METHODS FOR TREATING CANCER WITH ACYLDEPSIPEPTIDE ANALOGS

Information

  • Patent Application
  • 20210171576
  • Publication Number
    20210171576
  • Date Filed
    June 03, 2019
    5 years ago
  • Date Published
    June 10, 2021
    2 years ago
Abstract
Methods and uses for treating cancer in a subject are provided. In particular, the present disclosure provides methods and uses relating to treating a subject with cancer by activating human mitochondrial CIpP (HsCIpP). The present disclosure further provides methods and uses of an acyldepsipeptide (ADEP) analog that activates HsCIpP in the subject in need thereof.
Description
FIELD

The disclosure relates to methods for treating cancer in a subject.


The disclosure provides methods and uses relating to treating a subject with cancer with an acyldepsipeptide analog.


BACKGROUND


The HsCIpXP complex is an ATP-dependent protease complex found in the mitochondrial matrix that plays an important role in mitochondrial protein quality control. HsCIpXP is composed of the serine protease HsCIpP and the AAA+ ATPase HsCIpX. Assembly of the complex involves the capping of the barrel-shaped HsCIpP tetradecamer on one or both ends by the HsCIpX hexamer (Kang et al., 2002). Protein degradation by CIpXP typically involves the recognition, binding, and unfolding of the substrate by CIpX. The unfolded polypeptide is then threaded through CIpX's central pore into the lumen of CIpP. Once inside, the polypeptide is hydrolysed by the 14 Ser-His-Asp proteolytic sites of CIpP, and the resultant fragments are expelled from CIpP (Olivares et al., 2016).


The interaction between CIpX and CIpP is partly stabilized by the dynamic docking of IGF loops of CIpX (E436 to G450 in HsCIpX; the triplet motif being L439-G440-F441) at specific hydrophobic pockets formed between neighbouring CIpP subunits (Amor et al., 2016). Small molecules that abrogate the CIpX-CIpP interaction have been characterized extensively for bacterial CIpXP. Among them, the acyldepsipeptides (ADEPs) (Brotz-Oesterhelt et al., 2005) bind CIpP with high affinity at the same site that normally accommodates the IGF loops of CIpX (Alexopoulos et al., 2012). The high-affinity binding of ADEP also keeps CIpP in a poorly understood activated conformation (Lee et al., 2010; Li et al., 2010). Collectively, these molecular changes allow free access for small peptides, molten globules and even folded proteins into the lumen of CIpP, causing an increase in degradation activity that is dysregulated, leading to bacterial cell death. Hence, the ADEPs are considered potential antibiotics.


In humans, HsCIpP has been shown to physically interact with numerous mitochondrial proteins involved in vital cellular processes such as energy metabolism, mitochondrial translation, mitochondrial protein import, metabolism of amino acids and cofactors, and maintenance of the mitochondrial proteome (Cole et al., 2015; Szczepanowska et al., 2016).


SUMMARY

The present disclosure shows that acyldepsipeptide (ADEP) analogs activate human mitochondrial CIpP (HsCIpP) in cancer cells, which triggers intrinsic apoptotic pathway, thereby killing the cancer cells.


Accordingly, the present disclosure provides a method for treating a subject having cancer, comprising using or administering a therapeutically effective amount of an ADEP analog to the subject in need thereof. Also provided herein is use of a therapeutically effective amount of ADEP analog for treating a subject with cancer. Further provided is use of a therapeutically effective amount of ADEP analog in the manufacture of a medicament for treating subject with cancer. Even further provided is a therapeutically effective amount of ADEP analog for use in treating a subject with cancer.


In an embodiment, the ADEP analog is ADEP-01, ADEP-02, ADEP-03, ADEP-04, ADEP-05, ADEP-06, ADEP-07, ADEP-08, ADEP-09, ADEP-10, ADEP-11, ADEP-12, ADEP-13, ADEP-14, ADEP-15, ADEP-16, ADEP-17, ADEP-18, ADEP-19, ADEP-20, ADEP-21, ADEP-22, ADEP-23, ADEP-24, ADEP-25, ADEP-26, ADEP-27, ADEP-28, ADEP-29, ADEP-30, ADEP-31, ADEP-32, ADEP-33, ADEP-34, ADEP-35, ADEP-36, ADEP-37, ADEP-38, ADEP-39, ADEP-40, ADEP-41, ADEP-42, ADEP-43, ADEP-44, ADEP-45, or ADEP-46.


In another embodiment, the ADEP analog is ADEP-01, ADEP-02, ADEP-04, ADEP-06, ADEP-10, ADEP-13, ADEP-14, ADEP-15, ADEP-17, ADEP-20, ADEP-25, ADEP-28, ADEP-29, ADEP-30, ADEP-32, ADEP-37, ADEP-38, or ADEP-41.


In another embodiment, the ADEP analog is ADEP-14, ADEP-17, ADEP-28, ADEP-29, ADEP-30, ADEP-32, ADEP-37, or ADEP-38.


In another embodiment, the ADEP analog activates protease activity HsCIpP, and wherein the protease activity is at least 0.87 as measured by the relative degradation (RD) index at 25 μM of the ADEP analog, optionally at least 0.9, optionally at least 0.95.


In another embodiment, the protease activity is at least 0.74 as measured by the RD index at 5 μM of the ADEP analog, optionally at least 0.8, optionally at least 0.85.


In another embodiment, the protease activity is at least 0.2 as measured by the RD index at 1 μM of the ADEP analog, optionally at 0.5, optionally at least 0.75, optionally at least 0.8.


In another embodiment, the cancer is breast cancer, prostate cancer, colon cancer, liver cancer, uterus cancer, thyroid cancer, lung cancer, lymph nodes cancer, bladder cancer, ovary cancer, stomach cancer, testis cancer, brain cancer, skin cancer, pancreatic cancer, acute myeloid leukemia, kidney cancer, cervical cancer, osteosarcoma, or neuroblastoma.


In another embodiment, the cancer is breast cancer, kidney cancer, cervical cancer, osteosarcoma, or neuroblastoma.


In an embodiment, the cancer is metastatic.


In an embodiment, the metastatic cancer is breast cancer.


In another embodiment, the subject is human.


In another embodiment, the ADEP analog is administered subcutaneously, intraperitoneally, intravenously, topically, or orally.


Also provided is a compound for use in treating a subject having a cancer, wherein the compound is a therapeutically effective amount of an ADEP analog.


Also provided is an acyldepsipeptide (ADEP) analog, wherein the ADEP analog is ADEP-01, ADEP-02, ADEP-03, ADEP-04, ADEP-05, ADEP-06, ADEP-07, ADEP-08, ADEP-09, ADEP-10, ADEP-11, ADEP-12, ADEP-13, ADEP-14, ADEP-15, ADEP-16, ADEP-17, ADEP-18, ADEP-19, ADEP-20, ADEP-21, ADEP-22, ADEP-23, ADEP-24, ADEP-25, ADEP-26, ADEP-27, ADEP-28, ADEP-29, ADEP-30, ADEP-31, ADEP-32, ADEP-33, ADEP-34, ADEP-35, ADEP-36, ADEP-37, ADEP-38, ADEP-39, ADEP-40, ADEP-41, ADEP-42, ADEP-43, ADEP-44, ADEP-45, or ADEP-46, or a variant or a derivative thereof.


Other features and advantages of the present disclosure will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific Examples while indicating preferred embodiments of the disclosure are given by way of illustration only, since various changes and modifications within the spirit and scope of the disclosure will become apparent to those skilled in the art from this detailed description.





BRIEF DESCRIPTION OF THE DRAWINGS

Embodiments are described below in relation to the drawings in which:



FIG. 1A-O show graphical representation of activity of ADEP analogs against HsCIpP. FIG. 1A shows RD25 scores of the 46 ADEP analogs tested. Standard deviations (SDs) are shown as error bars. FIG. 1B shows RD1 scores of 18 ADEP analogs showing the strongest activation of HsCIpP. Names of the five ADEP analogs selected for further experiments are bolded. FIG. 1C shows chemical structures of the five ADEP analogs selected for further experiments. FIG. 1D shows chemical structures of ADEP-01, ADEP-02, ADEP-03, and ADEP-04. FIG. 1 E shows chemical structures of ADEP-05, ADEP-06, ADEP-07, and ADEP-08. FIG. 1F shows chemical structures of ADEP-09, ADEP-10, ADEP-11, and ADEP-12. FIG. 1G shows chemical structures of ADEP-13, ADEP-14, ADEP-15, and ADEP-16. FIG. 1H shows chemical structures of ADEP-17, ADEP-18, ADEP-19, and ADEP-20. FIG. 1I shows chemical structures of ADEP-21, ADEP-22, ADEP-23, and ADEP-24. FIG. 1J shows chemical structures of ADEP-25, ADEP-26, ADEP-27, and ADEP-28. FIG. 1K shows chemical structures of ADEP-29, ADEP-30, ADEP-31, and ADEP-32. FIG. 1L chemical structures of ADEP-33, ADEP-34, ADEP-35, and ADEP-36. FIG. 1M chemical structures of ADEP-37, ADEP-38, ADEP-39, and ADEP-40. FIG. 1N chemical structures of ADEP-41, ADEP-42, ADEP-43, and ADEP-44. FIG. 1O Chemical structures of ADEP-45 and ADEP-46.



FIG. 2A-G show graphical representation of ADEPs enhancing both the peptidase and protease activity of HsCIpP. FIG. 2A shows peptidase activity of 8 μM HsCIpP (monomeric concentration) against different fluorogenic peptides (at 500 μM final concentration). FIG. 2B shows Michaelis-Menten kinetic analysis of Ac-WLA-AMC cleavage by 6 μM of HsCIpP. The kinetic parameters shown were derived from non-linear regression analysis of the initial rates. SDs of the activity data are shown as error bars. FIG. 2C shows Enhancement of the peptidase activity of 6 μM HsCIpP by ADEP-28 (left panel) and ADEP-41 (right panel). Ac-WLA-AMC was used at a final concentration of 100 μM. FIG. 2D shows enhancement of the protease activity of 3 μM HsCIpP by ADEP-28 (left panel) and ADEP-41 (right panel). FIG. 2E shows Vmax, Hill coefficient (h), and the microscopic apparent dissociation constant (K0.5) of all five ADEP analogs tested, derived from the non-linear regression analyses of data collected for the peptidase (left side of table) and protease (right side of table) activity of HsCIpP. Errors shown are from curve-fitting. FIG. 2F shows HsCIpP protease activity profiles while interacting with HsCIpX in the presence of ADEP-28, ADEP-06 and ADEP-02. Solid lines represent simple traces of the kinetic data. FIG. 2G shows observed minimal rates (mean±SD) (Vmin Observed) of casein-FITC degradation and the corresponding ADEP concentration at Vmin Observed. The kinetic data shown in each panel in FIG. 2C and FIG. 2D were derived from three independent replicates. SDs are shown as error bars.



FIG. 3A-G show graphical representation of enhancement of peptidase and protease activity of HsCIpP by ADEP-06, ADEP-02 and ADEP-04. FIG. 3A shows Michaelis-Menten kinetic analysis of Ac-WLA-AMC cleavage by 1.5 μM HsCIpP in the presence of 4.5 μM HsCIpX (upper panel) and by 2 μM EcCIpP (lower panel). In both experiments, the kinetic parameters shown were derived from non-linear regression analysis of the initial rates. Error bars shown correspond to standard deviations (SDs) derived from at least three independent replicates. Statistical errors shown for the calculated kinetic parameters correspond to curve-fitting errors. FIG. 3B shows enhancement of the peptidase activity of HsCIpP by ADEP-06, ADEP-02 and ADEP-04. FIG. 3C shows enhancement of the peptidase activity of HsCIpP by ADEP-06, ADEP-02 and ADEP-04. FIG. 3D shows enhancement of the protease activity of HsCIpP by ADEP-06, ADEP-02 or ADEP-04. FIG. 3E shows kinetic analysis of the degradation of casein-FITC by HsCIpP in the presence of HsCIpX at different molar ratios. Data points were analyzed by non-linear regression using the Hill equation. Relevant kinetic parameters derived from the analysis are as shown. FIG. 3F shows degradation of unlabelled casein by HsCIpX and HsCIpP in the absence (DMSO panels) and presence of ADEP-28 at 0.1 μM (middle panels) and 2 μM (right panels), observed on SDS-PAGE gels. The arrow to the right of creatine kinase panels indicates the correct band for creatine kinase. FIG. 3G shows densitometry analysis of casein illustrated in (F). Data points were derived from 3 independent replicates. The kinetic data shown in each panel in FIG. 3B to FIG. 3D were derived from three independent replicates.



FIG. 4A-F show graphical representation of ADEP inducing HsCIpP-dependent cytotoxicity in HEK293 T-REx cells. FIG. 4A shows cytotoxicity profiles of HEK293 T-REx WT (CLPP+/+) and HEK293 T-REx CLPP cells treated with ADEP-28 (left panel) and ADEP-41 (right panel). Relative cell viability±SDs (shown as error bars) was calculated by normalizing data collected from 4 independent replicates to the DMSO-only control. FIG. 4B shows quantification of the cytotoxicity data collected for the five ADEP analogs tested. For WT, IC50 values±curve-fitting errors were derived from non-linear regression analysis using a standard dose response equation. For CLPP−/−, the lack of a minimum plateau in the cytotoxicity profiles prohibited non-linear regression analysis. FIG. 4C shows IC50 values of the five ADEP analogs tested strongly correlate with their respective apparent binding affinity (K0.5) to HsCIpP in vitro (shown in FIG. 2E) that were derived from the protease activity data. FIG. 4D shows Western blots on whole cell lysates of HEK293 T-REx WT, CLPP−/−, HEK293 T-REx+CLPP-FLAG, and HEK293 T-REx+CLPPS153A-FLAG. The absence or presence of Dox in the tissue culture is indicated as “−Dox” or “+Dox”, respectively. The primary antibody used in each blot is indicated to the right of each panel. Protein molecular weight markers are shown on the left. For the α-HsCIpP blot, band I corresponds to HsCIpP (WT or S153A mutant)-FLAG, band II corresponds to HsCIpP (WT or S153A mutant) that is missing the C-terminal FLAG-tag, and band Ill corresponds to endogenous HsCIpP. FIG. 4E shows ADEP-41 cytotoxicity profiles of HEK293 T-REx WT, CLPP−/−, and WT cells that overexpress the C-terminal FLAG-tagged HsCIpP (+CLPP-FLAG) or the proteolytically inactive mutant (+CLPPS153A-FLAG). Data collected on cells grown without Dox are shown on the left panel; data collected in the presence of 0.4 μg/mL Dox are shown on the right panel. FIG. 4F shows IC50 values derived from the cytotoxicity profiles of (E). For HEK293 T-REx CLPP−/−. IC50 in FIG. 2C and FIG. 2F were estimated and are expressed in numerical ranges (denoted with *).



FIGS. 5A-D show graphical representation of ADEP-induced cytotoxicity partially dependent on cell type. FIG. 5A shows cytotoxicity profiles of HEK293 T-REx WT (CLPP+/+) and HEK293 T-REx CLPP−/− cells treated with


ADEP-06 (left panel), ADEP-02 (middle panel) and ADEP-04 (right panel). The relative cell viability was calculated by normalizing all data points to the DMSO-only control for each data set. FIG. 5B shows ADEP-41 cytotoxicity profiles of HeLa (regular) and HeLa T-REx (left panel), U2OS (middle panel) and undifferentiated SH-SYSY cells (right panel). Relative cell viability was calculated by normalizing all data points to the DMSO-only control for each data set. FIG. 5C shows IC50 derived from the cytotoxicity profiles shown in FIG. 5B by non-linear regression analysis using the standard dose response equation. The IC50 for HEK293 T-REx WT is identical to the one shown in FIG. 4B. FIG. 5D shows Western blot for endogenous HsCIpP expression in HEK 293 T-REx WT, HEK 293 T-REx CLPP−/−, HeLa (regular), HeLa T-REx, U2OS, and undifferentiated SH-SY5Y. GAPDH is used as the loading control. The numbers at the bottom represent intracellular HsCIpP level in each cell line, relative to HEK293 T-REx WT. Data points in FIG. 5A and FIG. 5B were collected from 4 independent replicates.



FIGS. 6A-D show images and graphical representation of ADEP inducing intrinsic, caspase-dependent apoptosis. FIG. 6A shows light (DIC) and fluorescence (DAPI, TUNEL) microscopy images of HEK293 T-REx WT and HEK293 T-REx CLPP−/− treated with 10 μM of ADEP-41 or DMSO for 72 hours. The DAPI + TUNEL (Merge) images are shown on the far-right panels. WT cells showing fragmentation of chromosomal DNA and presence of DNA strand breaks (i.e. TUNEL-positive) are marked with white arrows. The scale bars in the DIC panels represent a length of 50 μm. FIG. 6B shows quantification of WT and CLPP−/− cells treated with ADEP-41 or DMSO that have acquired DNA strand breaks as observed in the TUNEL assay. SDs of three independent replicates are shown. FIG. 6C shows Western blots for HsCIpP, HsCIpX and major marker proteins for caspase-dependent apoptosis, on whole cell lysates of WT and CLPP−/− cells treated with 20 μM of ADEP-41 for 24 hours (left panels) or 10 μM of the compound for 72 hours (right panels). “pro” denotes the proform of the caspases. “*” denotes a cross-reacting protein band with the α-Caspase-9 antibody; “**” denotes a possibly non-activated intermediate of Caspase-3 processing. GAPDH was used as a loading control. The primary antibodies used in each blot are indicated to the right of each panel. Protein molecular weight markers are shown on the left. FIG. 6D shows 3DSIM analysis of mitochondrial morphology in WT and CLPP cells during apoptosis. Cells were treated with 2 μM of ADEP-41 or DMSO for 24 or 72 hours prior to imaging. Mitochondria were stained with MitoTracker Red CMXRos and are observable as web-like structures that are indicated with the capital letter M. Fragmentation of mitochondria is indicated with arrows pointing at selected fragments. Nuclei were stained with Hoechst 33342 and are observable as large oval entities that are indicated with the lower-case letter n. Representative images of at least three independent replicates are shown. Scale bars shown correspond to a length of 5 μm.



FIGS. 7A-F show images and Western blots showing activation of the intrinsic, caspase-dependent apoptosis in ADEP-induced cytotoxicity. FIG. 7A shows light microscopy on HEK293 T-REx WT (top 2 rows) and HEK293 T-REx CLPP−/− cells (bottom row) treated with 10 μM of ADEP-41 (panels on left column) or DMSO (panels on right column) over 72 hours. The white bars shown are 100 μm. FIG. 6B shows Western blot for HsCIpX, HsCIpP and GAPDH on whole cell lysates of WT, custom-characterCLPX*, CLPP−/− and CLPP−/−custom-characterCLPX* cells. The blot for HsCIpX is shown with normal exposure time (upper panel of α-HsCIpX) and long exposure time (lower panel of α-HsCIpX). FIG. 6C shows cytotoxicity profiles of HEK293 T-REx WT and CLPP−/− cells (upper left panel) and of custom-characterCLPX* and CLPP−/−custom-characterCLPX* cells (upper right panel) treated with ADEP-28. The IC50 values are shown in the table below. Relative cell viability was calculated by normalizing all data points to the DMSO-only control for each data set. Data points were collected from 4 independent replicates. For WT and ΔCLPX*, IC50 values were derived from non-linear regression analysis using a standard dose response equation (see Methods). For CLPP−/− and CLPP−/-31 ΔCLPX*, the lack of a minimum plateau in the cytotoxicity profiles prohibited the derivation of IC50 by non-linear regression analysis. Instead, IC50 values were estimated and are expressed in numerical ranges (denoted by **). FIG. 6D shows Western blot for Caspase-8 on whole cell lysates of WT and CLPP−/− cells treated with 10 μM ADEP-41 for 72 hours. “pro” denotes the pro form of Caspase-8. Protein molecular weight markers are shown on the left. FIG. 6E shows respiratory (OXPHOS) profiles of WT and CLPP−/− cells during apoptosis. Cell were treated with 1 μM of ADEP-28 or DMSO prior to metabolic measurements. Cellular oxygen consumption rate (OCR) associated with basal respiration, ATP synthase activity, proton leak and non-mitochondrial oxygen consumption (Non-mito O2 consumption) were calculated using normalized OCR data from at least three independent replications shown in FIG. 6F (upper panel) and are shown with ±SDs. FIG. 6F shows original OXPHOS data of WT and CLPP−/−0 cells after a 24-hour treatment with DMSO or ADEP-28. Injection of oligomycin (Oligo), FCCP and the mixture of antimycin A and rotenone (AA/Rtn) to each sample during the experiment was done at the specific time points as shown. Both OCR (upper panel) and ECAR (lower panel) were normalized by Hoechst 33342 nuclear stain fluorescence that reflects cell count. Errors shown correspond to the SDs of individual data sets collected from at least three independently prepared replicates. GAPDH is used as the loading control in FIG. 6B and FIG. 6D.



FIGS. 8A-F show representation of ADEP-binding inducing conformational changes in the HsCIpP tetradecamer. FIG. 8A show the ADEP-28-bound (left) and apo form (right) of the HsCIpP tetradecamer, shown in side views (top) and top views (bottom). The bound ADEP-28 are shown as sticks. The distances shown were measured between the Cα atoms of E109 (helix αB) located on the surface of two apposing CIpP ring. FIG. 8B shows the sequence and the secondary structure of HsCIpP in the presence and absence of ADEP-28. The catalytic triad residues (S153, H178, and D227) are indicated with diamonds. Note that Ser57 was introduced due to cloning. FIG. 8C shows 2Fo-Fc electron density map of ADEP-28 contoured at 1.0 σ. FIG. 8D shows superposition of the monomeric structures of ADEP28-bound HsCIpP (PDB 6BBA, chain A) and apo HsCIpP (PDB 1TG6, chain A), viewed from within the HsCIpP lumen. FIG. 8E shows cartoon putty representation of a single ring of ADEP-28-HsCIpP complex with thicker tubes indicating higher B-factors. FIG. 8F shows 2Fo-Fc map (mesh) contoured at 1.0 σ, and Fo-Fc map (mesh indicted with white arrow) contoured at 3.5 σ showing covalent modification on the thiol group of Cys86 that is observed in each HsCIpP subunit.



FIGS. 9A-C show representation of ADEP-binding causing structural rearrangements within the HsCIpP cylinder. FIG. 9A shows local molecular interactions at the binding pocket between adjacent HsCIpP subunits F and G in the absence of ADEP are shown in (i). Local molecular interactions between ADEP-28 and residues in the binding pocket between two adjacent HsCIpP subunits F and G are shown in (ii). ADEP-28 and the amino acid residues of interest are shown as sticks. Non-covalent bonds are indicated with dotted lines. The axial loop is indicated. FIG. 9B shows ADEP-28 binding induces the ordering of N-terminal axial pore loops into β-hairpins due to interactions shown in the three panels. FIG. 9C shows molecular interactions at the equatorial plane of apo and ADEP-28-bound HsCIpP. The different HsCIpP subunits are distinguished by different shades of grey. All the relevant amino acid residues are shown as sticks. The catalytic triad residues are S153, H178 and D227. Secondary structures of each HsCIpP subunit are labelled as shown, with subunits from the opposite ring denoted with the prime mark (′). Non-covalent bonds between residues are indicated with dotted lines.



FIGS. 10A and B show representation of structural details of the ADEP-28 binding pocket in HsCIpP. FIG. 10A shows a cartoon representation based on LigPlot of the ADEP-28 interactions with amino acid residues from the hydrophobic binding pocket between the two adjacent F and G subunits in HsCIpP. FIG. 10B show superposition of the amino acid residues lining the ADEP-binding pockets of CIpP from: human mitochondria (HsCIpP; this disclosure), Bacillus subtilis (BsCIpP, PDB 3KTI (Lee et al., 2010)), Neisseria meningitidis (NmCIpP, PDB 5DKP (Goodreid et al., 2016)), Staphylococcus aureus (SaCIpP, PDB 5VZ2), Mycobacterium tuberculosis (MtClpP2; PDB 4U0G (Schmitz et al., 2014)) and Escherichia coli (EcCIpP; PDB 3MT6 (Li et al., 2010)). All amino acid residues are shown as sticks and are listed from top to bottom in the same order as their respective CIpP proteins. The ADEPs are shown as very thin sticks.



FIG. 11 shows representation of molecular interactions at the ring-ring interface of different compact and compressed CIpP structures. The structures shown are from E. coli (EcCIpP, 3HLN; contains an A153C mutation introducing a disulfide bridge between two apposing αE helices), L. monocytogenes (LmCIpP1, 4JCQ), M. tuberculosis (MtCIpP1, 2CE3), P. falciparum (PfCIpP, 2F6I), S. aureus (SaCIpP, 4EMM), S. aureus (SaCIpP, 3ST9), and S. pneumoniae (SpCIpP, 1Y7O, contains an A153P mutation). Four neighbouring subunits are represented as cartoons and different shades of grey in the same way as in FIGS. 9A-C, with the relevant residues shown as stick models.



FIGS. 12A and B show representation of the ADEP-28-HsCIpP complex showing the presence of equatorial side pores in different CIpP structures. FIG. 12A shows the structures are shown as viewed from the inside of the HsCIpP tetradecamer. Top left panel—In the compact ADEP-28-HsCIpP complex, the catalytic triad is distorted such that H178 and D227 form hydrogen bonding and/or electrostatic interactions with D227 and H178 residues of the apposing ring, respectively, securing the compact conformation. Distortion of the catalytic triad causes S153 to move away from H178 and D227. The side pore is located in close proximity to the catalytic site. Top right panel—In the extended apo-HsCIpP (PDB 1TG6), the active site surface is significantly different from that of the compact ADEP-28-HsCIpP structure. The catalytic triads are ordered and poised for catalysis of bound peptide substrates, here shown as sticks occupying the substrate-binding cavity. The peptide substrates are shown by superimposing the structure of H. pylori CIpP with bound NVLGFTQ peptide (PDB 2ZL2) (Kim and Kim, 2008) to HsCIpP structure. Bottom panels—Cartoon representation of the compact ADEP-28-HsCIpP complex and extended apo-HsCIpP, showing the relative positions of the catalytic triads and peptide substrates. Four HsCIpP subunits that form the interface are in shades of grey the same way as in FIGS. 9A-C and FIG. 11. In the compact ADEP-28-HsCIpP structure, the unraveling of two N-terminal turns of the αE helix forms a structured loop that impinges on the substrate binding site (bottom left panel). In contrast, in the apo-HsCIpP structure, the β7 strand and αE helix are fully formed and present a cavity for substrate peptide binding. FIG. 12B shows surface representation of compact and compressed CIpP structures from human (HsCIpP, this study), E. coli (EcCIpP, PDB 3HLN), L. monocytogenes (LmCIpP, PDB 4JCQ), M. tuberculosis (MtCIpP1, PDB 2CE3), P. falciparum (PfCIpP, PDB 2F6I), S. aureus (SaCIpP, PDB 4EMM), S. aureus (SaCIpP, PDB 3ST9), and S. pneumoniae (SpCIpP, PDB 1Y7O). Equatorial side pores of distinct sizes and shapes are indicated by black surfaces that are from residues forming the rims of these pores.



FIG. 13 shows representation of putative intermediates in the CIpP functional cycle represented by various CIpP structures from different species. Shown are the relative positions of the Ser-His-Asp catalytic triads in the extended and compact structures of CIpP from different species. The PDB IDs for the extended HsCIpP, EcCIpP, LmCIpP1, and SaCIpP are: 1TG6, 1YG6, 4JCR, and 3STA, respectively; while those for the compact are: this study, 3HLN, 4JCQ, and 4EMM, respectively. Also, there is a compressed structure of SaCIpP with PDB ID 3ST9. Note that the compact state of EcCIpP was the result of the introduction of disulfide bond between two αE helices of apposing rings due to the mutation of A153 to Cys. For LmCIpP1, the WT protein has Asn instead of Asp in the catalytic triad (residue 165) and is in the compact state. The N165D mutant adopts the extended state. The Cα atoms of the catalytic Ser, His, and Asp are shown as spheres in black or different shades of grey. The distances between the different Cα atoms are shown as lines in the. The numbers on the left refer to the distance between the plane formed by the Ser Cα atoms of one heptamer relative to the plane formed by the Ser Cα atoms in the apposing heptamer.



FIGS. 14A-C show graphical representation of small angle X-ray scattering data for HsCIpP. FIG. 14A shows scattering profiles (Log of scattering intensity as a function of the scattering vector q) of apo HsCIpP in buffer, in DMSO-containing buffer and in the presence of ADEP-28. Experimental curves (open circles) were fitted using the GNOM program (solid lines). HsCIpP+DMSO and HsCIpP+ADEP-28 curves were divided by 10 and 100, respectively, for visualization purposes. FIG. 14B shows particle distance distribution functions of HsCIpP in buffer, in DMSO-containing buffer, and in the presence of ADEP-28. The vertical lines indicate maximum p(r) and arrows indicate Dmax. FIG. 14C shows the X-ray structures of apo HsCIpP (PDB: 1TG6) and ADEP-28-bound HsCIpP superimposed on the DAMs of HsCIpP+DMSO and HsCIpP+ADEP-28.



FIG. 15A and B show ADEP-14 are less toxic to cells with lower level of CIpP. FIG. 15A shows Western blots for HsCIpP. FIG. 15B shows cytotoxicity of ADEP-14 on MDA-MB-231 cells.



FIGS. 16A and B show ADEP impairing MDA-MB-231 cell migration in 2D culture. FIG. 16A shows scratch wound healing on MDA-MB-231 WT and CLPP-KD cells in the presence or absence of ADEP-14. FIG. 16B shows quantification of scratch wound closure data by measuring the area of scratch wounds.





DETAILED DESCRIPTION

Unless otherwise indicated, the definitions and embodiments described in this and other sections are intended to be applicable to all embodiments and aspects of the present disclosure herein described for which they are suitable as would be understood by a person skilled in the art.


In understanding the scope of the present disclosure, the term “comprising” and its derivatives, as used herein, are intended to be open ended terms that specify the presence of the stated features, elements, components, groups, integers, and/or steps, but do not exclude the presence of other unstated features, elements, components, groups, integers and/or steps. The foregoing also applies to words having similar meanings such as the terms, “including”, “having” and their derivatives. The term “consisting” and its derivatives, as used herein, are intended to be closed terms that specify the presence of the stated features, elements, components, groups, integers, and/or steps, but exclude the presence of other unstated features, elements, components, groups, integers and/or steps. The term “consisting essentially of”, as used herein, is intended to specify the presence of the stated features, elements, components, groups, integers, and/or steps as well as those that do not materially affect the basic and novel characteristic(s) of features, elements, components, groups, integers, and/or steps.


