The invention, in some aspects relates to compositions and methods for preparing and using signaling reporter islands for spatially multiplexed imaging in cells and subjects.
Biological signals can contain dozens to hundreds of different biomolecular building blocks, which interact in complex ways within cells. For example, intracellular Ca2+ dynamics, cyclic AMP (cAMP) levels, and protein kinase activities can all interact within single cells—with consequences for the proliferation and differentiation of cancer cells (Cho-Chung, 1990) and for plasticity and learning in the nervous system (Frey, Huang and Kandel, 1993; Trudeau, Emery and Haydon, 1996; Averaimo and Nicol, 2014), amongst many key biological phenomena. Accordingly, there has been a push by many biologists to create genetically encoded fluorescent indicators of each of these biological signals (DiPilato and Zhang, 2009; Depry, Allen and Zhang, 2011; Chen et al., 2013; Hackley, Mazzoni and Blau, 2018), as well as of many other biological signals (Violin et al., 2003; Belousov et al., 2006; Berg, Hung and Yellen, 2009; Vinkenborg et al., 2009; Oliveira and Yasuda, 2013; Shimozono et al., 2013; Ding et al., 2015, and US Patent Publication US2016/0305939). Because such biomolecular building blocks can exist in different quantities and in different functional states in different cells, but prior methods do not permit simultaneously imaging of multiple biological signals at the same time in individual cells, and so are not adequate for assessing multiple signals in a single physiological cascade (Mehta et al., 2018). On a conventional fluorescence microscope, reporters with different fluorescent spectra can be used simultaneously, but not reporters based on the same fluorophore, because the signals of the same fluorophores mix and result in ambiguous data. Thus the number of reporters used is limited to the number of spectrally resolvable channels on the microscope.
According to one aspect of the invention, a composition for RNA-protein-binding-based (RPB) clustering of independently selected reporter molecules in a cell is provided, the composition including: (a) a fusion protein component that includes: (i) one or more independently selected reporter protein elements, (ii) one or more independently selected RNA binding protein elements, (iii) zero, one, or more independently selected epitope tag elements, (iv) zero, one, or more independently selected localization protein motif elements, (v) zero, one, or more independently selected protein linker elements, wherein when present, each of the protein linker elements is positioned between two of the elements of (i), (ii), (iii), and (iv); and (b) an RNA component that includes: (v) a plurality of independently selected RNA molecules including a plurality of independently selected binding sequence elements recognized by the one or more RNA binding protein elements of (a)(ii). In some embodiments, the plurality of binding sequence elements in (b)(vi) includes ten or more binding sequences. In some embodiments, the RNA binding protein elements include one or more of a non-programmable RNA binding element. In certain embodiments, the non-programmable RNA binding elements include one or more of: an MS2 coat protein, a PP7 coat protein, a Lambda N protein, a Q-beta coat protein, a BglG protein, a U1Ap protein, HTLV-1 Rex protein, a TAT protein, an REV protein, and an eiF4A protein. In some embodiments, the RNA binding protein elements include one or more of a programmable RNA binding element. In some embodiments, the programmable RNA binding elements include one or more of a Pumilio homology domain (PumHD) and a Pumilio-based assembly (Pumby). In certain embodiments, when present, the epitope tag elements comprise one or more of an Xpress tag, an HA tag, and a V5 tag, an OLLAS tag, a VSVg tag, an S1 (Strep I) tag, an NWS (Strep II) tag, an E epitope (E tag), a FLAG tag, an E2 tag, an AU1 tag, an AU5 tag, an Myc tag, a Spot tag, a NE tag, an AviTag, a C-tag, a Calmodulin-tag, a polyglutamate tag, an Rho1D4-tag, an S-tag, an SBP-tag, a Softag 1, a Softag 3, a TC tag, and a Ty tag. In some embodiments, the localization protein motif element includes one or more of a subcellular localization protein and a trafficking protein. In some embodiments, the localization protein motif element includes one or more of a nucleus localization motif, a plasma membrane localization motif, and a synapse localization motif. In certain embodiments, when present the protein linker element(s) include one or more glycine-rich linkers. In some embodiments, the glycine-rich linkers include one or more of: a GSG linker, a (GGSGGT)×2 linker, a (GGSGGT)×4 linker, a (GGSGGSGGT)×3 linker, a (GGGS)×2 linker, and a (GGGS)×4 linker. In certain embodiments, when present the protein linker elements include one or more non-glycine-rich linkers. In some embodiments, the non-glycine-rich linkers include one of (EAAAK)×2 and APAPAP. In some embodiments, when two or more protein linker elements are present they include at least one each of a glycine rich linker and a non-glycine rich linker. In certain embodiments, the binding sequence elements include one or more of: an MS2 binding sequence, a PP7 binding sequence, and a lambda N binding sequence (BoxB), a Q-beta binding sequence, a BglG binding sequence, an UiAp aptamer sequence, an HTLV-1 Rex responsive element (RxRE), a TAR sequence, an RRE sequence, and an eiF4A aptamer sequence. In some embodiments, the binding sequence elements include one or more of a sequence that recognizes and selectively binds the programmable binding protein. In some embodiments, the independently selected reporter protein element is a voltage indicator polypeptide, a calcium indicator polypeptide, a potassium indicator polypeptide, a pH indicator polypeptide, or a magnesium indicator polypeptide. In some embodiments, the independently selected reporter protein element is GCaMP6, GCaMP6f, GCaMP6m, GCaMP6s, jGCaMP7, jGCaMP7f, jGCaMP7m, jGCaMP7s, jGCaMP7b, jGCaMP7c, GCaMP-X, jRGECO1, jRCaMP1, NIRGECO, BCaMP, ICUE, ICUE3, cAMPr, Epac-based cAMP indicator, AKAR, AKAR4, ExRai-AKAR, ExRai-AKAR2, CKAR, ExRai-CKAR, EKARev, or RAB-EKARev. In certain embodiments, the independently selected reporter protein element is a GCaMP6f polypeptide or an ICUE3 polypeptide.
According to another aspect of the invention a cell that includes any embodiment of an aforementioned composition.
According to another aspect of the invention, a cell that includes two or more of any embodiments of an aforementioned composition are provided and one or more of the elements of (i)-(iv) and the RNA binding sequence elements are different in each of the two or more of the compositions.
According to another aspect of the invention a vector that encodes a fusion protein of any embodiment of any aforementioned composition of the invention is provided. In some aspects of the invention a cell that includes any embodiment of any aforementioned vector of the invention is provided.
According to another aspect of the invention, a composition for protein self-assembly (PSA) based clustering of independently selected reporter molecules in a cell is provided, the composition included a fusion protein that includes: (a) one or more independently selected reporter protein elements, (b) one or more independently selected self-assembly protein elements, (c) zero, one, or more independently selected epitope tag elements, (d) zero, one, or more localization protein motif elements, and (e) zero, one, or more independently selected protein linker elements, wherein when present, each of the protein linker elements is positioned between two of the elements of (a), (b), (c), and (d). In certain embodiments, the self-assembly protein elements include one or more of a polyhedron-forming protein, a coiled-coil forming protein, a supramolecular self-assembly protein, and a protein oligomer. In some embodiments, the polyhedron-forming protein is I3-01, O3-33, ATC-HL3, or 3VDX. In some embodiments, the coiled-coil forming protein is HexCoil-Ala, 5H2L_2, EE, or RR. In some embodiments, the supramolecular self-assembly protein is 2AN9 or 1M3U. In certain embodiments, the protein oligomer is 5L6HC3_1 or 2L8HC4_15. In some embodiments, when present, the epitope tag elements includes one or more of an Xpress tag, an HA tag, and a V5 tag, an OLLAS tag, a VSVg tag, an S1 (Strep I) tag, an NWS (Strep II) tag, an E epitope (E tag), a FLAG tag, an E2 tag, an AU1 tag, an AU5 tag, an Myc tag, a Spot tag, a NE tag, an AviTag, a C-tag, a Calmodulin-tag, a polyglutamate tag, an Rho1D4-tag, an S-tag, an SBP-tag, a Softag 1, a Softag 3, a TC tag, and a Ty tag. In certain embodiments, the localization protein motif element includes one or more of a subcellular localization protein and a trafficking protein. In some embodiments, the localization protein motif element includes one or more of a nucleus localization motif, a plasma membrane localization motif, and a synapse localization motif. In some embodiments, when present the protein linker elements include one or more glycine-rich linkers. In some embodiments, the glycine-rich linkers include one or more of: a GSG linker, a (GGSGGT)×2 linker, a (GGSGGT)×4 linker, a (GGSGGSGGT)×3 linker, a (GGGS)×2 linker, and a (GGGS)×4 linker. In certain embodiments, when present the protein linker elements includes one or more non-glycine-rich linkers. In some embodiments, the non-glycine-rich linkers include one of (EAAAK)×2 and APAPAP. In some embodiments, when two or more protein linker elements are present they include at least one each of a glycine rich linker and a non-glycine rich linker. In certain embodiments, the independently selected reporter protein element is a voltage indicator polypeptide, a calcium indicator polypeptide, a potassium indicator polypeptide, a pH indicator polypeptide, or a magnesium indicator polypeptide. In some embodiments, the independently selected reporter protein element is GCaMP6, GCaMP6f, GCaMP6m, GCaMP6s, jGCaMP7, jGCaMP7f, jGCaMP7m, jGCaMP7s, jGCaMP7b, jGCaMP7c, GCaMP-X, jRGECO1, jRCaMP1, NIRGECO, BCaMP, ICUE, ICUE3, cAMPr, Epac-based cAMP indicator, AKAR, AKAR4, ExRai-AKAR, ExRai-AKAR2, CKAR, ExRai-CKAR, EKARev, or RAB-EKARev. In certain embodiments, the independently selected reporter protein element is a GCaMP6f polypeptide or an ICUE3 polypeptide.
According to another aspect of the invention a cell that includes any embodiment of an aforementioned composition.
According to another aspect of the invention, a cell that includes two or more of any embodiments of an aforementioned composition are provided and one or more of the elements of (a)-(e) are different in each of the two or more of the compositions.
According to another aspect of the invention a vector that encodes a fusion protein of any embodiment of any aforementioned composition of the invention is provided. In some aspects of the invention a cell that includes any embodiment of any aforementioned vector of the invention is provided.
According to another aspect of the invention a cell is provided that includes one or more of an embodiment of a composition for RNA-protein-binding-based (RPB) clustering of independently selected reporter molecules in a cell and one or more of an embodiment of a composition for protein self-assembly (PSA) based clustering of independently selected reporter molecules in a cell.
