.279
149.207
182.644
162.62


8082
N
N
GLY
1061
61.149
147.134
186.092
157.95


8083
C
CA
GLY
1061
62.197
146.199
186.449
157.95


8084
C
C
GLY
1061
63.443
146.243
185.597
157.95


8085
O
O
GLY
1061
64.35
145.431
185.82
157.95


8086
N
N
TYR
1062
63.529
147.154
184.634
158.59


8087
C
CA
TYR
1062
64.717
147.238
183.793
158.59


8088
C
C
TYR
1062
65.838
147.862
184.613
158.59


8089
O
O
TYR
1062
65.927
149.085
184.751
158.59


8090
C
CB
TYR
1062
64.414
148.03
182.526
158.59


8091
C
CG
TYR
1062
63.459
147.295
181.614
158.59


8092
C
CD1
TYR
1062
63.201
145.944
181.805
158.59


8093
C
CD2
TYR
1062
62.808
147.945
180.575
158.59


8094
C
CE1
TYR
1062
62.33
145.261
180.993
158.59


8095
C
CE2
TYR
1062
61.93
147.263
179.749
158.59


8096
C
CZ
TYR
1062
61.698
145.919
179.966
158.59


8097
O
OH
TYR
1062
60.831
145.216
179.163
158.59


8098
N
N
ASN
1063
66.697
147.01
185.162
149.24


8099
C
CA
ASN
1063
67.651
147.463
186.163
149.24


8100
C
C
ASN
1063
68.742
148.324
185.538
149.24


8101
O
O
ASN
1063
69.173
148.103
184.4
149.24


8102
C
CB
ASN
1063
68.257
146.275
186.917
149.24


8103
C
CG
ASN
1063
68.888
145.249
186
149.24


8104
O
OD1
ASN
1063
68.927
145.419
184.786
149.24


8105
N
ND2
ASN
1063
69.389
144.168
186.586
149.24


8106
N
N
LEU
1064
69.174
149.328
186.299
148.93


8107
C
CA
LEU
1064
70.152
150.31
185.841
148.93


8108
C
C
LEU
1064
71.554
149.788
186.116
148.93


8109
O
O
LEU
1064
72.132
150.039
187.175
148.93


8110
C
CB
LEU
1064
69.935
151.655
186.527
148.93


8111
C
CG
LEU
1064
68.883
152.625
185.984
148.93


8112
C
CD1
LEU
1064
69.27
153.071
184.588
148.93


8113
C
CD2
LEU
1064
67.49
152.027
185.996
148.93


8114
N
N
GLU
1065
72.114
149.062
185.149
146.78


8115
C
CA
GLU
1065
73.533
148.72
185.186
146.78


8116
C
C
GLU
1065
74.321
149.974
184.815
146.78


8117
O
C
GLU
1065
74.729
150.19
183.672
146.78


8118
C
CB
GLU
1065
73.842
147.553
184.26
146.78


8119
C
CG
GLU
1065
73.48
146.193
184.825
146.78


8120
C
CD
GLU
1065
72.072
145.77
184.478
146.78


8121
O
OE1
GLU
1065
71.406
146.494
183.709
146.78


8122
O
OE2
GLU
1065
71.637
144.708
184.968
146.78


8123
N
N
ALA
1066
74.505
150.823
185.811
144.43


8124
C
CA
ALA
1066
75.123
152.123
185.638
144.43


8125
C
C
ALA
1066
76.524
152.142
186.239
144.43


8126
O
O
ALA
1066
76.804
151.426
187.204
144.43


8127
C
CB
ALA
1066
74.273
153.203
186.299
144.43


8128
N
N
PRO
1067
77.421
152.953
185.688
145.31


8129
C
CA
PRO
1067
78.74
153.119
186.295
145.31


8130
C
C
PRO
1067
78.689
154.096
187.461
145.31


8131
O
O
PRO
1067
77.736
154.858
187.634
145.31


8132
C
CB
PRO
1067
79.58
153.677
185.144
145.31


8133
C
CG
PRO
1067
78.601
154.498
184.37
145.31


8134
C
CD
PRO
1067
77.268
153.781
184.478
145.31


8135
N
N
ASP
1068
79.743
154.057
188.268
145.93


8136
C
CA
ASP
1068
79.905
154.978
189.385
145.93


8137
C
C
ASP
1068
80.904
156.065
189.003
145.93


8138
O
O
ASP
1068
81.915
155.79
188.347
145.93


8139
C
CB
ASP
1068
80.317
154.235
190.661
145.93


8140
C
CG
ASP
1068
81.716
153.631
190.591
145.93


8141
O
OD1
ASP
1068
82.297
153.525
189.492
145.93


8142
O
OD2
ASP
1068
82.241
153.254
191.66
145.93


8143
N
N
GLN
1069
80.592
157.305
189.368
145.39


8144
C
CA
GLN
1069
81.451
158.443
189.052
145.39


8145
C
C
GLN
1069
82.436
158.621
190.199
145.39


8146
O
O
GLN
1069
82.098
159.181
191.245
145.39


8147
C
CB
GLN
1069
80.622
159.701
188.821
145.39


8148
C
CG
GLN
1069
81.454
160.945
188.555
145.39


8149
C
CD
GLN
1069
82.577
160.693
187.567
145.39


8150
O
OE1
GLN
1069
82.348
160.212
186.457
145.39


8151
N
NE2
GLN
1069
83.801
161.013
187.97
145.39


8152
N
N
ASP
1070
83.659
158.138
190.004
148.64


8153
C
CA
ASP
1070
84.686
158.279
191.023
148.64


8154
C
C
ASP
1070
85.1
159.739
191.161
148.64


8155
O
O
ASP
1070
84.97
160.537
190.229
148.64


8156
C
CB
ASP
1070
85.901
157.417
190.68
148.64


8157
C
CG
ASP
1070
85.666
155.945
190.951
148.64


8158
O
OD1
ASP
1070
85.912
155.503
192.093
148.64


8159
O
OD2
ASP
1070
85.235
155.23
190.023
148.64


8160
N
N
HIS
1071
85.591
160.087
192.348
164.22


8161
C
CA
HIS
1071
86.035
161.451
192.598
164.22


8162
C
C
HIS
1071
87.214
161.797
191.698
164.22


8163
O
O
HIS
1071
88.188
161.044
191.61
164.22


8164
C
CB
HIS
1071
86.42
161.619
194.068
164.22


8165
C
CG
HIS
1071
86.729
163.032
194.452
164.22


8166
N
ND1
HIS
1071
87.984
163.585
194.313
164.22


8167
C
CD2
HIS
1071
85.944
164.007
194.969
164.22


8168
C
CE1
HIS
1071
87.959
164.838
194.729
164.22


8169
N
NE2
HIS
1071
86.733
165.12
195.132
164.22


8170
N
N
ALA
1072
87.12
162.94
191.027
178.88


8171
C
CA
ALA
1072
88.154
163.405
190.114
178.88


8172
C
C
ALA
1072
88.796
164.668
190.67
178.88


8173
O
O
ALA
1072
88.094
165.609
191.055
178.88


8174
C
CB
ALA
1072
87.576
163.671
188.724
178.88


8175
N
N
ALA
1073
90.125
164.684
190.713
194.36


8176
C
CA
ALA
1073
90.842
165.86
191.184
194.36


8177
C
C
ALA
1073
90.707
167
190.183
194.36


8178
O
O
ALA
1073
90.858
166.806
188.973
194.36


8179
C
CB
ALA
1073
92.316
165.528
191.413
194.36


8180
N
N
ARG
1074
90.419
168.194
190.694
201.94


8181
C
CA
ARG
1074
90.271
169.39
189.877
201.94


8182
C
C
ARG
1074
91.183
170.475
190.43
201.94


8183
O
O
ARG
1074
91.153
170.764
191.631
201.94


8184
C
CB
ARG
1074
88.811
169.855
189.85
201.94


8185
C
CG
ARG
1074
88.539
171.055
188.957
201.94


8186
C
CD
ARG
1074
87.057
171.407
188.957
201.94


8187
N
NE
ARG
1074
86.578
171.813
190.273
201.94


8188
C
CZ
ARG
1074
86.686
173.039
190.764
201.94


8189
N
NH1
ARG
1074
87.237
174.018
190.065
201.94


8190
N
NH2
ARG
1074
86.224
173.293
191.986
201.94


8191
N
N
ALA
1075
91.995
171.07
189.551
202.15


8192
C
CA
ALA
1075
93.01
172.021
189.992
202.15


8193
C
C
ALA
1075
92.4
173.268
190.621
202.15


8194
O
O
ALA
1075
93.055
173.921
191.442
202.15


8195
C
CB
ALA
1075
93.909
172.411
188.818
202.15


8196
N
N
GLU
1076
91.18
173.633
190.216
190.56


8197
C
CA
GLU
1076
90.386
174.687
190.843
190.56


8198
C
C
GLU
1076
90.947
176.073
190.528
190.56


8199
O
O
GLU
1076
90.291
177.091
190.778
190.56


8200
C
CB
GLU
1076
90.29
174.428
192.359
190.56


8201
C
CG
GLU
1076
89.915
175.613
193.241
190.56


8202
C
CD
GLU
1076
88.448
175.98
193.136
190.56


8203
O
OE1
GLU
1076
87.635
175.095
192.8
190.56


8204
O
OE2
GLU
1076
88.11
177.156
193.386
190.56






1atom site ID number as assigned in mmCIF file for RCSB PBD structure 7UA1;




2element symbol representing atom species;




3atom identifier assigned in mmCIF file for RCSB PBD structure 7UA1;




4encompassing residue type;




5encompassing residue number;




6-8Atom-site coordinates in angstroms specified according to a set of orthogonal Cartesian axes related to the cell axes; Isotropic atomic displacement parameter, or equivalent isotropic atomic displacement parameter, Beq, calculated from the anisotropic displacement parameters, where: Beq = (1/3) sumi[sumj(Bij Ai Aj a*i a*j)]; A = the real space cell lengths; a* = the reciprocal space cell lengths; and Bij = 8 pi2 Uij.