As used herein, the singular forms “a”, “an” and “the” include plural references unless the content clearly dictates otherwise. The modifier “about” used in connection with a quantity is inclusive of the stated value and has the meaning dictated by the context (e.g., it includes the degree of error associated with measurement of the particular quantity). When referring to a period such as about a year or annually, it includes a range from 9 months to 15 months. All ranges disclosed herein are inclusive of the endpoints, and the endpoints are independently combinable with each other.


As used herein, the term “treating” and its derivatives, refer to improving the condition, such as reducing or alleviating symptoms associated with the condition or improving the prognosis or survival of the subject.


As used herein, the term “effective amount” or “therapeutically effective amount” means an amount effective, at dosages and for periods of time necessary to achieve the desired result. For example, in the context or treating cancer, an effective amount is an amount that, for example, induces remission, reduces tumor burden, and/or prevents tumor spread (e.g. spread by metastasis) or growth compared to the response obtained without administration of the compound. Effective amounts may vary according to factors such as the disease state, age, sex, weight of the subject. The amount of a given polypeptide that will correspond to such an amount will vary depending upon various factors, such as the given drug or compound, the pharmaceutical formulation, the route of administration, the type of disease or disorder, the identity of the subject or host being treated, and the like, but can nevertheless be routinely determined by one skilled in the art.


The present inventors have identified acyldepsipeptide (ADEP) analogs as treatment for cancer. The ADEP analogs activate human mitochondrial CIpP (HsCIpP) in cancer cells, which triggers intrinsic apoptotic pathway, thereby killing the cancer cells.


Accordingly, the present disclosure provides a method for treating a subject having cancer, comprising using or administering a therapeutically effective amount of an ADEP analog to the subject in need thereof. Also provided herein is use of a therapeutically effective amount of ADEP analog for treating a subject with cancer. Further provided is use of a therapeutically effective amount of ADEP analog in the manufacture of a medicament for treating subject with cancer. Even further provided is a therapeutically effective amount of ADEP analog for use in treating a subject with cancer.


The disclosure describes a number of ADEP analogs that are useful for treating cancer. In an embodiment, the methods and uses described herein comprise administering or using an ADEP analog, wherein the ADEP analog is ADEP-01, ADEP-02, ADEP-03, ADEP-04, ADEP-05, ADEP-06, ADEP-07, ADEP-08, ADEP-09, ADEP-10, ADEP-11, ADEP-12, ADEP-13, ADEP-14, ADEP-15, ADEP-16, ADEP-17, ADEP-18, ADEP-19, ADEP-20, ADEP-21, ADEP-22, ADEP-23, ADEP-24, ADEP-25, ADEP-26, ADEP-27, ADEP-28, ADEP-29, ADEP-30, ADEP-31, ADEP-32, ADEP-33, ADEP-34, ADEP-35, ADEP-36, ADEP-37, ADEP-38, ADEP-39, ADEP-40, ADEP-41, ADEP-42, ADEP-43, ADEP-44, ADEP-45, or ADEP-46. In another embodiment, the ADEP analog is ADEP-01, ADEP-02, ADEP-04, ADEP-06, ADEP-10, ADEP-13, ADEP-14, ADEP-15, ADEP-17, ADEP-20, ADEP-25, ADEP-28, ADEP-29, ADEP-30, ADEP-32, ADEP-37, ADEP-38, or ADEP-41. In another embodiment, the ADEP analog is ADEP-14, ADEP-17, ADEP-28, ADEP-29, ADEP-30, ADEP-32, ADEP-37, or ADEP-38. In an aspect, the ADEP analogs described herein are useful for activating protease activity of HsCIpP


Usefulness of structural analogs of ADEP in cancer treatment was assessed by their ability to activate and dysregulate the protease activity of HsCIpP by measuring their relative degradation (RD) index. All ADEP analogs were first screened at 25 μM, and their RD25 scores were calculated as described in the Example. The analogs that achieved RD25≥0.87 were screened a second time at 5 μM, followed by calculation of their RD5 scores. The top analogs with RD5≥0.74 were screened a third time at 1 μM, followed by calculation of their RD1 scores.


In an embodiment, the methods and uses described herein comprise administering or using an ADEP analog, wherein the ADEP analog activates protease activity of HsCIpP, wherein the protease activity is at least 0.2, 0.3, 0.4, 0.5, 0.55, 0.6, 0.65, 0.7, 0.71, 0.72, 0.73, 0.74, 0.75, 0.76, 0.77, 0.78, 0.79, 0.8, 0.85, 0.86, 0.87, 0.88, 0.89, 0.90, 0.91, 0.92, 0.93, 0.94, 0.95, 0.96, 0.97, 0.98, or 0.99 as measured by the RD index at 25 μM, 5 μM or 1 μM of the ADEP analog, preferably at least 0.2, 0.74, or 0.87, optionally at least 0.5, 0.75, 0.8, 0.85, 0.9 or 0.95. In another embodiment, the protease activity is at least 0.87 as measured by the RD index at 25 μM of the ADEP analog, optionally at least 0.9, optionally at least 0.95. In another embodiment, the protease activity is at least 0.74 as measured by the RD index at 5 μM of the ADEP analog, optionally at least 0.8, optionally at least 0.85. In another embodiment, the protease activity is at least 0.2 as measured by the RD index at 1 μM of the ADEP analog, optionally at 0.5, optionally at least 0.75, optionally at least 0.8.


The analogs described herein are useful for treating cancer. For example, the analogs are useful for treating cancer expressing high level of HsCIpP, such as when a cancer cell expresses at least 1.5-fold level of HsCIpP mRNA or protein compared to normal cells. Breast cancer, prostate cancer, colon cancer, liver cancer, uterus cancer, thyroid cancer, lung cancer, lymph nodes cancer, bladder cancer, ovary cancer, stomach cancer, testis cancer, kidney cancer, cervical cancer, osteosarcoma, skin cancer, pancreatic cancer, brain cancer such as neuroblastoma, as well as non-solid cancer such as acute myeloid leukemia, all exhibit high HsCIpP expression. In an embodiment, the methods and uses described herein comprise administering or using an ADEP analog for treating cancer, wherein the cancer is breast cancer, prostate cancer, colon cancer, liver cancer, uterus cancer, thyroid cancer, lung cancer, lymph nodes cancer, bladder cancer, ovary cancer, stomach cancer, testis cancer, brain cancer, skin cancer, pancreatic cancer, acute myeloid leukemia, kidney cancer, cervical cancer, osteosarcoma, or neuroblastoma. In another embodiment, the cancer is non-solid cancer. In another embodiment, the non-solid cancer is acute myeloid leukemia. In another embodiment, the cancer is breast cancer, kidney cancer, cervical cancer, osteosarcoma, or neuroblastoma. In an embodiment, the cancer is breast cancer. In an embodiment, the breast cancer is Invasive ductal carcinoma. In another embodiment, the brain cancer is neuroblastoma. In an embodiment, the cancer is metastatic. In an embodiment, the metastatic cancer is breast cancer. In another embodiment, the cancer cell expresses at least 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 2.5, 3, 3.5, 4, 4.5, 5, 6, 7, 8, 9 or 10-fold level of HsCIpP mRNA or protein compared to a normal cell, optionally at least 1.5-fold.


The term “breast cancer”, as used herein, refers to a cancer that develops from breast tissue. Breast cancers are classified by several grading systems. Breast cancer can be classified by its histological appearance. Breast cancers can be derived from the epithelium lining the ducts or lobules, and these cancers are classified as ductal or lobular carcinoma. Carcinoma in situ is growth of low-grade cancerous or precancerous cells within a particular tissue compartment such as the mammary duct without invasion of the surrounding tissue. In contrast, invasive carcinoma does not confine itself to the initial tissue compartment. Grading is also used for classification. Grading compares the appearance of the breast cancer cells to the appearance of normal breast tissue. Normal cells in an organ like the breast become differentiated, such that they take on specific shapes and forms that reflect their function as part of that organ. Cancerous cells lose that differentiation. In breast cancer, the cells that would normally line up in an orderly fashion make up the milk ducts become disorganized. Cell division becomes uncontrolled. Cell nuclei become less uniform. Breast cancer can also be classified by stages.


Breast cancer staging using the TNM system is based on the size of the tumor (T), whether the tumor has spread to the lymph nodes (N) in the armpits, and whether the tumor has metastasized (M) (i.e. spread to a more distant part of the body). Larger size, nodal spread, and metastasis have a larger stage number and a worse prognosis. The main stages are: Stage 0 is a pre-cancerous or marker condition, either ductal carcinoma in situ (DCIS) or lobular carcinoma in situ (LCIS); Stages 1-3 are within the breast or regional lymph nodes; and Stage 4 is “metastatic” cancer that has a less favorable prognosis since it has spread beyond the breast and regional lymph nodes. The ADEP analogs described herein are useful for treating breast cancer. The ADEP analogs described herein are also useful for treating or preventing metastatic cancer. Further, the ADEP analogs described herein are also useful for treating or preventing metastatic breast cancer.


The term “subject”, as used herein, refers to any individual who is the target of administration or treatment. The subject can be an animal, for example, a mammal, optionally a human. The term “patient” refers to a subject under the care or treatment of a health care professional. In an embodiment, the subject is human. In another embodiment, the subject is non-human animal. In another embodiment, the non-human animal is a pet or a farm-animal. In another embodiment, the non-human animal is an ovine, a bovine, an equine, a caprine, a porcine, a canine, a feline, a rabbit, a rodent or a non-human primate.


The ADEP analog of the present disclosure may be formulated into a pharmaceutical composition, such as by mixing with a suitable excipient, carrier, and/or diluent, by using techniques that are known in the art. For example, the ADEP analog can be administered or used in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavouring or colouring agents, for immediate-, delayed-, modified-, sustained-, pulsed- or controlled-release applications.


The tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycollate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxypropylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included.


Solid compositions of a similar type may also be employed as fillers in gelatin capsules. Preferred excipients in this regard include lactose, starch, a cellulose, milk sugar or high molecular weight polyethylene glycols. For aqueous suspensions and/or elixirs, the inhibitor may be combined with various sweetening or flavouring agents, colouring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.


As used herein, the term “pharmaceutically acceptable carrier” is intended to include any and all solvents, dispersion media, coatings, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical use or administration. Suitable carriers are described in the most recent edition of Remington's Pharmaceutical Sciences, a standard reference text in the field, which is incorporated herein by reference. Optional examples of such carriers or diluents include, but are not limited to, water, saline, ringer's solutions, dextrose solution, and 5% human serum albumin.


In one embodiment, the active ingredient is prepared with a carrier that will protect it against rapid elimination from the body, such as a sustained/controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art.


A pharmaceutical composition is formulated to be compatible with its intended route of use or administration. The use or administration of compound to a subject comprises ingestion, inhalation, injection, or topical application. The route of injection includes but not limited to intradermal, subcutaneous, intramuscular, intravenous, intraosseous, intraperitoneal, intrathecal, epidural, intracardiac, intraarticular, intracavernous, intravitreal, intracerebral, intracerebroventricular, or intraportal. Topical use or administration also includes transdermal application. In an embodiment, the methods and uses described herein comprises an ADEP analog, wherein the ADEP analog is injected, administered, or used via intradermal, subcutaneous, intramuscular, intravenous, intraosseous, intraperitoneal, intrathecal, epidural, intracardiac, intraarticular, intracavernous, intravitreal, intracerebral, intracerebroventricular, or intraportal route. In another embodiment, the ADEP analog is administered or used topically, optionally transdermally. In another embodiment, the ADEP analog is administered or used subcutaneously, intraperitoneally, intravenously, topically, or orally. In a specific embodiment, wherein the cancer is skin cancer, the ADEP analog is administered or used topically, optionally transdermally.


The methods of treating cancer described herein also include selecting a subject determined to have cancer cells comprising an increased level of expression of an HsCIpP gene product relative to a reference expression level. The reference expression level is an expression level of HsCIpP in a non-cancerous cell. The person skilled in art can readily recognize methods for measuring expression level of an HsCIpP gene product relative to the reference expression level, for example, by using Enzyme-linked immunosorbent assay (ELISA), protein microarray, immunoelectrophoresis, Western blotting, immunohistochemistry, High-performance liquid chromatography (HPLC), and/or mass spectrometry.


Also provided is a compound for use in treating a subject having a cancer, wherein the compound is a therapeutically effective amount of an acyldepsipeptide (ADEP) analog. In an embodiment, the ADEP analog activates protease activity of human mitochondrial CIpP (HsCIpP), and wherein the protease activity is at least 0.87 as measured by the relative degradation (RD) index at 25 μM of the ADEP analog, optionally at least 0.9, optionally at least 0.95. In another embodiment, the protease activity is at least 0.74 as measured by the RD index at 5 μM of the ADEP analog, optionally at least 0.8, optionally at least 0.85. In another embodiment, the protease activity is at least 0.2 as measured by the RD index at 5 μM of the ADEP analog, optionally at 0.5, optionally at least 0.75, optionally at least 0.8. In another embodiment, the ADEP analog is ADEP-01, ADEP-02, ADEP-04, ADEP-06, ADEP-10, ADEP-13, ADEP-14, ADEP-15, ADEP-17, ADEP-20, ADEP-25, ADEP-28, ADEP-29, ADEP-30, ADEP-32, ADEP-37, ADEP-38, or ADEP-41. In another embodiment, the ADEP analog is ADEP-14, ADEP-17, ADEP-28, ADEP-29, ADEP-30, ADEP-32, ADEP-37, or ADEP-38.


In an embodiment, the compound is for use in treating a cancer wherein the cancer is breast cancer, prostate cancer, colon cancer, liver cancer, uterus cancer, thyroid cancer, lung cancer, lymph nodes cancer, bladder cancer, ovary cancer, stomach cancer, testis cancer, brain cancer, skin cancer, pancreatic cancer, acute myeloid leukemia, kidney cancer, cervical cancer, osteosarcoma, or neuroblastoma. In another embodiment, the cancer is non-solid cancer. In another embodiment, the non-solid cancer is acute myeloid leukemia. In another embodiment, the cancer is breast cancer, kidney cancer, cervical cancer, osteosarcoma, or neuroblastoma. In an embodiment, the cancer is breast cancer. In an embodiment, the breast cancer is Invasive ductal carcinoma. In another embodiment, the brain cancer is neuroblastoma. In an embodiment, the cancer is metastatic. In an embodiment, the metastatic cancer is breast cancer. In another embodiment, the subject is human. In another embodiment, the subject is non-human animal. In another embodiment, the non-human animal is a pet or a farm-animal. In another embodiment, the non-human animal is an ovine, a bovine, an equine, a caprine, a porcine, a canine, a feline, a rabbit, a rodent or a non-human primate. In another embodiment, the ADEP analog is administered or used subcutaneously, intraperitoneally, intravenously, topically, or orally.


In another aspect, also provided is an acyldepsipeptide (ADEP) analog, wherein the ADEP analog is ADEP-01, ADEP-02, ADEP-03, ADEP-04, ADEP-05, ADEP-06, ADEP-07, ADEP-08, ADEP-09, ADEP-10, ADEP-11, ADEP-12, ADEP-13, ADEP-14, ADEP-15, ADEP-16, ADEP-17, ADEP-18, ADEP-19, ADEP-20, ADEP-21, ADEP-22, ADEP-23, ADEP-24, ADEP-25, ADEP-26, ADEP-27, ADEP-28, ADEP-29, ADEP-30, ADEP-31, ADEP-32, ADEP-33, ADEP-34, ADEP-35, ADEP-36, ADEP-37, ADEP-38, ADEP-39, ADEP-40, ADEP-41, ADEP-42, ADEP-43, ADEP-44, ADEP-45, or ADEP-46, or a variant or a derivative thereof.


The following non-limiting Examples are illustrative of the present disclosure:


EXAMPLES
Example 1: Identification of Acyldepsipeptide Analogs Dysregulate Human Mitochondrial CIpP (HsCIpP) Protease Activity and Cause Apoptotic Cell Death in Cancer Cells
Introduction

The present Example describes the identification of ADEP analogs that target HsCIpP. These analogs increase both the peptidase and protease activity of HsCIpP in vitro and displace HsCIpX from HsCIpP at low compound concentrations. Importantly, treatment of immortalized and cancer cell lines with ADEPs was found to induce cell death in an HsCIpP-dependent manner. A cell line deleted of HsCIpP showed high tolerance to all ADEP analogs tested. ADEP induced cytotoxicity via activating the intrinsic, caspase-dependent apoptosis, leading to cell death. A co-crystal structure of ADEP-HsCIpP was obtained and, unexpectedly, revealed an unusual compacted CIpP conformation.


Materials and Methods
Bacterial Cell Cultures

All Escherichia coli strains used for DNA propagation and protein expression (see Table 1) were grown in Luria-Bertani Broth (LB; 10 g/L bio-tryptone+5 g/L yeast extract+10 g/L NaCl) supplemented with the appropriate antibiotics, unless stated otherwise. For DNA propagation, cells were grown at 37° C. with shaking. For protein expression, cells in pre-cultures were grown 16-18 hours at 37° C. with shaking. Cells in protein expression cultures were also grown at 37° C. with shaking until induction of protein expression with 1 mM IPTG. The cultures were then maintained at 37° C. for 3-5 hours or at 18° C. for 16-18 hours.









TABLE 1







Key Resources









REAGENT or RESOURCE
SOURCE
IDENTIFIER










Antibodies









Mouse monoclonal anti-HsClpP
Abcam
RRID: AB_941071





Rabbit polyclonal anti-HsClpX
Abcam
RRID: AB_11000791





Mouse monoclonal anti-GAPDH
Abcam
RRID: AB_2107448





Rabbit polyclonal anti-Caspase-8
Cell Signaling
RRID: AB_10545768



Technology






Rabbit polyclonal anti-Caspase-9
Cell Signaling
RRID: AB_2068621



Technology






Rabbit monoclonal anti-PUMA
Cell Signaling
RRID: AB_2064551



Technology






Goat polyclonal anti-Caspase-3
R&D Biosystems
RRID: AB_354518





Mouse monoclonal anti-Mcl-1
EMD Millipore
RRID: AB_10806351





Mouse monoclonal anti-Bcl2
EMD Millipore
RRID: AB_11210561





Mouse monoclonal anti-FLAG-tag
Sigma-Aldrich
RRID: AB_262044





HRP-conjugated goat anti-rabbit
BioRad
RRID: AB_11125142


IgG







HRP-conjugated goat anti-mouse
BioRad
RRID: AB_11125547


IgG







HRP-conjugated rabbit anti-goat
Sigma-Aldrich
RRID: AB_258242


IgG












Chemicals, Peptides, and Recombinant Proteins









ADEP-28
(Goodreid et al.,
N/A



2016)






ADEP-41
(Goodreid et al.,
N/A



2016)






ADEP-06
(Goodreid et al.,
N/A



2016)






ADEP-02
(Goodreid et al.,
N/A



2016)






ADEP-04
(Goodreid et al.,
N/A



2016)






Other ADEP analogs, see Table 3
(Goodreid et al.,
N/A



2016)







N-acetyl-Trp-Leu-Ala-7-amido-4-

R&D Biosystems
Cat#S-330-02M


methylcoumarin







Benzyloxycarbonyl-Arg-Arg-7-
Sigma-Aldrich
Cat#C5429


amido-4-methylcoumarin








N-acetyl-Leu-Glu-His-Asp-7-amido-

Sigma-Aldrich
Cat#SCP0104


4-methylcoumarin








N-acetyl-Val-Glu-Thr-Asp-7-amido-

Sigma-Aldrich
Cat#SCP0139


4-methylcoumarin







Benzyloxycarbonyl-Leu-Leu-Leu-7-
Sigma-Aldrich
Cat#C0608


amido-4-methylcoumarin








N-succinyl-Leu-Tyr-7-amido-4-

MP Biochemicals
Cat#03AMC117


methylcoumarin







C-terminal His6-tagged Human
(Kimber et al.,
N/A


mitochondrial ClpP (HsClpP-His6)
2010)






Untagged human mitochondrial
This disclosure
N/A


ClpP (HsClpP)







Untagged human mitochondrial
This disclosure
N/A


ClpX (HsClpX)








Escherichia coli ClpA (EcClpA)

(Lo et al., 2001)
N/A






Escherichia coli ClpP (EcClpP)

(Wojtyra et al.,
N/A



2003)






Yeast SUMO protease
(Lee et al., 2008)
N/A





Casein fluorescein isothiocyanate
Sigma-Aldrich
Cat#C3777-25MG


from bovine milk (Casein-FITC)







Bovine milk α-casein
Sigma-Aldrich
Cat#C6780-1G





Creatine phosphate
Sigma-Aldrich
Cat#27920





Creatine phosphokinase
Sigma-Aldrich
Cat#C3755





Adenosine 5′-triphosphate disodium
Sigma-Aldrich
Cat#A26209


salt hydrate (ATP)







Sulforhodamine B (SRB)
Sigma-Aldrich
Cat#230162










Critical Commercial Assays









In Situ Cell Death Detection Kit,
Roche
Cat#Roche-


Fluorescein

11684795910





MitoTracker Red CMXRos
Molecular Probes
Cat#M7512





Gateway LR Clonase II Enzyme Mix
Thermo-Fisher
Cat#11791020



Scientific






Gateway BP Clonase II Enzyme
Thermo-Fisher
Cat#11789020


Mix
Scientific






Seahorse XF Cell Mitochondrial
Agilent
Cat#103015-100


Stress Kit












Deposited Data









X-ray structure: HsClpP in complex
This disclosure
PDB: 6BBA


with ADEP-28







X-ray structure: HsClpP
(Kang et al., 2004)
PDB: 1TG6





X-ray structure: BsClpP in complex
(Lee et al., 2010)
PDB: 3KTI


with ADEP1







X-ray structure: EcClpP
(Bewley et al.,
PDB: 1YG6



2006)






X-ray structure: EcClpP A153C
(Kimber et al.,
PDB: 3HLN



2010)






X-ray structure: EcClpP in complex
(Li et al., 2010)
PDB: 3MT6


with ADEP1







X-ray structure: HpClpP in complex
(Kim and Kim,
PDB: 2ZL2


with peptide NVLGFTQ
2008)






X-ray structure: LmClpP1 N65D
(Zeiler et al., 2013)
PDB: 4JCR





X-ray structure: LmClpP1
(Zeiler et al., 2013)
PDB: 4JCQ





X-ray structure: MtClpP1
(Ingvarsson et al.,
PDB: 2CE3



2007)






X-ray structure: MtClpP1P2 in
(Schmitz et al.,
PDB: 4U0G


complex with ADEP
2014)






X-ray structure: NmClpP in complex
(Goodreid et al.,
PDB: 5DKP


with ADEP A54556
2016)






X-ray structure: PfClpP
(El Bakkouri et al.,
PDB: 2F6I



2010)






X-ray structure: SaClpP (extended) 
(Zhang et al., 2011)
PDB: 3STA





X-ray structure: SaClpP (compact)
(Ye et al., 2013)
PDB: 4EMM





X-ray structure: SaClpP
(Zhang et al., 2011)
PDB: 3ST9


(compressed)







X-ray structure: SaClpP in complex
This disclosure
PDB: 5VZ2


with ADEP







X-ray structure: SpClpP A153P
(Gribun et al.,
PDB: 1Y7O



2005)











Experimental Models: Cell Lines









HEK293 T-REx
Gift from A.
RRID: CVCL_U427



Trifunovic






HEK293 T-REx CLPP-/-
(Seiferling et al.,
N/A



2016)






HEK293 T-REx + CLPP-FLAG
This disclosure
N/A





HEK293 T-REx + CLPPS153A-FLAG
This disclosure
N/A





HEK293 T-REx ΔCLPX*
This disclosure
N/A





HEK293 T-REx CLPP-/- ΔCLPX*
This disclosure
N/A





HeLa
Gift from P. Kim
RRID: CVCL_0030





HeLa T-REx
Gift from L.
RRID: CVCL_D587



Attisano






U2OS
Gift from A.
RRID: CVCL_0042



Palazzo






SH-SY5Y
Gift from M. Babu
RRID: CVCL_0019










Experimental Models: Organisms/Strains










Escherichia coli DH5α

Lab stock
NCBI: txid668369






Escherichia coli BL21(DE3)

(Kimber et al.,
N/A


ΔclpP::cat (SG1146)
2010)











Oligonucleotides









CLPX Exon1 pX330 F:
This disclosure
N/A


CACCGCGGTGCTTGTACTTGCG




GCG (SEQ ID NO: 1)







CLPX Exon1 pX330 R:
This disclosure
N/A


AAACCGCCGCAAGTACAAGCAC




CGC (SEQ ID NO: 2)







CLPX Exon1 ssODN:
This disclosure
N/A


GGCCTCGCGGAGATGCCCAGCT




GCGGTGCTTGTACTTGCGGCGC




GGCGTAGGTCCGGCTCATCACC




TCCTCACTCGCCTCCGCGCAGA




GA (SEQ ID NO: 3)












Recombinant DNA









pDT1668-LhclpP
(Kimber et al.,
N/A



2010)






pET9a-EcClpA M168T
(Lo et al., 2001)
N/A





pET9a-EcClpP
(Wojtyra et al.,
N/A



2003)






pET28-Ulp1
(Lee et al., 2008)
N/A





pETSUMO
(Lee et al., 2008)
N/A





pETSUMO2-CLPP(-MTS)
This disclosure
N/A





pST39
(Selleck and Tan,
N/A



2008)






pST39-SUMO2-CLPX(-MTS)
This disclosure
N/A





pDEST-MTS-HsCLPP-3xFLAG
This disclosure
N/A





pDEST-MTS-HsCLPPS153A-3xFLAG
This disclosure
N/A





pOG44
Thermo-Fisher
Cat#V600520



Scientific






pX330
(Cong et al., 2013)
N/A





pX330-CLPX KO
This disclosure
N/A










Software and Algorithms









HKL2000
(Otwinowski and
http://www.hkl-



Minor, 1997)
xray.com/download-




instructions-hkl-2000





PHENIX
(Adams et al.,
https://www.phenix-



2010)
online.org/





COOT
(Emsley and
http://www2.mrc-



Cowtan, 2004)
lmb.cam.ac.uk/Personal/




pemsley/coot/





ATSAS 2.7.2 software package
(Petoukhov et al.,
https://www.embl-



2012)
hamburg.de/biosaxs/




manuals/atsas-




2.7/install.html





UCSF Chimera software 
(Pettersen et al.,
https://www.cgl.ucsf.edu/



2004)
chimera/





MaxQuant software 1.6.1.0
Max Planck
http://www.coxdocs.org/



Institute
doku.php?id=maxquant:




common:download_and_




installation#download_




and_installation_guide





NIS-Elements Basic Research
Nikon
https://www.nikoninstruments.com/


software

Products/Software/NIS-




Elements-Basic-Research





ZEN Black software 8.1
Carl Zeiss
https://www.zeiss.com/



Microscopy
microscopy/int/software-




cameras.html





Wave Desktop software
Agilent
https://www.agilent.com/




en/products/cell-




analysis-




(seahorse)/software-




download-for-wave-




desktop





Quantity One 1D Analysis software
BioRad
http://www.bio-




rad.com/en-




ca/product/quantity-one-




1-d-analysis-




software?ID=1de9eb3a-




1eb5-4edb-82d2-




68b91bf360fb










Other









Rigaku FR-E SuperBright rotating
Structural
https://www.thesgc.org/


anode generator
Genomics
science/crystallography



Consortium,




University of




Toronto, Toronto,




Canada






Pilatus 300K detector, SAXS1
Brazilian
http://www.lnls.cnpem.br/


beamline
Synchrotron Light
linhas-de-luz/saxs1-



Laboratory, LNLS,
en/overview/



CNPEM,




Campinas, Brazil






Proxeon EASY nanoLC 1000
Thermo-Fisher
Cat#LC120


System
Scientific






Orbitrap Elite mass spectrometer
Thermo-Fisher
Cat#IQLAAEGAAPFAD



Scientific
BMAZQ





Acclaim PepMap C18 column (15
Thermo-Fisher
https://www.thermofisher.com/


cm x 50 μm ID, 3 μm, 100 Å)
Scientific
order/catalog/product/




160321





Eclipse 80i fluorescence
Nikon
https://www.nikoninstruments.com/


microscope

Products/Upright-




Microscopes/Research/




Eclipse-80i





X-Cite Series 120Q excitation
Excelitas
http://www.excelitas.com/


light source
Technologies
Pages/Product/X-Cite-




120Q.aspx





ELYRA PS.1 superresolution
Carl Zeiss
https://www.zeiss.com/


microscope
Microscopy
microscopy/int/products/




superresolution-




microscopy.html





iXon 885 EMCCD camera
Andor Technology
http://www.andor.com/




cameras/ixon-emccd-




camera-series





Microscope coverslips for SIM
Electron
Cat#72230-01



Microscopy




Sciences






Microscope glass slides for SIM
VWR
Cat#48312-401


ChemiDoc XRS + System
BioRad
http://www.bio-




rad.com/en-




ca/product/chemidoc-




xrs-




system?ID=NINJHRKG4





Seahorse XFe96 Metabolic Flux
Agilent
https://www.agilent.com/


Analyzer

en/products/cell-




analysis-




(seahorse)/seahorse-




analyzers/seahorse-




xfe96-analyzer





Seahorse XFe96 Sensor Cartridge
Agilent
Cat#102416-100





Seahorse XF96 Cell Culture
Agilent
Cat#101085-004


Microplates







EnSpire 2300 Multilabel Reader
Perkin-Elmer
http://www.perkinelmer.com/




product/enspire-




base-unit-2300-0000





96-well black flat-bottom plate
Greiner Bio-one
Cat#655077



International









Mammalian Cell Cultures

All mammalian cell lines used (see Table 1) were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 2 mM L-glutamine and 100 U/mL penicillin-streptomycin, unless stated otherwise. Cells were grown and maintained in standard tissue culture plates or tissue culture flasks with ventilated caps. For DNA transfection, cells were kept in the same media but without any antibiotics at least 24 hours prior to the procedure. All cells were passaged by standard trypsinization procedures at least three times before use in any experiments.