According to another aspect of the invention, a method of determining a physiological process in a cell is provided, the method including (a) expressing in a cell a plurality of each of two or more independently selected reporter protein elements each capable of generating an identifiable signal; (b) forming one or more clusters from the plurality of the two or more independently selected reporter protein elements in the cell, wherein each of the two or more independently selected reporter protein elements are in different clusters; (c) positioning the one or more formed clusters in the cell such that a distance between a cluster formed with one of the independently selected reporter protein elements and a cluster formed with another of the independently selected reporter protein elements is sufficient to (i) resolve the signal generated by the two independently selected reporter protein elements in the positioned clusters and (ii) simultaneously determine the signal generated by each of the independently selected reporter protein elements in the positioned clusters; (d) detecting the identifiable signals generated from the independently selected reporter protein elements in the positioned clusters; and (e) analyzing the detected signals to determine one or more physiological processes of the cell. In some embodiments, the signals generated by the two or more independently selected reporter protein elements include fluorescent signals. In certain embodiments, at least two of the fluorescent reporter protein elements generate spectrally overlapping signals. In some embodiments, the analyzing includes assessing two or more different physiological processes in the cell. In some embodiments, the analyzing includes assessing two or more simultaneous physiological processes in the cell. In some embodiments, the physiological process includes one or more of: pH of the cell, voltage in the cell, and the presence of one or more of: calcium, magnesium, chloride, and potassium in the cell. In certain embodiments, the method also includes externally stimulating the cell that includes the positioned clusters and determining the signals generated by the independently selected reporter protein elements in the positioned clusters. In some embodiments, the externally stimulating includes contacting the cell with one or more of: forskolin, Tetradecanoylphorbol acetate (PMA), (S)-3,5-Dihydroxyphenylglycine (DHPG), N-Methyl-D-aspartic acid (NMDA), a cancer therapeutic agent, an antibody or active fragment thereof, a toxin, an agonist of a receptor, an antagonist of a receptor, an electrical field, a magnetic field, light, gas, a temperature change, a gravity change, a pH change, a whole-cell patch clamp. In some embodiments, the method does not include externally stimulating the cell that includes the positioned clusters and determining the signals generated by the independently selected reporter protein elements in the positioned clusters. In some embodiments, the physiological process includes one or more of: a function of the cell, a response of the cell, ion flux in the cell, a therapeutic response of the cell; and an activation of the cell. In certain embodiments, the method also includes (f) repeating steps (d)-(e) for two or more different clusters formed from the two or more independently selected reporter protein elements each capable of generating an identifiable signal and positioned as in step (c). In some embodiments, the distance between the cluster formed with one of the independently selected reporter protein elements and the cluster formed with the other of the independently selected reporter protein elements is about 1-2 microns, 1-3 microns, 1-4 microns, 1-5 microns, 1-6 microns, 1-7 microns, or 1-8 microns. In some embodiments, the distance between the cluster formed with one of the independently selected reporter protein elements and the cluster formed with the other of the independently selected reporter protein elements is 2-8 microns. In certain embodiments, the signals generated by the independently selected reporter protein elements are detected with one or more of: video microscopy, computerized microscopic imaging, fluorescence microscopy, and confocal microscopy, light microscopy, light microscopy, light sheet microscopy, light field microscopy, and endoscopy. In some embodiments, the analyzing includes one or more of: live cell imaging, immunostaining, RNA FISH, live cell recording, an immunostaining method, an in situ hybridization method, determining fluorescence intensity, subtracting background fluorescence, a cluster/puncta localization means, an expansion microscopy means, a fluorescence microscopy means, a light microscopy means, and a fluorescence-lifetime imaging microscopy means. In certain embodiments, the cell is in, or has been obtained from, a cell culture, tissue culture, or a subject. In some embodiments, the cell is a vertebrate cell. In some embodiments, the cell is a human cell. In certain embodiments, the cell also includes two or more of any embodiment of an aforementioned composition for RNA-protein-binding-based (RPB) clustering of independently selected reporter molecules and one or more of the elements (i)-(v) and the RNA binding sequences are different in each of the two or more of the composition. In some embodiments, the two or more of the composition each form and position the clusters of different independently selected reporter protein elements. In some embodiments, the cell also includes two or more of any embodiment of an aforementioned composition for protein self-assembly (PSA) based clustering of independently selected reporter molecules and one or more of the elements of (a)-(e) are different in each of the two or more of the composition. In certain embodiments, the two or more of the composition each form and position the clusters of different independently selected reporter protein elements. In some embodiments, the cell includes one or more of one or more of any embodiment of an aforementioned composition for RNA-protein-binding-based (RPB) clustering of independently selected reporter molecules in a cell and one or more of any embodiment of an aforementioned composition for protein self-assembly (PSA) based clustering of independently selected reporter molecules in a cell.
According to another aspect of the invention, a method of identifying an effect of a candidate agent on a physiological process in a cell is provided, the method including (a) preparing a cell that includes one or more of any embodiment of an aforementioned composition for RNA-protein-binding-based (RPB) clustering of independently selected reporter molecules in a cell, wherein the prepared cell includes formed and positioned clusters of the independently selected reporter protein element; (b) contacting the prepared cell with a candidate agent; (c) detecting the identifiable signals generated from the independently selected reporter protein elements in the positioned clusters; (d) analyzing the detected signals to determine one or more physiological processes of the contacted cell; and (e) comparing the determined physiological process of the contacted cell with the determined physiological process of a control cell, wherein a difference identifies an effect of the candidate agent on a physiological process in the contacted cell. In some embodiments, the control cell is a cell that includes the composition but is not contacted with the candidate agent. In certain embodiments, the cell includes two or more of the compositions and at least one of the elements (i)-(v) and the RNA binding sequences are different in each of the two or more of the compositions. In some embodiments, the two or more of the composition each form and position the clusters of different independently selected reporter protein elements. In certain embodiments, the candidate agent is one or more of a pharmaceutical agent, an electrical agent, a magnetic agent, a temperature change agent, an environmental agent, and a therapeutic agent.
According to another aspect of the invention, a method of identifying an effect of a candidate agent on a physiological process in a cell is provided, the method including: (a) preparing a cell that includes one or more of any embodiment of an aforementioned composition for protein self-assembly (PSA) based clustering of independently selected reporter molecules, wherein the prepared cell includes formed and positioned clusters of the independently selected reporter protein element; (b) contacting the prepared cell with a candidate agent; (c) detecting the identifiable signals generated from the independently selected reporter protein elements in the positioned clusters; (d) analyzing the detected signals to determine one or more physiological processes of the contacted cell; and (e) comparing the determined physiological process of the contacted cell with the determined physiological process of a control cell, wherein a difference identifies an effect of the candidate agent on a physiological process in the contacted cell. In some embodiments, the control cell is a cell including the composition but not contacted with the candidate agent. In certain embodiments, the cell includes two or more of the compositions and one or more of the elements of (a)-(e) are different in each of the two or more of the compositions. In some embodiments, the two or more of the composition each form and position the clusters of different independently selected reporter protein elements. In some embodiments, the candidate agent is one or more of a pharmaceutical agent, an electrical agent, a magnetic agent, a temperature change agent, an environmental agent, and a therapeutic agent.
According to another aspect of the invention, a method of identifying an effect of a candidate agent on a physiological process in a cell is provided, the method including (a) preparing a cell that includes one or more of any embodiment of an aforementioned composition for RNA-protein-binding-based (RPB) clustering of independently selected reporter molecules and one or more of any embodiment of an aforementioned composition for protein self-assembly (PSA) based clustering of independently selected reporter molecules, and the prepared cell includes formed and positioned clusters of the independently selected reporter protein elements; (b) contacting the prepared cell with a candidate agent; (c) detecting the identifiable signals generated from the independently selected reporter protein elements in the positioned clusters; (d) analyzing the detected signals to determine one or more physiological processes of the contacted cell; and (e) comparing the determined physiological process of the contacted cell with the determined physiological process of a control cell, wherein a difference identifies an effect of the candidate agent on a physiological process in the contacted cell. In certain embodiments, the control cell is a cell includes the two or more compositions but is not contacted with the candidate agent. In some embodiments, the cell includes two or more of any embodiment of an aforementioned composition for RNA-protein-binding-based (RPB) clustering of independently selected reporter molecules and one or more of the elements of (i)-(vi) are different in each of the two or more of the composition. In some embodiments, the two or more of the composition each form and position the clusters of different independently selected reporter protein elements. In certain embodiments the cell includes two or more of any embodiment of an aforementioned composition for protein self-assembly (PSA) based clustering of independently selected reporter molecules, and one or more of the elements of (a)-(e) are different in each of the two or more of the composition. In some embodiments, the two or more of the composition each form and position the clusters of different independently selected reporter protein elements. In certain embodiments, the candidate agent is one or more of a pharmaceutical agent, an electrical agent, a magnetic agent, a temperature change agent, an environmental agent, and a therapeutic agent. In some embodiments, the cell includes two or more of any embodiment of an aforementioned composition for RNA-protein-binding-based (RPB) clustering of independently selected reporter molecules and one or more of the elements of (i)-(vi) are different in each of the two or more of the composition and the cell also includes two or more of any embodiment of an aforementioned composition for protein self-assembly (PSA) based clustering of independently selected reporter molecules, and one or more of the elements of (a)-(e) are different in each of the two or more of the composition. In some embodiments, the two or more of the composition each form and position the clusters of different independently selected reporter protein elements. In certain embodiments, the candidate agent is one or more of a pharmaceutical agent, an electrical agent, a magnetic agent, a temperature change agent, an environmental agent, and a therapeutic agent.
The invention in some aspects relates to novel methods of imaging that permit imaging of the physiology and activity within single cells with subcellular, nanoscale precision. The methods, in some aspects include clustering and stochastically distributing two or more compositions comprising one or more independently selected reporter protein elements inside a cell to be imaged. In certain embodiments, the composition is an RNA-protein-based clustering composition and in some embodiments, the composition is a protein self-assembly clustering composition. Certain embodiments of the invention may include one or more of each of an RNA-protein-based clustering composition and a protein self-assembly clustering composition.
Embodiments of methods and compositions of the invention can be used to prepare and use cluster sensors in signaling reporter islands (SiRIs). As used herein the term “SiRIs” means the engineered RNA and protein scaffolds that comprise clusters of sensors. The term “sensor” as used herein is a composition or molecule capable of producing a signal in response to a condition. Non-limiting examples of sensors are reporter protein elements such as those described elsewhere herein. Embodiments of SiRIs of the invention can be delivered into a cell and one or more of a condition, activity, change, in a physiological characteristic of the cell can be determined and assessed based on the signal produced by one or more sensors in the SiRIs. A general schematic of an embodiment of the invention is shown in
Different strategies can be used to prepare SiRIs of the invention. For example, one strategy includes scaffolding of different sensors onto distinct RNA strands (
It has now been identified that it is possible to monitor different signals, non-limiting examples of which are Ca2+ and cAMP signals, from spectrally overlapping or even spectrally identical fluorophores, in single cells and in single neurons in culture. The ability to use simultaneous measurement of these signals in individual cells has revealed very complex relationships between these signals, including variations in the timing, amplitude, and shape of the signals in relation to each other. These results demonstrate the value and importance of performing multiple measurements at the same time in individual cells, because prior methods that included measuring the signals in separate cells and then averaging the data across populations before comparing them to each other resulted in loss of these relationships. The ability to simultaneously detect and assess signals from two or more spectrally overlapping or even spectrally identical fluorophores in single cells, which supports the broad utility of compositions and methods of the invention in numerous scientific areas.
Unlike prior imaging methods, some of which permitted imaging of clusters of reporter polypeptides, which were capable of detecting and comparing simultaneous physiological processes only in different cells, methods and compositions of the invention described herein can be used to image clusters of different compositions in a single cell and thus can be used to acquire information about multiple simultaneous physiological processes occurring within a single cell. Non-limiting examples of physiological processes that can be assessed using embodiments of compositions and methods of the invention are one or more of: pH of the cell, voltage in the cell, and the presence of one or more of: calcium, magnesium, chloride, and potassium in the cell.
Compositions and methods of the invention can be used to localize, via engineered RNA and protein scaffolds that are bioorthogonal to mammalian cells, spectrally similar or even spectrally identical fluorescent reporters of different biological signals at different points in space, which are referred to herein as signaling reporter islands (SiRIs). Non-limiting examples of the use of compositions and methods of the invention include live imaging of the cell comprising SiRIs, post hoc reconstruction of sensor identity in fixed cells comprising SiRIs via epitope immunostaining, and RNA FISH, or other highly multiplexed fixed cell imaging methods. Using compositions and methods of the invention it is possible to record multiple fluorescent signals at different points in space within a cell, with minimal crosstalk, in a fashion where the identity of the signal can be clearly defined. In short, SiRIs allow the high multiplexing capacity of fixed cell imaging to be translated to help the live cell imaging case, using the spatial dimension as an asset.