TABLE 4







Three-dimensional atomic coordinates of Compound 1.














type_
label_



B_iso_


Id1
symbol2
atom_id3
Cartn_x6
Cartn_y7
Cartn_z8
or_equiv9
















138400
C
C01
50.572
176.272
179.982
0.5


138401
C
C03
50.159
173.915
179.952
0.5


138402
C
C04
50.151
173.364
181.224
0.5


138403
C
C05
50.624
172.075
181.449
0.5


138404
C
C06
51.112
171.343
180.349
0.5


138405
C
C07
51.122
171.906
179.077
0.5


138406
C
C08
50.643
173.184
178.878
0.5


138407
C
C10
53.121
170.019
181.711
0.5


138408
C
C11
52.652
170.105
183.135
0.5


138409
C
C13
50.617
171.488
182.839
0.5


138410
C
C14
52.107
171.869
184.785
0.5


138411
C
C15
52.944
173.123
184.842
0.5


138412
C
C16
52.464
174.307
184.309
0.5


138413
C
C17
53.225
175.461
184.339
0.5


138414
C
C18
54.492
175.447
184.897
0.5


138415
C
C19
54.981
174.265
185.425
0.5


138416
C
C20
54.215
173.112
185.392
0.5


138417
C
C21
55.317
176.694
184.929
0.5


138418
N
N12
51.976
171.382
183.4  
0.5


138419
0
O02
49.681
175.185
179.755
0.5


138420
0
O22
55.932
177.008
183.895
0.5


138421
0
O23
55.341
177.346
185.986
0.5


138422
S
S09
51.724
169.708
180.612
0.5






1-3,6-9See description for TABLE 3 above.














TABLE 5







Three-dimensional atomic coordinates of ATP.














type_
label_



B_iso_


Id1
symbol2
atom_id3
Cartn_x6
Cartn_y7
Cartn_z8
or_equiv9
















138369
P
PG
57.947
176.674
179.663
139.1


138370
O
O1G
58.257
178.124
179.946
139.1


138371
O
O2G
58.591
176.144
178.405
139.1


138372
O
O3G
58.08
175.792
180.877
139.1


138373
P
PB
55.52
175.306
179.197
139.1


138374
O
O1B
54.28
175.621
178.396
139.1


138375
O
O2B
56.411
174.167
178.771
139.1


138376
O
O3B
56.375
176.666
179.323
139.1


138377
P
PA
53.956
173.944
181.032
139.1


138378
O
O1A
53.819
173.014
179.856
139.1


138379
O
O2A
52.75
174.679
181.516
139.1


138380
O
O3A
55.083
175.036
180.716
139.1


138381
O
O5′
54.615
173.193
182.284
139.1


138382
C
C5'
55.879
173.641
182.753
139.1


138383
C
C4′
56.876
172.499
182.649
139.1


138384
O
O4′
56.489
171.592
181.613
139.1


138385
C
C3′
58.263
173.017
182.302
139.1


138386
O
O3′
59.167
172.826
183.394
139.1


138387
C
C2′
58.716
172.217
181.1 
139.1


138388
O
O2
59.974
171.595
181.367
139.1


138389
C
C1′
57.633
171.177
180.863
139.1


138390
N
N9
57.35
171.147
179.412
139.1


138391
C
C8
58.24
171.501
178.47
139.1


138392
N
N7
57.716
171.381
177.229
139.1


138393
C
C5
56.46
170.936
177.373
139.1


138394
C
C6
55.361
170.597
176.454
139.1


138395
N
N6
55.545
170.724
175.122
139.1


138396
N
N1
54.2
170.162
176.986
139.1


138397
C
C2
54.043
170.048
178.314
139.1


138398
N
N3
55.006
170.333
179.203
139.1


138399
C
C4
56.217
170.782
178.809
139.1






1-3,6-9See description for TABLE 3 above.







Comparison of the pore region from all the structures showed no differences in the closed states and no differences in the open states. The comparison suggests that the methodology implemented is reproducible. FIGS. 6A-6C depict aligned atomic models of all structures focusing on the pore and TM domain. FIGS. 7A-7F show pore radii estimation for each structure calculated with HOLE. Channel coordinates are arbitrary and correlation among structures are approximate. Visual and HOLE analysis suggest that no differences are found in the pore of the closed states and no differences are found in the pore of the open states.


Of the three activators present in the sample, only ATP was detected in the closed state structures (FIG. 8A). All three activators Ca2+, ATP, and xanthine were present in the open state structures (FIG. 8B), suggesting that the binding of the additional activators, Ca2+ and xanthine, promotes opening of the channel. FIG. 8A and FIG. 8B depict models of RyR2 with the respective cryo-EM maps centered on the ligand binding site of the closed (FIG. 8A) and open (FIG. 8B) state of representative RyR2 structures.


Xanthine occupied the site formed by W4645, I4926, and Y4944. To confirm the functional role of xanthine, single-channel experiments were performed showing that, at presumed physiological concentrations during exercise (10 μM), a ˜25-fold increase was observed in the open probability of RyR2 in the presence of xanthine (see FIG. 22A, FIG. 22B, EXAMPLE 7).


To understand the mechanism underlying CPVT better, structures of PKA-phosphorylated RyR2 (open, closed), PKA-phosphorylated RyR2-R2474S (“primed”), PKA-phosphorylated RyR2-R2474S bound to Compound 1 (closed), and PKA-phosphorylated RyR2-R2474S bound to CaM (closed) were compared. (FIG. 9, FIG. 10).



FIGS. 9A-9D depict cryo-EM reconstructions of human RyR2 showing that the CPVT mutant RyR2-R2474S predisposes the channel to assume the primed state, and treatment with the ryanodine receptor channel modulator Compound 1 and CaM restores the channel back toward the closed state.



FIG. 9A shows overlapped models of open PKA-phosphorylated RyR2 (P-RyR2-0, PDB: 7U9R, yellow) and closed PKA-phosphorylated RyR2 (P-RyR2-C, PDB: 7U9Q, gray). The arrows show that the cytosolic shell of the PKA-phosphorylated RyR2 shifted downward and outward when going from the closed to the open state. To facilitate visualization, only the front protomer is shown in colors, while the other three protomers are shown as gray transparent volumes. The positions of the sarcoplasmic reticular membranes are shown as black discs. Conditions included 10 mM ATP, 150 nM free Ca2+, and 500 μM xanthine.



FIG. 9B shows overlapped models of closed PKA-phosphorylated RyR2 (P-RyR2-C, PDB: 7U9Q, gray) and primed PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S-Pr, PDB: 7U9X, magenta). The arrows show that the cytosolic shell of RyR2-R2474S shifted downward and outward compared to closed PKA-phosphorylated RyR2, similar to the structural changes observed for PKA-phosphorylated RyR2 going from the closed state to the open state. This intermediate between closed and open states is defined as the primed state.



FIG. 9C shows overlapped models of primed PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S-Pr, PDB: 7U9X, magenta) and closed PKA-phosphorylated RyR2-R2474S+Compound 1 (P-RyR2-R2474S+Cpd1-C, PDB: 7UA1, cyan). The arrows show that the cytosolic shell of PKA-phosphorylated RyR2-R2474S+Compound 1 cytosolic domain shifted upward and inward compared to the RyR2-R2474S reversing the primed state back toward the closed state.



FIG. 9D shows overlapped models of primed PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S-Pr, PDB: 7U9X, magenta) and closed PKA-phosphorylated RyR2-R2474S+CaM (P-RyR2-R2474S+CaM-C, PDB: 7UA3, cyan). Similar to the effects of Compound 1, CaM reversed the primed state back toward the closed state.



FIGS. 10A-10K depict pairwise comparisons of the cytosolic domains of all structures. Domains are labelled. Conformational changes are shown with arrows. Sizes of the arrows represent the amount of changes observed.


As seen, the cytosolic shell of the PKA-phosphorylated RyR2 was shifted downward and outward when going from the closed state (P-RyR2-C, PDB: 7U9Q) to the open state (P-RyR2-0 PDB: 7U9R) (FIG. 9A and FIG. 10B).


The cytosolic shell of primed PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S-Pr, PDB: 7U9X) was shifted downward and outward compared to closed PKA-phosphorylated RyR2. This observation suggests that the CPVT mutant RyR2-R2474S is in a primed state (FIG. 9B and FIG. 10F). This primed state presents a structure that is approximately halfway between the closed and open states of PKA-phosphorylated RyR2. The same is true for the open state; the cytosolic shell of open PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S-O, PDB: 7U9Z) was also shifted downward and outward compared to open PKA-phosphorylated RyR2 (P-RyR2-0, FIG. 10G). This observation suggests that the CPVT-related cytosolic shell destabilization is independent of the state of the pore.