For long-term cryogenic storage, cells were first trypsinized and re-suspended in media, followed by gradual addition of 80% FBS+20% DMSO to a final 1:1 (v:v) ratio. The cell stocks were then transferred into cryogenic storage vials and stored in foam storage racks at −80° C. for at least 24 hours before long-term storage in a liquid nitrogen storage tank. To recover cells from the frozen stocks, the frozen cells were thawed at 37° C. and re-suspended in media at 9× the frozen stock's volume. The media was applied gradually to the cells to minimize osmotic shock. Cells were then collected by centrifugation and re-suspended in fresh media after which, they were transferred to tissue culture plates or ventilated tissue culture flasks for growth and maintenance as described above.


Molecular Cloning and Protein Purification

For protein expression, CLPP gene was cloned without its mitochondrial targeting sequence (MTS) (residues M1-P57) into a modified pETSUMO vector (Lee et al., 2008) resulting in pETSUMO2-CLPP(-MTS) expressing 2×(His6-thrombin)-SUMO-CLPP(-MTS). CLPX gene was also cloned without its MTS (residues M1-F64) into a modified pST39 (Selleck and Tan, 2008) resulting in pST39-SUMO2-CLPX(-MTS) expressing 2x(His6-thrombin)-SUMO-CLPX(-MTS). All plasmids were propagated in E. coli DH5α and isolated using the PureLink Quick Plasmid Miniprep Kit (Thermo-Fisher Scientific). Linearized DNA and PCR amplification products were purified by gel extraction using the PureLink Quick Gel Extraction Kit (Thermo-Fisher Scientific).


HsCIpP was expressed with a N-terminal 2×(His6-thrombin)-SUMO tag in E. coli SG1146, which is BL21(DE3) ΔcIpP::cat (Kimber et al., 2010). Cells transformed with pETSUMO2-CLPP(-MTS) were grown aerobically in LB+50 μg/mL kanamycin at 37° C. until OD600 reached ˜0.6. Protein expression was then induced with 1.5 mM IPTG (Thermo-Fisher Scientific) for 3-5 hours. Cells were harvested by centrifugation, and then re-suspended in 25 mM TrisHCl, pH 7.5, 0.5 M NaCl, 10% glycerol, and 10 mM imidazole. Cells were then lysed by 2 passes on the French Press, and the cell debris was removed by centrifugation. The SUMO-tagged HsCIpP was purified on Ni-NTA beads (Thermo-Fisher Scientific) using standard protocols. Subsequently, SUMO protease (Lee et al., 2008) was added to remove the N-terminal 2×(His6-thrombin)-SUMO tag. The purified, untagged HsCIpP was concentrated with an Amicon Ultra-15 centrifugal filter unit (10000 MWCO) (EMD Millipore) at 4° C., aliquoted, flash-frozen in liquid nitrogen and stored at −80° C. until use. A similar protocol was used for HsCIpX. All fractions collected during the purification were analyzed by SDS-PAGE. The proteins were found to be >95% pure.


For high-throughput drug activity assays, C-terminally Hiss-tagged HsCIpP (HsCIpP-Hiss) was expressed in SG1146 transformed with pDT1668-LhclpP and purified as described in (Kimber et al., 2010). Untagged E. coli CIpP (EcCIpP) was expressed and purified as described in (Wojtyra et al., 2003). E. coli CIpA (EcCIpA) was expressed and purified as described in (Lo et al., 2001).


Screen of ADEP Analogs Against HsCIpP

Structural analogs of acyldepsipeptide (ADEP) (Goodreid et al., 2016) that were previously developed as potential antibiotics against bacterial CIpP were screened and assessed for their ability to activate and dysregulate the protease activity of HsCIpP, by measuring their relative degradation (RD) index. Note that ADEP-28 was also previously reported by Carney et al. (2014). Details of the experimental protocol have been described in Leung et al., 2011. The RD index is defined as follows:






RD
=





(

Δ






ϕ

ClpP
+
compound



)


after





6





hrs


-


(

Δ






ϕ
ClpP


)


after





6





hrs






(

Δ






ϕ


E
.




coli






ClpAP



)


after





6





hrs


-


(

Δ






ϕ


E
.




coli






ClpP



)


after





6





hrs




.





Δφ is the change in fluorescence after 6 hr of starting the reaction measured using 485 nm excitation and 535 nm emission, which primarily detects the signal from casein-FITC. E. coli CIpAP was used as a benchmark for maximum CIpP proteolytic activity. The CIpP in the numerator can be from any other organism. RD25 measurements refer to the measurement in the presence of 25 μM compound. All ADEP analogs were first screened at 25 μM, and their RD25 scores were calculated using the above formula and method as described in Leung et al., 2011 (FIG. 1A). Those that achieved RD25≥0.87 were screened a second time at 5 μM, followed by calculation of their RD5 scores. The top analogs with RD5≥0.74 were screened a third time at 1 μM, followed by calculation of their RD1 scores (FIG. 1B).


Peptidase Activity Assays

The peptidase activity of purified, untagged HsCIpP was assessed by monitoring the degradation of peptidyl substrates labelled with a C-terminal 7-amido-4-methylcoumarin (AMC) fluorophore. As previously reported (Kang et al., 2002), HsCIpP did not exhibit any observable peptidase activity towards the commonly used N-succinyl-Leu-Tyr-7-amido-4-methylcoumarin (Suc-LY-AMC) peptide (FIG. 2A). To identify a suitable peptidyl substrate, commercially available AMC-labelled peptides were purchased and tested for degradation by HsCIpP. These include N-acetyl-Trp-Leu-Ala-AMC (Ac-WLA-AMC) (R&D Systems) that was also previously used by Csizmadia et (Csizmadia et al., 2016), benzyloxycarbonyl-Arg-Arg-AMC (Z-RR-AMC) (Sigma-Aldrich), N-acetyl-Leu-Glu-His-Asp-AMC (Ac-LEND-AMC) (Sigma-Aldrich), N-acetyl-Val-Glu-Thr-Asp-AMC (Ac-VETD-AMC) (Sigma-Aldrich), and benzyloxycarbonyl-Leu-Leu-Leu-AMC (Z-LLL-AMC) (Sigma-Aldrich). Suc-LY-AMC (MP Biomedicals) was also tested as a negative control. All stock solutions of the AMC-labelled peptides were prepared by dissolving the lyophilized peptides in DMSO at 90 mM or a lower concentration recommended by the manufacturer. All assays were performed in 50 mM TrisHCl, pH 8, 10 mM MgCl2, 100 mM KCl, 0.02% Triton X-100 (v:v), 5% glycerol (v:v), and 1 mM DTT. HsCIpP was used at 8 μM (final monomeric concentration) for the initial screening for a suitable substrate, but at 6 μM in subsequent experiments. All AMC-labelled peptides were used at the final concentration of 500 μM in the initial screen. Ac-WLA-AMC was used at the final concentration of 100 μM in all other experiments. Addition of the peptides also raised the final concentration of DMSO in the reaction mix to 3% (v:v). All reactions were carried out with 150 μL of the reaction mix per well on a 96-well black flat-bottom plate (Greiner) at 37° C. Degradation of the peptidyl substrates and subsequent release of the free AMC moiety was monitored by fluorescence with λex=360 nm and λem=440 nm using the EnSpire 2300 Multilabel Reader (Perkin-Elmer) set to perform a reading every 30 s with 20 flashes. A total of 240 readings were collected for each of three independent replicates prepared for each peptidyl substrate.


Protease Activity Assays

The protease activity of purified HsCIpP was assessed by monitoring the degradation of bovine milk casein labelled with fluorescein isothiocyanate (casein-FITC) (Sigma-Aldrich) in the presence of ADEP and/or purified HsCIpX. All reactions were carried out in 25 mM HEPES, pH 7.5, 35 mM KCl, 25 mM MgCl2, 0.03% Tween-20, 10% glycerol, and 1 mM DTT, supplemented with 16 mM creatine phosphate and 300 μM ATP. HsCIpP was used at 3 μM (final monomeric concentration) and casein-FITC was added at the final concentration of 10 μM. 13 U of creatine phosphokinase (Sigma-Aldrich) was also added for ATP regeneration. DMSO was present at 1% (v:v) final concentration in the experiments that involve the use of ADEP analogs. All reactions were carried out with 150 μL of the reaction mix per well in a 96-well black flat-bottom plate at 37° C. Degradation of casein-FITC and subsequent release of the free FITC moiety was monitored by fluorescence with λex=494 nm and λem=518 nm using the EnSpire 2300 Multilabel Reader set to perform a reading every 40 s with 20 flashes. A total of 100 readings were collected in each experiment.


To investigate the casein-FITC degradation by HsCIpXP, kinetic analysis was performed with HsCIpP held constant at 3 μM (final monomeric concentration) and HsCIpX was titrated from 0 to 18 μM (final monomeric concentration). Initial rates (v0) were determined from data collected over three independent experiments. Non-linear regression analysis was then performed on the initial rates data using the Hill equation, as follows:







v
0

=




V
max



[

HsClpX





or





ADEP

]


h



K

0
.
5

h

+


[

HsClpX





or





ADEP

]

h







Vmax is maximal velocity, h is the Hill coefficient, and K0.5 is the microscopic apparent dissociation constant.


The effect of ADEP in activating HsCIpP was assessed via the degradation of casein-FITC by HsCIpP independent of HsCIpX. All reactions were carried out with HsCIpP at 3 μM (final monomeric concentration) in the same way as described above. ADEP analogs were titrated from 0-2 μM or 0-128 μM, depending on their potency. Initial rates data were collected and analyzed using the Hill equation above to determine Vmax, h and K0.5 for each ADEP analog. For experiments that require HsCIpX in the reaction, the protein was added at 9 μM (final monomeric concentration).


For gel-based degradation assays, unlabelled bovine milk α-casein (Sigma-Aldrich; C6780-1G) was used in place of casein-FITC in the same reaction condition as before. HsCIpP and HsCIpX were used at 6 μM and 18 μM (both final monomeric concentrations), respectively, to ensure that sufficient amount of casein was degraded to be observable by SDS-PAGE, while maintaining the same HsCIpP-to-HsCIpX ratio as previously used. ATP concentration was also raised to 3 mM. As a representative analog, ADEP-28 was used at 0.1 μM and 2 μM alongside a DMSO-only control. Samples were collected at designated time points and analyzed by SDS-PAGE. Casein degradation was quantified by densitometry using the Quantity One 1D Analysis Software (BioRad). Three independent experiments were performed for each concentration of ADEP-28 and the DMSO-only control.


X-Ray Structure Determination of ADEP-28-HsCIpP

Purified untagged HsCIpP was dialyzed against buffer containing 25 mM Bis-Tris, pH 6.5 and 3 mM dithiothreitol, concentrated to 10 mg/mL, and 750 μM of ADEP-28 was added. The solution was pre-incubated for 1 hr at 37° C. Subsequently, 0.5 μL of the protein-ADEP-28 solution was mixed with 0.5 μL of crystallization reagent (0.1 M sodium acetate trihydrate pH 4.6, 4% w/v Polyethylene glycol 4,000). The drops were equilibrated against the crystallization reagent in the reservoir at 21° C. Large crystals of HsCIpP were observed within two weeks and grew to maximal dimensions within a month.


Crystals of HsCIpP were cryoprotected by quick soaking in crystallization solution containing 20% glycerol and 1 mM ADEP-28, then flash-cooled in liquid nitrogen. Diffraction data were collected at the Structural Genomics Consortium-University of Toronto at 100 K and wavelength of 1.5418 Å using Rigaku FR-E SuperBright rotating anode generator. Diffraction data were indexed, integrated, and scaled in HKL2000 (Otwinowski and Minor, 1997). The structure of HsCIpP complex with ADEP-28 was determined by molecular replacement using Phaser in PHENIX (Adams et al., 2010) with the published apo-HsCIpP structure (PDB 1TG6) (Kang et al., 2004) as search model. The crystal asymmetric unit contained 7 subunits of HsCIpP forming a single heptameric ring. The structure was refined initially in PHENIX with simulated annealing and coordinate shaking to remove model bias. Subsequent model building and refinement were performed in COOT (Emsley and Cowtan, 2004) and PHENIX (Adams et al., 2010) using translation-libration-screw (TLS) parameters with individual coordinate, occupancy and B factor optimization. The final model was refined to an Rwork/Rfree of 0.19/0.24 up to a resolution of 2.80 Å. The model has good geometry with >97% of amino acid residues in favored and allowed regions of the Ramachandran plot. Data collection and refinement statistics are summarized in Table 2. The PDB accession number is 6BBA.









TABLE 2





Data collection and refinement statistics of


ADEP-28-HsCIpP structure Data Collection







Data Collection










Space group
P 32 2 1







Cell dimensions










a, b, c (Å)
172.40, 172.40, 135.96



α, β, γ (°)
90.0, 90.0, 120.0,



Wavelength (Å)
1.5418











Resolution (Å)
46.73-2.80
(2.90-2.80)










Rmeas/Rpim
0.188/0.056











|/σ|
15.4
(1.3)



Completeness (%)
99.9
(100.0)



Redundancy
11.1
(11.0)







Refinement










Resolution (Å)
46.73-2.80











No. reflections
57,813
(5,716)










Rwork/Rfree
0.1866/0.2346



No. atoms
10,846



Protein
10,246



Ligand
392



Water
208







B factors










Protein
62.9



Ligand
61.3



Water
57.7







r.m.s. deviations










Bond lengths (Å)
0.010



Bond angles (°)
1.18







Values in parentheses are for the highest resolution shell.



The highest resolution shell includes all reflections between 2.80 and 2.90 Å.



Rwork is Σ|Fo-Fc|/ΣFo.



Rfree is the cross-validation R-factor computed for a test set of reflections (3.5% of total).






Small Angle X-Ray Scattering (SAXS) Experiments

Data collection was performed using a Pilatus 300K detector in the SAXS1 beamline, located at the Brazilian Synchrotron Light Laboratory (LNLS, CNPEM, Campinas, Brazil). Measurements were done using a monochromatic X-ray beam (λ=1.488 Å) and sample-to-detector distance of ˜1,000 mm. The scattering intensity, I(q), was detected as a function of the scattering vector (q), where q=4π sinθ/λ and 2θ is the scattering angle. The range for q was between 0.013 Å−1 and 0.5 Å−1. Samples were prepared at 0.5 mg/mL in buffer of 50 mM TrisHCl, pH 7.5, 200 mM KCl, 25 mM MgCl2 and 10% glycerol. The final concentration of DMSO was kept at 2.3% in samples containing ADEP (0.6 mM final) or DMSO only. Ten frames of 10 seconds and one frame of 300 seconds were recorded for every sample at 20° C. to inspect for X-ray damage.


Data analysis was performed using software from the ATSAS 2.7.2 package (Petoukhov et al., 2012). All data curves were verified for X-ray damage and aggregation using the Guinier approximation and the PRIMUS software. Molecular weight determination was performed by comparing the forward scattering intensity 1(0)-values obtained from Guinier approximation and I(0)-values determined from standard BSA samples (Barbosa et al., 2013; Mylonas and Svergun, 2007). Curves were normalized by protein concentration. The pair distance distribution functions, p(r), were generated by the GNOM software. Generation of dummy atoms models (DAMs) was performed using the SAXS curve until q=0.2 and P72 symmetry in the simulated-annealing method implemented by the DAMMIF software. Ten DAM models were averaged using the DAMAVER package. Superimposition of crystallographic structures and DAMs was performed using the SUPCOMB program. Visualization of DAMs and generation of density maps were done using the UCSF Chimera software (Pettersen et al., 2004).


Testing the Effect of ADEPs on Different Cell Lines

Different cell lines were grown and maintained in Dulbecco's


Modified Eagle Media (DMEM) (Gibco) supplemented with 10% fetal bovine serum (Gibco), 100 U/mL penicillin-streptomycin (Gibco) and 2 mM L-glutamine (Gibco) at 37° C. under a moist atmosphere with 6% CO2. All cells were passaged at least 3 times prior to use.


Cell lines used in the experiments presented here were acquired from various sources. HEK293 T-REx wild-type (WT) and HEK293 T-REx CLPP−/− cells were obtained from Professor Aleksandra Trifunovic (University of Cologne, Germany). HeLa (regular) cells were obtained from Professor Peter Kim (Hospital for Sick Children, Canada). HeLa T-REx cells were obtained from Professor Lilianna Attisano (University of Toronto, Canada). U2OS cells were obtained from Professor Alex Palazzo (University of Toronto, Canada). Undifferentiated SH-SYSY cells were obtained from Professor Mohan Babu (University of Regina, Canada).


HEK293 T-REx+CLPP-FLAG and HEK293 T-REx+CLPPS153A-FLAG were generated using the Flp-In System (Sigma-Aldrich). Briefly, full length CLPP gene that includes its native MTS was cloned as pDEST-MTS-HsCLPP-3×FLAG using the Gateway system (Thermo Fisher Scientific). For expressing the proteolytically inactive HsCIpP, the S153A mutation was introduced into pDEST-MTS-HsCLPP-333 FLAG by QuikChange site-directed mutagenesis (Qiagen) to generate the pDEST-MTS-HsCLPPS153A-3×FLAG. To generate the required cell lines expressing the FLAG-tagged HsCIpP proteins, HEK293 T-REx WT cells were co-transfected with pDEST-MTS-HsCLPP-3×FLAG or pDEST-MTS-HsCLPP(S153A)-3×FLAG and pOG44 (Thermo-Fisher Scientific), using the jetPRIME In Vitro DNA Transfection Reagent (Polyplus Transfection). Stable cell populations were isolated by multiple rounds of selection in media supplemented with 200 μg/mL hygromycin.


HEK293 T-REx ΔCLPX* and HEK293 T-REx CLPP−/− ΔCLPX* were generated by disrupting the CLPX gene in HEK293 T-REx WT and CLPP−/− cells, respectively, using CRISPR-Cas9 methodology as detailed in Cong et al., 2013. Briefly, the required gRNA sequence targeting in proximity to exon 1 of CLPX was introduced into pX330 with the oligonucleotides CLPX Exon1 pX330 F and CLPX Exon1 pX330 R (sequence shown in Table 1) via designated BbsI restriction sites. Next, the modified pX330 plasmid and the oligonucleotide CLPX Exon1 ssODN (sequence shown in Table 1) were co-transfected into WT and CLPP−/− cells using Lipofectamine 2000 (Thermo-Fisher Scientific) following the manufacturer's protocol. CLPX Exon1 ssODN provides the necessary repair template after the Cas9-mediated DNA cleavage and causes omission of CLPX's start codon to inhibit protein expression. The efficacy of CLPX disruption in suppressing HsCIpX expression was assessed by Western blotting. Both WT and CLPP were subjected to two additional rounds of CRISPR-Cas9 treatment in sequence to maximize the suppression of HsCIpX expression.


To assess the cytotoxicity of ADEP analogs, cells were seeded at 1×103 or 2×103 cells in 50 μL per well on sterile 96-well flat-bottom tissue culture plates (VWR). Growth media without any cells was included as a control. Cells were grown for at least 24 hours to allow proper adherence to the growth surface. Afterwards, ADEP analogs were serially diluted and introduced to the tissue cultures via fresh growth media at 50 μL per well. Inclusion of activators/DMSO brought the final DMSO concentration of the growth media up to a maximum of 0.5% (v:v). For experiments that involve overexpression of FLAG-tagged HsCIpP (WT or S153A mutant), Dox (Sigma-Aldrich) was also applied at 0.4 μg/mL (final concentration) as needed. A total of four independent replicates were prepared for each cell line and for each growth condition used. Cells were grown in the presence of ADEP/DMSO for 72 hours.


To assess cell survival, live adherent cells were fixed with 10% (w:v) trichloroacetic acid (TCA) at 4° C. for 1-2 hours. The tissue culture plates were then rinsed with water to remove dead cells and cell debris and air-dried overnight. Fixed cells were stained with a solution of 0.4% (w:v) sulforhodamine B (SRB) (Sigma-Aldrich) dissolved in 1% (v:v) acetic acid at room temperature for 30 minutes, followed by plate-rinsing with 1% acetic acid and air-drying. The SRB retained was extracted with 10 mM of Tris base (200 μL per sample). Absorbance at λ=510 nm (A510) was then measured using the EnSpire 2300 Multilabel Reader. A510 is linearly proportional to the amount of cellular protein present in each sample.


To determine the IC50 for the ADEP analogs, relative cell viability (RCV) values were calculated by normalizing the A510 readings in ADEP-containing sample to the DMSO-only control. Plots of RCV vs. ADEP concentration were constructed, followed by non-linear regression analysis using a standard dose response equation, to determine the required IC50 values.






RCV
=


RCV
min

+



RCV
max

-

RCV
min



1
+

1


0


(


log






IC

5

0



-

log


[
ADEP
]



)


h










In this equation, RCVmin and RCVmax are the minimum and maximum RCV observed, respectively; [ADEP] is the molar concentration of


ADEP applied; and h is the Hill coefficient. For HEK293 T-REx CLPP−/− and HEK293 T-REx CLPP−/−custom-characterCLPX* cells, IC50 values were estimated by manual examination of the plotted data due to the lack of a minimum plateaus, which prohibited meaningful analysis using the Hill model.


Mitochondrial Proteome Profiling for WT and CLPP−/−

HEK293 T-REx WT and CLPP−/− cells were grown to ˜80% confluence, with two biological replicates prepared for WT and three for CLPP−/−. All cells were chemically crosslinked by treatment with 0.5 mM of the cell membrane-permeable crosslinker dithiobis succinimidyl propionate (DSP) for 30 min at room temperature. The crosslinking reagent was quenched from the samples by the addition and incubation with 100 mM Tris-HCl (pH 7.5) and 2 mM EDTA for 10 min at room temperature. Cells were suspended by gentle pipetting and harvested by centrifugation at 600×g for 5 min and then washed twice in ice-cold NKM buffer (10 mM TrisHCl pH 7.5, 130 mM NaCl, 5 mM KCl, and 7.5 mM MgCl2). The pellet was resuspended in ice-cold buffer containing 10 mM TrisHCl (pH 6.8), 130 mM NaCl, 10 mM KCl, 150 mM MgCl2, 1 mM PMSF, and 1 mM DTT. The resuspended cells were then allowed to swell for 10 min, followed by lysis via repeated passages through a syringe fitted with a 23-gauge needle. Cell lysates were added to 1 cell-pellet-volume of 2 M sucrose and centrifuged at 1,300×g for 5 min at 4° C. The resulting supernatant was further centrifuged at 7,000×g for 10 min at 4° C. to pellet the mitochondrial fraction.


To lyse the mitochondrial fraction and further alkylate the mitochondrial proteins, the pellet was dissolved in 8 M urea, 20 mM TrisHCl pH 7.5 and 8 mM chloroacetamide, and subjected to sonication for 30 s with 15-30 s cooling cycles for 3 min on ice. After incubating the lysate at room temperature for 30 min, 50 mM TrisHCl was added to dilute the urea concentration from 8 M to 2 M. To this mixture, 20 mM DTT and 5 μg/mg-of-sample of Trypsin Gold (Promega) were also added to trypsinize the proteins overnight at room temperature. Trypsin activity was stopped by adding 1 μL of trifluoroacetic acid until a pH of 2-3 was reached. After desalting the samples using the C18 packed tips (Glygen Corp), the bound peptides were eluted for the tips with 0.1% formic acid and 60% acetonitrile. Protein content for WT and CLPP−/− samples was assessed by Bradford assay and adjusted to 0.9 mg. These were then dried and resuspended in 1% formic acid for mass spectrometry analysis.


All samples were analyzed by a Proxeon EASY nanoLC 1000 System (Thermo-Fisher Scientific) coupled to an Orbitrap Elite mass spectrometer (Thermo-Fisher Scientific). For chromatographic separation, 5 μL of WT and CLPP−/− samples with same protein concentration was loaded onto an Acclaim PepMap C18 column (15 cm×50 μm ID, 3 μm, 100 A; Thermo-Fisher Scientific). Peptides were separated using the following elution gradients at time points in sequence: 0-3% B (0-2 min); 3-24% B (2-170 min); 24-100% B (172-190 min); 100% B (190-200 min). For this step, Buffer A refers to 0.1% formic acid and Buffer B refers to 0.1% formic acid in acetonitrile. Separation of peptides was achieved at a column flow rate of 0.30 μL/min. Eluted peptides were immediately ionized by positive electrospray ionization at an ion source temperature of 250° C. and an ion spray voltage of 2.1 kV. Full-scan MS spectra (m/z 350-2000) was acquired in the Orbitrap at mass resolution of 60000 (m/z 400) using the positive ion mode. The automatic gain control was set at 1 e6 for full FTMS scans and 5e4 for MS/MS scans. Fragmentation was performed with collision-induced dissociation (CID) in the linear ion trap with an ion intensity of >1500 counts. The 15 most intense ions isolated for ion trap CID with charge states ≥2 were sequentially fragmented using the normalized collision energy setting at 35%, activation Q at 0.250, and an activation time of 10 ms. Ions selected for MS/MS were dynamically excluded for 30 s.


The mass spectra files were then searched using MaxQuant ver. 1.6.1.0 (Max Planck Institute) against a UniProt human protein database of canonical sequences. The resulting data were filtered for contaminants and reverse matches. The protein intensities of the detected mitochondrial proteins were normalized against the pooled WT samples, and Student t-test was used to identify proteins with a 1.5-fold change (CLPP−/−/WT) at a p-value significance 0.05.


Light and Fluorescence Microscopy

HEK293 T-REx WT and CLPP−/− cells were grown on glass cover slips placed in a 6-well tissue culture plate. Prior to use, the cover slips were cleaned by treatment with 1 M HCl for 2-3 days, followed by thorough rinsing with distilled water and a second treatment with 95% ethanol for 16-18 hours. The cleaned cover slips were sterilized by autoclaving and dried under UV light. To ensure proper attachment of the cells, the sterilized cover slips were coated with gelatin by application of a 0.2% solution and incubation at 37° C. for at least 2 hours. Excess gelatin solution was then removed, and the cover slips were allowed to dry in a sterile environment for 2-4 hours before use.


WT cells treated with ADEP-41 were seeded at a density of 1×105 cells/mL (2 mL per well) to compensate for lack of cell growth and division resulting from the ADEP treatment. WT cells treated with DMSO and CLPP cells treated with ADEP-41 or DMSO were all seeded at 5×104 cells/mL (2 mL per well). After the cells have adhered properly, ADEP-41 (20 μM for a 24-hour treatment; 10 μM for a 72-hour treatment) or DMSO was applied via media exchange (final DMSO concentration at 0.2% for all samples). Cells were then grown for 24 or 72 hours prior to fixing and permeabilization for microscopy.


Cells were fixed in the presence of 4% formaldehyde at room temperature for 30-45 minutes, followed by membrane permeabilization with 0.1% Triton X-100 (in PBS from Gibco) supplemented with 0.1% sodium citrate at room temperature for 30 minutes. All samples were rinsed gently with PBS in between steps. For the TUNEL (Terminal deoxynucleotidyl transferase dUTP nick end labelling) assay, the fixed and permeabilized cells were labelled using the In Situ Cell Death Detection Kit, Fluorescein (Roche-11684795910), following the manufacturer's instructions. Cells were mounted onto standard glass microscope slides with DAPI Fluoromount-G (Southern Biotech) for visual examination using the Eclipse 80i fluorescence microscope (Nikon) equipped with the X-Cite Series 120Q excitation light source (Excelitas Technologies). Both light and fluorescence microscopic images were captured and analyzed using the NIS-Elements Basic Research Software (Nikon).


3D Structured Illumination Microscopy (3DSIM)

HEK293 T-REx WT and CLPP'1/− cells were seeded on coverslips (Electron Microscopy Sciences) pre-coated with poly-D-lysine hydrobromide (Sigma-Aldrich) in a 12-well culture plate (Falcon). Cells were grown for 24 hours to allow adhesion, followed by treatment with 2 μM ADEP-41 or DMSO in the same manner as described in previous sections, for 24 or 72 hours. At these time points, the original media was removed, and cells were incubated for 30 minutes at 37° C., 5% CO2 with 200 nM MitoTracker Red CMXRos (Molecular Probes) in Opti-MEM™ Reduced Serum Medium (Gibco). Cells were then washed once with PBS and fixed with 4% (v/v) paraformaldehyde (Electron Microscopy Sciences) in PBS for 20 minutes at room temperature.


The fixed cells were then washed twice with PBS, permeabilized with 0.2% (v/v) Triton X-100 (BioShop) in PBS for 10 minutes at room temperature and incubated with 2 μg/mL Hoechst 33342 (Molecular Probes) in PBS for 10 minutes at room temperature to counterstain nuclei. Coverslips were then washed three times with PBS+0.05% (v/v) Tween-20 and mounted on microscope glass slides (VWR) using 0.5% (w/v) propyl gallate (Sigma-Aldrich) dissolved in glycerol as the mounting medium.


3DSIM data were collected on the ELYRA PS.1 super resolution microscope (Carl Zeiss Microscopy) using a 63× (1.4 NA) Plan-Apochromatic oil immersion objective (Carl Zeiss Microscopy) and a 1.6× Optovar. For imaging, fluorophores were laser-excited at wavelengths 405 nm and 555 nm, and the emissions were collected with band-pass 420-480, 570-620 filters, respectively. Z-stacks were acquired over a 10-μm thickness with 101 nanometre-steps, using an iXon 885 EMCCD camera (Andor). For each image field, grid excitation patterns were acquired for five phases and three rotation angles (−75°, −15°, +45°). Raw data were reconstructed using the SIM module of ZEN Black software version 8.1 (Carl Zeiss Microscopy), with a Wiener noise filter value of −5. The final images were obtained from the reconstructed data by using a maximum intensity projection (MIP) algorithm, implemented by the ZEN Black software.


Western Blots

HEK293 T-REx WT, HEK293 T-REx CLPP−/−, HeLa (regular), HeLa T-REx, U2OS and undifferentiated SH-SYSY cells were grown in the presence of ADEP or DMSO as described in previous sections. Cells were harvested by the standard trypsinization protocol and counted with a hemocytometer. Cell lysates were prepared by first re-suspending the cells in PBS at 5×106 cells/mL, followed by sonication. Proteins in the lysates were analyzed by Western blotting using Immobilin-P PVDF membranes (EMD Millipore). Blots were imaged and analyzed with the ChemiDoc XRS+ System (BioRad). Quantification of HsCIpP expression was performed by densitometry using the Quantity One 1D Analysis Software (BioRad). HsCIpP expression levels were corrected for sample loading errors and normalized to the expression level in HEK293 T-REx WT. Primary antibodies for HsCIpP, HsCIpX and GAPDH were purchased from Abcam. Primary antibodies for Caspase-8, Caspase-9 and PUMA were purchased from Cell Signaling Technologies. The primary antibody for Caspase-3 was purchased from R&D Systems. Primary antibodies for Mcl-1 and Bcl-2 were purchased from EMD Millipore. The primary antibody for the FLAG-tag was purchased from Sigma-Aldrich. HRP-conjugated goat anti-rabbit IgG (for HsCIpX, Caspase-8, Caspase-9 and PUMA) and HRP-conjugated goat anti-mouse IgG (for HsCIpP, Mcl-1, Bcl-2, GAPDH and FLAG-tag) were purchased from BioRad. HRP-conjugated rabbit anti-goat IgG (for Caspase-3) was purchased from Sigma-Aldrich.