SiRIs of the invention have been found to be stationary over timescales appropriate for live cell imaging. Methods of using two or more different scaffolds of the invention with different kinds of spacing can permit a balance between the number of signals that can be simultaneously observed, and the spatial sampling that is permitted. As set forth elsewhere herein, multiple different candidate scaffolds have been prepare and used, each showing clustering. Sequences included in embodiments of modular SiRIs of the invention are shown in Table 1. Table 2 provides details of components included in RNA-based and protein-based scaffolds that have been prepared. Sequences of SiRI components listed in Table 2 are available in Table 1 and also from Addgene (Watertown, Mass.; addgene.org); GenBank (NCBI, Bethesda, Md.; ncbi.nlm.nih.gov/genbank); and FPbase (Lambert, T J (2019) Nature Methods. 16, 277-278; fpbase.org). In Tables 1 and 2 UBC is UBC promoter “human ubiquitin C promoter”; CAG is CAG promoter “CMV early enhancer/chicken β actin promoter”; PBS is “PP7 binding sequence”, and PBS1 and PBS2 are functionally identical; MBS is “MS2 binding sequence”; KGC is “golgi export trafficking signal KGC”; SYN is “human synapsin I promoter”; and WPRE is “WHP Posttranscriptional Response Element”.
underlined; non-coding
UBC-NLS-HA-tdMCP-
CAG-miRFP-ActbUTR-
48xMBS
UBC-NLS-V5-tdPCP-Linker24-
CAG-miRFP-ActbUTR-
72xPBS
UBC-NLS-HA-tdPCP-
CAG-miRFP-ActbUTR-
72xPBS
UBC-NLS-HA-tdMCP-
CAG-miRFP-ActbUTR-
48xMBS
UBC-NLS-HA-tdPCP-
CAG-miRFP-ActbUTR-
72xPBS
UBC-HexCoil_Ala-Linker48-
UBC-1M3U-Linker48-cAMPr-
UBC-3VDX-Linker50-
UBC-I32_06B-Linker29-
UBC-1M3U-Linker48-
UBC-3VDX-Linker50-
UBC-O3_33-Linker48-HA-
UBC-HexCoil_Ala-Linker12-
UBC-HexCoil_Ala-Linker48-
UBC-HexCoil_Ala-Linker12-
Syn-I3_01-Linker12-Xpress-
Syn-HexCoil_Ala-Linker48-
UBC-O3_33-Linker48-cAMPr-
UBC-O3_33-Linker48-cAMPr-
UBC-ATC_HL3-Linker48-
UBC-T3_10-Linker48-cAMPr-
UBC-3VDX-Linker48-cAMPr-
UBC-ATC_HL3-Linker48-
UBC-T32_28-Linker48-cAMPr-
UBC-p3Z_11-Linker24-Xpress-
CAG-Xpress-GCaMP6f-
CAG-Xpress-GCaMP6f-
CAG-Xpress-GCaMP6f-
CAG-Xpress-GCaMP6f-
CAG-Xpress-GCaMP6f-
CAG-Xpress-GCaMP6f-
CAG-Xpress-GCaMP6f-
CAG-Xpress-GCaMP6f-
CAG-Xpress-GCaMP6f-
CAG-Xpress-GCaMP6f-
CAG-Xpress-GCaMP6f-
CAG-Xpress-GCaMP6f-
CAG-Xpress-GCaMP6f-
UBC-Xpress-GCaMP6f-
UBC-Xpress-GCaMP6f-
UBC-V5-Linker3-ICUE3-
UBC-V5-Linker3-ICUE3-
UBC-Xpress-GCaMP6f-
UBC-Xpress-GCaMP6f-
UBC-Xpress-GCaMP6f-
UBC-Xpress-GCaMP6f-
CAG-2AN9 mutantl-Linker12-
UBC-Xpress-GCaMP6f-
UBC-1FRW-Linker12-Xpress-
UBC-1L6W-Linker12-Xpress-
UBC-1YAC-Linker12-Xpress-
UBC-2AN9-Linker12-Xpress-
UBC-2AN9_mutant1-Linker12-
UBC-2AN9_mutant2-Linker12-
UBC-2IV1-Linker12-Xpress-
UBC-1M3U-Linker12-Xpress-
UBC-Xpress-GCaMP6f-
UBC-NLS-Xpress-GCaMP6f-
UBC-NLS-1FRW-Linker12-
UBC-NLS-1L6W-Linker12-
UBC-NLS-1YAC-Linker12-
UBC-NLS-2AN9-Linker12-
UBC-NLS-2AN9_mutant1-
UBC-NLS-2AN9_mutant2-
UBC-NLS-2IV1-Linker12-
UBC-NLS-1M3U-Linker12-
UBC-NLS-Xpress-GCaMP6f-
UBC-LNP-HA-NLS-Linker24-
CAG-miRFP-ActbUTR-
75xBoxB
UBC-LNP-FLAG-NLS
CAG-miRFP-ActbUTR-
75xBoxB
UBC-LNP-HA-NLS-Linker24-
CAG-miRFP-ActbUTR-
75xBoxB
UBC-NLS-HA-tdPCP-
UBC-miRFP-ActbUTR-72xPBS
UBC-NLS-Xpress-GCaMP6f-
UBC-miRFP-ActbUTR-72xPBS
UBC-Xpress-GCaMP6f-
UBC-miRFP-ActbUTR-72xPBS
UBC-3VDX-Linker50-
Certain embodiments of spatial multiplexing compositions and methods of the invention may be used to image fast cellular dynamics, such as those in neurons, under inexpensive single-camera microscopes with fluorescent sensors that can all be imaged in one shared optical channel (e.g. using all GFP-based sensors), because the imaging speed would then not be limited by the number of cameras available or any mechanical filter switching required to record from multiple channels. Use of spatial multiplexing compositions and methods of the invention can also free up optical channels for use for other purposes, such as cellular control. As a non-limiting example, the GFP channel may be used to observe cellular activity readout from multiple GFP-based sensors and then the red channel of the microscope used to operate red-light driven optogenetic tools (Chuong et al., 2014; Klapoetke et al., 2014).
An SiRI of the invention includes multiple components, each which may be independently selected. As used herein in reference to elements of SiRIs of the invention, the term “independently selected” means that each of a given type of element may differ from others of the same type of element in an SiRI and/or in multiple SiRIs in a cell. For example, though not intended to be limiting, an SiRI may include more than one reporter protein element and in instances when there are more than one, each may be selected so as to be different or the same as one or more other reporter protein elements in that SiRI. Similarly, in a cell that includes two or more SiRIs, each independently selected reporter protein element may be selected so as to be different than one or more other reporter protein elements in the cell, or may be selected so as to be the same as one or more other reporter protein elements in the cell. The independent election of elements in SiRIs of the invention, including reporter protein elements, RNA-binding protein elements, localization protein motif elements, protein linker elements, binding sequence elements may all be independently selected, thus permitting differences between SiRIs of the invention and permitting different SiRIs to be included in a single cell.
Different methods can be used to prepare SiRIs of the invention as is described herein using compositions capable of producing one or a plurality of SiRIs in a living cell. As used herein the term plurality means at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, or 1000. One such composition of the invention that can be used to prepare an SiRI using RNA-protein-binding-based clustering methods, is referred to herein as an RNA-protein binding (RPB) composition. Another composition of the invention that can be used to prepare an SiRI using protein self-assembly-based clustering of reporter molecules and to prepare an SiRI, is referred to herein as a protein self-assembly binding (PSA) composition. In certain embodiments of the invention, one or more different (PSA) compositions can be present in a live cell and produce one or more different SiRIs in that cell. In some embodiments of the invention, one or more different PSA compositions can be present in a live cell and produce one or more different SiRIs in that cell. In certain embodiments of the invention, one or more different RPB compositions and one or more different PSA compositions may be present in a live cell and produce one or more different SiRIs in that cell.
In certain embodiments of the invention an RPB composition comprises a fusion protein component and an RNA component. A fusion protein component of an RPB of the invention comprises independently selected elements such as (i) one or more independently selected reporter protein elements, (ii) one or more independently selected RNA binding protein elements; (iii) zero, one, or more independently selected epitope tag elements; (iv) zero, one, or more localization protein motif elements; and (v) zero, one, or more independently selected protein linker elements, wherein when present, each of the protein linker elements is positioned between two of the elements of (i), (ii), (iii), and (iv). An RNA component of an RPB composition of the invention comprises a plurality of independently selected RNA molecules, and each of the independently selected RNA molecules comprises a plurality of binding sequence elements that are recognized by the one or more RNA binding protein elements of (ii). It will be understood that in some embodiments, a composition of the invention comprises the sequences that encode the fusion protein of the RPB composition. For example, in some embodiments of the invention a composition of the invention includes a vector comprising sequences encoding an RPB fusion protein.
A PSA composition of the invention comprises a fusion protein component comprising independently selected elements, such as (a) one or more independently selected reporter protein element(s); (b) one or more independently selected self-assembly protein element(s); (c) zero, one, or more independently selected epitope tag element(s); (d) zero, one, or more protein localization motif element(s); and (e) zero, one, or more independently selected protein linker element(s), wherein when present, each of the protein linker elements is positioned between two of the elements of (a), (b), (c), and (d). It will be understood that in some embodiments, a composition of the invention comprises the sequences that encode the fusion protein of the PSA composition. For example, in some embodiments of the invention a composition of the invention includes a vector comprising sequences encoding a PSA fusion protein.
In both RPB and PSA compositions of the invention, the number of each type of element can differ or be the same in different prepared SiRIs. For example, an RPB and a PSA composition may each include one or more independently selected reporter protein elements, and so in some embodiments of the invention, each RPB and PSA has one, two, or other of the same number of independently selected reporter protein elements, and in certain embodiments of the invention each RPB and PSA has a different number of independently selected reporter protein elements. It will be understood that both situations may occur in a single cell, meaning that a cell may have the same number of independently selected reporter protein elements in two or more RPBs and/or PSAs, and may also have different independently selected reporter protein elements in each RPB and/or PSA in that cell. In some embodiments of RPB and PSA compositions of the invention, one or more of an independently selected element may be present. In some embodiments of the invention, the number of a particular element in an RPB and/or PSA composition is zero, meaning the element is not present in that component.
It will be understood that certain embodiments of the invention comprise nucleic acid sequences that encode protein elements as described herein. Compositions of the invention may be prepared using a nucleic acid sequence encoding one or more of: a reporter protein element, a localization protein element, an epitope tag element, a protein linker element, a binding sequence element, a self-assembly protein element, or a variant thereof.
The invention in part, includes methods with which to perform simultaneous readout of a plurality of reporter proteins that can each be independently selected by the practitioner. As a result, a practitioner can select a plurality different reporters to include in methods and compositions of the invention and when present in a cell, a signal from each of the plurality of reporter proteins can be detected singly or can be detected simultaneously with one or more other of the reporter proteins in the cell. As a non-limiting example, a practitioner can select three different reporter proteins, reporters A, B, and C, and include the reporters in different compositions in a cell, and read the signals of the reporter proteins one at a time, two at a time, or all three simultaneously.
RPBs and PSAs of the invention comprise a fusion protein comprising one or more independently selected reporter protein elements. Non-limiting examples of a reporter protein element that can be included in compositions and methods of the invention are: a voltage indicator polypeptide, a calcium indicator polypeptide, a potassium indicator polypeptide, a pH indicator polypeptide, and a magnesium indicator polypeptide. Additional non-limiting examples of reporter protein elements that can be included in certain embodiments of methods and compositions of the invention are: GCaMP6, GCaMP6f, GCaMP6m, GCaMP6s, jGCaMP7, jGCaMP7f, jGCaMP7m, jGCaMP7s, jGCaMP7b, jGCaMP7c, GCaMP-X, jRGECO1, jRCaMP1, NIRGECO, BCaMP, ICUE, ICUE3, cAMPr, Epac-based cAMP indicator, AKAR, AKAR4, ExRai-AKAR, ExRai-AKAR2, CKAR, ExRai-CKAR, EKARev, and RAB-EKARev. In some embodiments of the invention an independently selected reporter protein element is a GCaMP6f polypeptide, and in certain embodiments of the invention an independently selected reporter protein element is an ICUE3 polypeptide.
It will be understood that the methods and compositions of the invention may include one or more of numerous additional reporter proteins and that a practitioner will be able to select and include one or more independently selected reporter proteins such as those described herein and those otherwise available. Non-limiting examples of fluorescence reporter proteins that can be included in certain embodiments of methods and compositions of the invention are described in the ‘Fluorescent Biosensor Database’ (available at //biosensordb.ucsd.edu/), which lists numerous reporter proteins that can be used in compositions and methods of the invention.