The PKA-phosphorylated RyR2-R2474S in the presence of Compound 1 (P-RyR2-R2474S+Cpd1-C; PDB: 7UA1)—a benzothiazepine derivative that effectively lessens a likelihood of ventricular tachycardiac and sudden cardiac death in murine models of CPVT-exhibited an upward and inward shift of the cytosolic shell compared to primed PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S-Pr), thus reversing the primed state back toward the closed state of the channel (FIG. 9C and FIG. 10H). The PKA-phosphorylated RyR2-R2474S in the presence of CaM (P-RyR2-R2474S+CaM-C; PDB: 7UA3) exhibited a similar but less pronounced shift of the cytosolic shell upward and inward compared to primed PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S-Pr), also reversing the primed state back toward the closed state of the channel similar to the effects of Compound 1 (FIG. 9D and FIG. 10J). A comparison of the cytosolic shell of primed PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S-Pr, PDB: 7U9X) and the cytosolic shell of open PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S-O, PDB: 7U9Z) are depicted in FIG. 10I.


Compound I Stabilization of Closed State of the PKA-Phosphorylated RyR2-R2474S CPVT Variant.


FIG. 11A depicts cryo-EM maps of closed PKA-phosphorylated RyR2 (gray) and primed PKA-phosphorylated RyR2-R2474S (magenta) from the side (left) and top (right) views. Conformation changes are shown with arrows. FIG. 13A depicts normalized differences in RMSD of the primed PKA-phosphorylated RyR2-R2474S. FIG. 11C provides a close-up view of the region around residue 2474 of closed PKA-phosphorylated RyR2 (left) and primed PKA-phosphorylated RyR2-R2474S (right). Distances are measured between Cβ atoms. FIG. 11B depicts aligned models of closed PKA-phosphorylated RyR2 (PDB: 7U9Q, gray), open PKA-phosphorylated RyR2 (PDB: 7U9R, yellow), and primed PKA-phosphorylated RyR2-R2474S (PDB: 7U9X, magenta). Conformational changes are shown with arrows. Distances between closed PKA-phosphorylated RyR2 and primed PKA-phosphorylated RyR2-R2474S, and between closed and open PKA-phosphorylated RyR2 (in parentheses) are labeled.



FIG. 12A and FIG. 12B are identical to FIG. 11A and FIG. 11B respectively, except that each includes closed PKA-phosphorylated RyR2-R2474S+Compound 1 (P-RyR2-R2474S+Cpd1-C, PDB: 7UA1, cyan). In FIG. 12B, Distances between primed PKA-phosphorylated RyR2-R2474S and closed PKA-phosphorylated RyR2-R2474S+Compound 1 are labeled. Changes introduced by the R2474S mutation are partially reversed by the addition of Compound 1, shifting the structure towards a closed state. In FIG. 12A, right panel, the densities of Compound 1 and BSol1-RY1&2 interface are highlighted. FIG. 13B depicts normalized differences in RMSD of the closed PKA-phosphorylated RyR2-R2474S+Cpd1.



FIG. 12C shows aligned models of closed PKA-phosphorylated RyR2 (P-RyR2-C, PDB: 7U9Q, gray), primed PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S-Pr, PDB: 7U9X, magenta), and closed PKA-phosphorylated RyR2-R2474S+Compound 1 (P-RyR2-R2474S+Cpd1-C, PDB: 7UA1, cyan). Conformational changes of the RY1&2 and BSol domains are shown with arrows.


Visual inspection of the cryo-EM maps shows conformation changes that suggest a primed state of the PKA-phosphorylated RyR2-R2474S channels (FIG. 11A). To quantify the structural changes in the primed PKA-phosphorylated RyR2-R2474S channels, the normalized differences in root mean square deviation (RMSD) between the primed PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S-Pr, PDB: 7U9X) and closed (P-RyR2-R2474S-C PDB: 7U9Q) and open (P-RyR2-0, PDB: 7U9R) PKA-phosphorylated RyR2 (FIG. 13A and TABLE 6) were measured.


TABLE 6 shows the differences in normalized RMSD analysis between primed PKA RyR2-R2474S vs. closed/open PKA RyR2.














TABLE 6





RMSD Cα (Å)
BSol2
BSol
NTD
SPRY
CSol







primed P-RyR2-R2474S
5.17 ±
3.28 ±
1.49 ±
1.38 ±
0.50


(PDB:7U9X) vs closed
0.50
0.50
0.50
0.50


P-RyR2-C (PDB:7U9Q)


primed P-RyR2-R2474S
2.23 ±
1.55 ±
0.87 ±
0.77 ±
0.40


(PDB:7U9X) vs P-RyR2-O
0.40
0.40
0.40
0.40


(PDB:7U9R)


Normalized difference
0.70 ±
0.68 ±
0.63 ±
0.64 ±


in RMSD
0.09
0.14
0.27
0.30









TABLE 7 shows the differences in normalized RMSD analysis between Closed PKA RyR2-R2474S+Compound 1 vs. closed/open PKA RyR2.














TABLE 7





RMSD Cα (Å)
BSol2
BSol
NTD
SPRY
CSol







P-RyR2-R2474S + Cpd1*
3.08 ±
1.61 ±
1.20 ±
0.88 ±
0.53


(PDB:7UA1) vs P-RyR2-C
0.53
0.53
0.53
0.53


(PDB:7U9Q)


P-RyR2-R2474S + Cpd1*
4.63 ±
3.36 ±
1.66 ±
1.25 ±
0.41


(PDB:7UA1) vs P-RyR2-O
0.41
0.41
0.41
0.41


(PDB:7U9R)


Normalized difference
0.40 ±
0.32 ±
0.42 ±
0.41 ±


in RMSD
0.08
0.11
0.21
0.28





*Cpd 1 shifts the primed P-RyR2-R2474S toward the closed state. Thus, P-RyR2-R2474S + Cpd1 can be referred to herein as having a closed channel pore.






TABLE 8 shows the differences in normalized RMSD analysis between Closed PKA RyR2 vs. closed/open dephosphorylated RyR2.














TABLE 8





RMSD Cα (Å)
BSol2
BSol
NTD
SPRY
CSol







P-RyR2-C (PDB:7U9Q) vs
1.73 ±
0.74 ±
0.58 ±
1.29 ±
0.50


DeP-RyR2-C (PDB:7UA5)
0.50
0.50
0.50
0.50


P-RyR2-C (PDB:7U9Q) vs
5.38 ±
2.19 ±
5.09 ±
6.24 ±
0.77


DeP-RyR2-O (PDB:7UA9)
0.77
0.77
0.77
0.77


Normalized difference
0.24 ±
0.25 ±
0.10 ±
0.17 ±


in RMSD
0.08
0.19
0.09
0.07









TABLE 9 shows the differences in normalized RMSD analysis between closed state with stabilized RY3&4 vs. closed state with destabilized RY3&4/open state.














TABLE 9





RMSD Cα (Å)
SPRY3
calstabin-2
JSol
CSol
BSol1







closed state with
1.80 ±
2.40 ±
1.94 ±
1.66 ±
0.36


destabilized RY3&4 vs
0.36
0.36
0.36
0.36


closed state with


stabilized RY3&4


open state vs closed
3.90 ±
3.69 ±
2.65 ±
3.75 ±
0.61


state with stabilized
0.61
0.61
0.61
0.61


RY3&4


Normalized difference
0.32 ±
0.39 ±
0.42 ±
0.31 ±


in RMSD
0.07
0.07
0.10
0.08









In TABLES 6-9, RMSD difference values close to 0 indicated that the conformation is similar to the closed state, whereas values close to 1 indicated that the conformation is similar to the open state. Normalization allowed the direct comparison between different domains. RMSD analysis showed an average value of ˜0.7, which indicates that the cytosolic domains of the primed PKA-phosphorylated RyR2-R2474S were in nearly open positions, reducing the energetic barrier of the primed state to adopt a fully open state. Thus, the primed PKA-phosphorylated RyR2-R2474S is more readily activated and promotes an SR Ca2+ leak during diastole when the activating [Ca2+]cyt is very low (˜150 nM) and the WT channels are tightly closed.


Analyzing in detail the region of the mutation, the Arg to Ser mutation was readily identified because of the shortened side-chain density for the mutant residue S2474 compared to the WT residue R2474 (FIG. 11C). R2474 stabilizes the surrounding structure via interactions with S2312 and E2405. In the mutant channel, this network is disrupted and the distance between residues S2312 and E2405 increases from 5.8 to 6.9 Å (FIG. 11C). The increased distances observed in the RyR2-R2474S mutant channel affects the helix-loop-helix motif formed by residues 2406 to 2418, which propagates to the adjacent NTD-A domain (FIG. 11B). Therefore, a single mutation destabilizes the interaction between the NTD and the BSol, which leads to destabilization of the entire cytosolic shell (NTD, SPRY, JSol, and BSol domains).



FIG. 14A and FIG. 14B depict cryo-EM maps of local refinement cryoSPRAC jobs before 3D variability of primed PKA RyR2-R2474S (magenta), and closed PKA RyR2-R2474S+Compound 1 (cyan) from different views and map levels. For better interpretation, a softening gaussian was applied to the cryo-EM maps that make evident the different size of the densities inside the RY1&2 cleft. ATP and Compound 1 potential densities are labelled.