Cellular Respiration Analysis

HEK293 T-REx WT and CLPP31 /− cells were seeded onto Seahorse XF96 Cell Culture Microplates (Agilent) pre-coated with poly-D-lysine (Sigma-Aldrich). Adherent cells were then grown for 24 hours in the presence of 1 μM of ADEP-41 or DMSO as described in previous sections. Cellular respiration was assessed using the Seahorse XF Cell Mitochondrial Stress Kit (Agilent) following the manufacturer's protocol. Oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) were monitored using the Seahorse XFe96 Analyzer (Agilent) fitted the XFe96 sensor cartridge (Agilent). Carbonyl cyanide-4-(trifluoromethoxy)phenylhydrazone (FCCP) that was included in the Mitochondrial Stress Kit was applied to the cell cultures at a final concentration of 0.25 μM. Application of other reagents were done as outlined in the manufacturer's experimental guidelines. Data analysis was performed with the Wave Desktop program (Agilent) following the manufacturer's instructions. For OCR and ECAR normalization, cells were first fixed with 4% formaldehyde as described in previous sections. Nuclear DNA of the fixed cells was stained with Hoechst 33342 (Invitrogen) dissolved in PBS at 5 μg/mL for 30 minutes at room temperature, followed by three rounds of washes with PBS. To measure Hoechst 33342 fluorescence, 50 μL of PBS was applied to each well, followed by fluorescence measurements (λex=350 nm; λem=461 nm) using the EnSpire 2300 Multilabel Reader. Fluorescence data were expressed arbitrarily in 104 HFU (Hoechst fluorescence units) to keep the normalized OCR and ECAR within conventional numerical ranges. Operation of the Seahorse XFe96 Analyzer and data collection was performed by the SPARC BioCentre (Hospital for Sick Children, Toronto, Canada).


Quantification and Statistical Analysis

Statistical analyses and software that were used for specific experiments have been described in the relevant subsections under materials and methods. In general, all quantitative data were collected from at least three independent replicates and are presented as means±SD. All enzyme kinetics experiments and cytotoxicity assays on mammalian cells were repeated multiple times to ensure data reproducibility. Quantitative microscopy data for TUNEL were derived from examining at least 50 cells per sample replicate per repeated experiment.


Data and Software Availability

The structure of the ADEP-28-HsCIpP complex has been deposited in the Protein Data Bank (PDB) under the accession number 6BBA. All software used for the experiments presented here are available at the sources listed in the Table 1.


Results

Identification of ADEP Analogs that Dysregulate HsCIpP


To identify ADEP analogs that allow HsCIpP to degrade folded proteins independent of HsCIpX+ATP (i.e., that activate or dysregulate HsCIpP), the inventors employed the in vitro screening method that was used for identifying small molecule activators of Escherichia coli CIpP (EcCIpP) (Leung et al., 2011). In this approach, the ability of HsCIpP to degrade FITC-labeled casein (casein-FITC) was measured and compared to the ability of EcCIpAP to degrade casein-FITC. This is quantitatively expressed as relative degradation (RD) index (Leung et al., 2011).


Forty-six ADEP analogs (Goodreid et al., 2016) were tested against HsCIpP (see FIGS. 1D-1I for structures). Initially, the analogs were screened at a final concentration of 25 μM and their RD25 scores (i.e., RD at 25 μM of compound) were measured (FIG. 1A). Top hits were then screened at 5 μM and then at 1 μM compound concentration (FIG. 1B and Table 3). For follow-up experiments, five ADEP analogs (FIG. 10) were chosen based on their diverse RD1 scores to provide a good representation across the potency spectrum. Furthermore, the selected ADEP analogs have distinct structural differences (FIG. 1C).









TABLE 3





RD25. RD5 and RD1 scores of ADEP analogs in HsCIpP



















Hs
Hs



Compound
RD25
SD
SMILES





ADEP-02
1.08
0.08
O═C([C@H]C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC═CC═C3)





NC(/C═C/CC═OCC)═O)═O)CO4)═O)CCC2)═O)═O)





N5CCC[C@@]5([H])C4═O


ADEP-10
1.07
0.03
O═([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC═CC═C3)





NC(/C═C/CCCC)═O)═O)CO4)═O)CCC2)═O)═O)





N5C[C@H](C)C[C@@]5([H])C4═O


ADEP-18
1.07
0.04
O═C([C@H](C)NC([C@H](C)N(C)C([C@@]





1([H])N(C([C@@H](NC([C@H](CC2═CC═CC═C2)





NC(/C═C/CCCCC)═O)═O)CO3)═O)CCC1)═O)═O)





N4CCC[C@@]4([H])C3═O


ADEP-06
1.05
0.04
O═C([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])





N(C([C@@H](NC([C@H](CC2═CC(F)═CC(F)═C2)





NC(/C═C/CCCC)═O)═O)CO3)═O)CCC1)═O)═O)





N4C[C@H](C)C[C@@]4([H])C3═O


ADEP-15
1.05
0.02
O═C([C@H](C)NC([C@@](CCC1)([H])N1C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)NC





(/C═C/CCCC)═O)═O)CO4)═O)CCC2)═O)═O)N5C[C@H]





(C)C[C@@]5([H])C4═O


ADEP-20
1.04
0.03
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(F)═CC═C3)





NC(/C═C/CCC)═O)═O)CO4)═O)CCC2)═O)═O)N5C





[C@N(C)C[C@@]5([H])C4═O


ADEP-32
1.03
0.11
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)NC





(/C═C/CCCC)═O)═O)CN4)═O)CCC2)═O)═O)N5C[C@N(C)





C[C@@]5([H])C4═O


ADEP-04
1.03
0.02
O═C([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N(C([C@@H]





(NC([C@H](CC2═CC═CC═C2)NC(/C═C/C═C/CCC)═O)═O)





CO3)═O)CCC1)═O)═O)N4C[C@N(C)C[C@@]4([H])C3═O


ADEP-13
1.02
0.02
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)





NC(/C═C/CC4CC4)═O)═O)[C@N(C)O5)═O)CCC2)═O)═O)





N6C[C@N(C)C[C@@]6([H])C5═O


ADEP-17
1.02
0.02
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)NC





(/C═C/CC4CCCCC4)═O)═O)[C@H](C)O5)═O)CCC2)═O)═O)





N6C[C@N(C)C[C@@]6([H])C5═O


ADEP-21
1.00
0.03
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3CCCCC3)NC





(/C═C/CCCC)═O)═O)CO4)═O)CCC2)═O)═O)N5C





[C@H](C)C[C@@]5([H])C4═O


ADEP-14
1.00
0.03
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)NC





(/C═C/C═C/CCC)═O)═O)[C@H](C)O4)═O)CCC2)═O)═O)N5C





[C@H](C)C[C@@]5([H])C4═O


ADEP-30
0.98
0.05
O═C([C@H](C)NC([C@@]1([H])N(CC[C@H](C)C1)C([C@@]2





([H])N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)





NC(/C═C/CCCC)═O)═O)CO4)═O)CCC2)═O)═O)N5C[C@H](C)





C[C@@]5([H])C4═O


ADEP-05
0.98
0.02
O═([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N





(C([C@@H]( N C([C@H](CC2═CC═CC═C2)NC





(/C═C/C ═C/C ═C/C)═O)═O)CO3)═O)CCC1)═O)═O)





N4C[C@N(C)C[C@@]4([H])C3═O


ADEP-25
0.97
0.04
O═C([C@H(CC)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)NC





(/C═C/CCCC)═O)═O)CO4)═O)CCC2)═O)═O)N5C[C@H](C)C





[C@@]5([H])C4═O


ADEP-08
0.97
0.03
O═([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N





(C([C@@H](NC([C@H](CC2═CC═CC═C2)NC





(/C═C/C═C/C═C/C)═O)═O)CO3)═O)CCC1)═O)═O)





N4CCC[C@@]4([H])C3═O


ADEP-41
0.96
0.04
O═([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2





([H])N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)NC





(/C═C/CCCC)═O)═O)[C@N(C)O4)═O)CCC2)═O)═O)N5CCC





[C@@]5([H])C4═O


ADEP-01
0.96
0.04
O═([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N





(C([C@@H](NC([C@H](CC2═CC═CC═C2)NC





(/C═C/CCCC)═O)═O)CO3)═O)CCC1)═O)═O)N4CCC





[C@@]4([H])C3═O


ADEP-28
0.95
0.04
O═([C@H](C)NC([C@@]1([H])N(CC[C@H](C)C1)C





([C@@]2([H])N(C([C@@H](NC([C@H]





(CC3═CC(F)═CC(F)═C3)NC(/C═C/CCCC)═O)═O)





[C@H](C)O4)═O)CCC2)═O)═O)N5CCC[C@@]5([H])C4═O


ADEP-38
0.95
0.04
O═([C@H](C)NC([C@@]1([H])N(CC[C@H](C)C1)C([C@@]2





([H])N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3





NC(/C═C/CCCC)═O)═O)[C@H](C)O4)═O)CCC2)═O)═O)N





5C[C@N(C)C[C@@]5([H])C4═O


ADEP-37
0.92
0.04
O═C([C@H](C)NC([C@@]1([H])N(CCC2═C1C═CC═C2)





C([C@@]3([H])N(C([C@@H](NC([C@H](CC4═CC(F)═CC





(F)═C4)NC(/C═C/CCCC)═O)═O)CO5)═O)CCC3)═O)═O)N6C





[C@H](C)C[C@@]6([H])C5═O


ADEP-29
0.92
0.04
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)NC





(/C═C/CCCC)═O)═O)[C@N(C)O4)═O)CCC2)═O)═O)





N5C[C@H](C)C[C@@]5([H])C4═O


ADEP-43
0.87
0.03
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(C═CC═C4)═C4C═C3)





NC(/C═C/CCCC)═O)═O)CO5)═O)CCC2)═O)═O)N6C[C@H]





(C)C[C@@]6([H])C5═O


ADEP-42
0.87
0.02
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CSC4═C3C═CC═C4)NC





(/C═C/CCCC)═O)═O)CO5)═O)CCC2)═O)═O)N6C[C@N(C)





C[C@@]6([H])C5═O


ADEP-26
0.86
0.02
O═C([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])





N(C([C@@H](NC([C@H](CC2═CC═CC═C2)NC





(/C═C/C═C/C)═O)═O)CO3)═O)CCC1)═O)═O)





N4CCC[C@@]4([H])C3═O


ADEP-39
0.78
0.04
O═C([C@H](C)NC([C@@]1([H])N(CC[C@H](C)C1)





C([C@@]2([H])N(C([C@@H](NC([C@H]





(CC3═CC(F)═CC(F)═C3)NC(/C═C/C4═CC═CC





(C(F)(F)F)═C4)═O)═O)[C@H](C)O5)═O)CCC2)═O)═O)





N6C[C@N(C)C[C@@]6([H])C5═O


ADEP-11
0.77
0.03
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)NC





(/C═C/CCCC)═O)═O)CO4)═O)CCC2)═O)═O)N5C





[C@N(C)C[C@@]5([H])C4═O


ADEP-09
0.76
0.03
O═C([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N





(C([C@@H](NC([C@H](CC2═CC═CC═C2)NC





(/C═C/CCC)═O)═O)CO3)═O)CCC1)═O)═O)





N4CCC[C@@]4([H])C3═O


ADEP-03
0.66
0.03
O═C([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])





N(C([C@@H](NC([C@H](CC2═CC═CC═C2)NC





(/C═C/C═C/C)═O)═O)CO3)═O)CCC1)═O)═O)





N4C[C@N(C)C[C@@]4([H])C3═O


ADEP-27
0.64
0.02
O═C([C@H](C)NC([C@H](C)N(C)C([C@@]1





([H])N(C([C@@H](NC([C@H](CC2═CC═CC═C2)





NC(C#CCCCCC)═O)═O)CO3)═O)CCC1)═O)═O)





N4CCC[C@@]4([H])C3═O


ADEP-07
0.61
0.02
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)NC





(/C═C/CCCCC(O)═O)═O)═O)[C@N(C)O4)═O)CCC2)═O)═O)





N5C[C@N(C)C[C@@]5([H])C4═O


ADEP-44
0.42
0.02
O═([C@H](C)NC([C@@]1([H])N(CC[C@H](C)C1)C([C@





@]2([H])N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)





NC(/C═C/C4═CC═CC═C4C(F)(F)F)═O)═O)





[C@N(C)O5)═O)CCC2)═O)═O)





N6C[C@H](C)C[C@@]6([H])C5═O


ADEP-12
0.36
0.02
O═([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N





(C([C@@H](NC([C@H](CC2═CC═CC═C2)NC





(C#CCCCC)═O)═O)CO3)═O)CCC1)═O)═O)





N4CCC[C@@]4([H])C3═O


ADEP-19
0.34
0.02
O═([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N





(C([C@@H](NC([C@H](CC2═CC═CC═C2)NC





(/C═C\CCCC)═O)═O)CO3)═O)CCC1)═O)═O)





N4CCC[C@@]4([H])C3═OO═([C@H](C)NC





([C@@]1([H])N(CCCC1)C([C@@]2([H])


ADEP-45
0.29
0.01
N(C([C@@H](NC([C@H](CC(C(F)═C3F)═C(F)C(F)═C3F)





NC(/C═C/CCCC)═O)═O)CO4)═O)CCC2)═O)═O)





N5C[C@H](C)C[C@@]5([H])C4═O


ADEP-23
0.12
0.02
O═([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N





(C([C@@H](NC([C@H](CC2═CC═CC═C2)NC





(C#CCCC)═O)═O)CO3)═O)CCC1)═O)═O)





N4CCC[C@@]4([H])C3═O


ADEP-33
0.07
0.01
O═([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N





(C([C@@H](NC([C@H](CC2═CC═CC═C2)NC





(/C═C\CCC)═O)═O)CO3)═O)CCC1)═O)═O)





N4CCC[C@@]4([H])C3═O


ADEP-16
0.06
0.13
O═([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])





N(C([C@@H]( N C([C@H](CC2═CC═CC═C2)NC





(OCC3═CC═CC═C3)═O)═O)CO4)═O)CCC1)═O)═O)





N5C[C@N(C)C[C@@]5([H])C4═O


ADEP-46
0.02
0.02
O═([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N





(C([C@@H]( N C([C@H](CC2═CC ═CC ═C2)NC





(/C═C/C ═C/CC3(H])OCCCC3)═O)═O)CO4)═O)





CCC1)═O)═O)N5C[C@H](C)C[C@@]5([H])C4═O


ADEP-36
0.00

O═([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N





(C([C@@H](NC([C@H](CC2═CC═CC═C2)





NC(/C═C/C═C/CO)═O)═O)CO3)═O)CCC1)═O)═O)





N4C[C@H](C)C[C@@]4([H])C3═O


ADEP-24
0.00

O═([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC═CC═C3)NC





(C4═CC═C([N+]([O−])═O)C═C4)═O)═O)





CO5)═O)CCC2)═O)═O)N6CCC[C@@]6([H])C5═O


ADEP-34
0.00

O═([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N(C





([C@@H](NC([C@H](CC2═CC═CC═C2)NC





(OC(C)(C)C)═O)═O)CO3)═O)CCC1)═O)═O)





N4CCC[C@@]4([H])C3═O


ADEP-35
0.00

O═([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N(C([C@@H]





(NC([C@H](CC2═CC═CC═C2)NC(OC(C)(C)C)═O)═O)





CO3)═O)CCC1)═O)═O)N4C[C@H](C)C[C@@]4([H])C3═O


ADEP-31
0.00

O═([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N(C([C@@H]





(NC([C@H](CC2═CC═CC═C2)N)═O)CO3)═O)CCC1)═O)═O)





N4C[C@N(C)C[C@@]4([H])C3═O


ADEP-22
0.00

O═([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N





(C([C@@H](NC([C@H](CC2═CC═CC═C2)NC





(OCC3═CC═CC═C3)═O)═O)CO4)═O)CCC1)═O)═O)





N5CCC[C@@]5([H])C4═O


ADEP-40
0.00

O═([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N(C([C@@H]





(NC([C@H](CC2═CC═CC═C2)N)═O)CO3)═O)





CCC1)═O)═O)N4CCC[C@@]4([H])C3═O






Hs
Hs



Compound
RD5
SD
SMILES





ADEP-32
0.96
0.03
O═([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)NC





(/C═C/CCCC)═O)═O)CN4)═O)CCC2)═O)═O)N5C





[C@N(C)C[C@@]5([H])C4═O


ADEP-06
0.94
0.02
O═([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N





(C([C@@H](NC([C@H](CC2═CC(F)═CC(F )═C2)NC





(/C═C/CCCC)═O)═O)CO3)═O)CCC1)═O)═O)N4C





[C@N(C)C[C@@]4([H])C3═O


ADEP-30
0.93
0.02
O═([C@H](C)NC([C@@]1([H])N(CC[C@H](C)C1)C





([C@@]2([H])N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)





NC(/C═C/CCCC)═O)═O)CO4)═O)CCC2)═O)═O)





N5C[C@H](C)C[C@@]5([H])C4═O


ADEP-41
0.93
0.03
O═([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)NC





(/C═C/CCCC)═O)═O)[C@N(C)O4)═O)CCC2)═O)═O)





N5CCC[C@@]5([H])C4═O


ADEP-28
0.93
0.05
O═C([C@H](C)NC([C@@]1([H])N(CC[C@H](C)C1)C





([C@@]2([H])N(C([C@@H](NC([C@H]





(CC3═CC(F)═CC(F)═C3)NC(/C═C/CCCC)═O)═O)





[C@H](C)O4)═O)CCC2)═O)═O)N5CCC[C@@]5([H])C4═O


ADEP-25
0.92
0.02
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([CC@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)NC





(/C═C/CCCC)═O)═O)CO4)═O)CCC2)═O)═O)





N5C[C@H](C)C[C@@]5([H])C4═O


ADEP-14
0.92
0.02
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)NC





(/C═C/C═C/CCC)═O)═O)[C@H](C)O4)═O)CCC2)═O)═O)N5





C[C@H](C)C[C@@]5([H])C4═O


ADEP-15
0.92
0.02
O═C([C@H](C)NC([C@@](CCC1)([H])N1C([C@@]2([H])N





(C([C@@H](NC([CC@I-INC3═CC(F)═CC(F)═C3)NC(/C═C/





CCCC)═O)═O)CO4)═O)CCC2)═O)═O)N5C[C@H](C)C[C@





@]5([H])C4═O


ADEP-13
0.91
0.03
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)NC





(/C═C/CC4CC4)═O)═O)[C@N(C)O5)═O)CCC2)═O)═O)





N6C[C@N(C)C[C@@]6([H])C5═O


ADEP-17
0.91
0.02
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)NC





(/C═C/CC4CCCCC4)═O)═O)[C@H](C)O5)═O)CCC2)═O)═O)





N6C[C@N(C)C[C@@]6([H])C5═O


ADEP-20
0.90
0.03
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(F)═CC═C3)NC





(/C═C/CCC)═O)═O)CO4)═O)CCC2)═O)═O)N5C





[C@N(C)C[C@@]5([H])C4═O


ADEP-38
0.89
0.03
O═C([C@H](C)NC([C@@]1([H])N(CC[C@H](C)C1)C([C@@]2





([H])N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)NC





(/C═C/CCCC)═O)═O)[C@H](C)O4)═O)CCC2)═O)═O)N





5C[C@N(C)C[C@@]5([H])C4═O


ADEP-02
0.88
0.02
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC═CC═C3)NC





(/C═C/CCCC)═O)═O)CO4)═O)CCC2)═O)═O)





N5CCC[C@@]5([H])C4═O


ADEP-29
0.87
0.03
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(F)═CC(F)═C3)NC





(/C═C/CCCC)═O)═O)[C@N(C)O4)═O)CCC2)═O)═O)





N5C[C@H](C)C[C@@]5([H])C4═O


ADEP-37
0.86
0.03
O═C([C@H](C)NC([C@@]1([H])N(CCC2═C1C═CC═C2)





C([C@@]3([H])N(C([C@@H](NC([C@H]





(CC4═CC(F)═CC(F)═C4)NC





(/C═C/CCCC)═O)═O)CO5)═O)CCC3)═O)═O)





N6C[C@H](C)C[C@@]6([H])C5═O


ADEP-10
0.85
0.03
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC═CC═C3)NC





(/C═C/CCCC)═O)═O)CO4)═O)CCC2)═O)═O)





N5C[C@N(C)C[C@@]5([H])C4═O


ADEP-O4
0.78
0.02
O═C([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N





(C([C@@H]( N C([C@H](CC2═CC═CC═C2)NC





(/C═C/C═C/CCC )═O)═O)CO3)═O)CCC1)═O)═O)





N4C[C@N(C)C[C@@]4([H])C3═O


ADEP-18
0.72
0.02
O═C([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])





N(C([C@@H](NC([C@H](CC2═CC═CC═C2)NC





(/C═C/CCCCC)═O)═O)CO3)═O)CCC1)═O)═O)





N4CCC[C@@]4([H])C3═O


ADEP-21
0.70
0.02
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3CCCCC3)





NC(/C═C/CCCC)═O)═O)CO4)═O)CCC2)═O)═O)





N5C[C@H](C)C[C@@]5([H])C4═O


ADEP-01
0.70
0.02
O═C([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])





N(C([C@@H](NC([C@H](CC2═CC═CC═C2)





NC(/C═C/CCCC)═O)═O)CO3)═O)CCC1)═O)═O)





N4CCC[C@@]4([H])C3═O


ADEP-05
0.69
0.02
O═C([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N





(C([C@@H](NC([C@H](CC2═CC═CC═C2)





NC(/C═C/C═C/C═C/C)═O)═O)CO3)═O)CCC1)═O)═O)





N4C[C@N(C)C[C@@]4([H])C3═O


ADEP-43
0.55
0.02
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CC(C═CC═C4)═C4C═C3)





NC(/C═C/CCCC)═O)═O)CO5)═O)CCC2)═O)═O)N6C[C@H]





(C)C[C@@]6([H])C5═O


ADEP-08
0.52
0.02
O═C([C@H](C)NC([C@H](C)N(C)C([C@@]1([H])N





(C([C@@H](NC([C@H](CC2═CC═CC═C2)





NC(/C═C/C═C/C═C/C)═O)═O)CO3)═O)CCC1)═O)═O)





N4CCC[C@@]4([H])C3═O


ADEP-42
0.49
0.01
O═C([C@H](C)NC([C@@]1([H])N(CCCC1)C([C@@]2([H])





N(C([C@@H](NC([C@H](CC3═CSC4═C3C═CC═C4)NC





(/C═C/CCCC)═O)═O)CO5)═O)CCC2)═O)═O)N6C[C@N(C)





C[C@@]6([H])C5═O






Hs
Hs



Compound
RD1
SD
SMILES





ADEP-17
0.86
0.02
O═C(N1C[C@@H](C[C@]12[H])CAC@@H](NC([C@]





3(N(C([C@]4(N(CCC4)C([C@H]([C@@H](OC2═O)C)





NC([C@@H](NC(/C═C/CC5CCCCC5)═O)





CC6═CC(F)═CC(F)═C6)═O)═O)[H])═O)CCCC3)[H])═O)C


ADEP-28
0.82
0.03
O═C(N1CCC[C@]12[H])[C@@H](NC([C@]3(N(C([C@]





4(N(CCC4)C([C@H]([C@@H](OC2═O)C)NC([C@@H]





(NC(/C═C/CCCC)═O)CC5═CC(F)═CC(F)═C5)═O)═O)[H])═O)





CC[C@@H](C3)C)[H])═O)C


ADEP-14
0.79
0.02
O═C(N1C[C@@N(C[C@]12[H])C)[C@@H](NC([C@]





3(N(C([C@]4(N(CCC4)C([C@H]([C@@H](OC2═O)C)





NC([C@@H](NC(/C═C/C═C/CCC)═O)





CC5═CC(F)═CC(F)═C5)═O)═O)[H])═O)CCCC3)[H])═O)C


ADEP-41
0.79
0.02
O═C(N1CCC[C@]12[H])[C@@H](NC([CC@]3(N(C([C@]





4(N(CCC4)C([C@H]([C@@H](OC2═O)C)NC([CC@@H]





(NC(/C═C/CCCC)═O)CC5═CC(F)═CC(F)═C5)═O)═O)





[H])═O)CCCC3)[H])═O)C


ADEP-32
0.79
0.02
O═C(N1C[C@@N(C[C@]12[H])C)[C@@H](NC([C@]





3(N(C([C@]4(N(CCC4)C([C@H](CNC2═O)NC([C@@H]





(NC(/C═C/CCCC)═O)CC5═CC(F)═CC(F)═C5)═O)═O)[H])═O)





CCCC3)[H])═O)C


ADEP-29
0.76
0.02
O═C(N1C[C@@N(C[C@]12[H])C)[C@@H](NC([C@]





3(N(C([CC@]4(N(CCC4)C([C@H]([C@@H](OC2═O)C)





NC([C@@H](NC(/C═C/CCCC)═O)





CC5═CC(F)═CC(F)═C5)═O)═O)H])═O)CCCC3)[H])═O)C


ADEP-38
0.76
0.03
O═C(N1C[C@@N(C[C@]12[H])C)[C@@H](NC([C@]





3(N(C([C@]4(N(CCC4)C([C@H]([C@@H](OC2═O)C)





NC([C@@H](NC(/C═C/CCCC)═O)





CC5═CC(F )═CC(F)═C5)═O)═O)H])═O)





CC[C@@N(C3)CAN)═O)C


ADEP-30
0.69
0.02
O═C(N1C[C@@N(C[C@]12[H])C)[C@@H](NC([C@]





3(N(C([C@]4(N(CCC4)C([C@H](COC2═O)NC([C@@H]





(NC(/C═C/CCCC)═O)CC5═CC(F)═CC(F)═C5)═O)═O)





[H])═O)CC[C@@H](C3)C)[H])═O)C


ADEP-37
0.68
0.03
O═C(N1C[C@@N(C[C@]12[H])C)[C@@H](NC([C@]





3(N(C([C@]4(N(CCC4)C([C@H](COC2═O)NC([C@@H]





(NC(/C═C/CCCC)═O)CC5═CC(F)═CC(F)═C5)═O)═O)





[H])═O)CCC6═C3C═CC═C6)[H])═O)C


ADEP-06
0.64
0.02
O═C(N1C[C@@N(C[C@]12[H])C)[C@@H](NC([C@@H]





(N(C([CC@]3(N(CCC3)C([C@H](COC2═O)NC([CC@@H]





(NC(/C═C/CCCC)═O)CC4═CC(F)═CC(F)═C4)═O)═O)





[H])═O)C)C)═O)C


ADEP-25
0.61
0.02
O═C(N1C[C@@N(C[C@]12[H])C)[C@@H](NC([C@]





3(N(C([C@]4(N(CCC4)C([C@H](COC2═O)NC([C@@H]





(NC(/C═C/CCCC)═O)CC5═CC(F)═CC(F)═C5)═O)═O)





[H])═O)CCCC3)[H])═O)CC


ADEP-13
0.59
0.02
O═C(N1C[C@@N(C[C@]12[H])C)[C@@H](NC([C@]





3(N(C([C@]4(N(CCC4)C([C@H]([C@@H](OC2═O)C)





NC([C@@H](NC(/C═C/CC5CC5)═O)





CC6═CC(F)═CC(F)═C6)═O)═O)[H])═O)CCCC3)[H])═O)C


ADEP-15
0.58
0.02
O═C(N1C[C@@H](C[C@]12[H])C)[C@@H](NC([C@]





3([H])CCCN3C([C@]4(N(CCC4)C([C@H](COC2═O)





NC([C@H])NC(/C═C/CCCC)═O)CC5═CC





(F)═CC(F)═C5)═O)═O)[H])═O)═O)C


ADEP-20
0.53
0.02
O═C(N1C[C@@H](C[C@]12[H])CAC@@H](NC([C@]





3(N(C([C@]4(N(CCC4)C([C@H](COC2═O)NC([C@H]





(NC(/C═C/CCC)═O)CC5═CC(F)═CC═C5)═O)═O)[H])═O)





CCCC3)[HO═O)C


ADEP-02
0.52
0.02
O═C(N1CCC[C@]12[H])[C@@H](NC([C@]3(N(C([C@]





4(N(CCC4)C([C@H](COC2═O)NC([C@H]





(NC(/C═C/CCCC)═O)CC5═CC═CC═C5)═O)═O)





[H])═O)CCCC3)[H])═O)C


ADEP-10
0.44
0.02
O═C(N1C[C@@H](C[C@]12[H])CAC@@H](NC([C@]





3(N(C([C@]4(N(CCC4)C([C@H](COC2═O)NC([C@H]





(NC(/C═C/CCCC)═O)CC5═CC═CC═C5)═O)═O)[H])═O)





CCCC3)[H])═O)C


ADEP-O4
0.32
0.01
O═C(N1C[C@@H](C[C@]12[H])CAC@@H](NC([C@@H]





(N(C([C@]3(N(CCC3)C([C@H](COC2═O)NC([C@H]





(NC(/C═C/C═C/CCC)═O)CC4═CC═CC═C4)═O)═O)





[H])═O)C)C)═O)C


ADEP-01
0.22
0.01
O═C(N1CCC[C@]12[H])[C@@H](NC([C@@H]





(N(C([C@]3(N(CCC3)C([C@H](COC2═O)





NC([C@H](NC(/C═C/CCCC)═O)CC4═CC═CC═C4)═O)═O)





[H])═O)C)C)═O)C





RD25 refers to the RD value of the ADEP analog at 25 μM as a measure of its ability to activate HsCIpP.


RDS refers to the RD value of the ADEP analog at 5 μM as a measure of its ability to activate HsCIpP.


RD1 refers to the RD value of the ADEP analog at 1 μM as a measure of its ability to activate HsCIpP.


SD refers to the standard deviation of the RD value.