RPBs and PBAs of the invention comprise a fusion protein comprising zero, one, or more independently selected epitope tag elements, which may also be referred to herein as protein tags. In some embodiments of compositions and methods of the invention, a plurality of protein tags are included in a single fusion protein. The inclusion of a plurality of protein tags in one fusion protein results in tags capable for use as protein barcodes. Non-limiting examples of epitope tags that may be included in an embodiment of an RPB or a PBA composition of the invention are: an Xpress tag, an HA tag, and a V5 tag, an OLLAS tag, a VSVg tag, an S1 (Strep I) tag, an NWS (Strep II) tag, an E epitope (E tag), a FLAG tag, an E2 tag, an AU1 tag, an AU5 tag, an Myc tag, a Spot tag, a NE tag, an AviTag, a C-tag, a Calmodulin-tag, a polyglutamate tag, an Rho1D4-tag, an S-tag, an SBP-tag, a Softag 1, a Softag 3, a TC tag, and a Ty tag. As indicated elsewhere herein, in some embodiments of compositions of the invention the number of epitope tags in a fusion protein of an RPB or PBA of the invention is zero. Protein tags are polypeptide sequences that have been genetically added onto a recombinant protein. The preparation and use of epitope tags are routinely practiced in the art and numerous protein epitope tags and their encoding sequences are known and are suitable for use in compositions and methods of the invention.
RPBs and PBAs of the invention comprise a fusion protein comprising zero, one, or more independently selected localization protein motif elements. In some embodiments of compositions and methods of the invention, a plurality of localization protein motifs elements are included in a single fusion protein. As indicated elsewhere herein, in some embodiments of compositions of the invention the number of localization protein motif elements included in a fusion protein of an RPB or PBA of the invention is zero. Localization protein motifs are proteins that are capable of delivering fusion proteins of RPBs and PBAs of the invention from their site of synthesis in a cell to their intended location in the cell and may be referred to as trafficking sequences/motifs and targeting sequences/motifs. Methods of preparing and using localization protein motif elements are routinely practiced in the art and numerous types of known localization protein motifs and their encoding sequences, are suitable for use in compositions and methods of the invention. In certain embodiments of methods and compositions of the invention a localization protein motif comprises one or more of a subcellular localization protein and a trafficking protein. Non-limiting types of localization protein motifs that may be used in certain embodiments of the invention are nucleus localization motif, plasma membrane localization motifs, and synapse localization motifs. Further information available in: Martin, K. C. & a. Ephrussi (2009) Cell 136, 719-730; Negi, S. et al. (2015) Database Vol. 2015: article ID bav003; Donnes, P. & A. Hoglund (2004) Geno. Prot. Bioinfo. Vol. 2 No. 4: 209-215, each of which is incorporated by reference herein.
RPBs and PBAs of the invention comprise a fusion protein comprising zero, one, or more independently selected protein linker elements, which may also be referred to herein as “linkers”. Linkers are polypeptides that, when present, are positioned between other types of elements of an RPB and/or PBA of the invention. Non-limiting examples of positioning of linkers in a fusion protein of the invention are: a linker positioned between a reporter protein element and an epitope tag element, a linker between two epitope tag elements, two or more linkers between an RNA binding protein element and a reporter protein element, and two or more linkers between two RNA binding protein elements. Different types of linkers can be used in embodiments of the invention. For example, in some embodiments of compositions and methods of the invention a protein linker element comprises one or more glycine-rich linkers. Non-limiting examples of glycine-rich linkers comprise one or more of: a GSG linker, a (GGSGGT)×2 linker (SEQ ID NO: 13), a (GGSGGT)×4 linker (SEQ ID NO: 14), a (GGSGGSGGT)×3 linker (SEQ ID NO: 15), a (GGGS)×2 linker (SEQ ID NO: 67), and a (GGGS)×4 linker (SEQ ID NO: 68). Other glycine-rich linkers are known and routinely used in the art and a skilled artisan will recognize additional glycine-rich linkers suitable for use in compositions and methods of the invention. In some embodiments of the invention, a protein linker element comprises one or more non-glycine-rich linkers. Non-limiting examples of non-glycine-rich linkers that may be used in embodiments of compositions and methods of the invention are: (EAAAK)×2 (SEQ ID NO: 71), and APAPAP (SEQ ID NO: 69). Other non-glycine-rich linkers are known and routinely used in the art and a skilled artisan will recognize additional non-glycine-rich linkers suitable for use in compositions and methods of the invention.
A linker protein used in an embodiment of the invention may be a flexible linker or a rigid linker and may include at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 25, or 40 amino acids. Additional linker proteins and their encoding polynucleotides are known and routinely used in the art and may be included in some embodiments of compositions and methods of the invention, see for example: Chichili, V. P. R, et al., Protein Science 2013 Vol 22:153-167; Chen, X. et al., Adv Drug Deliv Rev. 2013 Oct. 15; 65(10): 1357-1369; Arai et al. Protein Eng. 2001 August; 14(8):529-32; and Klein, J. S., et al. (2014) Protein Engineering, Design & Selection vol. 27 no. 10 pp. 325-330, each of which is incorporated herein by reference.
RNA binding protein elements
RPBs of the invention comprise a fusion protein comprising one or more independently selected RNA binding protein elements. Each independently selected RNA binding protein element recognizes and selectively binds its partner binding sequence, which is present in the cell as an RNA component of the RPB. Non-limiting examples of RNA binding protein element that may be used in embodiments of the invention are non-programmable RNA binding elements and programmable RNA binding elements. Non-limiting examples of independently selected non-programmable RNA binding elements that can be included in compositions and methods of the invention are an MS2 coat protein, a PP7 coat protein, a Lambda N protein, a Q-beta coat protein, a BglG protein, a U1Ap protein, HTLV-1 Rex protein, a TAT protein, an REV protein, and an eiF4A protein. Non-limiting examples of independently selected programmable RNA binding elements that can be included in compositions and methods of the invention are one or more of a Pumilio homology domain (PumHD) and a Pumilio-based assembly (Pumby).
It will be understood that the methods and compositions of the invention may include one or more of other known RNA binding protein elements and that a practitioner will be able to select and include one or more independently selected non-programmable and/or programmable RNA binding protein elements such as those described herein and those otherwise available. Non-limiting examples of programmable RNA-binding proteins that can be included in certain embodiments of methods and compositions of the invention are described in Adamala, K. et al., (2016) PNAS; E2579-E2588, www.pnas.org/cgi/doi/10.1073/pnas.1519368113.
An RPB of the invention comprises a plurality of independently selected RNA molecules comprising a plurality of independently selected binding sequence elements. Each independently selected binding sequence recognizes and selectively binds its partner RNA binding protein element, which is present in the cell in the fusion protein component of the RPB. In some embodiments of the invention the plurality of the independently selected binding sequence elements includes ten or more binding sequences. In certain embodiments of the invention the plurality of the independently selected binding sequence elements includes at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 40, or 50 binding sequences. A composition of the invention may include one or a plurality of independently selected binding sequences. Non-limiting examples of binding sequence elements that may be included in an embodiment of the invention are: an MS2 binding sequence, a PP7 binding sequence, and a lambda N binding sequence (BoxB), a Q-beta binding sequence, a BglG binding sequence, an U1Ap aptamer sequence, an HTLV-1 Rex responsive element (RxRE), a TAR sequence, an RRE sequence, and an eiF4A aptamer sequence. Other binding sequences are known and routinely used in the art and a skilled artisan will recognize additional non-glycine-rich linkers suitable for use in certain compositions and methods of the invention. Further information is present in Chen, X., et al. Adv Drug Deliv Rev. 2013 Oct. 15; 65(10): 1357-1369 and Klein, J. S., et al. (2014) Protein Engineering, Design & Selection vol. 27 no. 10 pp. 325-330, each of which is incorporated herein by reference.
Binding protein elements: binding sequence elements are also referred to herein as “binding pairs”. A number of binding pairs are disclosed herein and known and routinely used in the art. Non-limiting examples of binding pairs that may be used in compositions and methods of the invention are: MS2 coat protein: MS2 binding sequence, PP7 coat protein: PP7 binding sequence, Lambda N protein: lambda N binding sequence (BoxB), Q-beta coat protein: Q-beta binding sequence, BglG protein: BglG binding sequence, U1Ap protein: U1Ap aptamer sequence, HTLV-1 Rex protein: HTLV-1 Rex responsive element (RxRE), TAT protein: TAR sequence, REV protein: RRE sequence, and eiF4A protein: eiF4A aptamer sequence. Other binding pairs and their encoding sequences are known and routinely used in the art and a skilled artisan will recognize additional binding pairs suitable for use in certain compositions and methods of the invention.
Additional details, such as a binding sequence and a binding partner name are provided after the following non-limiting examples of protein tags that may be included in compositions and methods of the invention are: AviTag, which permits biotinylation by the enzyme BirA, thereby permitting the protein to be isolated by streptavidin (GLNDIFEAQKIEWHE, SEQ ID NO: 82); C-tag, Calmodulin-tag, which comprises a peptide binds calmodulin KRRWKKNFIAVSAANRFKKISSSGAL, SEQ ID NO: 83); a polyglutamate tag, which binds anion-exchange resins such as Mono-Q (EEEEEE, SEQ ID NO: 84); an E-tag, which is a peptide that binds an antibody comprising the sequence: GAPVPYPDPLEPR (SEQ ID NO: 79, which is also referred to herein as “E_epitope); a FLAG-tag, which binds antibody comprising the sequence: DYKDDDDK (SEQ ID NO: 80); an HA-tag, which binds a sequence set forth as YPYDVPDYA (SEQ ID NO: 75); a His-tag; a Myc-tag; an NE-tag, which comprises an 18-amino-acid synthetic peptide TKENPRSNQEESYDDNES (SEQ ID NO: 85) that binds a monoclonal IgG1 antibody; a Rho1D4-tag, which comprises nine amino acids of the intracellular C-terminus of bovine rhodopsin (TETSQVAPA, SEQ ID NO: 86); an S-tag, which is derived from Ribonuclease A (KETAAAKFERQHMDS, SEQ ID NO: 87); an SBP-tag, which binds streptavidin (MDEKTTGWRGGHVVEGLAGELEQLRARLEHHPQGQREP, SEQ ID NO: 88); Softag 1, for mammalian expression (SLAELLNAGLGGS, SEQ ID NO: 89); Softag 3, for prokaryotic expression (TQDPSRVG, SEQ ID NO: 90); Spot-tag, which binds a nanobody, set forth as: PDRVRAVSHWSS (SEQ ID NO: 91); Strep-tag, which binds to streptavidin or the modified streptavidin called streptactin (Strep-tag II: WSHPQFEK, SEQ ID NO: 92); TC tag, which binds FlAsH and ReAsH biarsenical compounds (CCPGCC, SEQ ID NO: 93); a Ty tag (EVHTNQDPLD, SEQ ID NO: 94); a V5 tag, which binds an antibody (GKPIPNPLLGLDST, SEQ ID NO: 76); a VSV-tag, which binds an antibody (YTDIEMNRLGK, SEQ ID NO: 78); and an Xpress tag (DLYDDDDK, SEQ ID NO: 74).
A PSA of the invention comprises one or more of an independently selected self-assembly protein element. Self-assembly proteins are capable of take a defined physical arrangement without outside guidance. Some embodiments of the invention comprise self-assembly proteins that are intramolecular self-assembly proteins. Certain embodiments of the invention comprise self-assembly proteins that are intermolecular self-assembly proteins.
Non-limiting examples of self-assembly proteins that can be used in embodiments of the invention are: a polyhedron-forming protein, a coiled-coil forming protein, a supramolecular self-assembly protein, and a protein oligomer. Non-limiting examples of a polyhedron-forming protein that may be used include: I3-01, O3-33, ATC-HL3, and 3VDX. Non-limiting examples of a coiled-coil forming protein include HexCoil-Ala, 5H2L_2, EE, and RR. Non-limiting examples of a supramolecular self-assembly protein that can be used are: 2AN9 and 1M3U. Non-limiting examples of a protein oligomer that can be used are: 5L6HC3_1 and 2L8HC4_15. Other self-assembly proteins are known and routinely used in the art and a skilled artisan will recognize additional self-assembly proteins suitable for use in certain compositions and methods of the invention.