FIG. 14C shows the closed PKA RyR2 model (PDB:7U9Q) with the aligned cryo-EM map centered on the RY1&2 domain from the top (top) and side (middle) views. Clear density is observed in the cleft of the RY1&2 domain. A molecule of ATP was fitted in the density as previously observed in RyR1. (bottom) Atomic model centered on the RY1&2 domain showing the residues that are potentially involved in ATP binding.



FIG. 14D shows a close up of closed PKA R2474S RyR2+Compound 1 (P-RyR2-R2474S-C, PDB:7UA1) centered on the ryanodine receptor channel modulator binding site. The precise placement of ATP and Compound 1 was hampered due to local low resolution.



FIG. 14E shows closed PKA RyR2-R2474S+Compound 1 (P-RyR2-R2474S-C, PDB:7UA1) centered on the BSol1-RY1&2 interface. Candidate residues involved in the BSol1-RY1&2 interaction are labeled. FIG. 14F depicts the same comparison provided in FIG. 14E but with distances between sidechains of candidate residues labeled in yellow.


Incubation with Compound 1 partially reversed the primed state of PKA-phosphorylated RyR2-R2474S, placing the channel in a state closer to that of the closed PKA-phosphorylated RyR2 (FIG. 12A and FIG. 12B). In this case, the RMSD analysis of closed PKA-phosphorylated RyR2-R2474S+Compound 1 (P-RyR2-R2474S-C, PDB: 7UA1) showed a reduction of more than 50% toward the closed state in some domains compared to primed PKA-phosphorylated RyR2-R2474S (P-RyR2-R2474S-Pr) (FIG. 13B and TABLE 7). Analyzing in detail the cryo-EM map of closed PKA-phosphorylated RyR2-R2474S in the presence of Compound 1, a clear density in the cleft of the RY1&2 domain was detected (FIG. 12A, FIG. 14A, and FIG. 14B). Therefore, Compound 1, when bound to PKA-phosphorylated RyR2-R2474S channels, shifts the “primed” state of the P-RyR2-R2474S channel pores at least partially towards a closed state.


Analysis of the rest of the cryo-EM map showed no additional densities that could correspond to Compound 1. Compound 1 density was absent in the particles in the open state. This observation suggests that Compound 1 and congeners thereof act by stabilizing the closed state of RyR2. In the absence of Compound 1, the RY1&2 domain of RyR2 showed a weak density that was attributed to one ATP molecule (FIG. 14A and FIG. 14C) (PDB: 6UHH). In the presence of Compound 1, this density was stronger and larger than one ATP molecule and suggests that at least two molecules are present (ATP and Compound 1; FIG. 14D). In addition, the RY1&2 domain adopts a conformation that closes around both ATP and Compound 1 (FIG. 12C). In comparing PKA-phosphorylated RyR2-R2474S atomic models in the absence and presence of Compound 1 (PDB: 7U9X versus PDB: 7UA1), the largest changes were observed not in the contiguous SPRY1 domain, but instead in the adjacent BSol domain (FIG. 12C). The interface between the BSol1 and the RY1&2 domains showed enhanced density. This observation suggests that the transduction of the signal from the Compound 1-loaded RY1&2 domain is through the stabilization of the BSol1 domains (FIG. 12A, FIG. 14B, and FIG. 14E). On the basis of the model, the interface between the BSol1 and RY1&2 domains would be stabilized by His2995 in the BSol1 domain and Asp1070 in the RY1&2 domain. Arg2988 (BSol1) and His1071 (RY1&2) are also available to be involved in strengthening the BSol1-RY1&2 interaction (FIG. 14E and FIG. 14F).


CaM Stabilization of the Closed PKA-Phosphorylated RyR2-R2474S CPVT Variant.

To ascertain the role of CaM in CPVT, the structure of closed PKA-phosphorylated RyR2+CaM (P-RyR2-C, PDB: 7U9T) and closed PKA-phosphorylated RyR2-R2474S+CaM (P-RyR2-R2474S+CaM-C, PDB: 7UA3) was analyzed. FIG. 15, Panel A shows aligned cryo-EM maps of closed PKA-phosphorylated RyR2 (P-RyR2-C, gray) and closed PKA-phosphorylated RyR2+CaM (cyan). FIG. 15, Panel B depicts JSol and CSol models of closed PKA-phosphorylated RyR2 (PDB: 7U9Q, gray) and open PKA-phosphorylated RyR2 (PDB: 7U9R, yellow). Conformation changes are shown with arrows. FIG. 15, Panel C depicts JSol and CSol models of closed PKA-phosphorylated RyR2 (PDB: 7U9Q, gray) and closed PKA-phosphorylated RyR2+CaM (PDB: 7U9T, cyan). Conformation changes are shown with arrows. FIG. 15, Panel D shows aligned cryo-EM maps of primed PKA-phosphorylated RyR2-R2474S (magenta) and closed PKA-phosphorylated RyR2-R2474S+CaM (cyan). Conformation changes are shown with arrows. CaM reverses the changes in the BSol2 domain introduced by the mutation by stabilizing the BSol3 domain. FIG. 15, Panel E depicts a model with cryo-EM map of closed PKA-phosphorylated RyR2-R2474S+CaM (PDB: 7UA3) centered on the BSol3 domain that is stabilized by CaM.


At a free Ca2+ concentration of 150 nM, CaM binds to PKA-phosphorylated RyR2 and PKA-phosphorylated RyR2-R2474S in the extended conformation that corresponds to the apo-CaM state (FIG. 15, Panel A and FIG. 10E). In the closed PKA-phosphorylated RyR2+CaM (PDB: 7U9T), the apo-CaM N-lobe binds to the BSol1 domain but does not substantially affect the BSol1 conformation. This observation suggests an exclusive binding role for the N-lobe. The C-lobe binds to CaMBD2 or helix α-1 (3594 to 3604) and pushes the JSol domain outward, although this change is not propagated to the rest of the domain. CaM-bound helix α-1 also interacts with the α9 helix of the CSol (3806 to 3816), inducing small changes to the CSol domain in the opposite direction of the open state (FIG. 15, Panel B and Panel C). Although small, the changes observed in the CSol domain show that apo-CaM may counteract signals from the cytosolic domains, such as the phosphorylation of the RY3&4 domain and the changes resulting from CPVT-causing mutations found in the NTD and BSol domains. Not surprisingly, no particles in the open state showed substantial density of bound apo-CaM, confirming that binding of apo-CaM stabilizes the closed state.


In the case of closed PKA-phosphorylated RyR2-R2474S+CaM (P-RyR2-R2474S+CaM-C, PDB: 7UA3), apo-CaM reverses the changes in the BSol2 domain of the closed state introduced by the RyR2-R2474S mutation (FIG. 15, Panel D and FIG. 10J). This observation may be a result of apo-CaM stabilizing the BSol3 domain (FIG. 15, Panel D and Panel E), which interacts with the SPRY2, JSol, and CSol domains stabilizing the whole BSol domain. This finding is surprising because the BSol3 domain was not detected in any previous RyR2 structures. Unlike PKA-phosphorylated RyR2+CaM, apo-CaM bound to the open PKA-phosphorylated RyR2-R2474S+CaM (P-RyR2-R2474S+CaM-0, PDB: 7UA4), where the cytosolic domains were further shifted downward and outward enhancing the open state (FIG. 10K). This observation suggests that, in the presence of the activator xanthine, apo-CaM could also act as an activator of PKA-phosphorylated RyR2-R2474S, in agreement with an increased number of particles found for the open state in this dataset (60% compared to 10 to 20% for the other conditions). The JSol of open PKA-phosphorylated RyR2-R2474S+CaM (P-RyR2-R2474S+CaM-0) (FIG. 10K), which is in direct contact with apo-CaM, was shifted downward, enhancing and stabilizing the open state.


PKA Phosphorylation of RyR2 and the Structure-Function of the RY3&4 Domain.

To determine the effect that PKA treatment has on the structures of RyR2 and the phosphorylation RY3&4 domain, WT RyR2 was pretreated with phosphatase X to obtain a completely dephosphorylated control (FIG. 1A). The structures of the dephosphorylated (DeP-RyR2) and PKA-phosphorylated (P-RyR2) RyR2 channels were compared.