SMILES refers to the structural formulas of the ADEP analogs in SMILES format. Configuration at tetrahedral carbon is specified by @ or @@. Consider the four bonds in the order in which they appear, left to right, in the SMILES form. Looking toward the central carbon from the perspective of the first bond, the other three are either clockwise or counter-clockwise. These cases are indicated with @@ and@, respectively. The structures described by the SMILES format can be drawn using a software like ChemDraw.






ADEPs Enhance the Peptidase and Protease Activities of HsCIpP

The effects of the ADEPs on the peptidase and protease activities of HsCIpP were investigated using N-acetyl-Trp-Leu-Ala-AMC (Ac-WLA-AMC) and casein-FITC, respectively. Ac-WLA-AMC was the best peptide substrate for HsCIpP among those tested (FIG. 2A). The degradation of Ac-WLA-AMC by 6 μM of HsCIpP (monomeric concentration) has an apparent kcat, KM and kcat/KM lower than that obtained for the degradation of the peptide with 1.5 μM of HsCIpP in the presence of 4.5 μM HsCIpX (compare FIG. 2B with 3A, top panel). Similar values were obtained for the degradation of Ac-WLA-AMC using 2 μM of EcCIpP (FIG. 3A, lower panel). In contrast, while there was no detectable degradation of Suc-LY-AMC by HsCIpP (FIG. 2A), the reported apparent kcat of Suc-LY-AMC degradation by 10 μM EcCIpP (protomer concentration) is 2.4 s−1 with KM of 4.4×103 μM, yielding a kcat/KM of 5.5×10−4 s−1.μM−1 (Bewley et al., 2006). Hence, while HsCIpP and EcCIpP have similar general peptidase activities against Ac-WLA-AMC, EcCIpP seems to have broader substrate preferences compared to HsCIpP.


Initial degradation rates data were analyzed using the Hill model. The results for ADEP-28 and ADEP-41 are shown in FIGS. 2C and D, and those for ADEP-06, ADEP-02 and ADEP-04 in FIGS. 3B and C. The kinetic parameters derived are listed in FIG. 2E. With all five ADEP analogs, the peptidase activity of HsCIpP was similarly enhanced, with Vmax increased by about 1.8-fold. The Hill coefficients (h), ranging from 0.8 for ADEP-04 to 1.5 for ADEP-41 (FIG. 2E), seem to suggest that the binding of ADEP and the subsequent stimulation of HsCIpP's peptidase activity exhibits only marginal positive cooperativity. The apparent dissociation constants (K0.5) of the ADEP analogs are closely correlated to their respective RD1 scores. Specifically, the higher the RD1 score of a given analog, the lower is its K0.5 (FIG. 2E).


Similar to their effects on HsCIpP's peptidase activity, all five ADEP analogs activate and enhance the protease activity of HsCIpP independently of HsCIpX. Notably, the Vmax measured in the presence of the five ADEP analogs shows little variation (between 10.9 and 14.0 RFU.s−1; FIG. 2E). Their K0.5, determined by measuring the protease activity of HsCIpP, correlates with their respective RD1 scores. Both ADEP-28 and ADEP-41 exhibit the lowest K0.5 (0.12 μM), followed by ADEP-06, ADEP-02, and ADEP-04 (FIG. 2E). In this assay, unlike in the peptidase activity assay, the binding of ADEP and the subsequent activation and enhancement of HsCIpP's protease activity exhibits strong, positive cooperativity. Specifically, enhancement by ADEP-28 has the highest Hill coefficient (FIG. 2E).


By comparison, titration of HsCIpX and fitting the data to the Hill model yields a Vmax of 6.88 RFU.s−1, a K0.5 of 5.7 μM, and mild positive cooperativity (h=1.25) in HsCIpP-binding (FIG. 3D). These results indicate that the ADEPsexhibit stronger binding to HsCIpP than does HsCIpX under the conditions of these experiments.


Low Concentration of ADEPs Displace HsCIpX From HsCIpXP Complexes

To assess the effects of ADEPs on the interaction between HsCIpX and HsCIpP, the degradation of casein-FITC by HsCIpXP was monitored in the presence of increasing concentrations of ADEP-28, ADEP-06 or ADEP-02. At low ADEP concentrations, the rate of casein-FITC degradation was decreased, down to an observed minimum (Vmin) that ranges from 0.9 to 1.3 RFU.s−1 (FIGS. 2F and G). Notably, the ADEP concentration needed to minimize the protease activity is dependent on the apparent binding affinity of each analog. As ADEP concentration was further increased, the protease activity increased likely as the population of ADEP-activated HsCIpP increased. Accordingly, the Vmax achieved with all three ADEP analogs at saturation is identical to the Vmax previously observed in the absence of HsCIpX (FIGS. 2E and F). The initial decrease of protease activity at low ADEP concentrations was not caused by the degradation of HsCIpX, HsCIpP or creatine phosphokinase as shown by SDS-PAGE gels (FIGS. 3E-G).


Taken together, the results clearly illustrate that ADEP can induce the dissociation of the HsCIpXP complex at low concentrations, resulting in a net loss of protease activity. At higher ADEP concentrations, the population of ADEP-activated HsCIpP increases, resulting in the restoration and subsequent increase of protease activity.


ADEPs Induce HsCIpP-Dependent Cytotoxicity in HEK293 T-REx Cells

To assess the biological impact of ADEPs in targeting HsCIpP in vivo, HEK293 T-REx WT and CLPP−/− cells were treated with serially diluted ADEP-28, ADEP-41, ADEP-06, ADEP-02 and ADEP-04, followed by an assessment of their survival 72 hours post treatment. For HEK293 T-REx WT cells, all five ADEP analogs were found to be cytotoxic with 1050 values between 0.36 μM and 8.20 μM (FIGS. 4A, B and 5A). In comparison, IC50 of cisplatin on HEK293 cells has been reported at 0.43 μM and that of the dihydrofolate reductase-targeting Metoprine at 0.80 μM (Bram et al., 2006).


Importantly, the cytotoxicity of ADEPs showed a strong, linear correlation with their respective apparent binding affinity for HsCIpP (FIG. 4C), revealing a direct link between the potency of ADEPs in activating HsCIpP and their cytotoxicity. Furthermore, ADEP-induced cytotoxicity is HsCIpP-dependent, as CLPP−/− cells, which lack endogenous HsCIpP, are highly resistant to all five ADEP analogs (FIGS. 4A, B and 5A). Hence, without wishing to be bound by theory, these ADEPs are likely acting on-target in cells, although interactions with other proteins that do not cause cytotoxicity cannot yet be ruled out.


It should be noted that, using deep proteomics profiling of mitochondrial extracts isolated from WT and CLPP HEK293 T-REx cells, 281 distinct mitochondrial proteins were identified covering 24% of the estimated human mitochondrial proteome (281 of 1158 human mitochondrial proteins from MitoCarta2.0 inventory (Calvo et al., 2016)). Of these, only 19 mitochondrial proteins in CLPP−/− were significantly (p≤0.05) altered with more than 1.5-fold change compared to WT cells (Table 4). This indicates that there are no drastic changes in the mitochondrial proteome upon CLPP deletion under the conditions tested.









TABLE 4







Relative fold change of mitochondrial proteins identified in the wild type versus CLPP knockouts















Total peptide ion intensity detected by
Avg.






MaxQuant in wild type (WT) and CLPP
fold






knockout (KO)
change

















UniProt
Protein
Protein


CLPP
CLPP
CLPP
(CLPP
P-Value


IDs
names
description
WT-1
WT-2
KO-1
KO-2
KO-3
KO/WT)
significance



















Q9UDR5
AASS
Alpha-aminoadipic
4.45E+06
1.27E+06
0.00E+00
0.00E+00
6.37E+05
−5.95
0.023




semialdehyde











synthase,











mitochondrial;











Lysine











ketoglutarate











reductase;











Saccharopine











dehydrogenase









Q9NRK6
ABCB10
ATP-binding
6.39E+06
5.31E+05
0.00E+00
9.28E+05
0.00E+00
−6.65
0.040




cassette sub-family











B member 10,











mitochondrial









P09110
ACAA1
3-ketoacyl-CoA
1.18E+06
4.49E+05
0.00E+00
0.00E+00
3.83E+04
−16.43
0.018




thiolase,











peroxisomal









P33121
ACSL1
Long-chain-fatty-
7.15E+05
3.90E+05
0.00E+00
8.11E+03
0.00E+00
−18.71
0.042




acid--CoA ligase 1









O14874
BCKDK
[3-methyl-2-
8.36E+05
0.00E+00
5.32E+06
4.94E+06
9.96E+05
20.55
0.045




oxobutanoate











dehydrogenase











[lipoamide]] kinase,











mitochondrial









Q9HA77
CARS2
Probable cysteine--
4.64E+06
5.63E+05
3.55E+06
2.73E+06
1.92E+06
1.97
0.045




tRNA ligase,











mitochondrial









Q16740
CLPP
ATP-dependent
4.80E+06
1.52E+06
0.00E+00
0.00E+00
0.00E+00
−86.23
0.007




Clp protease











proteolytic











subunit,











mitochondrial









P23786
CPT2
Carnitine O-
7.36E+05
2.36E+05
0.00E+00
0.00E+00
0.00E+00
−13.35
0.011




palmitoyltransferase











2, mitochondrial









Q9BX68
HINT2
Histidine triad
5.14E+06
7.84E+05
0.00E+00
1.29E+06
6.01E+05
−2.66
0.038




nucleotide-binding











protein 2,











mitochondrial









Q16891
IMMT
MICOS
4.74E+07
9.75E+06
3.98E+06
3.62E+06
6.76E+06
−3.26
0.014




complex











subunit MIC60









Q9GZY8
MFF
Mitochondrial
2.32E+06
6.78E+05
0.00E+00
0.00E+00
3.53E+05
−5.44
0.026




fission











factor









Q9BPX6
MICU1
Calcium uptake
3.14E+06
1.22E+06
0.00E+00
3.18E+05
1.35E+05
−10.74
0.022




protein 1,











mitochondrial









Q8N5N7
MRPL50
39S ribosomal
7.05E+06
2.07E+06
0.00E+00
1.55E+06
6.12E+0
−4.67
0.017




protein L50,











mitochondrial









Q9BQC6
MRPL57
Ribosomal
2.16E+06
7.32E+05
0.00E+00
0.00E+00
1.22E+05
−13.50
0.014




protein 63,











mitochondrial









Q9Y676
MRPS18B
28S ribosomal
6.24E+06
1.56E+06
2.00E+05
0.00E+00
1.03E+06
−4.54
0.030




protein S18b,











mitochondrial









Q96TC7
RMDN3
Regulator of
6.57E+06
3.30E+05
4.96E+06
3.75E+06
2.62E+06
2.51
0.049




microtubule











dynamics











protein 3









Q99595
TIMM17A
Mitochondrial
1.18E+06
5.56E+05
0.00E+00
0.00E+00
0.00E+00
−27.78
0.030




import











inner membrane











translocase











subunit











Tim17-A









Q9Y5J7
TIMM9
Mitochondrial
3.79E+06
4.82E+05
0.00E+00
0.00E+00
6.70E+04
−21.32
0.003




import











inner membrane











translocase subunit











Tim9









P49411
TUFM
Elongation
1.19E+08
1.65E+07
1.77E+07
2.86E+07
1.45E+07
−1.90
0.015




factor Tu,











mitochondrial









Q5JTZ9
AARS2
Alanine--tRNA
1.36E+07
7.20E+05
8.12E+05
2.44E+06
5.95E+05
−2.75
0.119




ligase,











mitochondrial









P11310
ACADM
Medium-chain
1.46E+07
1.26E+06
2.59E+07
1.15E+07
8.28E+06
3.88
0.083




specific acyl-CoA











dehydrogenase,











mitochondrial









P53396
ACLY
ATP-citrate
1.82E+06
6.51E+05
0.00E+00
5.69E+05
6.62E+05
−1.96
0.173




synthase









Q4G176
ACSF3
Acyl-CoA
1.20E+06
6.33E+05
0.00E+00
0.00E+00
5.83E+05
−2.65
0.177




synthetase











family member 3,











mitochondrial









O95573;
ACSL3;
Long-chain-fatty-
6.15E+06
0.00E+00
3.15E+06
0.00E+00
1.55E+06
1.57
0.341


O60488
ACSL4
acid--CoA ligase 3;











Long-chain-fatty-











acid--CoA ligase 4









Q9Y4W6
AFG3L2
AFG3-like
7.33E+06
1.34E+06
1.07E+05
2.91E+06
1.55E+06
−1.72
0.161




protein 2









P27144
AK4
Adenylate kinase 4,
7.31E+06
5.38E+06
4.51E+06
4.08E+06
1.75E+06
−2.01
0.175




mitochondrial









P30837
ALDH1B1
Aldehyde
3.29E+07
1.18E+07
5.95E+05
1.87E+07
5.88E+06
−2.26
0.114




dehydrogenase X,











mitochondrial









P05091
ALDH2
Aldehyde
2.77E+07
8.69E+06
0.00E+00
4.29E+06
8.41E+06
−2.53
0.120




dehydrogenase,











mitochondrial









P51648
ALDH3A2
Fatty aldehyde
2.87E+06
1.52E+06
0.00E+00
2.01E+06
1.46E+06
−1.61
0.269




dehydrogenase









Q02252
ALDH6Al
Methylmalonate-
1.06E+07
2.54E+06
0.00E+00
1.92E+06
2.46E+06
−2.55
0.081




semialdehyde











dehydrogenase











[acylating],











mitochondrial









P78540
ARG2
Arginase-2,
2.30E+06
4.54E+05
0.00E+00
0.00E+00
3.33E+06
1.99
0.361




mitochondrial









Q5T9A4
ATAD3B
ATPase family AAA
3.85E+07
1.12E+07
0.00E+00
5.14E+06
1.35E+07
−2.21
0.170




domain-containing











protein 3B









P25705
ATP5A1
ATP synthase
3.29E+08
2.62E+07
2.45E+08
9.28E+07
6.85E+07
1.57
0.263




subunit alpha,











mitochondrial









P36542
ATP5C1
ATP synthase
2.14E+07
1.09E+07
8.61E+06
1.97E+07
2.17E+06
−1.70
0.215




subunit gamma,











mitochondrial









P30049
ATP5D
ATP synthase
2.20E+06
8.62E+05
1.09E+06
5.80E+06
8.91E+05
1.78
0.265




subunit delta,











mitochondrial









P56134
ATP5J2
ATP synthase
1.57E+06
7.44E+05
2.94E+05
6.54E+05
8.33E+05
−1.50
0.277




subunit f,











mitochondrial









O75964
ATP5L
ATP synthase
3.65E+06
8.98E+05
0.00E+00
0.00E+00
8.39E+05
−3.45
0.073




subunit g,











mitochondrial









Q9Y276
BCS1L
Mitochondrial
1.51E+06
5.30E+04
0.00E+00
0.00E+00
4.62E+04
−7.41
0.095




chaperone











BCS1









Q86WA6
BPHL
Valacyclovir
1.61E+06
1.22E+05
4.32E+06
3.16E+06
6.60E+04
5.23
0.162




hydrolase









Q99832
CCT7
T-complex protein 1
2.85E+07
0.00E+00
5.91E+04
0.00E+00
1.38E+04
−189.33
0.136




subunit eta









Q9NYJ1
COA4
Cytochrome c
3.80E+06
1.80E+06
1.94E+06
0.00E+00
1.03E+06
−2.17
0.154




oxidase assembly











factor 4 homolog,











mitochondrial









P10606
COX5B
Cytochrome c
1.64E+07
3.20E+06
0.00E+00
0.00E+00
4.73E+06
−2.52
0.160




oxidase subunit 5B,











mitochondrial









P12074
COX6A1
Cytochrome c
2.54E+07
8.04E+06
8.52E+05
1.02E+08
1.25E+07
2.54
0.278




oxidase subunit 6A1,











mitochondrial









P14854
COX6B1
Cytochrome c
7.85E+06
5.56E+06
6.04E+06
5.29E+06
1.11E+06
−1.84
0.208




oxidase subunit 6B1









P09669
COX6C
Cytochrome c
4.46E+06
1.99E+05
0.00E+00
0.00E+00
0.00E+00
−36.69
0.061




oxidase subunit 6C









O75390
CS
Citrate synthase,
1.15E+08
7.45E+06
1.01E+08
3.56E+07
1.93E+07
1.78
0.262




mitochondrial









P51398
DAP3
28S ribosomal
7.97E+06
6.12E+06
0.00E+00
1.81E+06
5.85E+06
−2.17
0.238




protein S29,











mitochondrial









P11182
DBT
Lipoamide
0.00E+00
1.80E+05
1.27E+06
0.00E+00
3.91E+04
1.96
0.357




acyltransferase











component of











branched-chain











alpha-keto acid











dehydrogenase











complex,











mitochondrial









P09622
DLD
Dihydrolipoyl
1.19E+07
6.40E+05
6.51E+05
1.27E+06
0.00E+00
−5.34
0.077




dehydrogenase,











mitochondrial









Q9NVH1
DNAJC11
DnaJ homolog
9.16E+05
6.32E+05
0.00E+00
4.66E+06
7.46E+05
1.96
0.305




subfamily C











member 11









O00429
DNM1L
Dynamin-1-like
1.09E+06
5.14E+05
0.00E+00
0.00E+00
3.31E+05
−3.72
0.097




protein









P30084
ECHS1
Enoyl-CoA
1.38E+08
6.41E+06
1.14E+07
2.39E+07
7.50E+06
−2.37
0.147




hydratase,











mitochondrial









O75521
ECI2
Enoyl-CoA delta
2.71E+06
1.45E+06
0.00E+00
0.00E+00
7.36E+05
−4.83
0.084




isomerase 2,











mitochondrial









Q96GK7;
FAHD2A;
Fumarylacetoacetate
1.32E+06
4.59E+05
0.00E+00
0.00E+00
2.77E+05
−4.28
0.053


Q6P2I3
FAHD2B
hydrolase domain-











containing protein 2A;











Fumarylacetoacetate











hydrolase











domain-containing











protein 2B









P49327
FASN
Fatty acid
1.91E+07
2.54E+06
3.49E+06
2.64E+07
4.88E+06
1.68
0.282




synthase;











[Acyl-carrier-protein]











S-acetyltransferase;











[Acyl-carrier-protein]











S-malonyltransferase;











3-oxoacyl-[acyl-











carrier-protein]











synthase;











3-oxoacyl-











[acyl-carrier-protein]











reductase;











3-hydroxyacyl-[acyl-











carrier-protein]











dehydratase;











Enoyl-











[acyl-carrier-protein]











reductase;











Oleoyl-











[acyl-carrier-protein]











hydrolase









P10109
FDX1
Adrenodoxin,
6.94E+06
3.01E+06
0.00E+00
0.00E+00
4.54E+06
−1.81
0.301




mitochondrial









P22830
FECH
Ferrochelatase,
2.73E+06
1.80E+06
1.48E+06
1.15E+07
1.37E+06
1.75
0.308




mitochondrial









Q14318
FKBP8
Peptidyl-prolyl
1.74E+07
2.54E+06
1.46E+07
1.06E+07
6.63E+06
1.86
0.058




cis-trans











isomerase











FKBP8









Q8TAE8
GADD4
Growth arrest and
1.16E+06
2.23E+06
0.00E+00
9.57E+05
1.57E+06
−2.29
0.246



5G1P1
DNA damage-











inducible proteins-











interacting protein 1









P04406
GAPDH
Glyceraldehyde-3-
1.45E+07
8.03E+05
7.34E+06
3.20E+07
5.87E+06
3.84
0.127




phosphate











dehydrogenase









O43716
GATC
Glutamyl-tRNA(Gln)
1.01E+06
4.77E+05
0.00E+00
4.62E+06
1.26E+06
2.75
0.217




amidotransferase











subunit C,











mitochondrial









O75323
GBAS
Protein NipSnap
1.20E+07
2.38E+06
0.00E+00
9.87E+05
3.37E+06
−2.28
0.154




homolog 2









Q96RP9
GFM1
Elongation factor G,
1.67E+06
2.76E+05
0.00E+00
4.10E+06
3.82E+05
2.11
0.312




mitochondrial









P32189;
GK; GK3P
Glycerol kinase;
1.00E+06
7.78E+05
0.00E+00
2.46E+06
0.00E+00
−1.51
0.370


Q14409

Putative glycerol











kinase 3









P00367;
GLUD1;
Glutamate
6.67E+07
1.95E+07
6.48E+06
2.39E+07
2.29E+07
−1.60
0.202


P49448
GLUD2
dehydrogenase 1,











mitochondrial;











Glutamate











dehydrogenase 2,











mitochondrial









P00505
GOT2
Aspartate
2.79E+06
1.29E+06
0.00E+00
1.72E+06
9.15E+05
−1.96
0.176




aminotransferase,











mitochondrial









Q12849
GRSF1
G-rich
7.52E+05
5.11E+05
3.61E+06
2.31E+06
0.00E+00
2.65
0.234




sequence











factor 1









Q16836
HADH
Hydroxyacyl-
7.75E+06
3.92E+06
2.87E+06
7.33E+06
1.48E+06
−1.53
0.236




coenzyme A











dehydrogenase,











mitochondrial









P55084
HADHB
Trifunctional enzyme
1.22E+07
2.44E+06
3.60E+06
2.38E+07
3.51E+06
1.89
0.261




subunit beta,











mitochondrial;











3-ketoacyl-CoA











thiolase









Q9Y241
HIGD1A
HIG1 domain
2.35E+07
4.99E+06
1.17E+06
2.87E+06
5.27E+06
−2.44
0.077




family member 1A,











mitochondrial









P19367
HK1
Hexokinase-1
6.33E+06
1.00E+06
0.00E+00
3.52E+06
5.19E+05
−1.86
0.193


Q99714
HSD17B10
3-hydroxyacyl-CoA
6.22E+07
1.44E+07
3.69E+07
1.10E+08
1.95E+07
1.90
0.179




dehydrogenase











type-2









P51659
HSD17B4
Peroxisomal
1.13E+07
4.14E+06
8.99E+05
4.36E+06
3.55E+06
−1.94
0.141




multifunctional











enzyme type 2;











(3R)-hydroxyacyl-











CoA dehydrogenase;











Enoyl-CoA











hydratase 2









Q9NSE4
IARS2
Isoleucine--tRNA
9.19E+06
2.71E+05
0.00E+00
2.31E+06
8.82E+05
−1.87
0.258




ligase, mitochondrial









P50213
IDH3A
Isocitrate
8.80E+06
4.38E+05
0.00E+00
4.07E+06
6.56E+05
−1.50
0.332




dehydrogenase











[NAD] subunit alpha,











mitochondrial









Q96AB3
ISOC2
Isochorismatase
1.36E+06
7.22E+05
0.00E+00
1.78E+06
1.51E+05
−1.80
0.268




domain-containing











protein 2,











mitochondrial









Q96GW9
MARS2
Methionine--tRNA
2.25E+05
1.16E+05
0.00E+00
2.94E+05
0.00E+00
−1.59
0.294




ligase, mitochondrial









Q8IVS2
MCAT
Malonyl-CoA-acyl
1.10E+06
1.06E+06
0.00E+00
2.30E+06
4.52E+05
−1.52
0.330




carrier protein











transacylase,











mitochondrial









Q96RQ3
MCCC1
Methylcrotonoyl-
2.45E+06
1.13E+06
0.00E+00
1.92E+06
4.37E+05
−2.19
0.159




CoA











carboxylase subunit











alpha, mitochondrial









P23368
ME2
NAD-dependent
4.23E+07
3.64E+06
1.20E+07
8.22E+06
2.31E+06
−1.63
0.191




malic enzyme,











mitochondrial









Q8IYU8
MICU2
Calcium uptake
3.15E+06
2.92E+05
0.00E+00
4.08E+05
7.57E+05
−1.70
0.258




protein 2,











mitochondrial









Q9Y3B7
MRPL11
39S ribosomal
2.41E+07
7.95E+06
2.80E+06
3.14E+06
9.26E+06
−1.91
0.189




protein L11,











mitochondrial









P52815
MRPL12
39S ribosomal
4.41E+07
2.75E+06
4.46E+07
3.30E+07
7.43E+06
2.41
0.149




protein L12,











mitochondrial









Q9BYD1
MRPL13
39S ribosomal
5.94E+06
9.42E+05
1.24E+07
4.94E+06
1.89E+06
3.01
0.172




protein L13,











mitochondrial









Q9P015
MRPL15
39S ribosomal
5.88E+06
1.16E+06
2.49E+06
6.85E+06
1.81E+06
1.53
0.181




protein L15,











mitochondrial









Q9NRX2
MRPL17
39S ribosomal
1.53E+06
3.70E+05
0.00E+00
2.25E+06
2.15E+06
2.59
0.230




protein L17,











mitochondrial









Q9H0U6
MRPL18
39S ribosomal
2.98E+06
3.05E+05
0.00E+00
0.00E+00
7.78E+05
−2.02
0.241




protein L18,











mitochondrial









P49406
MRPL19
39S ribosomal
3.64E+06
9.49E+05
6.42E+05
7.74E+06
1.00E+06
1.64
0.326




protein L19,











mitochondrial









Q16540
MRPL23
39S ribosomal
2.00E+06
2.71E+06
9.38E+05
1.00E+06
1.77E+06
−2.10
0.233




protein L23,











mitochondrial









Q96A35
MRPL24
39S ribosomal
2.04E+06
1.63E+06
0.00E+00
0.00E+00
1.62E+06
−2.32
0.240




protein L24,











mitochondrial









Q9P0M9
MRPL27
39S ribosomal
1.13E+06
2.78E+06
0.00E+00
0.00E+00
1.68E+06
−3.36
0.207




protein L27,











mitochondrial









Q96DV4
MRPL38
39S ribosomal
1.53E+06
8.48E+05
0.00E+00
1.23E+06
2.98E+05
−2.35
0.155




protein L38,











mitochondrial









Q9NYK5
MRPL39
39S ribosomal
7.18E+06
2.35E+06
1.34E+07
7.98E+06
5.73E+06
2.51
0.064




protein L39,











mitochondrial









Q9BYD3
MRPL4
39S ribosomal
6.93E+06
1.70E+06
9.44E+06
8.48E+06
1.80E+06
2.03
0.140




protein L4,











mitochondrial









Q8IXM3
MRPL41
39S ribosomal
3.73E+06
3.92E+05
7.86E+06
1.28E+07
5.69E+05
5.52
0.122




protein L41,











mitochondrial









Q8N983
MRPL43
39S ribosomal
4.40E+06
9.42E+04
7.32E+06
1.57E+06
1.12E+06
3.62
0.200




protein L43,











mitochondrial









Q9H2W6
MRPL46
39S ribosomal
5.90E+06
3.16E+06
0.00E+00
0.00E+00
1.08E+06
−7.30
0.060




protein L46,











mitochondrial









Q96EL3
MRPL53
39S ribosomal
2.21E+06
5.90E+05
0.00E+00
1.33E+06
4.44E+05
−1.72
0.177




protein L53,











mitochondrial









O60783
MRPS14
28S ribosomal
3.75E+06
2.49E+06
2.60E+06
1.41E+06
1.84E+06
−1.49
0.268




protein S14,











mitochondrial









Q9Y3D93
MRPS2
28S ribosomal
8.27E+06
2.42E+06
2.33E+06
1.30E+07
4.17E+06
1.56
0.251




protein S23,











mitochondrial









Q9BYN86
MRPS2
28S ribosomal
1.97E+06
5.25E+05
8.97E+06
2.58E+06
1.07E+06
4.54
0.178




protein S26,











mitochondrial









Q9Y2Q98
MRPS2
28S ribosomal
3.25E+06
1.88E+06
6.17E+05
0.00E+00
1.60E+06
−2.41
0.182




protein S28,











mitochondrial









Q926651
MRPS3
28S ribosomal
4.51E+06
9.43E+05
6.41E+05
5.99E+05
1.13E+06
−1.90
0.108




protein S31,











mitochondrial









Q9Y2913
MRPS3
28S ribosomal
1.27E+07
5.08E+05
0.00E+00
0.00E+00
4.99E+05
−10.23
0.082




protein S33,











mitochondrial









P829304
MRPS3
28S ribosomal
1.20E+07
3.59E+06
1.82E+07
1.13E+07
3.93E+06
1.83
0.182




protein S34,











mitochondrial









P82932
MRPS6
28S ribosomal
5.47E+06
9.09E+05
1.24E+06
0.00E+00
1.25E+06
−1.84
0.159




protein S6,











mitochondrial









Q9Y2R9
MRPS7
28S ribosomal
6.44E+06
2.04E+05
0.00E+00
6.43E+05
1.73E+05
−5.18
0.116




protein S7,











mitochondrial









P00403
MT-CO2
Cytochrome c
7.20E+06
2.34E+06
0.00E+00
7.56E+05
2.80E+06
−2.22
0.184




oxidase subunit 2









P11586
MTHFD1
C-1-tetrahydrofolate
2.18E+06
4.41E+05
0.00E+00
1.97E+06
0.00E+00
−1.64
0.316




synthase,











cytoplasmic;











Methyl-











enetetrahydrofolate











dehydrogenase;











Methenyl-











tetrahydrofolate











cyclohydrolase;











Formyl-











tetrahydrofolate











synthetase;











C-1-tetrahydrofolate











synthase,











cytoplasmic,











N-terminally











processed









P13995
MTHFD2
Bifunctional
2.60E+06
9.36E+05
0.00E+00
3.52E+06
7.12E+06
3.24
0.257




methylenetetra-











hydrofolate











dehydrogenase/











cyclohydrolase,











mitochondrial;











NAD-dependent











methylenetetra-











hydrofolate











dehydrogenase;