SiRIs of the invention can be prepared and used in methods of the invention in individual cells. Compositions that may be used in embodiments of the invention to prepare SiRIs in a cell comprise RPB and PSA compositions. The terms “PSA-based SiRI” and RPB-based SiRI” mean an SiRI prepared from a PSA composition or an RPB composition, respectively. In some embodiments of the invention one, two, or more independently selected RPB compositions that are delivered into a cell produce one or more SiRIs in that cell. In some embodiments of the invention one, two, or more independently selected PSA compositions that are delivered into a cell produce one or more SiRIs in that cell. In certain embodiments of the invention one, two, or more independently selected RPB compositions and one, two, or more PSA compositions are delivered into a cell and in the cell, the RPB and PSA compositions produce SiRIs in that cell. In some embodiments, methods of the invention may include preparing two or more different clusters that are formed from two or more independently selected reporter protein elements and positioned in the cell, wherein each reporter protein element is capable of generating an identifiable signal.
In some embodiments of the invention, two or a plurality of RPB compositions can be delivered into a cell and form SiRIs that include components of the RPB compositions. The SiRIs may include different elements because of the independent selection of elements in the RPB compositions. In some embodiments of the invention, two or a plurality of PAS compositions can be delivered into a cell and form SiRIs that include components of the PSA compositions. The SiRIs may include different elements because of the independent selection of elements in the PSA compositions. In some embodiments of the invention, two or a plurality of RPB compositions and PSA compositions can be delivered into a cell and form SiRIs that include components of the RPB composition and SiRIs that include components of the PSA compositions. There may be differences between the SiRIs prepared using RPB compositions, differences between the SiRIs prepared using PSA compositions, and differences between SiRIs prepared using PSA compositions and SiRIs prepared using RPB compositions, and the differences can be predetermined because of the independent selection of elements in the RPB and PSA compositions.
SiRIs prepared and present in a cell can be used in methods of the invention to determine one or more physiological process or characteristic of the cell. Because of the options to include a plurality of independently selected elements in the SiRIs prepared using RPB and/or PSA compositions of the invention, it is possible to determine two or more simultaneous physiological processes or characteristics of the same cell. In some embodiments the invention one, two, or more RPB and/or PSA compositions of the invention can be placed into a cell and used in methods to determine one or more physiological processes in the cell.
In a non-limiting example, preparation of one or a plurality of SiRIs in a cell comprises the delivery into a cell of at least one RPB and/or PSA composition. Using either an RPB or PSA composition, fusion proteins are expressed in the cell. The expressed fusion proteins include a plurality of each of two or more independently selected reporter protein elements each capable of generating an identifiable signal. As used herein the term “identifiable signal” means that at least by using methods of the invention a signal generated by a reporter protein indicates the identity of the reporter protein generating the signal. The method also includes forming one or more clusters from the plurality of the two or more independently selected reporter protein elements in the cell, wherein each of the two or more independently selected reporter protein elements are in different clusters. The presence of the independently selected elements in the RPB and/or PSA compositions present in the cell result in the formation of clusters in the cell that include the reporter protein elements of that RPB or PSA, respectively.
As a non-limiting example, if one or more RPB compositions are present in a cell, the individually selected RNA binding protein elements bind with the independently selected RNA molecules of the RNA component that is also in the cell. As described elsewhere herein, each independently selected binding sequence recognizes and selectively binds its partner RNA binding protein element present in the fusion protein component of the RPB. The binding of the binding partners results in the formation of clusters that include the fusion protein component(s) and the RNA component (s) of one or a plurality of the RPB composition(s). In another non-limiting example, if a PSA composition is present in a cell, the independently selected self-assembly protein elements in the fusion protein component assemble to form clusters of the fusion protein components of the PSA composition.
Certain embodiments of methods of the invention include positioning the one or more formed clusters in the cell such that a distance between a cluster formed with one of the independently selected reporter protein elements and a cluster formed with another of the independently selected reporter protein elements is sufficient to (i) resolve the signal generated by the two independently selected reporter protein elements in the positioned clusters and (ii) simultaneously determine the signal generated by each of the independently selected reporter protein elements in the positioned clusters. In some embodiments of the invention a distance between a cluster formed that includes one of the independently selected reporter protein elements and a cluster formed that includes another of the independently selected reporter protein elements is about 1-2 microns, 1-3 microns, 1-4 microns, 1-5 microns, 1-6 microns, 1-7 microns, or 1-8 microns. In certain embodiments the distance between clusters that include two different independently selected reporter proteins is 2-8 microns.
Elements included in the RPB and PSA compositions can be independently selected to position the SiRIs that are prepared from the compositions. As a non-limiting example, in RPB and PSA compositions, a localization protein motif may be selected and included to assist with positioning the formed clusters in a cell. As another non-limiting example, the number and length of linkers included in a fusion protein of an RPB and/or PSA composition can assist in determining a physical conformation/size of a formed cluster, thereby assisting in positioning the cluster. Certain embodiments of methods of the invention include resolving signals generated by the independently selected reporter proteins in positioned clusters and detecting changes in the signals as measure of changes in physiological processes in the cell. Non-limiting examples of combinations of linkers, localization elements, and other elements that can be selected at least in part to determine size and position of SiRI clusters of the invention are described herein. Additional combinations of linkers, localization elements, and other elements of SiRIs of the invention are also suitable for use in methods of the invention and in view of the teaching provided herein can be prepared and utilized using routine methods.
Methods and components for preparing and using SiRIs of the invention include a number of independently selected elements, thereby permitting flexibility in SiRI design. For example, it will be understood that the individually selected binding pairing generated by a first RPB in a cell may be different from the independently selected binding pairing generated by a second RPB in the same cell, thereby resulting in the formation of different clusters, one including components of the first RPB and another including components of the second RPB. Similarly, it will be understood that the individually selected self-assembly protein elements in a first PSA composition in a cell may be different from the independently selected self-assembly protein elements in a second PSA composition in the same cell, thereby resulting in the formation of different clusters, one including components of the first PSA and another including components of the second PSA.
As described herein, different elements in one or more components in RPBs and PSAs in a cell result in one or more of: the same cluster formation, different cluster formation, the same reporter signal, different reporter signal, the same epitope tags, different epitope tags, etc. One of skill in the art will recognize the variations of elements in components of RPBs and PSAs of the invention and methods of their use for imaging in a cell, including simultaneous imaging of different physiological processes in a single cell.
Embodiments of methods of the invention may also include detecting the identifiable signals generated from the independently selected reporter protein elements in the positioned clusters; and the analysis of the detected signals as a determination of one or more physiological processes of the cell.
Certain methods of the invention include detecting a signal generated reporter protein elements that are present in one or more SiRIs in a cell. In some aspects of the invention a signal generated by a reporter protein is a fluorescent signal. An advantage of the invention described herein is the ability to detect signals from two or more different fluorescent reporter protein elements and to be able to distinguish between the two. Using methods and compositions of the invention it is possible to distinguish between the signals of two different fluorescent reporter protein elements even when the two reporters generate spectrally overlapping signals. In some embodiments of the invention a signal is a fluorescent signal. In certain embodiments of the invention a signal is a luminescent signal.
A reporter protein signal may be generated as a result of stimulation of the cell comprising the reporter protein. In some embodiments of the invention the stimulation of the cell is an external stimulation. Non-limiting examples of agents that can be used to externally stimulate a cell comprise: comprise one or more of tetradecanoylphorbol acetate (PMA), (S)-3,5-Dihydroxyphenylglycine (DHPG), N-Methyl-D-aspartic acid (NMDA), forskolin, a cancer drug, an antibody, a toxin, an agonist of a receptor, an antagonist of a receptor, an electrical field, a magnetic field, light, gas, a thermal change, a gravity change, a pH change, whole cell patch claim of the cell. Other agents that can be used to stimulate a cell comprising one or more SiRIs of the invention and a skilled artisan will be able to apply agents described herein as well as other agents to stimulate the cell. Factors that may be involved in applying stimulation to a cell that comprises one or more SiRIs include, but are not limited to one or more of: stimulation intensity, stimulation frequency, pattern of stimulation, and combinations of stimulating agents. These factors and their application are known and routinely practiced in the art and can be applied to embodiments of methods of the invention. As described herein, embodiments of methods of the invention include detecting and/or monitoring physiological response(s) to a stimulation of a cell comprising one or more SiRIs of the invention, and methods and compositions of the invention also permit simultaneous imaging of two or more different physiological processes in a single cell. In some embodiments of the invention one or more different agents can be used to stimulate a cell and can be administered simultaneously or sequentially. In some aspects of the invention the cell is not externally stimulated.
Methods and compositions of the invention can be used to detect and analyze physiological processes in a cell that comprises one of more SiRIs of the invention. The flexibility of components in RPB and PSA compositions and in SiRIs of the invention permit detection and analysis of two or more different physiological processes in the same cell, and also permit detection and analysis of two or more simultaneous physiological processes in the same cell. To detect and assess a physiological process in a cell that includes one or more SiRIs of the invention a practitioner may externally stimulate the cell and determine the signals generated by the independently selected reporter protein elements in the positioned SiRIs. In certain embodiments of the invention a practitioner may determine signals generated by the independently selected reporter protein elements in the positioned SiRIs in the absence of stimulation. Non-limiting examples of physiological processes that can be assessed using compositions and methods of the invention include: a function of the cell, a response of the cell, ion flux in the cell, a therapeutic response of the cell; and an activation of the cell, etc.
An element in detection and analysis methods of the invention is detecting the signals generated by the independently selected reporter protein elements that are present in SiRIs in a cell. Detection methods may include, but are not limited to one or more of: video microscopy, computerized microscopic imaging, fluorescence microscopy, and confocal microscopy, light microscopy, light sheet microscopy, light field microscopy, and endoscopy. Methods used to detect physiological processes in a cell comprising one or more SiRIs of the invention included, but are not limited to: live cell imaging, immunostaining, RNA FISH, live cell recording, an immunostaining method, an in situ hybridization method, determining fluorescence intensity, subtracting background fluorescence, a cluster/puncta localization means, expansion microscopy, fluorescence microscopy, light microscopy, and fluorescence-lifetime imaging microscopy. It will be understood that in addition to detection methods described herein, additional art-known detection methods can also be used to detect a signal generated by a reporter protein, an epitope tag, or another element of an SiRI of the invention.
Methods for analysis of signals detected from one or more SiRIs in a cell may include data analysis, analysis programs, algorithms, etc. Information is provided herein regarding suitable methods of data analysis and a skilled artisan will recognize additional resources and programs suitable to evaluate data generated using compositions and methods of the invention.
SiRIs of the invention can be used in single cells to detect and assess physiological processes, activities, changes, etc. in the cell. Compositions and methods of the invention may be used in prokaryotic and eukaryotic cells. Compositions and methods of the invention can also be used in artificial cells. Certain embodiments of the invention include preparing one or more SiRIs in a cell that is a mammalian cell; including but not limited to a human cell, a non-human primate cell, a cell of a dog, a cell of a cat, a cell of a rodent, etc. In some embodiments of the invention, compositions and methods of the invention may be used in non-mammalian cells; including but not limited to insect cells, avian cells, fish cells, invertebrate cells, single-cell organisms, plant cells, etc. Compositions and methods of the invention may be used in non-excitable cells and in excitable cells, the latter of which includes cells able to produce and respond to electrical stimulation/signals. Examples of excitable cell types include but are not limited, to neurons, muscle cells, cardiac cells, and secretory cells (such as pancreatic cells, adrenal medulla cells, pituitary cells, etc.). As used herein the term “plurality” when used in context of cells, means two or more cells.
Non-limiting examples of cells that may be used in methods of the invention include: neuronal cells, nervous system cells, cardiac cells, circulatory system cells, connective tissue cells, visual system cells, auditory system cells, secretory cells, endocrine cells, and muscle cells.
In some embodiments, a cell used in conjunction with a method of the invention is a healthy normal cell, which is not known to have a disease, disorder, or abnormal condition. In some embodiments, a cell used in conjunction with a method of the invention is an abnormal cell, for example, a cell that is believed to have, or has been has been diagnosed as having, a disorder, disease, or condition, including, but not limited to a degenerative cell, a neurological disease-bearing cell, a cell model of a disease or condition, an injured cell, etc. In some embodiments of the invention, a cell is a control cell.