FIG. 16, Panel A illustrates a RMSD analysis of the closed PKA phosphorylated RyR2 (P-RyR2-C, PDB:7U9Q) vs. the closed (DeP-RyR2-C, PDB:7UA5) and open (DeP-RyR2-0, PDB:7UA9) states of dephosphorylated RyR2. The value of the normalized difference in RMSD is shown inside each bar. The analysis was performed on “primed” PKA RyR2-R2474S (P-RyR2-R2474S-P) because this dataset presents the most particles and best local refinement resolution. FIG. 16, Panel B depicts a 3D variability analysis of the RyR2 structures showing that the dynamic behavior of the RY3&4 domain is independent of the phosphorylation state, pore state, and mutation state. FIG. 16, Panel C shows views of the primed PKA RyR2-R2474S model (P-RyR2-R2474S-Pr, PDB:7U9X) with the aligned cryo-EM map centered on the RY3&4 domain. Connecting linkers to the BSol1 domain and missing phosphorylation loop are labeled. FIG. 16, Panel D shows the primed PKA RyR2-R2474S model (P-RyR2-R2474S-Pr, PDB:7U9X) centered on the interface between the RY3&4 and BSol1/SPRY3 domains. Residues that are close enough to generate salt bridges or hydrogen bonds are highlighted. The primary residues involved in the RY3&4-BSol1 interaction are charged residues of the RY3&4 region 2875-2881. The primary residue involved in the RY3&4-SPRY3 interaction is the polar N2830. FIG. 16, Panel E shows the atomic model of the RY3&4 domain of P-RyR2-R2474S-Pr centered on the phosphorylation loop. Distances between the terminal residues of the phosphorylation loop (N2802 and G2820) and the nearest positive residues (R1500 and K1525) are shown in yellow. RyR2-N2802, the closest detectable residue to the phosphorylation site, was 13 Å and 14 Å from RyR2-R1500 and RyR2-K1525, respectively. These distances allowed RyR2-pS2808 to interact with either residue, strengthening the interaction between RY3&4 and SRPY3 and stabilizing this conformation. FIG. 16, Panel F depicts cryo-EM maps of the particles in the closed state with destabilized RY3&4 domain (gray) and stabilized RY3&4 domain (magenta). A downward shift in surrounding domains can be observed. Individual domains are labelled. FIG. 16, Panel G depicts cryo-EM maps of the particles in the open state with destabilized RY3&4 domain (gray) and stabilized RY3&4 domain (magenta). A smaller downward shift in surrounding domains was observed, except for the deP RyR2 where no changes are observed. Individual domains are labelled.


Analysis of the global structure showed that the cytosolic shell of the closed PKA-phosphorylated RyR2 (P-RyR2-C, PDB: 7U9Q) showed a small shift outward and downward compared to the closed dephosphorylated RyR2 (DeP-RyR2-C, PDB: 7UA5; FIGS. 10A-10D). RMSD analysis showed values of ˜0.2 mainly in the BSol and SPRY domains, which are adjacent to the RY3&4 phosphorylation domain where the residue 52808 is located (FIG. 16, Panel A, and TABLE 8). This result suggests that PKA phosphorylation has a priming effect. These changes would reduce the global energetic barrier for reaching the open state, thus sensitizing the channel to CICR. This conformational change is smaller than the one introduced by the CPVT mutation (˜0.2 versus ˜0.7). The cytosolic shell conformation remains closer to the closed state for PKA phosphorylation than for the CPVT mutation, in agreement with the physiological role of PKA phosphorylation and the pathological role of the CPVT mutation.


The RY3&4 phosphorylation domain has not been previously resolved in the cryo-EM structures of any RyRs due to the intrinsic dynamic behavior. Under the hypothesis that this dynamic nature of the RY3&4 domain would be necessary for interacting with modulator enzymes such as PKA and protein phosphatase 1 (PP1), 3D variability analysis centered on the BSol containing the RY3&4 domain was performed.



FIG. 17, Panel A and Panel B are cryo-EM maps of the closed particles with destabilized (gray) and stabilized (magenta) RY3&4 domain. A downward shift in surrounding domains was observed. Individual domains are labeled. FIG. 17, Panel C and Panel D provide different points of view of the cryo-EM maps depicted in Panel A and Panel B, respectively. FIG. 18 shows aligned models of the closed state with destabilized (gray) and stabilized (magenta) RY3&4 domain, and open state (yellow). Models were aligned at the BSol1 domains to facilitate interpretation of conformational changes. When the RY3&4 domain is stabilized, the changes are in the same direction as the open state. Shown with arrows are the distance between the closed state with destabilized RY3&4 domain and the closed state with stabilized RY3&4 domain, and between the closed state with destabilized RY3&4 domain and the open state (in parentheses). FIG. 19 is a chart illustrating an RMSD analysis of the closed state with stabilized RY3&4 domain. This analysis was performed on primed PKA-phosphorylated RyR2-R2474S because this dataset presents the most particles and best local refinement resolution.


This 3D variability analysis revealed two distinct populations: one where the RY3&4 domain was stabilized and could be resolved, and one where there was no substantial density, suggesting that RY3&4 is detached from the BSol domain (FIG. 17, Panels A-D). In other words, in the same RyR2 particle, which has four RY3&4 domains, some of the RY3&4 domains are stabilized and some are destabilized. This behavior was observed in all the structures. This observation suggests that the stabilities are independent of the phosphorylation state, closed/open state, and WT/CPVT mutation (FIG. 16, Panel B). The distribution of stabilized versus destabilized RY3&4 domains is roughly 50/50, but the limitations of this methodology prevent a precise measurement of the particle distribution.


To improve the local cryo-EM map of the RY3&4 domain, symmetry expanded particles with the stabilized RY3&4 domain were clustered and analyzed. The resulting cryo-EM map with a local resolution of 2.90 Å allowed an atomic model of this domain to be constructed with high confidence (FIG. 16, Panel C). On the basis of the model, weak interactions between the RY3&4 and BSol1 domains and almost no interaction between the RY3&4 and SPRY3 domains were observed (FIG. 16, Panel D). Phosphorylation of RyR2-S2808 may strengthen the interaction between the RY3&4 and SPRY3 domains due to the introduction of a salt bridge between the negatively charged RyR2-pS2808 and either the positively charged RyR2-R1500 or RyR2-K1525 in SPRY3 (FIG. 16, Panel E). The same conclusion can be obtained when analyzing the residue RyR2-S2814, which is preferentially phosphorylated by CaM-dependent protein kinase II (CaMKII). Hence, phosphorylation of either RyR2-S2808 or RyR2-S2814 may stabilize this conformation of the RY3&4 phosphorylation domain. However, no extra densities around these residues were found. Thus, the interaction is still very dynamic.


The interaction between the stabilization of the RY3&4 domain and the surrounding RyR2 structure was also analyzed. Comparison to the destabilized RY3&4 domain in the closed state showed that stabilization of the RY3&4 domain increased the distance between the adjacent BSol1 and SPRY3 domains, adopting a conformation that is closer to the open conformation (FIG. 17 and FIG. 16, Panel E). This movement is propagated from the SPRY3 domain to the adjacent JSol and CSol domains. To quantify those changes, normalized difference in RMSD between whole domains (FIG. 18, FIG. 19, and TABLE 9) was compared. The RMSD analysis revealed values in the range of 0.3 to 0.4, confirming that the stabilization of the RY3&4 domain had a priming effect. In the case of the open channels, stabilization of the RY3&4 domain showed no effect on open dephosphorylated RyR2 but exhibited a small effect on open PKA-phosphorylated RyR2 and open PKA-phosphorylated RyR2-R2474S. The observation confirmed that the adopted conformation is similar to the open state and that PKA phosphorylation still has a priming effect on the open states (FIG. 16, Panel G). Binding of accessory proteins to the phosphorylation loop could shift the stabilization distribution of the RY3&4 domain to either completely destabilized or completely stabilized. Phosphomimic substitution at RyR2-S2814 has been shown to induce the formation of an a helix, and this could be a substrate for protein-protein interaction that could further stabilize or destabilize the RY3&4 domain.


Auxiliary Intramembrane Helices.

Two intramembrane helices laterally positioned and encircling the TM domain were observed. FIG. 20A and FIG. 20B show a model with overlapped cryo-EM map of PKA-phosphorylated RyR2 (PDB: 7U9Q) highlighting the auxiliary helices (Sx) helices from the side view (FIG. 20A) and bottom view (FIG. 20B). Auxiliary helices and pocket with extra densities are labeled.


Previous cryo-EM analyses of RyR2 attributed these densities to detergent and lipids. These auxiliary helices (Sx) were predicted to exist in the RyR2 sequence upstream of the six transmembrane helices that form the pore (S1 to S6). The Jpred4 algorithm was used to predict the secondary structure of the fragment, which was previously reported to encompass these helices. FIG. 21A shows sequence alignment of residues 4231-4320 between the secondary structure predicted by Jpred and the secondary structure from the cryo-EM-resolved model. Aromatic residues used for model building are highlighted in bold. CaMBD3 sequence is underlined. FIG. 21B depicts the RyR2 model highlighting the Sx helices. Sidechains are displayed to show the good fitting of the model. Pocket with extra densities are labelled. FIG. 21C depicts RyR2 model highlighting the interaction between the Sx helices and the neighbor helical elements. FIG. 21D depicts RyR2 model highlighting the lysine rich linker. FIG. 21E depicts Cryo-EM maps of closed PKA RyR2+CaM (gray), open PKA RyR2 (yellow), open PKA RyR2-R2474S (magenta), and open PKA RyR2-R2474S+CaM (cyan) centered on the Sx helices.


Predicted Sx helices were modeled because of the presence of well-resolved densities for large side chains, including W4288 as well as several tyrosine and phenylalanine residues (FIG. 21B). Since the helices are upstream of the S1 to S6 helices, they were named S-1 (4238 to 4259), S-1/S0 linker (4262 to 4271), and S0 (4277 to 4309). In addition, a pocket formed by the interactions with helices S1, S2, and S3 was detected, where two densities were found that could correspond to detergent molecules or protein fragments (FIG. 20B). The S-1/S0 linker contained several positively charged lysine residues and is positioned at the cytosolic surface of the SR membrane, where these lysine residues would be able to interact with the negatively charged phospholipid head groups or other transmembrane proteins (FIG. 21D). The Sx density is strong in the closed states but weaker in the open states (FIG. 21E). This observation suggests that the Sx density plays a role in stabilizing the RyR2 closed state. The CaM binding motif CaMBD3 is also contained in this sequence. In the presence of CaM, the Sx density was absent only in the open state. This finding suggests that, in the closed state of RyR2, the CaMBD3 motif would be inaccessible to CaM, but in the open state of RyR2, it would interact with CaM and completely destabilize the Sx helices.