Methenyl-











tetrahydrofolate











cyclohydrolase









Q13505
MTX1
Metaxin-1
1.70E+06
1.00E+06
0.00E+00
0.00E+00
6.15E+05
−3.88
0.115


O75431
MTX2
Metaxin-2
2.47E+06
0.00E+00
1.37E+06
4.21E+06
1.14E+05
3.29
0.212


O95299
NDUFA10
NADH
3.03E+06
1.47E+05
9.30E+06
3.12E+06
1.17E+06
6.13
0.152




dehydrogenase











[ubiquinone] 1 alpha











subcomplex











subunit 10,











mitochondrial









Q9P0J0
NDUFA13
NADH
3.42E+06
1.08E+06
2.03E+06
1.19E+07
1.82E+06
2.65
0.196




dehydrogenase











[ubiquinone] 1 alpha











subcomplex











subunit 13









P51970
NDUFA8
NADH
3.69E+06
8.74E+05
0.00E+00
1.98E+06
3.89E+05
−2.18
0.113




dehydrogenase











[ubiquinone] 1 alpha











subcomplex











subunit 8









Q16795
NDUFA9
NADH
5.96E+06
1.74E+06
0.00E+00
3.65E+06
1.18E+06
−1.84
0.165




dehydrogenase











[ubiquinone] 1 alpha











subcomplex











subunit 9,











mitochondrial









Q8N183
NDUFAF2
Mimitin,
1.03E+07
1.25E+06
0.00E+00
1.43E+06
1.62E+06
−2.59
0.080




mitochondrial









P28331
NDUFS1
NADH-ubiquinone
2.19E+07
4.62E+06
6.80E+05
8.69E+06
2.35E+06
−2.41
0.053




oxidoreductase











75 kDa











subunit,











mitochondrial









O75306
NDUFS2
NADH
2.54E+06
8.48E+05
8.05E+06
2.24E+06
9.31E+05
2.73
0.226




dehydrogenase











[ubiquinone] iron-











sulfur protein 2,











mitochondrial









O00217
NDUFS8
NADH
2.95E+06
8.49E+05
0.00E+00
1.51E+06
4.39E+04
−3.34
0.076




dehydrogenase











[ubiquinone] iron-











sulfur protein 8,











mitochondrial









Q9BPW8
NIPSNAP1
Protein NipSnap
9.70E+06
5.63E+06
1.08E+06
9.00E+05
3.40E+06
−3.19
0.118




homolog 1









Q9H857
NT5DC2
5-nucleotidase
2.48E+05
2.09E+05
0.00E+00
7.48E+05
8.23E+05
2.48
0.242




domain-containing











protein 2









Q02218
OGDH
2-oxoglutarate
7.59E+06
7.92E+05
1.74E+06
3.27E+05
1.07E+06
−1.85
0.138




dehydrogenase,











mitochondrial









Q9Y3D7
PAM16
Mitochondrial
1.91E+06
1.15E+06
7.76E+05
0.00E+00
1.37E+06
−1.57
0.312




import inner











membrane











translocase subunit











TIM16









P05166
PCCB
Propionyl-CoA
8.76E+05
5.67E+05
0.00E+00
0.00E+00
0.00E+00
−26.16
0.052




carboxylase











beta chain,











mitochondrial









Q16822
PCK2
Phospho-
3.33E+06
1.48E+06
0.00E+00
0.00E+00
7.47E+05
−5.13
0.064




enolpyruvate











carboxykinase











[GTP],











mitochondrial









Q6L8Q7
PDE12
2,5-
2.06E+06
6.39E+05
0.00E+00
1.01E+06
0.00E+00
−3.68
0.067




phosphodiesterase











12









O00330
PDHX
Pyruvate
1.76E+06
1.18E+06
0.00E+00
1.80E+06
3.32E+05
−2.36
0.179




dehydrogenase











protein X











component,











mitochondrial









Q96HS1
PGAM5
Serine/threonine-
1.36E+07
1.88E+05
0.00E+00
0.00E+00
2.59E+06
−1.97
0.312




protein phosphatase











PGAM5,











mitochondrial









P35232
PHB
Prohibitin
2.35E+08
8.14E+07
3.00E+08
2.80E+08
5.31E+07
1.54
0.234


Q5JRX3
PITRM1
Presequence
9.57E+06
2.02E+06
0.00E+00
1.62E+07
3.49E+06
1.55
0.338




protease,











mitochondrial









Q10713
PMPCA
Mitochondrial-
6.70E+06
1.21E+06
0.00E+00
5.46E+05
1.45E+06
−2.71
0.088




processing











peptidase subunit











alpha









O75439
PMPCB
Mitochondrial-
2.26E+07
8.76E+05
2.89E+06
4.00E+07
3.06E+06
2.47
0.283




processing











peptidase subunit











beta









Q8TCS8
PNPT1
Polyribonucleotide
1.23E+07
4.46E+05
3.78E+05
2.75E+05
2.99E+06
−1.56
0.351




nucleotidyl-











transferase 1,











mitochondrial









Q9Y2S7
POLDIP2
Polymerase delta-
8.87E+06
4.93E+05
1.41E+07
1.97E+07
2.00E+06
4.89
0.092




interacting protein 2









O00411
POLRMT
DNA-directed
2.60E+06
0.00E+00
1.47E+06
4.81E+05
5.76E+05
1.79
0.253




RNA polymerase,











mitochondrial









P32119
PRDX2
Peroxiredoxin-2
1.64E+06
9.81E+05
0.00E+00
0.00E+00
2.30E+05
−9.17
0.059


Q13162
PRDX4
Peroxiredoxin-4
2.34E+06
9.28E+05
0.00E+00
0.00E+00
1.02E+06
−2.49
0.182


P30044
PRDX5
Peroxiredoxin-5,
1.03E+07
3.34E+05
1.24E+07
3.31E+06
1.09E+06
2.38
0.264




mitochondrial









Q9H7Z7
PTGES2
Prostaglandin E
3.90E+05
4.93E+05
0.00E+00
8.96E+05
0.00E+00
−2.27
0.251




synthase 2;











Prostaglandin E











synthase 2











truncated form









Q96C36
PYCR2
Pyrroline-5-
1.55E+07
7.07E+05
0.00E+00
0.00E+00
2.46E+06
−2.75
0.197




carboxylate











reductase 2









Q9HOR6
QRSL1
Glutamyl-tRNA(Gln)
1.84E+06
7.62E+05
0.00E+00
0.00E+00
8.28E+05
−2.45
0.187




amidotransferase











subunit A,











mitochondrial









Q8IX12
RHOT1
Mitochondrial Rho
1.09E+06
6.62E+05
0.00E+00
0.00E+00
4.88E+05
−3.16
0.147




GTPase 1









P62906
RPL10A
60S ribosomal
1.55E+07
3.36E+06
0.00E+00
7.71E+06
3.25E+06
−1.72
0.175




protein L10a









P62263
RPS14
40S ribosomal
3.29E+07
1.15E+07
0.00E+00
4.77E+06
7.50E+06
−3.44
0.065




protein S14









P62244
RPS15A
40S ribosomal
3.39E+06
7.27E+05
0.00E+00
5.95E+05
1.06E+06
−1.89
0.191




protein S15a









P62269
RPS18
40S ribosomal
1.19E+07
5.80E+06
1.17E+06
2.63E+06
1.79E+06
−4.09
0.058




protein S18









Q6P087
RPUSD3
RNA
1.34E+06
7.03E+05
0.00E+00
3.32E+05
3.30E+05
−3.49
0.086




pseudouridylate











synthase domain-











containing protein 3









Q9NP81
SARS2
Serine--tRNA ligase,
1.95E+07
1.90E+06
0.00E+00
1.03E+07
1.83E+06
−1.56
0.288




mitochondrial









P31040
SDHA
Succinate
1.37E+06
8.56E+05
1.44E+07
2.43E+06
1.74E+06
5.48
0.195




dehydrogenase











[ubiquinone]











flavoprotein subunit,











mitochondrial









P21912
SDHB
Succinate
1.05E+07
1.28E+06
4.22E+05
1.85E+07
1.37E+06
1.70
0.354




dehydrogenase











[ubiquinone] iron-











sulfur subunit,











mitochondrial









P53007
SLC25Al
Tricarboxylate
2.18E+06
4.50E+05
0.00E+00
6.21E+06
1.22E+06
2.58
0.246




transport protein,











mitochondrial









O75746
SLC25A12
Calcium-binding
1.51E+06
8.50E+05
0.00E+00
6.19E+05
3.85E+05
−3.03
0.105




mitochondrial carrier











protein Aralar1









P05141
SLC25A5
ADP/ATP
1.05E+08
8.05E+06
1.13E+08
5.38E+07
2.75E+07
2.31
0.155




translocase 2;











ADP/ATP











translocase 2, N-











terminally processed









P12236
SLC25A6
ADP/ATP
2.00E+07
2.11E+06
3.42E+07
4.48E+06
1.16E+07
3.10
0.173




translocase 3;











ADP/ATP











translocase 3, N-











terminally processed









Q9GZT3
SLIRP
SRA stem-loop-
1.78E+07
2.97E+06
0.00E+00
6.31E+06
1.60E+06
−2.58
0.058




interacting RNA-











binding protein,











mitochondrial









Q8IYB8
SUPV3L1
ATP-dependent
5.14E+06
8.42E+05
0.00E+00
6.92E+06
2.06E+06
1.57
0.317




RNA helicase











SUPV3L1,











mitochondrial









Q9BSH4
TACO1
Translational
8.87E+06
1.33E+06
0.00E+00
1.71E+06
1.44E+06
−2.50
0.063




activator of











cytochrome c











oxidase 1









Q9BW92
TARS2
Threonine--
6.82E+06
3.42E+06
0.00E+00
5.41E+06
2.21E+06
−1.86
0.205




tRNA ligase,











mitochondrial









Q9Y5L4
TIMM13
Mitochondrial
4.55E+06
5.90E+05
0.00E+00
1.23E+06
9.94E+05
−1.70
0.195




import inner











membrane











translocase subunit











Tim13









O43615
T1MM44
Mitochondrial
2.22E+07
9.31E+05
0.00E+00
3.69E+06
1.25E+06
−3.17
0.137




import inner











membrane











translocase subunit











TIM44









O60220
TIMM8A
Mitochondrial
2.17E+07
1.01E+07
6.06E+06
9.25E+06
6.92E+06
−1.81
0.146




import inner











membrane











translocase subunit











Tim8 A









Q15388
TOMM20
Mitochondrial
2.23E+06
7.19E+05
0.00E+00
1.40E+07
1.46E+06
3.78
0.250




import receptor











subunit TOM20











homolog









O94826
TOMM70A
Mitochondrial
2.82E+07
1.35E+06
0.00E+00
1.65E+06
1.30E+06
−5.96
0.081




import receptor











subunit











TOM70









P14927
UQCRB
Cytochrome b-c1
8.50E+05
5.87E+05
0.00E+00
0.00E+00
5.33E+04
−15.37
0.060




complex subunit 7









P31930
UQCRC1
Cytochrome b-c1
1.10E+07
1.85E+06
0.00E+00
5.34E+07
3.59E+06
3.92
0.266




complex subunit 1,











mitochondrial









P22695
UQCRC2
Cytochrome b-c1
1.17E+07
2.88E+06
0.00E+00
2.63E+07
1.13E+07
2.47
0.213




complex subunit 2,











mitochondrial









P45880
VDAC2
Voltage-dependent
2.62E+07
4.16E+06
9.80E+04
1.92E+06
5.46E+06
−2.69
0.097




anion-selective











channel protein 2









P40939
HADHA
Trifunctional enzyme
3.95E+07
9.56E+06
2.59E+07
3.16E+07
1.43E+07
1.40
0.020




subunit alpha,











mitochondrial;











Long-chain











enoyl-CoA











hydratase;











Long chain











3-hydroxyacyl-











CoA











dehydrogenase









P04181
OAT
Ornithine
1.08E+08
2.95E+07
7.16E+07
9.65E+07
4.92E+07
1.46
0.031




aminotransferase,











mitochondrial;











Ornithine











aminotransferase,











hepatic form;











Ornithine











aminotransferase,











renal form









P42765
ACAA2
3-ketoacyl-CoA
4.37E+07
9.50E+06
6.95E+06
3.21E+07
7.06E+06
−1.27
0.313




thiolase,











mitochondrial









P45954
ACADSB
Short/branched
3.24E+06
1.98E+06
0.00E+00
4.39E+06
2.50E+06
−1.07
0.466




chain specific











acyl-CoA











dehydrogenase,











mitochondrial









P24752
ACAT1
Acetyl-CoA
1.32E+08
2.41E+07
2.07E+07
6.41E+07
5.41E+07
1.04
0.469




acetyltransferase,











mitochondrial









Q99798
ACO2
Aconitate hydratase,
4.15E+07
1.88E+06
0.00E+00
9.92E+06
1.27E+07
−1.05
0.481




mitochondrial









O95831
AIFM1
Apoptosis-inducing
3.14E+07
4.18E+06
3.29E+06
1.10E+07
5.73E+06
−1.48
0.155




factor 1,











mitochondrial











Adenylate kinase 2,











mitochondrial;









P54819
AK2
Adenylate
2.88E+07
7.81E+06
1.32E+07
1.71E+07
1.19E+07
1.11
0.332




kinase 2,











mitochondrial,











N-terminally











processed









Q92667
AKAP1
A-kinase anchor
2.97E+06
3.92E+05
0.00E+00
2.32E+06
2.83E+05
−1.27
0.393




protein 1,











mitochondrial









P54886
ALDH18A1
Delta-1-pyrroline-
1.35E+08
2.60E+07
5.16E+07
5.80E+07
3.60E+07
−1.03
0.383




5-carboxylate











synthase;











Glutamate











5-kinase;











Gamma-











glutamyl phosphate











reductase









P30038
ALDH4A1
Delta-1-pyrroline-
6.19E+05
1.09E+05
0.00E+00
1.55E+05
1.95E+05
−1.41
0.274




5-carboxylate











dehydrogenase,











mitochondrial









P49419
ALDH7A1
Alpha-aminoadipic
7.80E+06
9.70E+05
0.00E+00
8.35E+06
1.01E+06
1.09
0.468




semialdehyde











dehydrogenase









Q9NVI7
ATAD3A
ATPase family
6.21E+07
2.29E+07
6.86E+06
9.35E+07
2.58E+07
1.13
0.434




AAAd omain-











containing











protein 3A









P06576
ATP5B
ATP synthase
9.49E+08
2.13E+08
7.81E+08
5.50E+08
3.42E+08
1.45
0.134




subunit beta,











mitochondrial









P24539
ATP5F1
ATP synthase F(0)
3.56E+06
7.58E+04
3.21E+06
0.00E+00
0.00E+00
1.40
0.423




complex subunit B1,











mitochondrial









O75947
ATP5H
ATP synthase
3.25E+07
1.23E+07
1.99E+07
3.12E+07
1.11E+07
1.09
0.389




subunit d,











mitochondrial









P18859
ATP5J
ATP synthase-
1.16E+07
4.58E+06
9.35E+06
5.64E+06
4.57E+06
1.00
0.497




coupling factor 6,











mitochondrial









P48047
ATP5O
ATP synthase
6.83E+07
5.94E+06
1.47E+07
1.62E+07
1.52E+07
−1.12
0.397




subunit O,











mitochondrial









Q07021
C1QBP
Complement
2.76E+07
1.58E+07
4.00E+07
3.36E+07
7.57E+06
1.19
0.385




component 1 Q











subcomponent-











binding protein,











mitochondrial









Q4VC31
CCDC58
Coiled-coil
3.81E+06
3.22E+06
4.47E+05
1.02E+07
2.88E+06
1.07
0.471




domain-containing











protein 58









Q9BXW7
CECR5
Cat eye
7.73E+06
2.46E+06
5.92E+05
7.56E+06
3.11E+06
−1.06
0.465




syndrome











critical region











protein 5









P12532
CKMT1A
Creatine kinase
1.94E+07
8.62E+05
9.48E+05
9.54E+06
8.08E+05
−1.44
0.355




U-type,











mitochondrial









O76031
CLPX
ATP-dependent
9.68E+06
2.00E+06
1.73E+06
4.26E+06
3.57E+06
−1.09
0.419




CIp protease











ATP-binding











subunit cIpX-like,











mitochondrial









Q5HYK3
COQ5
2-methoxy-6-
9.71E+05
4.62E+05
0.00E+00
1.29E+05
9.38E+05
−1.19
0.440




polyprenyl-1,4-











benzoquinol











methylase,











mitochondrial









P20674
COX5A
Cytochrome c
1.07E+08
5.62E+06
6.44E+06
5.78E+07
1.15E+07
−1.13
0.440




oxidase subunit 5A,











mitochondrial









P00387
CYB5R3
NADH-cytochrome
5.74E+06
4.61E+06
9.35E+06
1.35E+04
3.40E+06
−1.20
0.419




b5 reductase 3;











NADH-cytochrome











b5 reductase 3











membrane-bound











form; NADH-











cytochrome b5











reductase 3











soluble form









P08574
CYC1
Cytochrome c1,
1.39E+06
1.87E+06
0.00E+00
9.24E+06
9.12E+05
1.32
0.417




heme protein,











mitochondrial









Q6PI48
DARS2
Aspartate--tRNA
1.21E+07
2.71E+06
9.29E+05
1.29E+07
2.21E+06
−1.07
0.466




ligase,











mitochondrial









Q9NUL7
DDX28
Probable ATP-
1.09E+06
4.84E+05
0.00E+00
0.00E+00
9.29E+05
−1.36
0.397




dependent RNA











helicase DDX28









Q7L2E3
DHX30
Putative ATP-
1.44E+07
7.43E+06
1.61E+06
2.05E+07
5.50E+06
−1.19
0.406




dependent RNA











helicase DHX30









P10515
DLAT
Dihydrolipoyllysine-
1.23E+07
3.62E+06
7.53E+06
1.29E+06
2.73E+06
−1.43
0.265




residue











acetyltransferase











component of











pyruvate











dehydrogenase











complex,











mitochondrial









P36957
DLST
Dihydrolipoyllysine-
3.89E+06
6.08E+05
6.98E+05
2.72E+06
2.01E+06
1.46
0.277




residue











succinyltransferase











component of 2-











oxoglutarate











dehydrogenase











complex,











mitochondrial









Q13011
ECH1
Delta(3,5)-
2.88E+07
5.35E+06
1.17E+07
2.35E+07
8.92E+06
1.33
0.121




Delta(2,4)-dienoyl-











CoA isomerase,











mitochondrial









P13804
ETFA
Electron transfer
3.78E+07
6.40E+06
2.29E+07
1.30E+07
7.21E+06
1.06
0.436




flavoprotein subunit











alpha, mitochondrial









P38117
ETFB
Electron transfer
2.68E+07
3.43E+06
1.75E+06
7.77E+06
6.23E+06
−1.46
0.234




flavoprotein subunit











beta









P22570
FDXR
NADPH:
2.58E+06
6.77E+05
0.00E+00
2.57E+06
6.19E+05
−1.17
0.413




adrenodoxin











oxidoreductase,











mitochondrial









P07954
FH
Fumarate hydratase,
1.18E+07
2.88E+06
0.00E+00
7.11E+06
3.56E+06
−1.39
0.294




mitochondrial









Q92947
GCDH
Glutaryl-CoA
3.64E+06
4.55E+05
0.00E+00
2.71E+06
1.19E+06
1.16
0.419




dehydrogenase,











mitochondrial









Q86SX6
GLRX5
Glutaredox
9.15E+05
3.26E+05
0.00E+00
2.81E+06
0.00E+00
1.46
0.407




in-related











protein 5,











mitochondrial









P43304
GPD2
Glycerol-3-
5.37E+06
4.68E+05
0.00E+00
2.76E+06
7.83E+05
−1.31
0.354




phosphate











dehydrogenase,











mitochondrial









Q9HAV7
GRPEL1
GrpE protein
1.12E+07
9.67E+05
0.00E+00
1.16E+07
1.45E+06
1.21
0.433




homolog 1,











mitochondrial









Q9Y2Q3
GSTK1
Glutathione S-
3.17E+06
5.02E+05
0.00E+00
4.52E+06
4.90E+05
1.29
0.414




transferase kappa 1









P31937
HIBADH
3-hydroxyisobutyrate
2.71E+06
6.14E+05
0.00E+00
3.84E+06
3.56E+05
1.04
0.486




dehydrogenase,











mitochondrial









P38646
HSPA9
Stress-70 protein,
3.15E+08
7.14E+07
1.28E+08
2.33E+08
7.98E+07
1.08
0.352




mitochondrial









P10809
HSPD1
60 kDa heat shock
8.21E+08
2.84E+08
7.93E+08
5.91E+08
2.47E+08
1.21
0.324




protein,











mitochondrial









P61604
HSPE1
10 kDa heat shock
3.83E+07
9.74E+06
9.67E+06
3.12E+07
7.61E+06
−1.14
0.378




protein,











mitochondrial









Q5T440
IBA57
Putative transferase
3.15E+06
4.47E+05
0.00E+00
0.00E+00
1.48E+06
−1.31
0.411




CAF17,











mitochondrial









P48735
1DH2
Isocitrate
2.83E+06
6.13E+05
0.00E+00
2.06E+06
7.53E+05
−1.23
0.369




dehydrogenase











[NADP],











mitochondrial









O43837
IDH3B
Isocitrate
1.81E+07
1.61E+06
2.04E+06
1.58E+07
5.96E+05
−1.01
0.496




dehydrogenase











[NAD] subunit beta,











mitochondrial









P51553
IDH3G
Isocitrate
3.24E+06
9.38E+05
0.00E+00
7.33E+06
7.25E+05
1.45
0.386




dehydrogenase











[NAD] subunit











gamma,











mitochondrial









P28838
LAP3
Cytosol
8.29E+06
2.49E+06
0.00E+00
8.39E+06
2.74E+06
−1.14
0.427




aminopeptidase









P07195
LDHB
L-lactate
1.33E+07
3.58E+06
4.06E+06
1.19E+07
2.42E+06
−1.10
0.416




dehydrogenase B











chain









O95202
LETM1
LETM1 and
2.01E+07
5.77E+06
3.69E+06
1.84E+07
6.62E+06
−1.03
0.475




EF-hand











domain-containing











protein 1,











mitochondrial









P36776
LONP1
Lon protease
5.65E+07
1.22E+07
2.47E+07
2.87E+07
7.23E+06
−1.21
0.245




homolog,











mitochondrial









P42704
LRPPRC
Leucine-rich PPR
6.52E+07
1.49E+07
1.28E+07
3.50E+07
1.57E+07
−1.29
0.196




motif-containing











protein,











mitochondrial









Q9HCCO
MCCC2
Methylcrotonoyl-
1.16E+07
2.81E+06
5.86E+06
5.46E+06
2.78E+06
−1.06
0.363




CoA carboxylase











beta chain,











mitochondrial









P40926
MDH2
Malate
5.44E+07
1.79E+07
2.76E+07
1.83E+07
1.10E+07
−1.47
0.163




dehydrogenase,











mitochondrial









Q8IWA4
MFN1
Mitofusin-1
3.58E+05
6.80E+04
0.00E+00
3.40E+05
0.00E+00
−1.21
0.409


Q99797
MIPEP
Mitochondrial
7.26E+06
4.53E+05
0.00E+00
5.42E+06
1.22E+06
1.12
0.454




intermediate











peptidase









Q5T653
MRPL2
39S ribosomal
1.18E+07
3.84E+06
6.20E+06
3.71E+06
2.29E+06
−1.48
0.171




protein L2,











mitochondrial









Q9BYC9
MRPL20
39S ribosomal
3.84E+06
1.29E+06
0.00E+00
4.71E+06
4.53E+05
−1.40
0.358




protein L20,











mitochondrial









Q9NWU5
MRPL22
39S ribosomal
6.61E+06
2.91E+06
6.36E+06
6.58E+06
1.79E+06
1.11
0.408




protein L22,











mitochondrial









P09001
MRPL3
39S ribosomal
2.39E+06
1.24E+06
0.00E+00
6.21E+06
1.14E+06
1.28
0.407




protein L3,











mitochondrial









Q9BZE1
MRPL37
39S ribosomal
7.58E+06
2.51E+06
0.00E+00
3.59E+06
4.50E+06
−1.21
0.412




protein L37,











mitochondrial









Q9NQ50
MRPL40
39S ribosomal
2.03E+06
2.25E+05
0.00E+00
2.07E+06
4.24E+05
1.26
0.402




protein L40,











mitochondrial









Q9H9J2
MRPL44
39S ribosomal
1.07E+06
5.50E+05
0.00E+00
2.87E+06
5.80E+05
1.38
0.376




protein L44,











mitochondrial









Q9BRJ2
MRPL45
39S ribosomal
1.07E+07
7.85E+06
6.12E+06
1.44E+07
5.03E+06
−1.21
0.374




protein L45,











mitochondrial









Q9HD33
MRPL47
39S ribosomal
3.51E+06
1.12E+06
0.00E+00
6.29E+06
4.30E+05
1.03
0.491




protein L47,











mitochondrial









Q86TS9
MRPL52
39S ribosomal
4.12E+06
2.08E+06
0.00E+00
5.97E+06
1.73E+06
−1.18
0.417




protein L52,











mitochondrial









Q7Z7F7
MRPL55
39S ribosomal
2.55E+06
1.49E+06
5.06E+05
1.94E+06
2.58E+06
1.01
0.496




protein L55,











mitochondrial









Q9NP92
MRPS30
28S ribosomal
4.48E+06
0.00E+00
1.13E+06
1.39E+06
8.40E+04
−1.12
0.454




protein S30,











mitochondrial









P82673
MRPS35
28S ribosomal
6.01E+06
1.17E+06
9.11E+05
7.73E+06
1.44E+05
1.00
0.499




protein S35,











mitochondrial









P82675
MRPS5
28S ribosomal
7.28E+05
3.80E+05
9.49E+05
6.88E+05
2.17E+05
1.14
0.410




protein S5,











mitochondrial









P82933
MRPS9
28S ribosomal
2.53E+06
4.89E+05
0.00E+00
1.92E+06
2.94E+05
−1.47
0.302




protein S9,











mitochondrial











Monofunctional









Q6UB35
MTHFD1L
C1-tetrahydrofolate
1.67E+07
5.02E+06
4.90E+05
8.33E+06
7.60E+06
−1.31
0.348




synthase,











mitochondrial









P22033
MUT
Methylmalonyl-
2.10E+06
9.02E+05
0.00E+00
2.52E+06
1.08E+06
−1.07
0.464




CoA mutase,











mitochondrial









O43678
NDUFA2
NADH
1.74E+07
3.45E+06
2.73E+06
1.15E+07
3.55E+06
−1.19
0.331




dehydrogenase











[ubiquinone] 1 alpha











subcomplex











subunit 2









Q16718
NDUFA5
NADH
1.62E+07
2.94E+06
9.87E+06
4.25E+06
3.28E+06
−1.01
0.493




dehydrogenase











[ubiquinone] 1 alpha











subcomplex











subunit 5









Q9P032
NDUFAF4
NADH
6.24E+06
2.59E+06
4.51E+06
2.24E+06
1.82E+06
−1.28
0.307




dehydrogenase











[ubiquinone] 1 alpha











subcomplex











assembly











factor 4









O95168
NDUFB4
NADH
5.38E+06
9.26E+05
0.00E+00
4.67E+06
1.47E+06
1.01
0.493




dehydrogenase











[ubiquinone] 1 beta











subcomplex











subunit 4









O95139
NDUFB6
NADH
9.62E+06
1.14E+06
5.29E+06
0.00E+00
1.80E+06
−1.12
0.442




dehydrogenase











[ubiquinone] 1 beta











subcomplex











subunit 6









O75489
NDUFS3
NADH
1.47E+07
5.60E+06
4.74E+06
9.62E+06
5.50E+06
−1.23
0.298




dehydrogenase











[ubiquinone] iron-











sulfur protein 3,











mitochondrial









Q9Y697
NFS1
Cysteine
4.30E+06
1.23E+06
0.00E+00
4.21E+06
1.64E+06
−1.06
0.468




desulfurase,











mitochondrial









Q13423
NNT
NAD(P)
1.29E+07
2.53E+06
2.09E+06
2.91E+06
6.43E+06
−1.04
0.480




transhydrogenase,











mitochondrial









A8MXV4
NUDT19
Nucleoside
1.81E+07
4.58E+06
7.57E+05
1.12E+07
7.55E+06
−1.14
0.418




diphosphate-linked











moiety X motif 19,











mitochondrial









O60313
OPA1
Dynamin-like
1.50E+07
3.49E+06
2.11E+06
9.75E+06
4.69E+06
−1.13
0.387




120 kDa protein,











mitochondrial;











Dyna min-l











ike 120 kDa











protein, form S1









P07237
P4HB
Protein disulfide-
2.51E+07
6.39E+06
8.79E+06
1.00E+07
1.61E+07
1.24
0.375




isomerase









Q99497
PARK7
Protein deglycase
9.21E+05
5.71E+05
0.00E+00
1.97E+06
0.00E+00
−1.44
0.382




DJ-1









P11177
PDHB
Pyruvate
5.34E+07
2.01E+07
2.98E+07
2.55E+07
1.41E+07
−1.30
0.238




dehydrogenase











E1 component











subunit beta,











mitochondrial









Q15118
PDK1
[Pyruvate
9.61E+05
5.63E+05
0.00E+00
3.00E+06
6.33E+05
1.48
0.355




dehydrogenase











(acetyl-transferring)]











kinase isozyme 1,











mitochondrial









Q15120
PDK3
[Pyruvate
9.09E+05
4.36E+05
0.00E+00
0.00E+00
9.55E+05
−1.17
0.451




dehydrogenase











(acetyl-transferring)]











kinase isozyme 3,











mitochondrial









Q99623
PHB2
Prohibitin-2
4.36E+08
2.15E+07
1.22E+08
7.85E+07
6.77E+07
−1.09
0.436


Q9H2U2
PPA2
Inorganic
8.10E+05
8.64E+05
7.64E+05
2.12E+06
9.59E+05
1.25
0.359




pyrophosphatase 2,











mitochondrial









P30048
PRDX3
Thioredoxin-
7.24E+07
2.48E+07
6.04E+07
8.36E+07
1.36E+07
1.23
0.336




dependent











peroxide











reductase,











mitochondrial









Q96EY7
PTCD3
Pentatricopeptide
1.06E+07
1.49E+06
0.00E+00
6.97E+06
2.52E+06
−1.13
0.430




repeat domain-











containing protein 3,











mitochondrial









Q9Y606
PUS1
tRNA
2.83E+06
1.25E+06
1.75E+06
4.21E+06
1.23E+06
1.26
0.301




pseudouridine











synthase A,











mitochondrial









Q8IXI1
RHOT2
Mitochondrial Rho
7.10E+06
1.58E+06
0.00E+00
2.05E+06
2.81E+06
−1.45
0.311




GTPase 2









Q9H9B4
SFXN1
Sideroflexin-1
4.42E+07
8.56E+06
9.03E+05
1.63E+07
1.67E+07
−1.29
0.360


Q96NB2
SFXN2
Sideroflexin-2
7.68E+05
3.84E+05
0.00E+00
1.77E+06
4.53E+05
1.32
0.381


Q9UJS0
SLC25A13
Calcium-binding
1.99E+07
4.45E+06
2.32E+06
5.69E+06
8.30E+06
−1.26
0.360




mitochondrial carrier











protein Aralar2









Q7KZF4
SND1
Staphylococcal
4.16E+07
6.65E+06
5.39E+06
2.33E+07
7.37E+06
−1.26
0.284




nuclease domain-











containing protein 1









Q9UJZ1
STOML2
Stomatin-like
8.74E+07
1.99E+07
4.13E+07
3.23E+07
1.37E+07
−1.27
0.194




protein 2,











mitochondrial









Q9P2R7
SUCLA2
Succinyl-CoA
2.51E+06
4.49E+05
1.36E+06
1.01E+06
2.36E+04
−1.32
0.336




ligase











[ADP-forming]











subunit beta,











mitochondrial









Q96I99
SUCLG2
Succinyl-CoA ligase
6.19E+06
1.06E+06
0.00E+00
4.25E+06
7.86E+05
−1.49
0.292




[GDP-forming]











subunit beta,











mitochondrial









O14925;
TIMM23;
Mitochondrial import
2.03E+06
5.54E+05
0.00E+00
7.65E+05
1.63E+06
1.14
0.453


Q5SRD1
TIMM23B
inner membrane











translocase subunit











Tim23; Putative











mitochondrial import











inner membrane











translocase subunit











Tim23B









Q3ZCQ8
TIMM50
Mitochondrial import
3.88E+07
6.16E+06
1.24E+07
8.71E+06
6.61E+06
−1.40
0.086




inner membrane











translocase subunit











TIM50









Q9Y5J9
TIMM8B
Mitochondrial import
1.71E+07
9.53E+06
1.72E+07
9.77E+06
5.00E+06
−1.21
0.370




inner membrane











translocase subunit











Tim8 B









Q9NS69
TOMM22
Mitochondrial import
1.18E+07
6.60E+06
1.41E+07
1.33E+07
1.14E+06
−1.05
0.472




receptor subunit











TOM22 homolog









Q12931
TRAP1
Heat shock protein
1.48E+08
5.55E+07
1.10E+08
1.61E+08
5.80E+07
1.27
0.209




75 kDa,











mitochondrial









P43897
TSFM
Elongation factor Ts,
1.80E+07
1.18E+07
8.08E+05
4.43E+07
6.46E+06
−1.03
0.488




mitochondrial









Q16762
TST
Thiosulfate
1.43E+07
3.17E+06
1.16E+07
4.68E+06
1.89E+06
1.00
0.498




sulfurtransferase









A3KMH1
VWA8
von Willebrand
1.57E+06
4.00E+05
0.00E+00
9.08E+05
7.06E+05
−1.13
0.428




factor A domain-











containing protein 8









Q96TA2
YME1L1
ATP-dependent
8.87E+06
3.17E+06
3.52E+05
9.35E+06
4.49E+06
−1.01
0.495




zinc











metalloprotease











YME1L1












UniProt IDs: Uniprot accession IDs of identified mitochondrial proteins as defined by MitoCarta 2.0 (Calvo etal. 2016; PMID:26450961)


Protein names: Protein names of identified mitochondrial proteins


Protein description: Description of identified mitochondrial proteins


WT-1: Total peptide ion intensity determined by MaxQuant in replicate 1 of wild type (WT)


WT-2: Total peptide ion intensity determined by MaxQuant in replicate 2 of WT


CLPP KO-1: Total peptide ion intensity determined by MaxQuant in replicate 1 of CLPP knockout (KO)


CLPP KO-2: Total peptide ion intensity determined by MaxQuant in replicate 2 of CLPP KO


CLPP KO-3: Total peptide ion intensity determined by MaxQuant in replicate 3 of CLPP KO


Avg. fold change (CLPP KO/WT): Fold change of protein detected in CLPP knockout as compared to WT


p-Value significance: Proteins detected in CLPP KO versus WT using Students paired t test


Mitochondrial proteins identified with p-value 0.05 and |1.5| fold change are highlighted in bold.