Compositions and methods of the invention may be used in cells from culture, cells in solution, cells obtained from subjects, isolated cells, recombinant cells, and/or cells in a subject (in vivo cells). Compositions and method of the invention may be used in cultured cells, cultured tissues (e.g., brain slice preparations, organ preparations, etc.), and in living subjects, etc. As used herein, a the term “subject” is used to refer to a human, non-human primate, cow, horse, pig, sheep, goat, dog, cat, bird, rodent, insect, or other vertebrate or invertebrate organism in which a method of the invention is applied. In certain embodiments of the invention, a subject is a mammal and in certain embodiments of the invention a subject is a human.
One or more physiological processes in a single cell can be detected and analyzed using embodiments of methods of the invention. Methods of the invention can also be used to detect and analyze a change in a physiological process or a condition in a single cell. Certain embodiments of the invention permit determination of the presence or absence of one or more physiological processes, as well as changes or modulations of such processes in a cell. Some embodiments of methods of the invention can be used to identify an effect of candidate agent on a cell. For example, though not intended to be limiting, a cell comprising one or more SiRIs of the invention can be contacted with a candidate agent and one or more physiological processes of the cell analyzed. The results of the analysis can be compared with an analysis of a control cell that was not contacted with the candidate agent. In some embodiments of the invention one or more SiRIs can be prepared in a single cell and one or more of the same SiRIs can be prepared in other cells. These “identical” cells can then be used to assess an effect of candidate compounds on individual cells. A “test” cell may be a cell in which the activity in the cell may be tested or assayed. Results obtained using assays and tests of a test cell using a method of the invention may be compared results obtained from the assays and tests performed in other test cells or assays and/or may be compared to a control value.
As used herein a control value may be a predetermined value, which can take a variety of forms. It can be a single cut-off value, such as a median or mean. It can be established based upon comparative groups, such as cells or tissues that have been imaged under similar conditions using a method of the invention, but are not contacted with a candidate compound with which the test cell is contacted and imaged. Another example of comparative groups may include cells or tissues that have a disorder or condition and groups without the disorder or condition. Another comparative group may be cells from a group with a family history of a disease or condition and cells from a group without such a family history. A predetermined value can be arranged, for example, where a tested population is divided equally (or unequally) into groups based on results of testing. Those skilled in the art are able to select appropriate control groups and values for use in comparative methods of the invention.
As a non-limiting example of use of method of the invention to assess the presence or absence of a change in a cell as a means to identify a candidate compound, a physiological process in a cell may be determined using a method of the invention in culture or in a subject and the cell may then be contacted the candidate compound and re-determined using a method of the invention. Any change in the physiological process of the cell as determined using methods of the invention, may indicate an effect of the candidate compound on the cell. In some embodiments of the invention, methods of the invention may be used to determine and/or analyze a physiological process in one or more test cells before and after the one or more test cells is contacted with a candidate compound and the before and after results can be compared to determine whether or not contact with the candidate compound resulted in a change in the physiological process in the cell.
A cell, tissue, and/or subject that include a cell comprising one or more an SiRIs of the invention may be monitored for the presence or absence of a change that occurs in the cell. As a non-limiting example, in a cell, a change in a physiological process in the cell may include a change in depolarization of the cell, a change in a depolarization-mediated cell characteristic, response to stimuli, an action potential, pH change, release of a neurotransmitter, etc.
As described elsewhere herein, some embodiments of methods of the invention include preparing a cell that contains one type of RPB-based SiRIs, resulting in a cell in which all SiRIs are the same as each other. Certain embodiments of methods of the invention include preparing a cell that comprises two or more types of RPB-based SiRIs, resulting in a cell that includes two or more different SiRIs. Similarly, some embodiments of methods of the invention include preparing a cell that contains one type of PSA-based SiRIs, resulting in a cell in which all SiRIs are the same as each other. Certain embodiments of methods of the invention include preparing a cell that comprises two or more types of PSA-based SiRIs, resulting in a cell that includes two or more different SiRIs. In addition, some embodiments of methods of the invention include preparing a cell that contains one type of RPB-based SiRIs and one type of PSA-based SiRIs, resulting in a cell in which all SiRIs prepared from the RPB composition are the same as each other and the SiRIs prepared from the PSA composition are the same as each other. Certain embodiments of methods of the invention include preparing a cell that comprises two or more types of RPB-based SiRIs, resulting in a cell that includes two or more different SiRIs generated from RPB compositions and two or more different SiRIs generated from PSA compositions.
The resulting cell prepared using one of these combinations of RPB and/or PSA compositions can be used to identify the effect of a candidate agent on a physiological process in the cell. In such a method the cell would be prepared as described elsewhere herein and the RPB and/or PSA composition(s) would form and position clusters as described. Methods of the invention would then include contacting the cell with a candidate agent of interest, and detecting the presence or absence of an identifiable signal generated from the independently selected reporter protein elements present in the positioned clusters. The detected signals are then analyzed and compared to control results obtained in a cell not contacted with the candidate agent, and the comparison indicates whether or not the contact with candidate agent resulted in a change in a physiological process or processes in the contacted cell. Different candidate agents can be tested using methods of the invention. Non-limiting examples of candidate agents include: pharmaceutical agent, an electrical agent, a temperature change agent, an environmental agent, etc. A skilled artisan will recognize additional candidate agents that can be tested using compositions and methods of the invention.
Proteins and RNA molecules used in methods and compounds of the invention may include functional variants of molecules disclosed herein and functional variants art-known molecules. For example, functional variants of reporter protein elements, localization protein elements, epitope tag elements, protein linker elements, binding sequence elements, self-assembly protein elements—and their encoding sequences, and functional variants of RNA component molecules can be used in methods of the invention. As used herein, the term “parent” when used in the context of a variant element of the invention means the element molecule of which the variant element molecule is the variant. Based on the teaching provided herein regarding elements that can be used in embodiments of the invention, functional variants of protein elements that have sufficient amino acid sequence similarity/identity to a parent protein element sequence and have at least a portion of the function of the parent protein element molecule can be prepared and used in embodiments of methods of the invention. Based on teaching provided herein regarding RNA molecule elements that can be used in embodiments of the invention, functional variants of the RNA molecule elements that have sufficient sequence similarity/identity to a parent RNA molecule element sequence and have at least a portion of the function of the parent RNA element molecule can be prepared and used in embodiments of methods of the invention.
As used herein, the term “identity” refers to the degree of relatedness or similarity between two or more polypeptide sequences [or polynucleotide (nucleic acid) sequences]. Sequence identity may be determined by the alignment and match between the sequences using standard methods. The percentage is obtained as the percentage of identical amino acids in two or more sequences taking account of gaps and other sequence features. The identity between polypeptide sequences can be determined by means of art-known procedures. Algorithms and programs are available and routinely used by those in the art to determine identity between polypeptide sequences and to determine identity between nucleic acid sequences. Non-limiting examples of programs and algorithms include BLASTP, BLASTN and FASTA (Altschul et al., NCB NLM NIH Bethesda Md. 20894; Altschul et al., 1990), Online BLAST programs from the National Library of Medicine are available, for example, at blast.ncbi.nlm.nih.gov/Blast.cgi.
The presence of functionality of a variant, for example the ability to be used in a method of the invention, can be determined using testing methods described herein. Functional variants of protein elements and RNA molecule elements disclosed herein can be used in compositions and methods of the invention. It will be understood that the level of sequence identity with a protein element or an RNA element of the invention, and the level of functionality with respect to methods of the invention can be characteristics used to identify protein element and RNA molecule element variants using teaching provided herein in conjunction with standard procedures for sequence alignment, comparisons, and knowledge of sequence modifications in the protein and nucleic acid arts.
A variant of a protein element (or its encoding sequence), or an RNA element includes one or more sequence modifications. As used herein the term “modified” or “modification” in reference to a protein sequence refers to a change such as one or more of an insertion, deletion, or substitution of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more amino acids in the sequence as compared to the unmodified parent sequence. As used herein the term “modified” or “modification” in reference to a nucleic acid sequence refers to a change such as one or more of an insertion, deletion, or substitution of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 nucleic acids in a sequence. As used herein the term “nucleic acid” is used interchangeably with the term “polynucleotide”.
The sequence of a protein element or nucleic acid element can be modified with one or more substitutions, deletions, insertions, or other modifications and the resulting variant element can be tested using methods described herein for characteristics including, but not limited to: expression, cell localization, imaging, etc. Exemplary modifications include, but are not limited to conservative amino acid substitutions, which will produce molecules having functional characteristics similar to those of the parent molecule. Conservative amino acid substitutions are substitutions that do not result in a significant change in the activity or tertiary structure of a selected polypeptide or protein. Such substitutions typically involve replacing a selected amino acid residue with a different residue having similar physico-chemical properties. For example, substitution of Glu for Asp is considered a conservative substitution because both are similarly sized, negatively charged amino acids. Groupings of amino acids by physico-chemical properties are known to those of skill in the art. The following groups each contain amino acids that are conservative substitutions for one another: 1) Alanine (A), Glycine (G); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W); 7) Serine (S), Threonine (T); and 8) Cysteine (C), Methionine (M) (see, e.g., Creighton, Proteins (1984)). RPB and PSA variant elements that include modifications such as, but not limited to one, two, three, four, or more conservative amino acid substitutions can be identified and tested for characteristics including, but not limited to: expression, cell localization, imaging characteristics, etc., using methods disclosed herein.
A protein element variant may include modifications that result in an amino acid sequence having at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to its parent sequence. Some embodiments of compositions and methods of the invention may include a functional variant of one or more of a reporter protein element, a localization protein element, an epitope tag element, a protein linker element, a binding sequence element, a self-assembly protein element and the variant may include modifications that result in an amino acid sequence having at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity with the amino acid sequence of the parent molecule of the element. Some embodiments of the invention a composition and/or cell comprises one or more functional variants of a sequence encoding a protein element and/or one or more functional variants of an RNA molecule element.
It is understood in the art that the codon systems in different organisms can be slightly different, and that therefore where the expression of a given protein from a given organism is desired, the nucleic acid sequence can be modified for expression within that organism. Thus, in some embodiments, one or more of a reporter protein element, a localization protein element, an epitope tag element, a protein linker element, a binding sequence element, a self-assembly protein element or functional variant thereof, is encoded by a mammalian-codon-optimized nucleic acid sequence, which may in some embodiments be a human-codon optimized nucleic acid sequence.
Delivery of a RPB and/or PSA compositions into a cell can be done using art-known delivery means. It is well known in the art how to prepare and utilize fusion proteins that comprise one or a plurality of protein sequences. In certain embodiments of the invention, a fusion protein comprising elements of an RPB and/or a PSA composition can be delivered into a cell. In some embodiments, delivery of an RPB and/or a PSA composition can be targeted to a particular cell or cell type utilizing routine means such as targeting sequences for delivery to, and expression in, a desired cell, tissue or region. Genetic targeting methods can be used to deliver an RPB and/or a PSA composition of the invention into a cell of a predetermined type, location within a subject, etc. Genetic targeting also relates to the control of the amount of expression of an RPB and/or a PSA composition that is expressed, and the timing of the expression. Routine genetic procedures can be used to control parameters of expression, such as but not limited to: the amount and/or timing of expression of a fusion protein of a RPB and/or a PSA composition in a cell.
Some embodiments of the invention include a reagent for delivering an RPB and/or a PSA composition into a cell, wherein the reagent comprises a vector comprising nucleic acid sequences encoding the protein elements of the RPB and/or a PSA composition fusion proteins. As used herein, the term “vector” refers to a nucleic acid molecule capable of transporting between different genetic environments another nucleic acid to which it has been operatively linked. The term “vector” also refers to a virus or organism that is capable of transporting the nucleic acid molecule. One type of vector is an episome, i.e., a nucleic acid molecule capable of extra-chromosomal replication. Some useful vectors are those capable of autonomous replication and/or expression of nucleic acids to which they are linked. Vectors capable of directing the expression of genes to which they are operatively linked are referred to herein as “expression vectors”. Other useful vectors, include, but are not limited to viruses such as lentiviruses, retroviruses, adenoviruses, and phages. Vectors useful in some methods of the invention can genetically insert a sensor polypeptide and a soma-targeting polypeptide of the invention into dividing and non-dividing cells and can insert a sensor polypeptide and a soma-targeting polypeptide of the invention into a cell that is an in vivo, in vitro, or ex vivo cell.