RyR2 Leak in CPVT

In CPVT patients, SR Ca2+ leak occurs via mutant RyR2 channels during diastole when the heart is supposed to be electrically silent, resulting in afterdepolarizations, arrhythmias, and eventually sudden cardiac death. Intense exercise and adrenergic stimulation can cause two independent but synergistic events that affect RyR2. The β-adrenergic response to exercise (i) results in PKA phosphorylation of RyR2 mainly at S2808 and (ii) activates SR Ca2+ uptake via SERCA2a, thus increasing the SR Ca2+ load, and the driving force for Ca2+ leak out of the membrane via RyR2 channels. By virtue of being in a primed state, the CPVT variant RyR2-R2474S is likely more sensitive to channel-activating posttranslational modifications (e.g., phosphorylation, nitrosylation or oxidation), resulting in a diastolic SR Ca2+ leak that can trigger fatal cardiac arrhythmias during intense exercise (FIG. 26 A and B).



FIG. 26 illustrates the proposed mechanism of CPVT-related RyR2 variants, other gain-of-function mutants, and heart failure. FIG. 26, Panel A is a schematic representation of the normal function of RyR2. FIG. 26, Panel B is a schematic representation of the CPVT-related Ca2+ leak during diastole under intense exercise or stress conditions. In the case of CPVT variants, the resting state is already in a primed state. This state correlates with the higher open probability during exercise or stress conditions. Such conditions can result in opening during diastole, afterdepolarizations, arrhythmias, and sudden cardiac death. This pathological state can be reversed by treatment with Compound 1. This basal primed state scenario could be a shared mechanism among other RyR1 and RyR2 gain-of-function mutants. FIG. 26, Panel C is a schematic representation of the heart failure-related primed state and Ca2+ leak.


As demonstrated herein, the CPVT mutation R2474S can put the channel into a primed state that is independent of the binding of activators including Ca2+. Thus, the mutant CPVT channel RyR2-R2474S is able to be inappropriately activated during diastole when the [Ca2+]cyt is too low to activate the WT RyR2 channel. This inappropriate activation of the mutant channel results in diastolic SR Ca2+ leak that can trigger fatal cardiac arrhythmias. Stabilizing effects of Compound 1 and CaM through different mechanisms are demonstrated herein. One involves the stabilization of the BSol domain through the interaction with the Compound 1-bound RY1&2 domain, and the other occurs via the stabilization of the CSol and BSol3 domains through the binding of CaM to CAMBD2 and the BSol domain.


As demonstrated herein, the CPVT mutation can disrupt local interactions that destabilize the BSol domain and induces a primed state. This primed state leads to inappropriate opening of RyR2 channels that can be reversed by treatment with the RyR2 stabilizer Compound 1. Compound 1 binds to a cleft in the RY1&2 domain where can stabilize interactions between residues that are required to reduce flexibility between domains, particularly with the BSol1 domain, of the cytosolic shell. The net effect of Compound 1 binding is to stabilize the overall channel structure closer to the closed state. This renders the channel less likely to be inappropriately activated during diastole when the conditions favor the closed state of the channel (e.g., low non-activating [Ca2+]cyt).


Example 7: Single Channel Recordings

To confirm the functional role of xanthine in RyR2, single-channel experiments were performed. ER vesicles from HEK293 cells expressing RyR2 were prepared by homogenizing cell pellets on ice using a Teflon glass homogenizer with two volumes of solution containing 20 mM tris-maleate (pH 7.4), 1 mM EDTA, 1 mM DTT, and protease inhibitors (Roche). Homogenate was then spun by centrifuge at 4000 g for 15 min at 4° C., and the resulting supernatant was spun by centrifuge at 40,000 g for 30 min at 4° C. The final pellet, containing the ER fractions, was resuspended and aliquoted in 250 mM sucrose, 10 mM Mops (pH 7.4), 1 mM EDTA, 1 mM DTT, and protease inhibitors. Samples were frozen in liquid nitrogen and stored at −80° C.


ER vesicles were fused to planar lipid bilayers formed by painting a lipid mixture of phosphatidylethanolamine and PC (Avanti Polar Lipids) in a 5:3 ratio in decane across a 200-am hole in polysulfonate cups (Warner Instruments) separating two chambers. The trans chamber (1.0 ml), representing the intra-SR (luminal) compartment, was connected to the head stage input of a bilayer voltage clamp amplifier. The cis chamber (1.0 ml), representing the cytoplasmic compartment, was held at virtual ground. The following asymmetrical solutions were used: for the cis solution, 1 mM EGTA, 250/125 mM Hepes/tris, 50 mM KCl (pH 7.35); for the trans solution, 53 mM Ca(OH)2, 50 mM KCl, 250 mM Hepes (pH 7.35). The concentration of free Ca2+ in the cis chamber was calculated as previously described. ER vesicles were added to the cis side, and fusion with the lipid bilayer was induced by making the cis side hyperosmotic by the addition of 400 to 500 mM KCl. After the appearance of potassium and chloride channels, the cis side was perfused with the cis solution. At the end of each experiment, 10 μM ryanodine was added to block the RyR2 channel. Single-channel currents were recorded at 0 mV using Bilayer Clamp BC-525D (Warner Instruments), filtered at 1 kHz using Low-Pass Bessel Filter 8 Pole (Warner Instruments), and digitized at 4 kHz. All experiments were performed at room temperature (23° C.). Data acquisition was performed by using Digidata 1322A and Axoscope 10.1 software (Axon Instruments). The recordings were analyzed using Clampfit 10.1 (Molecular Devices) and GraphPad Prism software.


This experiment determined that at presumed physiological concentrations during exercise (10 μM), there was a ˜25-fold increase in the open probability of RyR2 in the presence of xanthine FIG. 22 shows single-channel current recordings traces from recombinant RyR2 at Ca2+ 150 nM, before (Panel A) and after (Panel B) the addition of xanthine 10 μM. Opening events were recorded as an upward deflection. Po: opening probability, To: meantime open, Tc: meantime closed.


Example 8: Telemetric Electrocardiogram Recordings in Ryr2R2474S/WT Mice

Ryr2R2474S/WT mice undergo sustained ventricular tachycardia and sudden cardiac death after exercise and epinephrine. This experiment evaluated the effect of Compound 1 treatment on frequency of ventricular tachycardia and sudden cardiac death in Ryr2R2474S/WT mice.


Ryr2R2474S/WT mice were implanted with radio telemetry transmitters (Data Sciences International). Briefly, the transmitter (PhysioTel, ETA-F10 transmitter) was inserted in mice subcutaneously along the back under general anesthesia (5% inhaled isoflurane). Two electrocardiogram (ECG) electrodes were placed hypodermically in the region of the right shoulder (negative pole) and toward the lower left chest (positive pole) to approximate lead II of the Einthoven surface ECG. During the procedure, respiratory and cardiac rhythm, adequacy of anesthetic depth, muscle relaxation, body temperature, and analgesia were monitored to avoid anesthesia-related complications. Postoperative pain was considered during a 1-week post-implantation period, and carprofen (5 mg/kg, subcutaneously) was given. A minimum period of 2 weeks was allowed for recovery from the surgery. Animals were housed in individual stainless-steel cages for telemetry recordings. Environmental parameters were recorded continuously and maintained within a fixed range: room temperature at 15° C. to 21° C. and 45 to 65% relative humidity. The artificial day/night cycle was 12-hour light/12-hour dark with light on at 0700 hours. Drinking water was provided ad libitum. Solid diet (300 g) was given daily in the morning. ECG waveforms were continuously recorded at a sampling rate of 2000 Hz using a signal transmitter-receiver (RPC-1) connected to a data acquisition system (Ponemah system, Data Sciences International). Compound 1 (50 mg/kg per day) was given in drinking water for 2 weeks before ECG recordings. Mice were recorded for 1 hour at baseline and then given epinephrine injection (1 mg/kg, intraperitoneally) and recorded for another 2 hours. The animals used in the study were maintained and studied according to protocols approved by the Institutional Animal Care and Use Committee of Columbia University (reference no. AC-AABP1551).


This experiment determined that treatment with Compound 1 reduced the frequency of ventricular tachycardia and prevented sudden cardiac death in Ryr2R2474S/WT mice. FIG. 23 depicts representative telemetric electrocardiogram (ECG) recordings of Ryr2R2474S/WT mice (n=4) during arrhythmia provocation stress testing by epinephrine injection (1 mg/kg epinephrine). Catecholamine injection and exercise resulted in rapid sustained ventricular tachycardia (sVT) and sudden cardiac death (SCD). Compound 1 (50 mg/kg/day in drinking water) treatment prevented sVT and SCD.