Cellular Sensitivity to ADEP Correlates with Intracellular HsCIpP Expression Levels


Given that ADEP-induced cytotoxicity depends on HsCIpP, the effect of intracellular HsCIpP levels on the cell's sensitivity to ADEP was examined. HsCIpP overexpression was achieved using HEK293 T-REx overexpressing a C-terminally FLAG-tagged, full length HsCIpP (CLPP-FLAG) upon induction with doxycycline (Dox). Similarly, HEK293 T-REx+CLPPS153A-FLAG overexpresses a C-terminally FLAG-tagged, full length inactive HsCIpP(S153A) mutant.


HsCIpP overexpression was confirmed by Western blot analysis. As shown in FIG. 4D, high expression of FLAG-tagged HsCIpP or FLAG-tagged HsCIpPS153A was detected in HEK293 T-REx+CLPP-FLAG and HEK293 T-REx+CLPPS153A-FLAG, respectively, in the presence of Dox (band I). Due to leakiness of the Dox-inducible promoter, expression of the FLAG-tagged HsCIpP proteins (WT or S153A mutant) was also detected in the absence of Dox, albeit at significantly lower levels (band I). Expression of endogenous HsCIpP was also observed (band III). Interestingly, an additional HsCIpP band (band II) that corresponds to a slightly larger HsCIpP protein but lacks the C-terminal FLAG-tag (α-FLAG panel), was observed in both HEK293 T-REx+CLPP-FLAG (with or without Dox) and in HEK293 T-REx+CLPPS153A-FLAG (with Dox only) cells. Band II is likely the result of the C-terminal FLAG-tag being cleaved via HsCIpP's own proteolytic activity.


To determine the effect of intracellular HsCIpP expression on the cell's sensitivity to ADEP, HEK293 T-REx WT, CLPP−/−, HEK293 T-REx+CLPP-FLAG and HEK293 T-REx+CLPPS153A-FLAG cells were grown in the presence of ADEP-41 with or without Dox. Without Dox, both HEK293 T-REx+CLPP-FLAG and HEK293 T-REx+CLPPS153A-FLAG were equally sensitive to ADEP-41 as WT, while CLPP−/− was resistant (FIG. 4E, −Dox panel). Accordingly, the IC50 for WT, HEK293 T-REx+CLPP-FLAG and HEK293 T-REx+CLPPS153A-FLAG were similar within statistical errors (FIG. 4F). In the presence of Dox, the increase in intracellular HsCIpP level rendered HEK293 T-REx+CLPP-FLAG highly sensitive to ADEP-41 compared to WT (FIG. 4E, +Dox panel), resulting in a significant decrease in IC50 (FIG. 4F). Importantly, HEK293 T-REx+CLPPS153A-FLAG, which expresses native HsCIpP at the WT level but can only overexpress the proteolytically inactive HsCIpP mutant, maintains WT-like sensitivity to ADEP-41 (FIG. 4E, +Dox panel) and shows no change in IC50 (FIG. 4F). These results clearly illustrate that the intracellular expression level of proteolytically active HsCIpP can modulate the cell's sensitivity to ADEP, with higher expression resulting in greater sensitivity.


Aside from HEK293 T-REx, the cellular sensitivity to ADEP-41 was investigated in several other commonly used cell lines. These include HeLa (regular), HeLa T-REx, U2OS and undifferentiated SH-SYSY (FIGS. 5B and C). The IC50 for all cell lines were similar with undifferentiated SH-SYSY having a slightly higher value (FIG. 5C). However, U2OS and undifferentiated SH-SYSY have lower levels of HsCIpP than the other cell lines (about 20% and 40% relative to HEK293 T-REx, respectively) (FIG. 5D). Hence, in addition to the intracellular level of HsCIpP, ADEP sensitivity varies slightly by cell type.


ADEP Activates the Intrinsic, Caspase-Dependent Apoptosis in HEK293 T-REx Cells

The cellular mechanism underlying ADEP's cytotoxicity on HEK293 T-REx cells was further investigated. First, morphological changes of HEK293 T-REx WT and CLPP−/− exposed to ADEP-41 for 72 hours were examined by microscopy. ADEP-41-treated WT cells appeared compact and spherical, and exhibited a significant loss of adherence to the growth surface (FIGS. 6A and 7A). This is in stark contrast to DMSO-treated WT cells as well as to CLPP−/− cells that are resistant to ADEP-induced cytotoxicity (FIGS. 6A and 7A). Furthermore, blebs were observed on the surface of affected WT cells (indicated with arrows in FIG. 6A).


Both cell shrinkage and surface blebs formation are strong indicators of apoptosis (Elmore, 2007). Apoptosis in HEK293 T-REx WT cells induced by ADEP-41 was confirmed by TUNEL assay indicating the fragmentation of chromosomal DNA that congregate into small, condensed bodies (appearing as multiple foci) upon compound treatment (FIG. 6A). In contrast, DMSO-treated WT cells have intact chromosomal DNA and were not TUNEL-labeled. Importantly, both ADEP-treated and DMSO-treated CLPP−/− cells have normal nuclei and intact chromosomal DNA (FIG. 6A). Quantification of cells with positive TUNEL-labelling revealed that about 20 times more of the ADEP-41-treated WT cells were TUNEL-positive compared to DMSO-treated ones, or compared to CLPP−/− cells treated with ADEP-41 or DMSO (FIG. 6B).


Subsequently, the intracellular expression of signature apoptotic protein markers was examined by Western blotting. The expression of both HsCIpP and HsCIpX was also examined. Treatment with ADEP for 24 or 72 hours did not change HsCIpP levels in WT cells compared to DMSO (FIG. 6C). In contrast, HsCIpX was no longer detected in WT cells treated with ADEP-41 for either duration (FIG. 6C), despite the fact that HsCIpX is not degraded by the ADEP-activated HsCIpP in vitro (FIGS. 3E-G). To determine whether the loss of HsCIpX has any potential role in ADEP-induced apoptosis, a CLPX gene disruption was obtained using CRISPR-Cas9, which reduces HsCIpX expression by ≥97% (ΔCLPX*; FIG. 7B). Significant reduction of HsCIpX levels had no statistically significant impact on the cells' sensitivity to ADEP-28 regardless of HsCIpP expression (FIG. 7C). Hence, without wishing to be bound by theory, the loss of HsCIpX in ADEP-treated HEK293 T-REx WT cells may be part of a stress response mechanism. Alternatively, HsCIpX could be degraded by other mitochondrial proteases.


After 24 hours of ADEP-41 treatment, no activation-specific cleavage was observed for Caspase-8, Caspase-9 or Caspase-3. However, the anti-apoptotic Mcl-1 protein was degraded in WT cells, but not in CLPP−/− cells (FIG. 6C). Conversely, the pro-apoptotic PUMA protein was significantly upregulated in WT cells in contrast to CLPP−/− cells (FIG. 6C). These changes in the intracellular levels of Mcl-1 and PUMA are strong indications for the initiation of the intrinsic apoptotic pathway.


After 72 hours of ADEP-41 treatment, the degradation of Mcl-1 in WT cells remained clearly observable (FIG. 6C). Notably, there was no change in the level of anti-apoptotic Bcl-2 protein, suggesting that the loss of Mcl-1 is a specific cellular response to the ADEP-41 treatment. Importantly, activation-specific cleavage was detected for both Caspase-9 and Caspase-3, which produced the characteristic p37 and p35 fragments for Caspase-9, and p19/p17 for Caspase-3 (FIG. 6C). Furthermore, no activation-specific cleavage was observed for Caspase-8 (FIG. 7D). Given that Caspase-9 is functionally associated with the intrinsic apoptotic pathway while Caspase-8 is linked to the extrinsic pathway (Mcllwain et al., 2013), it is concluded that treatment of HEK293 T-REx WT cells with ADEP activates the intrinsic apoptotic pathway, leading to cell death.


ADEP Induces Mitochondrial Fragmentation and Abolishes Oxidative Phosphorylation in HEK293 T-REx Cells

Given that mitochondrial fragmentation and outer membrane permeabilization (MOMP) are hallmarks of apoptosis (Landes and Martinou, 2011), the impact of ADEP on mitochondrial morphology and oxidative phosphorylation (OXPHOS) was further investigated. Mitochondria in HEK293 T-REx WT and CLPP−/− treated with ADEP-41 or DMSO for 24 and 72 hours were examined below the diffraction limit by super resolution 3D Structured Illumination Microscopy (3DSIM). As shown in FIG. 6D, HEK293 T-RExWT cells treated with ADEP-41 already showed clear signs of mitochondrial fragmentation after 24 hours compared to DMSO-treated cells. Furthermore, the fragmentation observed was very significant after 72 hours of treatment. In contrast, CLPP−/− cells treated with ADEP-41 showed no change in mitochondrial morphology for either duration.


MOMP is known to dissipate the mitochondrial electrochemical gradient (Δψ) during apoptosis (Kroemer and Reed, 2000), which abolishes OXPHOS. As such, OXPHOS in WT and CLPP−/− cells treated with ADEP-28 or DMSO for 24 hours was examined using the Seahorse extracellular flux (XF) analyzer. Notably, OXPHOS in ADEP-28-treated WT cells was largely abolished, as exemplified by the large reduction in oxygen consumption rate (OCR) associated with basal respiration (˜86%) and ATP synthase activity (˜91%), relative to DMSO-treated WT cells (FIG. 7E and F upper panel).


Furthermore, ADEP-treated WT cells showed a higher basal extracellular acidification rate (ECAR) that was largely unresponsive to ATP synthase inhibition (FIG. 7F lower panel), showing the cell's reliance on glycolysis for energy metabolism as a result of losing OXPHOS. Without wishing to be bound by theory, both events can be explained with Δψ dissipation as a result of MOMP.


The fragmentation of mitochondria and loss of OXPHOS from Δψ dissipation both provide confirmative evidence that ADEP induces apoptosis in HsCIpP-expressing cells.


ADEP-Binding Induces Conformational Changes in the HsCIpP Tetradecamer

To further understand the mechanism by which ADEP binding leads to HsCIpP dysregulation, the structure of HsCIpP co-crystallized with ADEP-28 was determined (FIG. 8A) at 2.8 Å resolution (Table 2). ADEP-28 has high binding affinity for HsCIpP (FIG. 2E). In the crystal, the asymmetric unit contains a single heptameric ring that forms the HsCIpP tetradecamer with a second ring related to the first by crystallographic symmetry (FIG. 8A-E and Table 2). Of the 277 residues in HsCIpP, no electron density was observed for residues 252-277 (chains A-D) or 251-277 (chains E-G) at the C-terminus, whereas residues 1-57 that contain the mitochondrial transport signal were omitted during cloning (FIG. 8B). It was also observed that extra electron density attached to the thiol group of Cys86 in each subunit, indicating covalent modification (FIG. 8F). Mass spectrometry analysis identified this modification as potentially being persulfidation of the cysteine thiol.


As observed for other CIpPs, ADEP-28 bound to the hydrophobic pocket formed by two neighbouring HsCIpP subunits (FIG. 8A). Clear electron densities for all seven ADEP-28 molecules per heptameric ring were observed (FIG. 8C). The N-terminal axial loops were ordered (FIGS. 8A, D, and E), as observed previously in the ADEP-bound EcCIpP (Li et al., 2010), N. meningitidis CIpP (NmCIpP) (Goodreid et al., 2016), and M. tuberculosis CIpP (MtCIpP) (Schmitz et al., 2014). ADEP-28 binding enlarged the HsCIpP's axial pore (FIG. 8A). Interestingly, ADEP-28-HsCIpP adopts a compact conformation (FIG. 8A), which has not been seen in other ADEP-bound CIpP structures characterized to date (Goodreid et al., 2016; Lee et al., 2010; Li et al., 2010; Schmitz et al., 2014); and PDB 5VZ2. The inventors will first describe the ADEP binding pocket and then discuss the distal molecular rearrangements observed.


The binding of ADEP-28 to HsCIpP induces local structural changes at and around the binding pocket between two neighbouring subunits, forming a highly complementary surface for the ADEP molecule (FIGS. 9A and 10). Normally, the N-terminal β-1-β0 hairpins forming the axial loops are disordered in the absence of ADEP (FIG. 8A). An ionic interaction between R78 of one subunit and E109 of the neighbouring subunit secures the interface between the subunits (FIG. 9Ai). Upon ADEP-28 binding, ordered axial loop formation places E64 between R78 of the same subunit and E109 of the neighbouring subunit, creating three new hydrogen bonds (H bonds) that also involves R78 interacting with the intrasubunit E82 (FIG. 9Aii). These interactions stabilize a widened surface with the bound ADEP-28 acting as a wedge.


At the ADEP-28/CIpP interface, two Tyr residues, Y118 of one subunit and Y138 of the neighbouring subunit, form H-bonds with carbonyl carbon atoms of the depsipeptide ring (FIGS. 9Aii and 10). Similar interactions have been reported for other bacterial CIpP-ADEP complexes (Goodreid et al., 2016; Li et al., 2010; Schmitz et al., 2014). In addition, a H2O-mediated H-bond is observed between Q107 and the amide carbonyl connecting the hydrophobic tail and 3,5-difluorophenyl moieties of ADEP-28. A Trp residue (W146) in


HsCIpP forms a hydrophobic stacking interaction with the 3,5-difluorophenyl moiety of ADEP-28 (FIGS. 9Aii and 10A). The corresponding residues in bacterial CIpPs all have smaller aliphatic side chains, which yield a smaller hydrophobic surface for the difluorophenyl group (FIG. 10B).


The depsipeptide ring is solvent-exposed but with its nitrogenous, heterocyclic rings forming stacking interactions with the aromatic or hydrophobic residues in the ADEP-binding pocket. In particular, the methyl piperidine ring of ADEP-28 (FIG. 6Aii on far right of ADEP-28 when viewed as shown) sits directly on top of H168 of β5. The relatively small side chain of the His residue is likely to provide additional space to accommodate the methyl piperidine ring and in turn strengthens the ADEP-28-HsCIpP interaction, resulting in the high apparent binding affinity observed (FIG. 2E). In contrast, the equivalent residue in bacterial CIpPs is either a Tyr, Phe or Met, all of which are bulkier than the His residue of HsCIpP (FIG. 10B).


As mentioned above, binding of ADEP-28 induces the formation of the N-terminal β-1-β0 hairpins (residues 58-74), creating ordered axial loops (FIGS. 8A, D and E). While the β-1 and β0 strands are highly structured in all subunits, segments of the β-hairpin loops formed by residues 65-69 in subunits C to G have ambiguous electron density insufficient to confidently assign side chain positions. These residues have significantly higher B-factors than those of the rest of the HsCIpP molecule (FIG. 8E), indicating greater flexibility. Each axial β-hairpin is anchored to the head domain of the same subunit through the interactions of E64 on β-1 strand and Y73 on β0 strand with R78 and R81 on aA helix, respectively (FIG. 9Bi, ii). The continuous stretch of 7 hydrophobic residues (L58-V63) comprising the short N-terminal loop and the β-1 strand form hydrophobic interactions with an extensive surface formed by Y73 (β0), Y76 (αA), L80 (αA), V95 (αB), L98 (αB), F105 (αB), L106 (αB) of a neighbouring subunit, and I75 (αA) and Y76 (αA) of the same subunit (FIG. 9Biii). These interactions result in the retraction of the N-terminus away from the 7-fold axis, leading to HsCIpP's axial pore widening. This has also been observed to occur in ADEP-bound EcCIpP (Li et al., 2010).


The compaction of the ADEP-28-HsCIpP complex was unexpected and is a direct consequence of molecular rearrangements at and around the handle region comprised mainly of β7 and αE resulting in significant reshuffling of the residues at the interface of the two apposing HsCIpP rings (FIG. 9Ci,ii). At the subunit level, residues 182-187 (182-191 in chain B) that are part of β7-strand of the handle region in apo HsCIpP show no clear electron density upon ADEP-28 binding, suggesting local structural disorder of the handle region (FIG. 9Cii). Furthermore, the ensuing αE-helix is shortened by the unwinding of two helical turns to an ordered loop that extends into HsCIpP's lumen due to a kink that starts at residue Q194 (FIG. 9Cii).


In apo HsCIpP, the interface of two heptameric rings consists of an intricate network of H-bonding and ionic interactions. The highly conserved oligomerization sensor R226 (αF-β8 loop) participates in intra-ring interactions with Q187 (β7-αE loop) and D190 (αE) of a neighbouring subunit and in inter-ring interactions with E225 (α′F) of the apposing subunit (FIG. 9Ci). Residues Q179 (β6-β7 loop) and K202 (αE) also form H-bonds with T189 (α′E) of the apposing subunit. Residue Q194 (αE) forms a H-bond with the catalytic D227 residue of a neighbouring subunit to further cement intra-ring subunit contacts (FIG. 9Ci). These interactions are conserved and highly symmetrical across the equatorial region of the HsCIpP tetradecamer and are likely essential for a properly aligned catalytic triad (S153, H178, D227).


In ADEP-28-bound HsCIpP, significant rearrangements of the intra- and inter-ring contacts occur as the long αF-β8 loops of apposing subunits come closer together to form part of the new interface. Remarkably, the catalytic triads become distorted as a result, such that H178 of one subunit now interacts with D227 of the apposing subunit and vice versa, while the original intra-subunit interaction between H178 and S153 and the inter-subunit interaction between D227 of one subunit with Q194 of the adjacent subunit are both lost (FIG. 9Cii). His178 forms a bond with Q194 of an apposing subunit, while D227 forms a bond with S181 of another apposing subunit. Furthermore, new intra-ring and inter-ring contacts are borne out of this structural rearrangement. These include the R174 (β6)-E197 (α′E) ionic interaction, the interactions of residues K202 and Y206 in α′E with Q194 and E196 of the apposing subunit, and the bond between S181 (β6-β7 loop) and the catalytic D227 of the apposing ring. Interestingly, despite these major changes at the interface, the inter-ring interaction of R226 in one subunit with E225 of the apposing subunit is preserved, albeit with a shift in the relative positions of the residues (FIG. 9Cii). The overall result of these molecular rearrangements is the compaction of the HsCIpP tetradecamer.


Compaction of HsCIpP Results in the Formation of Equatorial Side Pores

This structure of ADEP-bound HsCIpP highlights a novel role of the catalytic triad residues in stabilizing the compact conformation. Several distinct, compact conformations have been observed in the crystal structures of CIpP from E. coli (Kimber et al., 2010), L. monocytogenes (Zeiler et al., 2013), M. tuberculosis (Ingvarsson et al., 2007), P. falciparum (El Bakkouri et al., 2010), S. aureus (Ye et al., 2013), and S. pneumoniae (Gribun et al., 2005) (FIG. 11). In addition, SaCIpP was also crystallized in a compressed conformation (Zhang et al., 2011). Here the compact conformation is defined as a shortened CIpP cylinder losing one or more turns at the N-terminus of the αE helix and in many cases concomitant with the loss of density for the β7 strand. The compressed conformation refers to an even shorter CIpP cylinder caused by a kink in the αE helix resulting in the formation of two smaller helices.


These CIpP structures (FIG. 11) have varying degrees of compaction with the distorted catalytic triads engaging in distinct interactions with nearby residues. Among these, the LmCIpP1 and SaCIpP compact structures show engagement of one or more of the catalytic triad residues in inter-ring interactions (FIG. 11) as does the compact ADEP-28-HsCIpP structure (FIG. 9ii), albeit having different interacting residues. The compressed SaCIpP shows inter-ring interactions of the catalytic triad but they are mediated by a sulfate group (FIG. 11).


The compact structure of ADEP-28-HsCIpP exhibits small, equatorial side pores that presumably enlarge to facilitate the egress of cleaved peptides from the CIpP barrel (FIG. 12A). These are in close proximity to the peptide substrate binding site and are formed by residues in the vicinity of the hinge region. Other compact CIpP structures exhibit equatorial side pores of varying sizes and shapes depending on the degree of compaction (FIG. 12B). Interestingly, in the compact ADEP-28-HsCIpP, the unraveling of two N-terminal helical turns of the αE helix results in a structured loop (spanning residues 188-194), a segment of which occupies the location of the bound peptide substrate near the catalytic site and the side pore (FIG. 12A). A similar phenomenon is observed in the compact structure of PfCIpP, as well as in the compressed structure of SaCIpP, where the αE helix is kinked in such a way that the ensuing residues of a short loop occupy the peptide binding site. Therefore, the compact and compressed structures of CIpP are likely intermediates that are populated during CIpP's functional cycle in which a cleaved peptide is poised for release from the CIpP lumen through the equatorial side pores.


Analysis of the available CIpP structures from different species where both extended and compact conformations have been solved shows that the 7 catalytic Ser residues of a CIpP heptameric ring form a plane that is parallel to that formed by the other 7 catalytic Ser residues in the apposing heptamer (rotation angle, θ=−3° to +1.5°) (FIG. 13). The distance between each plane's center of mass (with each plane defined by the coordinates of the Ser Cα atoms) is ˜30 Å, and this ‘height’ decreases by as much as 8 Å upon compaction. Compaction is accompanied by rotation of the catalytic Ser planes relative to each other by as much as 23° and a concomitant, general increase in the distances between the 7 coplanar Ser residues of a ring (FIG. 13). The same is true for the other catalytic residues. For SaCIpP where a third, compressed structure is available, the ‘height’ is further reduced to ˜20 Å, while the rotation angle between catalytic Ser planes is reduced to just 6.4°, presumably to relieve the strain caused by compression (FIG. 13). Thus, collectively, these CIpP structures may constitute distinct structural intermediates in the conformational landscape of CIpP, whether as a result of activation by ADEP or CIpX or by natural breathing motions.


Solution Structure of ADEP-Bound HsCIpP Characterized by Small Angle X-Ray Scattering (SAXS)

The global conformational change in the HsCIpP structure upon ADEP binding was further examined in solution using SAXS (FIG. 14A). HsCIpP, HsCIpP+DMSO (control for DMSO's effect) and HsCIpP+ADEP-28 showed molecular mass (MM) values that correspond to tetradecamers (Table 5). The small increase in MM for HsCIpP+ADEP-28 suggested that ADEP-28 molecules were bound to HsCIpP. Pair distance distribution functions, p(r), were then generated (FIG. 14B) and the values for the radius of gyration (Rg) and maximum dimension (Dmax) were determined. HsCIpP and HsCIpP+DMSO showed similar p(r) profiles (FIG. 14B), and also similar Rg and Dmax values (Table 5). All values were in agreement with the calculated size and dimensions of HsCIpP's crystallographic structure. Importantly, the addition of ADEP-28 changed HsCIpP's p(r) profile, shifting the maximum p(r) and Dmax towards higher values (FIG. 14B).









TABLE 5







Size and dimension properties determined for HsCIpP by SAXS










SAXS
Crystallography6

















HsCIpP




HsCIpP +
HsCIpP +

+


Properties
HsCIpP
DMSO
ADEP-28
HsCIpP
ADEP-28















Rg (Å)1
44.5
44.3
45.9
42.9
42.7


Dmax (A)1
130
132
140
136
130


MMExperimental (kDa)2
337 ± 21
344 ± 11
376 ± 21




MMTheoretical (kDa)3
340
340
3514




Oligomeric state5
13.9
14.2
15.1
14-mer
14-mer






1Properties experimentally determined using final merged SAXS curves. Rg is radius of gyration. Dmax is maximum dimension of the molecule.




2MMExperimental is the average molecular mass determined from eleven SAXS curves.




3Calculated from the amino acid sequence using the Protparam program (http://web.expasy.org/protparam/).




4The MMTheoretical of compound-bound CIpP was calculated taking into account additional 14 molecules of ADEP-28 (MMTheoretical = 784.89 g/mol).




5For the SAXS data, the oligomeric state is obtained by dividing MMExperimental by the MMTheoretical of the monomer (24.2 kDa). For the crystallographic data, the oligomeric state refers to the established biological complex.




6Values calculated from the crystallographic structures by the program Hydropro (http://leonardo.inf.um.es/macromol/programs/hydropro/hydropro.htm).







Using SAXS data, low resolution dummy atoms models (DAMs) were generated for HsCIpP+DMSO and HsCIpP+ADEP-28 (FIG. 14C). Notably, in the presence of DMSO, HsCIpP adopts a cylindrical shape in solution that is compatible with the published structure of apo HsCIpP (Kang et al., 2004). The ADEP-bound HsCIpP DAM showed an increased radial and axial occupancy with dummy atoms models, adopting an ellipsoidal shape. The superposition of our ADEP-bound HsCIpP crystallographic structure on this DAM (FIG. 14C) showed a good fit at radial positions, supporting the existence of this compact conformation. Nevertheless, the empty spaces observed at the top and bottom regions of the ADEP-bound HsCIpP DAM, and the differences between properties calculated from the crystal structure and determined from the SAXS data (Table 5) indicate the likely presence of other HsCIpP conformers in solution.


Discussion

The present disclosure describes the first detailed biochemical and biophysical characterization of the molecular interactions between ADEP and mitochondrial HsCIpP, as well as the first report on the physiological impact of these interactions on human cancer cells. Lowth et al., 2012 had suggested earlier that some ADEP analogs that they tested may have affected HsCIpP, although they did not characterize those interactions further and provided no insight or evaluation on the use of ADEP in cancer treatment.


The present identification of ADEPs that activate HsCIpP was a fortuitous finding stemming from inventors' work on developing antibiotics targeting the bacterial CIpP.


The co-crystal structure of the ADEP-28 and HsCIpP recapitulates previous findings in ADEP-bound bacterial CIpPs, while providing new insights into the protease's functional cycle. Notably, the compact conformation of the ADEP-28-HsCIpP complex may constitute a putative intermediate state. While the degradation of substrates requires a properly aligned Ser-His-Asp catalytic triad that is found only in the extended conformation of the tetradecameric CIpP, additional conformations are also required to facilitate the other steps in the protein degradation process, such as the release of peptide fragments from inside the CIpP lumen. Without wishing to be bound by theory, biochemical, and biophysical experiments have thus pointed towards a dynamic CIpP tetradecamer that can extend, compact, and compress, with the flexible handle region acting as a hinge point for achieving these various conformations (Kimber et al., 2010; Liu et al., 2014). The structure of ADEP-28-HsCIpP highlights the essential role of the handle region to achieve a compact conformation whilst bound by ADEP-28 at the activator site, providing the first structural evidence that an ADEP-activated CIpP is not locked in the extended conformation, contrary to the proposal by Gersch and co-workers (Gersch et al., 2015). Instead, the ADEP-bound CIpP is sufficiently flexible and can dynamically assume conformations found in other stages of the CIpP degradation cycle. Remarkably, the X-ray structure also unveils a previously unknown structural role for the catalytic triad outside of proteolysis that appears essential for stabilizing the compact conformation.