Vectors useful in methods of the invention may include additional sequences including, but not limited to, one or more signal sequences and/or promoter sequences, or a combination thereof. In certain embodiments of the invention, a vector may be a lentivirus, adenovirus, adeno-associated virus, or other vector that comprises a gene encoding a sensor polypeptide and a gene encoding a soma-targeting polypeptide of the invention. An adeno-associated virus (AAV) such as AAV8, AAV1, AAV2, AAV4, AAV5, AAV9, are non-limiting examples of vectors that may be used to express a fusion protein of the invention in a cell and/or subject. Expression vectors and methods of their preparation and use are well known in the art. Non-limiting examples of suitable expression vectors and methods for their use are provided herein.
Promoters that may be used in methods and vectors of the invention include, but are not limited to, cell-specific promoters or general promoters. Non-limiting examples promoters that can be used in vectors of the invention are: ubiquitous promoters, such as, but not limited to: CMV, CAG, CBA, and EF1a promoters; and tissue-specific promoters, such as but not limited to: Synapsin, CamKIIa, GFAP, RPE, ALB, TBG, MBP, MCK, TNT, and aMHC promoters. Methods to select and use ubiquitous promoters and tissue-specific promoters are well known in the art. A non-limiting example of a tissue-specific promoter that can be used to express a fusion protein of the invention in a cell such as a neuron is a synapsin promoter, which can be used to express an RBP and/or a PSA fusion protein of the invention in embodiments of methods of the invention. Additional tissue-specific promoters and general promoters are well known in the art and, in addition to those provided herein, may be suitable for use in compositions and methods of the invention. Additional methods for generating fusion proteins and recombinant polypeptides are known in the art may include use of prokaryotic and eukaryotic expression systems including but not limited to bacterial and mammalian expression systems.
The present invention in some aspects includes one or more methods of preparing and using vectors encoding one or more RPB and/or PSA fusion proteins, including the delivery of the vectors encoding PRB and/or PSA fusion proteins into a cell, expression of the encoded fusion protein in the cell, delivery of additional components of one or more RPB compositions into the cell, detecting signals generated by one or more reporter protein elements in SiRIs generated from the RPB and/or PSA compositions delivered into the cell. The present invention enables simultaneous detection of two or more physiological processes in a single cell. The PRB and PSA compositions of the invention and their use, have broad-ranging applications for determining simultaneous physiological processes within cells, drug screening, therapeutic testing, and research applications, some of which are describe herein.
NEB Stable Competent E. coli (New England Biolabs, Ipswich, Mass.) were used for small-scale isolation of plasmids encoding repetitive RNA scaffolds and were grown in a shaking incubator (250 RPM) at 30° C. Small-scale isolations of all other plasmids were done with Z-Competent E. coli (Zymo Research, Ipswich, Mass.) grown in a shaking incubator (250 RPM) at 37° C.
All procedures involving animals at MIT were conducted in accordance with the US National Institutes of Health Guide for the Care and Use of Laboratory Animals and approved by the Massachusetts Institute of Technology Committee on Animal Care.
HeLa cells, passage 5-12 (ATCC) were maintained between 10% and 90% confluence at 37° C. with 5% CO2 in DMEM (Gibco, Waltham, Mass.) with the addition of 10% heat inactivated fetal bovine serum (HI-FBS) (Corning, Corning, N.Y.), 1% penicillin/streptomycin (Gibco, Waltham, Mass.), and 1% sodium pyruvate (Gibco, Waltham, Mass.). Cells were authenticated by the manufacturer and tested for mycoplasma contamination to their standard levels of stringency and were used herein because they are common cell lines for testing new tools. Transfection and recordings for experiments that did not involve post hoc RNA FISH proceeded in glass 24-well plates treated with 75 μL diluted Matrigel (250 μL Matrigel (Corning, Corning, N.Y.) diluted in 12 mL DMEM) per well at 37° C. for 30-60 minutes. Transfection and recordings for experiments involving post hoc RNA FISH proceeded in glass coverslips (Carolina Biological Supply, Burlington, N.C.) treated with the diluted Matrigel in glass 24-well plates.
Hippocampal neurons were prepared from postnatal day 0 or 1 Swiss Webster mice (Taconic Biosciences, Rensselaer, N.Y.) (both male and female mice were used) as previously described (Klapoetke et al., 2014) with the following modifications: dissected hippocampal tissue was digested with 50 units of papain (Worthington Biochem, Lakewood, N.J.) for 6-8 min, and the digestion was stopped with ovomucoid trypsin inhibitor (Worthington Biochem, Lakewood, N.J.). Cells were plated at a density of 20,000-30,000 per glass coverslip coated with diluted Matrigel in a 24-well plate. Neurons were seeded in 100 μL plating medium containing MEM (no glutamine, no phenol, Life Technologies, Carlsbad, Calif.), glucose (33 mM, Millipore Sigma, Burlington, Mass.), holo-Transferrin bovine (0.01%, Millipore Sigma, Burlington, Mass.), HEPES (10 mM, Millipore Sigma, Burlington, Mass.), glutaGRO (2 mM, Corning, Corning, N.Y.), insulin (0.13%, Millipore Sigma, Burlington, Mass.), B27 supplement (2%, Gibco, Waltham, Mass.), and HI-FBS. After cell adhesion, additional plating medium was added. AraC (0.002 mM, Millipore Sigma, Burlington, Mass.) was added when glial density was 50-70% of confluence. Neurons were grown at 37° C. and 5% CO2 in a humidified atmosphere.
The DNAs encoding the protein motifs (mammalian-codon optimized), the 24×MS2 binding sites (24×MBS), and the 24×PP7 binding sites (24×PBS) used in this study were synthesized by Epoch Life Science (Epoch Life Science, Sugar Land, Tex.) or GenScript (GenScript Biotech, Piscataway, N.J.). The 48×MS2 binding sites (48×MBS) and 72×PP7 binding sites (72×PBS) were cloned from 24×MS2 and 24×PP7 arrays using the restriction cloning method described in (Golding and Cox, 2004). The sensor vectors for RNA scaffold-based clustering strategies were cloned into the pAAV-UBC backbone. The scaffold vectors for RNA scaffold-based clustering strategies were cloned into the pAAV-CAG backbone. The vectors for protein scaffold-based clustering strategies were cloned into the pAAV-UBC, pAAV-Syn, or pAAV-CAG backbones. For control experiments with unassembled sensors in HeLa cells, pAAV-CAG-GCaMP6f and pcDNA3-CMV-ICUE3 were used. For control experiments with unassembled sensors in neurons, pAAV-UBC-GCaMP6f and pAAV-UBC-cAMPr were used. (Table 1, motif sequences; Table 2, all tested constructs and their Des.)
Small-scale isolation of plasmid DNA was performed with Plasmid Mini-Prep kits (Qiagen, Germantown, Md.), after transformation in either NEB Stable Competent E. coli (New England Biolabs, Ipswich, Mass.) for plasmids encoding repetitive RNA scaffolds or in Z-Competent DH5a E. coli (Zymo Research, Irvine, Calif.) for plasmids that did not encode repetitive RNA scaffolds, per manufacturers' protocols.
The DNA was transiently transfected into HeLa cells using a TransIT-X2 Dynamic Delivery System kit (Minis Bio, Madison, Wis.). The 250-500 ng of total plasmid DNA per well was transfected into HeLa cells according to the manufacturer's protocol. Cell culture media in the wells was changed to fresh media 24 hours after transfection. The cells were then incubated for another 24 hours before live cell imaging.
Cultured neurons were transfected at 4-5 days in vitro (DIV) with a commercial calcium phosphate transfection kit (Invitrogen, Waltham, Mass.) as previously described (Piatkevich et al., 2018). Briefly, for transfection in each coverslip/well, 250 ng of each plasmid of interest, 500 ng pAAV-Syn-miRFP plasmid as a cell morphology marker, and pUC19 plasmid as a ‘dummy’ DNA plasmid to bring the total amount of DNA to 1500 ng (to avoid variation in DNA-calcium phosphate co-precipitate formation) were used. The cells were washed with acidic MEM buffer (containing 15 mM HEPES with final pH 6.7-6.8 adjusted with acetic acid (Millipore Sigma, Burlington, Mass.) after 45-60 minutes of calcium phosphate precipitate incubation to remove residual precipitates. The neurons were then incubated for another 4-5 days before live cell imaging at 8-9 DIV.
HeLa cells were imaged 48 hours after transfection. For 10-15 minutes at 37° C. right before imaging, 10 μL of NucBlue Live ReadyProbes Reagent (Invitrogen, Waltham, Mass.) was added to the media to stain the cell nucleus after which the media was replaced with Fluorobrite DMEM supplemented with 15 mM HEPES. Live cell imaging of HeLa cells was performed on an inverted epi-fluorescence wide-field Nikon Eclipse Ti microscope (Nikon, Melville, N.Y.) with a 40×1.15 NA water immersion objective (Nikon MRD77410), a SPECTRA X light engine (LumenCor, Beaverton, Oreg.), and a Zyla 5.5 camera (Andor Technology, South Windsor, Conn.) controlled by NIS-Elements AR software. For imaging GFP intensity-based sensors (GCaMP6f and cAMPr), a 475/28 nm excitation filter (Semrock, Rochester, N.Y.) and a 527/50 nm emission filter (Semrock, Rochester, N.Y.) were used. For imaging the CFP/YFP FRET-based sensor (ICUE3 and AKAR4), a 438/24 nm donor excitation filter (Semrock, Rochester, N.Y.), a 483/32 nm CFP emission filter (Semrock, Rochester, N.Y.), and a 542/27 nm YFP emission filter (Semrock, Rochester, N.Y.) were used. For imaging miRFP, a 631/28 nm excitation filter (Semrock, Rochester, N.Y.) and a 664 long-pass emission filter (Semrock, Rochester, N.Y.) were used. Under the 40× objective, cells were recorded for 5-15 minutes in the GFP, CFP and/or YFP channels at 10 seconds per frame (0.1 Hz), during which the reagents for extracellular stimulation, 10 mM final concentration of calcium chloride (Millipore Sigma, Burlington, Mass.) or 20 μM final concentration of forskolin (Millipore Sigma, Burlington, Mass.), was added into the Fluorobrite media. After recording, images were taken at 40× and 10× in the GFP, CFP, YFP, miRFP, and/or NucBlue channels, and then a tiled image in the NucBlue channel at 4× covering the entire glass coverslip was taken to facilitate registration with the images from downstream immunostaining or RNA FISH. HeLa cells were fixed for 10 minutes in TissuePrep buffered 10% formalin (Electron Microscopy Sciences, Hatfield, Pa.), followed by washing with 1× phosphate buffered saline (PBS) three times, 5 minutes each. If the cells were for downstream immunostaining, cells were stored in 1×PBS at 4° C. If the cells were for downstream RNA FISH, cells were stored in 70% ethanol at 4° C.
Live neuron imaging was performed on a spinning disk confocal microscope (a Yokogawa CSU-W1 Confocal Scanner Unit (Yokogawa, Sugar Land, Tex.) on a Nikon Eclipse Ti microscope (Nikon, Melville, N.Y.) equipped with a 40×1.15 NA water immersion objective (Nikon MRD77410) and a Zyla PLUS 4.2 Megapixel camera (Andor Technology, South Windsor, Conn.) controlled by NIS-Elements AR software. The filter set for GFP was used for imaging GFP intensity-based sensors. For primary neuron cultures, 4-5 days after transfection, cells were recorded for 1-3 minutes in the GFP channel at 50 ms per frame (20 Hz), during which 5 μM final concentration of forskolin was added into the media. After recording, z stacks were taken at 40× in GFP and miRFP channels, and then a tiled image in GFP channel at 4× covering the entire glass coverslip was taken for registration of live images with the images from downstream immunostaining. Neurons were then fixed for 10 minutes in TissuePrep buffered 10% formalin, followed by washing with 100 mM glycine (Millipore Sigma, Burlington, Mass.) in 1×PBS for 10 minutes, and then washing with 1×PBS three times, 5 minutes each. Cells were stored in 1×PBS at 4° C.