Example 9: SR Ca2+ Leak Assay in Ryr2R2474S/WT Mice Heart Microsomes

Ca2+ leak was measured in microsomes from heart lysates isolated from control Ryr2R2474S/WT mice, Ryr2R2474S/WT mice treated with epinephrine, and Ryr2R2474S/WT mice treated with epinephrine and Compound 1 in EXAMPLE 8. 10704.1 Cardiac muscle SR microsomes were prepared by homogenizing heart samples on ice using a Teflon glass homogenizer (50 times) with 2 volumes of 20 mM tris-maleate (pH 7.4), 1 mM EDTA, 1 mM DTT, and protease inhibitors (Roche). Homogenate was then spun by centrifuge at 4000 g for 15 min at 4° C., and the resultant supernatant was spun by centrifuge at 50,000 g for 45 min at 4° C. Pellets were resuspended in lysis buffer containing 300 mM sucrose. Microsomes (5 g/ml) were diluted into a buffer (pH 7.2) containing 8 mM K-phosphocreatine, and creatine kinase (2 U/ml), mixed with 3 μM Fluo-4 and added to multiple wells of a 96-well plate. Ca2+ loading of the microsomes was initiated by adding 1 mM ATP. After Ca2+ uptake (50 s), 3 μM thapsigargin was added to inhibit the Ca2+ reuptake by SERCA. SR Ca2+ leak was measured by the increase in intensity of the Fluo-4 signal (measured in a Tecan fluorescence plate reader). Ca2+ leak was quantified as the difference between the average Fluo-4 signal before and after addition of thapsigargin. Graphs were plotted with GraphPad Prism software.


This experiment determined that Compound 1 treatment prevented SR Ca2+ leak via RyR2-R2474S channels. FIG. 24 shows SR Ca2+ leak measured in microsomes from Ryr2R2474S/WT mouse heart lysates. The Ca2+ leak was compared for hearts isolated from control Ryr2R2474S/WT mice (gray), Ryr2R2474S/WT mice treated with epinephrine (magenta), and Ryr2R2474S/WT mice treated with epinephrine and Compound 1 (cyan). Bar graphs represent the quantification of the increase in Fluo-4 signal/s over the first 5 seconds after addition of thapsigargin. N=2 in each group.


Example 10: Mass Spectrometry Analyses

Mass spectroscopic analysis of the hyperphosphorylated channels prepared in EXAMPLE 5 was conducted to determine phosphorylation sites.


ER vesicles from HEK293 cells expressing RyR2 were separated on 4 to 12% gradient SDS-PAGE and sent for mass spectrometry analysis to in-house Columbia Proteomics Shared Resource (HICCC). Protein gel slices were excised, and in-gel digestion was performed. Digested peptides were collected and further extracted from gel slices in extraction buffer (1:2 ratio by volume of 5% formic acid:acetonitrile) at high speed, shaking in an air thermostat. Peptides were separated within 80 min at a flow rate of 400 nl/min on a reversed-phase C18 column with an integrated CaptiveSpray Emitter (25 cm×75 m, 1.6 m, IonOpticks). Mobile phases A and B were with 0.1% formic acid in water and 0.1% formic acid in acetonitrile. The fraction of B was linearly increased from 2 to 23% within 70 min, followed by an increase to 35% within 10 min and a further increase to 80% before reequilibration. The timsTOF Pro was operated in parallel accumulation-serial fragmentation (PASEF) mode with the following settings: mass range, 100 to 1700 mass/charge ratio (m/z); 1/K0 start, 0.6 V s/cm2; end, 1.6 V s/cm2; ramp time, 100 ms; lock duty cycle to 100%; capillary voltage, 1600 V; dry gas, 3 l/min; and dry temperature, 200° C. PASEF settings: 10 Tandem Mass Spectrometry frames (1.16 s duty cycle); charge range, 0 to 5; active exclusion for 0.4 min; target intensity, 20,000; intensity threshold, 2500; and collision-induced dissociation collision energy, 59 eV. A polygon filter was applied to the m/z and ion mobility plane to select features most likely representing peptide precursors rather than singly charged background ions. Acquired PASEF raw files were analyzed using the MaxQuant environment v.2.0.1.0 and Andromeda for database searches. MaxQuant was configured to search with the reference human proteome database downloaded from UniProt. The following modifications were used for protein identification and quantification: Carbamidomethylation of cysteine residues (+57.021 Da) was set as static modifications, while the oxidation of methionine residues (+15.995 Da), deamidation (+0.984) on asparagine and glutamine, and phosphorylation (+79.966) on serine, threonine, and tyrosine were set as a variable modification. Results obtained from MaxQuant, Phospho (STY) sites table was used for RyR2 phospho-site quantification. Sequence coverage was obtained with the software Scaffold 5.


TABLE 10 and TABLE 11 are mass-spectroscopy tables indicating the phosphopeptides detected before and after PKA phosphorylation of recombinant human RyR2 samples.















TABLE 10











Score


Position





for


within
Localization
Score


Delta
local-


RyR2
probability
difference
log10PEP
Score
score
ization





















16
1.00
64.13
−6.34
67.66
51.88
67.66


1495
0.86
7.94
−3.18
62.49
53.34
62.49


1856
0.97
18.05
−14.55
64.40
52.48
64.40


1863
0.54
0.89
−4.61
60.59
56.99
40.37


1869
1.00
35.49
−3.87
82.73
77.89
69.82


2363
0.97
17.27
−4.17
93.50
53.12
93.50


2368
0.60
3.79
−4.66
75.69
59.00
75.69


2804
0.73
4.26
−34.70
167.56
158.97
167.56


2806
1.00
28.36
−80.37
285.07
257.49
157.38


2810
0.67
3.11
−8.92
102.55
97.74
102.55


2811
0.74
4.22
−2.73
81.90
80.43
81.90


2814
0.98
18.09
−2.48
100.27
94.94
100.27


2822
1.00
40.37
−5.53
121.74
119.20
121.74




















TABLE 11





Position
Intensity
Intensity
Intensity
Intensity


within RyR2
control 1
control 2
control 3
PKA



















 16
56,978

63,044
70,955


1495
12,130





1856



155,050


1863


81,407



1869
218,050
215,750
349,580
270,440


2363
68,221

38,508
33,431


2368
45,015





2804
530,280






2808

3,944,000
3,623,900
567,320
3,385,300


2810
1,332,700





2811


80,673



2814
29,077


55,669


2822
29,678

32,822
28,207









The analysis revealed that RyR2-S2808 is the main detected phosphorylated peptide before and after PKA phosphorylation. Other detected residues, with good localization probability and score difference, were considered secondary phosphorylation sites (TABLE 10 and TABLE 11, underlined values). Mass-spectrometry is a semi-quantitative technique, indicating that intensities can be only compared within the same sample (within the same column). FIG. 25 illustrates sequence coverage of RyR2 provided in this experiment. Sequence coverage of 75% was consistently obtained for all samples. Detected peptides are highlighted in orange. Detected residues with oxidative or phosphorylation modifications are highlighted in green. High-confidence phosphorylated residues are framed in black, RyR2-S2808 is framed in red, and RyR2-S2031 is framed in cyan.


The results showed that RyR2-S2808 was the major and only significantly PKA-phosphorylated site in RyR2 treated with PKA (coverage of 75%; FIG. 25). A secondary site with one order of magnitude lower intensity at RyR2-S1869 and other phosphorylation sites with even lower intensity at RyR2-T16, RyR2-S1856, RyR2-S2363, RyR2-S2814, and RyR2-S2822 (TABLE 10 and TABLE 11) were detected. No phosphorylation of RyR2-S2031 (FIG. 25) was detected.