Importantly, the interaction of ADEP with HsCIpP produces a cytotoxic effect on cells that manifests via the intrinsic, caspase-dependent apoptosis in cancer cells. An important utility from these results is that ADEPs are useful as novel therapeutic compounds for cancer treatment. Modulation of the cell's sensitivity to ADEPs by intracellular HsCIpP expression enables the fine-tuning of the ADEP chemical structure, such that new analogs can be developed with better abilities to distinguish and target cancer cells that express high levels of HsCIpP without affecting normal, healthy cells. The biophysical and structural data on the interaction between ADEP and HsCIpP are useful in identifying key chemical features in the ADEP structure that define its potency and will facilitate the design of better analogs.


The mitochondrion is a vital organelle in the human cell that has many essential biological functions. These include energy metabolism, signaling, and apoptosis. Consequently, a dysfunctional mitochondrion may give rise to a wide range of diseases including cancer. In this disclosure, the identification of compounds, i.e. ADEP analogs, that dysregulate the activity of a critical protease present in the mitochondrial matrix termed CIpP, i.e. HsCIpP, is described. These ADEP analogs dysregulate the protease activity of CIpP causing it to degrade proteins in the mitochondrion in a dysregulated manner, resulting in apoptotic cell death. The co-crystal structure of CIpP with an ADEP analog was obtained which revealed a novel conformation for the protease. The disclosure shows that ADEP analogs kill cancer cells which express high level of HsCIpP. Hence, ADEP analogs are useful for cancer treatment in targeting cancer cells expressing high levels of HsCIpP.


Example 2: Additional Experiments on ADEP Analogs Materials and Methods
Cell Line Maintenance and Propagation

HEK293 T-REx wild-type (WT) and HEK293 T-REx CLPP−/− cells were obtained from Professor Aleksandra Trifunovic (University of Cologne, Germany). MDA-MB-231 and MDA-MB-468 were obtained from Professor Lilianna Attisano (University of Toronto, Canada). MCF-7 cells were obtained from the lab of Professor Grant Brown (University of Toronto, Canada). MDA-MB-231 is an invasive ductal carcinoma cell line. It is triple-negative, i.e. MDA-MB-231 cells do not express estrogen receptors, progesterone receptors, and have no ERBB2 amplification. These cells have metastatic origin and was isolated from pleural effusion of a breast cancer patient. MCF-7 is another invasive ductal carcinoma cell line originated from pleural effusion. MCF-7 cells express estrogen receptors and do not express progesterone receptors and do not have ERBB2 amplification. MDA-MB-468 cells were extracted from a pleural effusion of mammary gland and breast tissues, and are useful for the study of metastasis, migration, and breast cancer proliferation.


Unless otherwise stated, both HEK293 T-REx WT and CLPP−/− were propagated and maintained with Dulbecco's Modified Eagle Media (DMEM) (Gibco) supplemented with 10% fetal bovine serum (FBS) (Gibco), 100 U/mL penicillin-streptomycin (Gibco) and 2 mM L-glutamine (Gibco), while MDA-MB-231, MDA-MB-468 and MCF-7 cells were grown in DMEM:F12 (Gibco) with the same supplements. All cultures were kept at 37° C. under a moist atmosphere with 6% CO2. All cells were passaged at least 3 times prior to use.


Genetic Knockdown of CLPP in MDA-MB-231 by CRISPR-Cas9

The genetic knockdown of CLPP in MDA-MB-231 was achieved via the CRISPR-Cas9 protocol as outlined in (Cong et al., 2013). Briefly, the single-strand DNA oligos CLPP Exon1 pX330 F (5′-caccggcgtgcggagggatgtggcc-3′) (SEQ ID NO:4) and CLPP Exon1 pX330 R (5′-aaacggccacatccctccgcacgcc-3′) (SEQ ID NO:5) were phosphorylated at their 5′-ends with T4 PNK (New England Biolabs) and annealed together by gradual cooling from 95° C. to 25° C. at a rate of 5° C./min in a thermocycler. The phorphorylated annealed oligo duplex was then ligated to a pX330 plasmid already digested with FastDigest BbsI (New England Biolabs) and dephosphorylated with FastAP (Thermo Scientific), using the T4 DNA ligase (New England Biolabs). The ligation reaction was then used to transform chemically competent Escherichia coli DH5a cells, followed by selection of ampicilin-resistant colonies on LB-agar plates supplemented with 100 μg/mL of the antibiotic. The resultant pX330-ΔCLPP plasmid was isolated using the PureLink Quick Plasmid Miniprep Kit (Invitrogen) and sequence-verified.


To generate MDA-MB-231 CLPP-KD, WT cells were first passaged in DMEM:F12 media without any antibiotics. These cells were then grown in 6-well tissue culture plates to near confluence in the absence of antibiotics. A DNA transfection mixture containing 1.7 μg of pX330-ΔCLPP and 100 nmol of the single-strand DNA oligo, CLPP Exon1 ssODN (5′-gtagttccgccatcggacggaagccgaccggggcgtgcggagggtgataataatgaatattggtagggg gggcccgggtggcgtcatgc-3′) (SEQ ID NO:6) was prepared using the JetPrime in vitro transfection reagent (Polyplus) following the manufacturer's protocol. Cells were incubated in the presence of the transfection mixture for 5 hours inside an incubator, after which a media change using fresh DMEM:F1 without antibiotics was performed and cells were maintained in the incubator for an additional 16-18 hours prior to passaging of cells to a larger tissue culture dish. At this time, some cells were kept for Western blotting to monitor the expression of CLPP. The transfection procedure was repeated to maximize the inhibition of CLPP expression.


Clonal populations of MDA-MB-231 CLPP-KD were generated by diluting the transfected cells and plating them in 96-well tissue culture plate at approximately 1 cell per 200 μL of media per well. Successful isolation of single cells was determined by visual examination of the wells, and their expansion into clonal populations was carried out for 1-2 weeks. Individual clonal populations were then transferred into larger tissue culture dishes for continued expansion. The successful knock down (KD) of CLPP was determined by Western blotting.


Western Blotting

The same procedure as detailed under Example 1 for Western blotting was used on MDA-MB-231 WT and CLPP-KD cells. GAPDH is blotted to provide a sample-loading control.


Cytotoxicity Test

Assessment of cytotoxicity of ADEP-14 on human breast cancer cell lines (MDA-MB-231 WT, MDA-MB-231 CLPP-KD, MDA-MB-468, and MCF-7) and HEK cells (HEK293 T-Rex and HEK293 T-REx CLPP) were determined using the methods as described above in Example 1. Briefly, cells were grown for at least 24 hours to allow proper adherence to the growth surface, and then ADEP-14 were serially diluted and introduced to the tissue cultures via fresh growth media. A total of four independent replicates were prepared for each cell line and for each growth condition used. Cells were grown in the presence of ADEP/DMSO for 72 hours. Cytotoxicity test with mitomycin-c, an antitumour antibiotic that inhibits DNA synthesis, was also conducted for comparison. Sulforhodamine B (SRB) staining method was used to quantify cell survival.


Scratch Wound Healing Assay

MDA-MB-231 WT and CLPP-KD cells were grown to near confluence in 6-well tissue culture plates. Prior to scratch wound generation, all cultures were serum-starved by growing in serum-free DMEM:F12 media for 24 hours to inhibit new rounds of cell division, so to ensure that all wound closure events must originate from cell migration alone. Scratch wounds were generated by manually scratching the cell monolayer using a sterile pipette tip in a single, unidirectional stroke while keeping the tilt of the pipette tip constant.


A sterile ruler was used to guide the scratching action to ensure consistency across cultures. The positions of the wounds were marked with a permanent marker on the bottom side of each culture as positional reference for repeated imaging of the same wound areas. Cell debris was removed by gently washing the cultures with fresh serum-free DMEM:F12. The cells were then incubated in fresh serum-free DMEM:F12 containing ADEP-14 at 0, 100 nM, 200 nM, 300 nM, 400 nM, and 500 nM over 48 hours, during which the wounds were imaged using a Nikon TMS inverted microscope equipped with an OMAX A3550S digital camera and the ToupView software (ver. X64, 3.7.13865.20190127; ToupTek). Image analysis and scratch wound quantification were performed using the Bowhead software package (Engel et al., 2018), by measuring the area of scratch wounds (their perimeters outlined) at 0-hr and 48-hr time points for both WT and CLPP-KD, with or without ADEP-14. Normalization of data was performed using the 0-hr scratch wound area as reference. The error bars shown refer to the standard deviation across three independently constructed replicates (see FIG. 16B).


Results and Discussion

ADEP-14 is Less Toxic to Cells with Lower Level of CIpP


The dependency of ADEP-induced cytotoxicity on HsCIpP levels was further determined in breast cancer cell lines. HsCIpP knock down in MDA-MB-231 CLPP-KD cells were confirmed by Western blot analysis (FIG. 15A). MDA-MB-231 CLPP-KD showed 10× lower response to ADEP-14 induced cytotoxicity comparing to MDA-MB-231 WT (FIG. 15B). ADEP-14 was also shown to be cytotoxic to breast cancer cell lines MDA-MB-468 and MCF-7 (Table 6). A summary of ADEP-induced cytotoxicity in various cell lines, including breast cancer cell lines, is shown in Table 6. The cytotoxic effects of ADEP-14 on breast cancer cell lines are comparable to antitumour agent mitomycin-C.









TABLE 6







ADEP-induced cytotoxicity in various cell lines












IC50 for
IC50 for




ADEP-14
Mitomycin-C



Cell Line
(μM)
(μM)







HEK293 T-REx
1.01 ± 0.05
n/d



HEK293 T-REx CLPP−/−
>10*
n/d



MDA-MB-231
1.35 ± 0.O4
0.8 ± 0.2



MDA-MB-231 CLPP-KD
>10*
n/d



MDA-MB-468
0.95 ± 0.06
1.5 ± 0.4



MCF-7
1.6 ± 0.2
0.4 ± 0.2







*IC50 estimated by visual examination, due to lack of minimum plateau in cell viability data that prohibits meaningful data analysis using the Hill model.



KD = knock down



n/d = not determined






ADEP-14 Impairs MDA-MB-231 Cell Migration in 2D Culture

The effects of ADEP on cell migration was determined in a scratch wound healing assay in MDA-MB-231 WT and CLPP-KD cells. As shown in FIG. 16A, the presence of ADEP-14 impaired 2D migration of MDA-MB-231 WT cells. MDA-MB-231 CLPP-KD cells appeared to be less efficient in 2D migration. The assay also showed that migration of MDA-MB-231 CLPP-KD cells were unaffected by the presence of ADEP-14. Normalized quantification of wound closure is shown in FIG. 16B.


Cell migration is involved in cancer metastases. The inhibition of cell migration in MDA-MB-231 WT cells by ADEP-14 show that ADEP analogs are useful for inhibiting, treating or preventing cancer metastasis, including breast cancer metastasis.


While the present disclosure has been described with reference to what are presently considered to be the preferred example, it is to be understood that the disclosure is not limited to the disclosed example. To the contrary, the disclosure is intended to cover various modifications and equivalent arrangements included within the spirit and scope of the appended claims.


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


REFERENCES

Adams, P. D., Afonine, P. V., Bunkoczi, G., Chen, V. B., Davis, I. W., Echols, N., Headd, J. J., Hung, L. W., Kapral, G. J., Grosse-Kunstleve, R. W., et al. (2010). PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta crystallographica Section D, Biological crystallography 66, 213-221.


Alexopoulos, J.A ., Guarne, A., and Ortega, J. (2012). CIpP: a structurally dynamic protease regulated by AAA+ proteins. J Struct Biol 179, 202-210.


Amor, A. J., Schmitz, K. R., Sello, J. K., Baker, T. A., and Sauer, R. T. (2016). Highly Dynamic Interactions Maintain Kinetic Stability of the CIpXP Protease During the ATP-Fueled Mechanical Cycle. ACS Chem Biol 11, 1552-1560.


Barbosa, L. R. S., Spinozzi, F., Mariani, P., and Itri, R. (2013). Small-Angle X-Ray Scattering Applied to Proteins in Solution. In Proteins in Solution and at Interfaces: Methods and Applications in Biotechnology and Materials Science (Wiley), pp. 49-72.


Bewley, M. C., Graziano, V., Griffin, K., and Flanagan, J. M. (2006). The asymmetry in the mature amino-terminus of CIpP facilitates a local symmetry match in CIpAP and CIpXP complexes. Journal of Structural Biology 153, 113-128.


Bram, E., Ifergan, I., Shafran, A., Berman, B., Jansen, G., and Assaraf, Y. G. (2006). Mutant Gly482 and Thr482 ABCG2 mediate high-level resistance to lipophilic antifolates. Cancer Chemother Pharmacol 58, 826-834.


Brotz-Oesterhelt, H., Beyer, D., Kroll, H. P., Endermann, R., Ladel, C., Schroeder, W., Hinzen, B., Raddatz, S., Paulsen, H., Henninger, K., et al. (2005). Dysregulation of bacterial proteolytic machinery by a new class of antibiotics. Nat Med 11, 1082-1087.


Calvo, S. E., Clauser, K. R., and Mootha, V. K. (2016). MitoCarta2.0: an updated inventory of mammalian mitochondrial proteins. Nucleic acids research 44, D1251-1257.


Carney, D. W., Schmitz, K. R., Truong, J. V., Sauer, R. T., and Sello, J. K. (2014). Restriction of the conformational dynamics of the cyclic acyldepsipeptide antibiotics improves their antibacterial activity. J Am Chem Soc 136, 1922-1929.


Cole, A., Wang, Z., Coyaud, E., Voisin, V., Gronda, M., Jitkova, Y., Mattson, R., Hurren, R., Babovic, S., Maclean, N., et al. (2015). Inhibition of the Mitochondrial Protease CIpP as a Therapeutic Strategy for Human Acute Myeloid Leukemia. Cancer Cell 27, 864-876.


Cong, L., Ran, F.A., Cox, D., Lin, S., Barretto, R., Habib, N., Hsu, P. D., Wu, X., Jiang, W., Marraffini, L. A., et al. (2013). Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819-823.


Csizmadia, V., Hales, P., Tsu, C., Ma, J., Chen, J., Shah, P., Fleming, P., Senn, J. J., Kadambi, V. J., Dick, L., et al. (2016). Proteasome inhibitors bortezomib and carfilzomib used for the treatment of multiple myeloma do not inhibit the serine protease HtrA2/Omi. Toxicology Research 5, 1619-1628.


El Bakkouri, M., Pow, A., Mulichak, A., Cheung, K. L., Artz, J. D., Amani, M., Fell, S., de Koning-Ward, T. F., Goodman, C. D., McFadden, G. I., et al. (2010). The CIp chaperones and proteases of the human malaria parasite Plasmodium falciparum. J Mol Biol 404, 456-477.


Elmore, S. (2007). Apoptosis: a review of programmed cell death. Toxicol Pathol 35, 495-516.


Emsley, P., and Cowtan, K. (2004). Coot: model-building tools for molecular graphics. Acta crystallographica Section D, Biological crystallography 60, 2126-2132.


Engel, M., Longden, J., Ferkinghoff-Borg, J., Robin, X., Saginc, G., and Linding, R. (2018). Bowhead: Bayesian modelling of cell velocity during concerted cell migration. PLoS Comput Biol 14, e100590.


Gersch, M., Famulla, K., Dahmen, M., Gobl, C., Malik, I., Richter, K., Korotkov, V. S., Sass, P., Rubsamen-Schaeff, H., Madl, T., et al. (2015). AAA+ chaperones and acyldepsipeptides activate the CIpP protease via conformational control. Nat Commun 6, 6320.


Goodreid, J. D., Janetzko, J., Santa Maria, J. P., Jr., Wong, K. S., Leung, E., Eger, B. T., Bryson, S., Pai, E. F., Gray-Owen, S. D., Walker, S., et al. (2016). Development and Characterization of Potent Cyclic Acyldepsipeptide Analogues with Increased Antimicrobial Activity. J Med Chem 59, 624-646.


Gribun, A., Kimber, M. S., Ching, R., Sprangers, R., Fiebig, K. M., and Houry, W. A. (2005). The CIpP double ring tetradecameric protease exhibits plastic ring-ring interactions, and the N termini of its subunits form flexible loops that are essential for CIpXP and CIpAP complex formation. The Journal of biological chemistry 280, 16185-16196.


Ingvarsson, H., Mate, M. J., Hogbom, M., Portnoi, D., Benaroudj, N., Alzari, P. M., Ortiz-Lombardia, M., and Unge, T. (2007). Insights into the inter-ring plasticity of caseinolytic proteases from the X-ray structure of Mycobacterium tuberculosis CIpP1. Acta crystallographica Section D, Biological crystallography 63, 249-259.


Kang, S. G., Maurizi, M. R., Thompson, M., Mueser, T., and Ahvazi, B. (2004). Crystallography and mutagenesis point to an essential role for the N-terminus of human mitochondrial CIpP. J Struct Biol 148, 338-352.


Kang, S. G., Ortega, J., Singh, S. K., Wang, N., Huang, N. N., Steven, A. C., and Maurizi, M. R. (2002). Functional proteolytic complexes of the human mitochondrial ATP-dependent protease, hCIpXP. The Journal of biological chemistry 277, 21095-21102.


Kim, D. Y., and Kim, K. K. (2008). The structural basis for the activation and peptide recognition of bacterial CIpP. J Mol Biol 379, 760-771.


Kimber, M. S., Yu, A. Y., Borg, M., Leung, E., Chan, H. S., and Houry, W. A. (2010). Structural and theoretical studies indicate that the cylindrical protease CIpP samples extended and compact conformations. Structure 18, 798-808.


Kroemer, G., and Reed, J. C. (2000). Mitochondrial control of cell death. Nat Med 6, 513-519.


Landes, T., and Martinou, J. C. (2011). Mitochondrial outer membrane permeabilization during apoptosis: the role of mitochondrial fission. Biochim Biophys Acta 1813, 540-545.


Lee, B. G., Park, E. Y., Lee, K. E., Jeon, H., Sung, K. H., Paulsen, H., Rubsamen-Schaeff, H., Brotz-Oesterhelt, H., and Song, H. K. (2010). Structures of CIpP in complex with acyldepsipeptide antibiotics reveal its activation mechanism. Nat Struct Mol Biol 17, 471-478.


Lee, C. D., Sun, H. C., Hu, S. M., Chiu, C. F., Homhuan, A., Liang, S. M., Leng, C. H., and Wang, T. F. (2008). An improved SUMO fusion protein system for effective production of native proteins. Protein Sci 17, 1241-1248.


Leung, E., Datti, A., Cossette, M., Goodreid, J., McCaw, S. E., Mah, M., Nakhamchik, A., Ogata, K., El Bakkouri, M., Cheng, Y. Q., et al. (2011). Activators of cylindrical proteases as antimicrobials: identification and development of small molecule activators of CIpP protease. Chem Biol 18, 1167-1178.


Li, D. H., Chung, Y. S., Gloyd, M., Joseph, E., Ghirlando, R., Wright, G. D., Cheng, Y. Q., Maurizi, M. R., Guarne, A., and Ortega, J. (2010). Acyldepsipeptide antibiotics induce the formation of a structured axial channel in CIpP: A model for the CIpX/CIpA-bound state of CIpP. Chem Biol 17, 959-969.


Liu, K., Ologbenla, A., and Houry, W. A. (2014). Dynamics of the CIpP serine protease: a model for self-compartmentalized proteases. Crit Rev Biochem Mol Biol 49, 400-412.


Lo, J. H., Baker, T. A., and Sauer, R. T. (2001). Characterization of the N-terminal repeat domain of Escherichia coli CIpA-A class I Clp/HSP100 ATPase. Protein Science 10, 551-559.


Lowth, B. R., Kirstein-Miles, J., Saiyed, T., Brötz-Oesterhelt, H., Morimoto, R. I., Truscott, K. N., and Dougan, D. A. (2012). Substrate recognition and processing by a Walker B mutant of the human mitochondrial AAA+ protein CLPX. Journal of Structural Biology 179, 193-201.


Mcllwain, D. R., Berger, T., and Mak, T. W. (2013). Caspase functions in cell death and disease. Cold Spring Harb Perspect Biol 5, a008656.


Mylonas, E., and Svergun, D. I. (2007). Accuracy of molecular mass determination of proteins in solution by small-angle X-ray scattering. Journal of Applied Crystallography 40, s245-s249.


Olivares, A. O., Baker, T. A., and Sauer, R. T. (2016). Mechanistic insights into bacterial AAA+ proteases and protein-remodelling machines. Nat Rev Microbiol 14, 33-44.


Otwinowski, Z., and Minor, W. (1997). Processing of X-ray Diffraction Data Collected in Oscillation Mode. In Macromolecular Crystallography, part A, C. W. Carter Jr., and R. M. Sweet, eds. (New York: Academic Press), pp. 307-326.


Petoukhov, M. V., Franke, D., Shkumatov, A. V., Tria, G., Kikhney, A. G., Gajda, M., Gorba, C., Mertens, H. D., Konarev, P. V., and Svergun, D. I. (2012). New developments in the ATSAS program package for small-angle scattering data analysis. J Appl Crystallogr 45, 342-350.


Pettersen, E. F., Goddard, T. D., Huang, C. C., Couch, G. S., Greenblatt, D. M., Meng, E. C., and Ferrin, T. E. (2004). UCSF Chimera—a visualization system for exploratory research and analysis. J Comput Chem 25, 1605-1612.


Schmitz, K. R., Carney, D. W., Sello, J. K., and Sauer, R. T. (2014). Crystal structure of Mycobacterium tuberculosis CIpP1P2 suggests a model for peptidase activation by AAA+ partner binding and substrate delivery. Proceedings of the National Academy of Sciences 111, E4587-E4595.


Selleck, W., and Tan, S. (2008). Recombinant protein complex expression in E. coli. Curr Protoc Protein Sci Chapter 5, Unit 5 21.


Szczepanowska, K., Maiti, P., Kukat, A., Hofsetz, E., Nolte, H., Senft, K., Becker, C., Ruzzenente, B., Hornig-Do, H. T., Wibom, R., et al. (2016). CLPP coordinates mitoribosomal assembly through the regulation of ERAL1 levels. EMBO J 35, 2566-2583.


Wojtyra, U. A., Thibault, G., Tuite, A., and Houry, W. A. (2003). The N-terminal zinc binding domain of CIpX is a dimerization domain that modulates the chaperone function. The Journal of biological chemistry 278, 48981-48990.


Ye, F., Zhang, J., Liu, H., Hilgenfeld, R., Zhang, R., Kong, X., Li, L., Lu, J., Zhang, X., Li, D., et al. (2013). Helix unfolding/refolding characterizes the functional dynamics of Staphylococcus aureus CIp protease. The Journal of biological chemistry 288, 17643-17653.


Zeiler, E., List, A., Alte, F., Gersch, M., Wachtel, R., Poreba, M., Drag, M., Groll, M., and Sieber, S. A. (2013). Structural and functional insights into caseinolytic proteases reveal an unprecedented regulation principle of their catalytic triad. Proceedings of the National Academy of Sciences of the United States of America 110, 11302-11307.


Zhang, J., Ye, F., Lan, L., Jiang, H., Luo, C., and Yang, C. G. (2011). Structural switching of Staphylococcus aureus CIp protease: a key to understanding protease dynamics. The Journal of biological chemistry 286, 37590-37601.

Claims
  • 1. A method for treating a subject having cancer, comprising administering a therapeutically effective amount of an acyldepsipeptide (ADEP) analog to the subject in need thereof.
  • 2. The method of claim 1 , wherein the ADEP analog is ADEP-01, ADEP-02, ADEP-03, ADEP-04, ADEP-05, ADEP-06, ADEP-07, ADEP-08, ADEP-09, ADEP-10, ADEP-11, ADEP-12, ADEP-13, ADEP-14, ADEP-15, ADEP-16, ADEP-17, ADEP-18, ADEP-19, ADEP-20, ADEP-21, ADEP-22, ADEP-23, ADEP-24, ADEP-25, ADEP-26, ADEP-27, ADEP-28, ADEP-29, ADEP-30, ADEP-31, ADEP-32, ADEP-33, ADEP-34, ADEP-35, ADEP-36, ADEP-37, ADEP-38, ADEP-39, ADEP-40, ADEP-41, ADEP-42, ADEP-43, ADEP-44, ADEP-45, or ADEP-46.
  • 3. The method of claim 1, wherein the ADEP analog is ADEP-01 , ADEP-02, ADEP-04, ADEP-06, ADEP-10, ADEP-13, ADEP-14, ADEP-15, ADEP-17, ADEP-20, ADEP-25, ADEP-28, ADEP-29, ADEP-30, ADEP-32, ADEP-37, ADEP-38, or ADEP-41.
  • 4. The method of claim 1, wherein the ADEP analog is ADEP-14, ADEP-17, ADEP-28, ADEP-29, ADEP-30, ADEP-32, ADEP-37, or ADEP-38.
  • 5. The method of claim 1, wherein the ADEP analog activates protease activity of human mitochondrial CIpP (HsCIpP), and wherein the protease activity is at least 0.87 as measured by the relative degradation (RD) index at 25 μM of the ADEP analog, optionally at least 0.9, optionally at least 0.95.
  • 6. The method of claim 1, wherein the protease activity is at least 0.74 as measured by the RD index at 5 μM of the ADEP analog, optionally at least 0.8, optionally at least 0.85.
  • 7. The method of claim 1, wherein the protease activity is at least 0.2 as measured by the RD index at 1 μM of the ADEP analog, optionally at 0.5, optionally at least 0.75, optionally at least 0.8.
  • 8. The method of claim 1, wherein the cancer is breast cancer, prostate cancer, colon cancer, liver cancer, uterus cancer, thyroid cancer, lung cancer, lymph nodes cancer, bladder cancer, ovary cancer, stomach cancer, testis cancer, brain cancer, skin cancer, pancreatic cancer, acute myeloid leukemia, kidney cancer, cervical cancer, osteosarcoma, or neuroblastoma.
  • 9. The method of claim 1, wherein the cancer is breast cancer, kidney cancer, cervical cancer, osteosarcoma, or neuroblastoma.
  • 10. The method of claim 1, wherein the cancer is metastatic.
  • 11. The method of claim 10, wherein the metastatic cancer is breast cancer.
  • 12. The method of claim 1, wherein the subject is human.
  • 13. The method of claim 1, wherein the ADEP analog is administered subcutaneously, intraperitoneally, intravenously, topically, or orally.
  • 14. A compound for use in treating a subject having a cancer, wherein the compound is a therapeutically effective amount of an acyldepsipeptide (ADEP) analog.
  • 15. The compound of claim 14, wherein the ADEP analog is ADEP-01, ADEP-02, ADEP-03, ADEP-04, ADEP-05, ADEP-06, ADEP-07, ADEP-08, ADEP-09, ADEP-10, ADEP-11, ADEP-12, ADEP-13, ADEP-14, ADEP-15, ADEP-16, ADEP-17, ADEP-18, ADEP-19, ADEP-20, ADE P-21, ADEP-22, ADEP-23, ADEP-24, ADEP-25, ADEP-26, ADEP-27, ADEP-28, ADEP-29, ADEP-30, ADEP-31, ADEP-32, ADEP-33, ADEP-34, ADEP-35, ADEP-36, ADEP-37, ADEP-38, ADEP-39, ADEP-40, ADEP-41, ADEP-42, ADEP-43, ADEP-44, ADEP-45, or ADEP-46.
  • 16. The compound of claim 14, wherein the ADEP analog is ADEP-01, ADEP-02, ADEP-04, ADEP-06, ADEP-10, ADEP-13, ADEP-14, ADEP-15, ADEP-17, ADEP-20, ADEP-25, ADEP-28, ADEP-29, ADEP-30, ADEP-32, ADEP-37, ADEP-38, or ADEP-41.
  • 17. The compound of claim 14, wherein the ADEP analog is ADEP-14, ADEP-17, ADEP-28, ADEP-29, ADEP-30, ADEP-32, ADEP-37, or ADEP-38.
  • 18. The compound of claim 14, wherein the ADEP analog activates protease activity of human mitochondrial CIpP (HsCIpP), and wherein the protease activity is at least 0.87 as measured by the relative degradation (RD) index at 25 μM of the ADEP analog, optionally at least 0.9, optionally at least 0.95.
  • 19. The compound of claim 14, wherein the protease activity is at least 0.74 as measured by the RD index at 5 μM of the ADEP analog, optionally at least 0.8, optionally at least 0.85.
  • 20. The compound of claim 14, wherein the protease activity is at least 0.2 as measured by the RD index at 1 μM of the ADEP analog, optionally at 0.5, optionally at least 0.75, optionally at least 0.8.
  • 21. The compound of claim 14, wherein the cancer is breast cancer, prostate cancer, colon cancer, liver cancer, uterus cancer, thyroid cancer, lung cancer, lymph nodes cancer, bladder cancer, ovary cancer, stomach cancer, testis cancer, brain cancer, skin cancer, pancreatic cancer, acute myeloid leukemia, kidney cancer, cervical cancer, osteosarcoma, or neuroblastoma.
  • 22. The compound of claim 14, wherein the cancer is metastatic.
  • 23-26. (canceled)
  • 27. An acyldepsipeptide (ADEP) analog, wherein the ADEP analog is ADEP-01, ADEP-02, ADEP-03, ADEP-04, ADEP-05, ADEP-06, ADEP-07, ADEP-08, ADEP-09, ADEP-10, ADEP-11, ADEP-12, ADEP-13, ADEP-14, ADEP-15, ADEP-16, ADEP-17, ADEP-18, ADEP-19, ADEP-20, ADEP-21, ADEP-22, ADEP-23, ADEP-24, ADEP-25, ADEP-26, ADEP-27, ADEP-28, ADEP-29, ADEP-30, ADEP-31, ADEP-32, ADEP-33, ADEP-34, ADEP-35, ADEP-36, ADEP-37, ADEP-38, ADEP-39, ADEP-40, ADEP-41, ADEP-42, ADEP-43, ADEP-44, ADEP-45, or ADEP-46, or a variant or a derivative thereof.
RELATED APPLICATIONS

This application claims priority to U.S. Provisional Patent Applications Nos. 62/679,448 filed on Jun. 1, 2018, and 62/680,302 filed on Jun. 4, 2018, the contents of which are hereby incorporated by reference in their entirety.

PCT Information
Filing Document Filing Date Country Kind
PCT/CA2019/050771 6/3/2019 WO 00
Provisional Applications (2)
Number Date Country
62679448 Jun 2018 US
62680302 Jun 2018 US