Claims
  • 1.-248. (canceled)
  • 249. A composition comprising a complex suspended in a solid medium comprising vitreous ice, wherein the complex comprises a protein and a synthetic compound, wherein the protein is a ryanodine receptor 2 protein (RyR2) or a mutant thereof.
  • 250. The composition of claim 249, wherein the composition is prepared by a process, the process comprising vitrifying an aqueous solution that is applied to an electron microscopy grid, wherein the aqueous solution comprises the complex.
  • 251. The composition of claim 250, wherein the aqueous solution further comprises one or more or each of a buffering agent, a phospholipid, a zwitterionic surfactant, a disulfide-reducing agent, a protease inhibitor, or a xanthine alkaloid.
  • 252. The composition of claim 250, wherein the aqueous solution further comprises one or more or each of a Ca2+ ion, sodium adenosine triphosphate (NaATP), cyclic adenosine monophosphate (cAMP), or calmodulin.
  • 253. The composition of claim 249, wherein the complex further comprises one or more or each of calmodulin, calstabin, a xanthine alkaloid or a Ca2+ ion.
  • 254. The composition of claim 249, wherein the protein is wild type RyR2, a mutant RyR2 or a post-translationally modified RyR2 protein, wherein the post-translationally modified RyR2 protein is a phosphorylated, oxidized or nitrosylated RyR2 or is associated with heart failure.
  • 255. The composition of claim 249, wherein the protein is a mutant RyR2 containing at least one mutation that is associated with Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT).
  • 256. The composition of claim 255, wherein the mutation is RyR2-R2474S, RyR2-R420Q, or RyR2-R420W.
  • 257. The composition of claim 249, wherein the protein is a mutant RyR2 or a post-translationally modified RyR2 protein, wherein the mutation or the post-translational modification destabilizes an interaction between NTD and BSol domains of the RyR2 protein; or wherein the mutation or the post-translational modification destabilizes a cytosolic shell of the RyR2 protein, wherein the cytosolic shell comprises NTD, SPRY, JSol and BSol domains of the RyR2 proteins.
  • 258. The composition of claim 249, wherein the protein is a tetramer of RyR2 monomers, wherein each RyR2 monomer is a peptide according to SEQ ID NO: 3 or SEQ ID NO: 4.
  • 259. The composition of claim 249, wherein the complex further comprises a nucleoside-containing molecule.
  • 260. The composition of claim 259, wherein the nucleoside-containing molecule and the synthetic compound bind a RYR domain of the protein, wherein the RYR domain is a RY1&2 domain.
  • 261. The composition of claim 260, wherein the RY1&2 domain has a three-dimensional structure according to TABLE 3.
  • 262. The composition of claim 259, wherein the nucleoside-containing molecule is a purine nucleoside-containing molecule, a nucleotide or nucleoside polyphosphate, or an adenosine triphosphate (ATP) molecule.
  • 263. The composition of claim 262, wherein the nucleoside-containing molecule is an adenosine triphosphate (ATP) molecule, wherein the ATP molecule forms a pi-stacking interaction with the synthetic compound or molecule has a three-dimensional conformation according to TABLE 5.
  • 264. The composition of claim 263, wherein the ATP molecule cooperatively binds the protein with the synthetic compound, or wherein the ATP molecule forms a pi-stacking interaction with the synthetic compound.
  • 265. The composition of claim 249, wherein the complex further comprises a second nucleoside-containing molecule bound to a C-terminal domain of the RyR1 protein, wherein the second nucleoside-containing molecule is a second ATP molecule.
  • 266. The composition of claim 249, wherein the synthetic compound comprises a benzazepane, benzothiazepane, or benzodiazepane moiety.
  • 267. The composition of claim 249, wherein the synthetic compound is a compound of Formula (I):
  • 268. The composition of claim 249, wherein the synthetic compound is a compound of Formula (I-k):
  • 269. The composition of claim 249, wherein the synthetic compound is:
  • 270. The composition of claim 269, wherein the synthetic compound has a three-dimensional conformation according to TABLE 4.
  • 271. A method of determining a binding site of a synthetic compound in a protein, the method comprising subjecting a composition of claim 249 to single-particle cryogenic electron microscopy analysis, wherein the structure of the of protein obtained by single-particle cryogenic electron microscopy analysis has a resolution from about 2 Å to about 3.5.
  • 272. A method for predicting a docked position of a target ligand in a binding site of a biomolecule, the method comprising: receiving a template ligand-biomolecule structure, the template ligand-biomolecule structure comprising a template ligand docked in the binding site of the biomolecule;comparing a pharmacophore model of the template ligand to a pharmacophore model of the target ligand;overlapping the pharmacophore model of the target ligand with the pharmacophore model of the template ligand while the template ligand is in the binding site of the biomolecule; andpredicting the docked position of the target ligand in the binding site of the biomolecule based on a position of the pharmacophore model of the target ligand when overlapped with the pharmacophore model of the template ligand,wherein the template ligand-biomolecule structure is obtained by a process comprising subjecting a complex of the biomolecule and the template ligand to single-particle cryogenic electron microscopy analysis,wherein the biomolecule is a ryanodine receptor 2 protein (RyR2) or a mutant thereof and the template ligand is a synthetic compound, andwherein the complex of the biomolecule and the template ligand is obtained by the process to prepare the composition of claim 250.
  • 273. The method of claim 272, wherein the biomolecule is a RY1&2 domain of RyR2, wherein the RY1&2 domain comprises a structure according to TABLE 3.
  • 274. The method of claim 272, wherein the template ligand has a three-dimensional conformation according to TABLE 4.
  • 275. The method of claim 273, wherein the RY1&2 domain further comprises an ATP molecule having a three-dimensional conformation according to TABLE 5.
  • 276. The method of claim 272, wherein the template ligand is
  • 277. A method of identifying a plurality of potential lead compounds, the method comprising the steps of: (a) analyzing, using a computer system, an initial lead compound known to bind to a biomolecular target, the analyzing comprising partitioning, by providing a database of known reactions, the initial lead compound into atoms defining partitioned lead compound comprising a lead compound core and atoms defining a lead compound non-core, wherein the initial lead compound is partitioned using a computational retrosynthetic analysis of the initial lead compound;(b) identifying, using the computer system, a plurality of alternative cores to replace the lead compound core in the initial lead compound, thereby generating a plurality of potential lead compounds each having a respective one of the plurality of alternative cores;(c) calculating, using the computer system, a difference in binding free energy between the partitioned lead compound and each potential lead compound;(d) predicting, using the computer system, whether each potential lead compound will bind to the biomolecular target and identifying a predicted active set of potential lead compounds based on the prediction;(e) obtaining a synthesized set of at least some of the potential leads of the predicted active set to establish a first of potential lead compounds; and(f) determining, empirically, an activity of each of the first set of synthesized potential lead compounds,wherein the biomolecular target is a ryanodine receptor 2 protein (RyR2) or a mutant thereof and the initial lead compound is a synthetic compound, andwherein the structure of the biomolecular target used in the predicting of (d) is obtained by a process comprising subjecting a complex of the biomolecular target and the initial lead compound to single particle cryogenic electron microscopy analysis, andwherein the complex of the biomolecular structure and the initial lead compound is obtained by the process to prepare the composition of claim 250.
  • 278. A method for pharmaceutical drug discovery, comprising: identifying an initial lead compound for binding to a biomolecular target;using the method of claim 278 to identify a predicted active set of potential lead compounds for binding to the biomolecular target based on the initial lead compound;selecting one or more of the predicted active set of potential lead compounds for synthesis; andassaying the one or more synthesized selected compounds to assess each synthesized selected compounds suitability for in vivo use as a pharmaceutical compound,wherein the biomolecular target is a RY1&2 domain of RyR2, and the structure of the biomolecular target used in the predicting of (d) is obtained by a process comprising subjecting a complex of the biomolecular target and the initial lead compound to single-particle cryogenic electron microscopy analysis.
  • 279. A computer-implemented method of quantifying binding affinity between a ligand and a receptor molecule, the method comprising: receiving by one or more computers, data representing a ligand molecule,receiving by one or more computers, data representing a receptor molecule domain,using the data representing the ligand molecule and the data representing the receptor molecule domain in computer analysis to identify ring structure within the ligand, the ring structure being an entire ring or a fused ring;using the data representative of the identified ligand ring structure to designate a first ring face and a second ring face opposite to the first ring face, and classifying the ring structure by: a) determining proximity of receptor atoms to atoms on the first face of the ligand ring; andb) determining proximity of receptor atoms to atoms on the second face of the ligand ring; andc) determining solvation of the first face of the ligand ring and solvation of the second face of the ligand ring;classifying the identified ligand ring structure as buried, solvent exposed or having a single face exposed to solvent based on receptor atom proximity to and solvation of the first ring face and receptor atom proximity to and solvation of the second ring face;quantifying the binding affinity between the ligand and the receptor molecule domain based at least in part on the classification of the ring structure; anddisplaying, via computer, information related to the classification of the ring structure,wherein the receptor molecule domain is a RY1&2 domain of RyR2 protein or a mutant thereof, wherein the data representing a ligand molecule and the data representing a receptor molecule domain are obtained by a process comprising subjecting a complex comprising the ligand molecule and the receptor molecule domain to single-particle cryogenic electron microscopy analysis, andwherein the ligand molecule is a synthetic compound, and wherein the complex is obtained by the process to prepare the composition of claim 250.
  • 280. A method of identifying a compound having RyR2 modulatory activity, the method comprising: (a) determining an open probability (Po) of a RyR2 protein;(b) contacting the RyR2 protein with a test compound;(c) determining an open probability (Po) of the RyR2 protein in the presence of the test compound; and(d) determining a difference between the Po of the RyR2 protein in the presence and absence of the test compound;wherein a reduction in the Po of the RyR2 protein in the presence of the test compound relative to the Po of the RyR2 protein in the absence of the test compound is indicative of the compound having RyR2 modulatory activity.
  • 281. The method of claim 280, wherein the RyR2 protein is a mutated or a post-translationally modified RyR2 protein, wherein the test compound preferentially binds to the mutated or post-translationally modified RyR2 relative to wild-type RyR2.
  • 282. A method for identifying a compound having RyR2 modulatory activity, comprising: (a) contacting a RyR2 protein with a ligand having known RyR2 modulatory activity to create a mixture, wherein the RyR2 protein is a leaky RyR2, the leaky RyR2 comprising mutant RyR2 protein, post-translationally modified RyR2, or a combination thereof,(b) contacting the mixture of step (a) with a test compound; and(c) determining the ability of the test compound to displace the ligand from the RyR2 protein.
  • 283. The method of claim 282, wherein the ligand is radiolabeled and generates a signal, wherein determining the ability of the test compound to displace the ligand from the RyR2 protein comprises determining a change in the signal.
  • 284. The method of claim 282, wherein the RyR2 protein is a mutant RyR2, wherein the mutant RyR2 contains at least one mutation that is associated with Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT).
CROSS REFERENCE

This application claims the benefit of U.S. Provisional Patent Application No. 63/286,861, filed on Dec. 7, 2021, the content of which is incorporated by reference herein in its entirety.

STATEMENT AS TO FEDERALLY SPONSORED RESEARCH

This present disclosure was made with government support under R01HL1 R01HTL145473, R01DK118240, R01HTL142903, R01HTL140934, R01AR070194, R25HL156002R25, R25NS076445 and T32 HL120826, awarded by the National Institutes of Health (NIH). The government has certain rights in the disclosure.

Provisional Applications (1)
Number Date Country
63286861 Dec 2021 US