All solutions described herein were made in 1×PBS unless otherwise specified, and incubations carried out at room temperature unless otherwise specified. Because it was observed that the miRFP fluorescence was not preserved after fixation, fluorescent antibodies that had spectral overlap with miRFP for immunostaining were used. Cells were permeabilized with 0.1% Triton X-100 for 15 minutes and then blocked with 5% normal donkey (ThermoFisher Scientific, Waltham, Mass.), horse (ThermoFisher Scientific, Waltham, Mass.), or bovine (Abcam, Cambridge, Mass.) serum for 15 minutes. Cells were incubated with primary antibodies in blocking buffer at 1:400 for 1 hour, and then washed in PBS three times for 5 minutes each. Cells were incubated with fluorescently-labeled secondary antibodies in blocking buffer at 1:400 for 2 hours for HeLa cells and 1 hour for cultured neurons, then washed in PBS three times. Nuclei were stained with DAPI at 1 μg/ml for 1-5 minutes followed by 1×PBS wash. Imaging was performed on the same spinning disk confocal microscope as in the live neuron imaging experiments. Antibodies used in certain of the experiments included: Xpress Monoclonal Antibody, Mouse, Invitrogen, Thermofisher Scientific, Waltham, Mass.; Anti-V5 Tag Antibody, Chicken, Abcam, Cambridge, Mass.; Anti-HA Tag Antibody, Rabbit, Abcam, Cambridge, Mass.; Goat anti-Mouse IgG1 Cross-Adsorbed Secondary Antibody, Alexa Fluor 546, Invitrogen, Thermofisher Scientific, Waltham, Mass.; Goat anti-Mouse IgG (H+L) Highly Cross-Adsorbed Secondary Antibody, Alexa Fluor 594, Invitrogen, Thermofisher Scientific, Waltham, Mass.; Goat anti-Chicken IgY (H+L) Secondary Antibody, Alexa Fluor 647, Invitrogen, Thermofisher Scientific, Waltham, Mass.; Goat anti-Rabbit IgG (H+L) Secondary Antibody, Alexa Fluor 647, Invitrogen, Thermofisher Scientific, Waltham, Mass.; and Donkey Anti-Rabbit IgG (H+L), Highly Cross-Adsorbed, CF543, Biotium, Fremont, Calif.
Acryloyl-X (6-((acryloyl)amino)hexanoic acid, succinimidyl ester (AcX) (ThermoFisher Scientific, Waltham, Mass.) powder was dissolved in anhydrous DMSO (ThermoFisher Scientific, Waltham, Mass.) at a concentration of 10 mg/ml, and stored in a desiccated environment at −20° C. Cell cultures on a round coverslip were then incubated in 300-500 μl of AcX at a concentration of 0.1 mg/ml in 1×PBS with 0.5% Triton-X for 30 minutes at 4° C. and then for 1.5 hours at 37° C. Then, cells were washed with 1×PBS three times for 5 minutes each.
A monomer solution composed of 2 M NaCl (Sigma-Aldrich, St. Louis, Mo.), 8.625% (w/v) sodium acrylate (Sigma-Aldrich, St. Louis, Mo.), 2.5% (w/v) acrylamide (Sigma-Aldrich, St. Louis, Mo.), and 0.10% (w/v) N,N′-methylenebisacrylamide (Sigma-Aldrich, St. Louis, Mo.) was then prepared in 1×PBS and aliquotted and stored it at −20° C. Next, a gelling solution composed of monomer solution and the chemicals 4-hydroxy-2,2,6,6-tetramethylpiperidin-1-oxyl (4HT) (Sigma-Aldrich, St. Louis, Mo.) was prepared as an inhibitor, and tetramethylethylenediamine (TEMED) (Sigma-Aldrich, St. Louis, Mo.) as an accelerator, and ammonium persulfate (APS) (Sigma-Aldrich, St. Louis, Mo.) was used as an initiator. 4HT, TEMED, and finally APS were then sequentially added to the monomer solution to prepare the gelling solution (final concentration, 0.01% (w/w) for 4HT, and 0.2% (w/w) for both APS and TEMED) Then, each coverslip containing a neuronal cell culture sample was placed on a glass slide with the cells facing up, and constructed a gel chamber by putting three No. 1.5 coverslips on top of each other unto the glass slide to function as spacers on either end of the neuronal coverslip to avoid compression. The sample was then covered with gelling solution and a coverslip placed over the sample and across the two spacers to ensure the sample was covered with gelling solution and no air bubbles were formed on the sample. Samples were first incubated at 4° C. for 30 minutes in a humidified atmosphere to prevent premature gelation and enable diffusion of solution into tissues, and subsequently incubated at 37° C. for 2.5 hours in a humidified atmosphere to complete gelation.
Afterward, the top coverslip was removed from the samples, and only the sample gel and original coverslip were removed and placed in a 50 ml large conical tube containing 5 ml of denaturation buffer, consisting of 20% (w/v) sodium dodecyl sulfate (SDS), 100 mM f3-mercaptoethanol, 25 mM ethylenediaminetetraacetic acid (EDTA) and 0.5% Triton-X in Tris 50 mM at pH 8. Samples were incubated in denaturation buffer for 30 minutes at 37° C. followed by 1 hour in an autoclave at 121° C. Samples were then cooled to RT for 30 minutes. At this stage, gels completely fell off the original coverslip during denaturation or immediately afterwards following gentle shaking and washing with 1×PBS 5 times for 3 min each at RT. The hydrogel embedded samples attained a final state of ˜2.3× linear expansion given the use of an expandable hydrogel.
Next, samples underwent blocking by incubating for 30 minutes at 37° C. in MAXBlock Blocking medium (Active Motif, Carlsbad, Calif.), followed by three washes for 5 minutes each at RT in MAXWash Washing medium (Active Motif, Carlsbad, Calif.). Next, samples were immunostained by diluting primary antibodies in MAXStain Staining medium (Active Motif, Carlsbad, Calif.) and incubating for 1.5 hours at 37° C. or overnight at 4° C. This was followed by three washes for 5 minutes each at RT in MAXWash Washing medium. Then, samples were incubated with secondary antibodies for 1.5 hours at 37° C. or overnight at 4° C. Samples where then washed three times for 5 minutes each at RT in MAXWash Washing medium and stored in 1×PBS.
Antibodies used in certain of the experiments included: Xpress Monoclonal Antibody, Mouse, Invitrogen, Thermofisher Scientific, Waltham, Mass.; Anti-VS Tag Antibody, Chicken, Abcam, Cambridge, Mass.; Anti-HA Tag Antibody, Rabbit, Abcam, Cambridge, Mass.; Goat anti-Mouse IgG1 Cross-Adsorbed Secondary Antibody, Alexa Fluor 546, Invitrogen, Thermofisher Scientific, Waltham, Mass.; Goat anti-Mouse IgG (H+L) Highly Cross-Adsorbed Secondary Antibody, Alexa Fluor 594, Invitrogen, Thermofisher Scientific, Waltham, Mass.; Goat anti-Chicken IgY (H+L) Secondary Antibody, Alexa Fluor 647, Invitrogen, Thermofisher Scientific, Waltham, Mass.; Goat anti-Rabbit IgG (H+L) Secondary Antibody, Alexa Fluor 647, Invitrogen, Thermofisher Scientific, Waltham, Mass.; and Donkey Anti-Rabbit IgG (H+L), Highly Cross-Adsorbed, CF543, Biotium, Fremont, Calif.
For spectral clearance to enable antibody stripping, samples were placed in a water tight container with denaturation buffer and incubated for 2 hours at 70° C. with shaking at 200 rpm. Then, excess denaturation buffer was removed, and samples were washed with 1×PBS at RT 5 times for 3 minutes. Samples then underwent immunostaining steps as noted in the previous paragraph for the subsequent rounds of staining.
Quasar 570 conjugated fluorescent oligonucleotides were purchased from LGC Biosearch Technologies (Biosearch Technologies, Petaluma, Calif.) as fluorescent RNA FISH probes including: MBS probe, gacatgggtgatcctcatgt (SEQ ID NO: 95); MBS/PBS Linker 1 probe, atctaatgaacccgggaata (SEQ ID NO: 96); MBS/PBS Linker 2 probe, ttctaggcaattaggtacct (SEQ ID NO: 97); PBS 1 probe, agcgacgccatatcgtctgc (SEQ ID NO: 98); and PBS 2 probe, agcgagcccatatgctctgc (SEQ ID NO: 99).
Cells were washed twice for 5 minutes each with wash buffer A (10% formamide in 2×SSC). Afterward, fluorescent RNA FISH probes were added into the hybridization buffer [10% formamide (Millipore Sigma, Burlington, Mass.) and 10% dextran sulfate (Millipore Sigma, Burlington, Mass.) in 2×SSC] to a total probe concentration of 50 nM. Cells were incubated in the FISH probe containing hybridization buffer for 16 hours at 37° C. in a humidity controlled incubator. The cells were then washed with wash buffer A twice at 37° C., each for 30 minutes, before nuclear staining with DAPI at 1 μg/ml for 1-5 minutes followed by a 1×PBS wash. The cells were then transferred to 24-well glass plates with 1×PBS for imaging. Imaging was performed on the same wide-field Nikon Eclipse Ti microscope as the live HeLa imaging experiments.
For each sample, the images and movies taken from live cell imaging and the images taken after immunostaining or RNA FISH were registered by the tiled images from the NucBlue channel (before fixation) and the DAPI channel (after immunostaining or RNA FISH), in each case taken under a 4× objective.
The analysis of the recorded movies from live cell imaging was performed in ImageJ (ImageJ, National Institutes of Health) and Excel (Microsoft, Redmond, Wash.). For the analysis of the movies from cells expressing fluorescent sensors assembled by RNA scaffolds or protein scaffolds, the time course of the fluorescence intensity from a punctum in the recorded optical channel was measured as the time course of the average fluorescence intensity within the apparent boundary of the punctum. Then the time course of the net fluorescence, F, from that punctum was obtained by subtracting the background fluorescence intensity measured from a region that had no cells in it from that movie. For the analysis of the movies from cells expressing non-assembled fluorescent sensors as control groups, the time course of the fluorescence intensity from a cell in the recorded optical channel was measured as the time course of the average fluorescence intensity within a random ROI in the cytosol of the cell with the area of that ROI similar to the size of individual puncta in the cells expressing assembled fluorescent sensors that these control groups were compared to, about 1 μm2 in HeLa cells, 5-10 μm2 in the somata of neurons, and 1 μm2 in the neurites of neurons. This was for keeping the noise from the camera contained in the measured fluorescence time courses at a comparable level between cells expressing assembled fluorescent sensors and cells expressing non-assembled fluorescent sensors as control groups. Then the time course of the net fluorescence, F, from that cell was obtained by subtracting the background fluorescent intensity measured from a region that had no cells in it from that movie. For each neurite-localized ROI in neurons, the cumulative distance between the ROI and the soma was calculated by measuring along the neurite.
To calculate the dF/F0 for GCaMP6f or cAMPr expressing HeLa cells or neurons, the baseline fluorescence, F0, was first calculated as the average net fluorescence during the 2-minute period (for movies from live HeLa imaging) or 5-second period (for movies from live neuron imaging) right before adding the extracellular stimulation. dF/F0 was then calculated as dF/F0=(F−F0)/F0. To calculate the signal-to-noise ratio (SNR), the maximum dF/F0 was divided by the standard deviation of the net fluorescence during the 2-minute period (HeLa) or 5-second period (neuron) right before adding the extracellular stimulation.
To calculate the change of CFP/YFP ratio, d(C/Y)/(C/Y), for ICUE3 expressing HeLa cells, the CFP/YFP ratio, C/Y, was first calculated by dividing the net fluorescence from the CFP channel by the net fluorescence from the YFP channel. Then the baseline of the CFP/YFP ratio, (C/Y)0, was calculated as the average of the CFP/YFP ratio during the 2-minute period right before adding the extracellular stimulation. d(C/Y)/(C/Y) was then calculated as d(C/Y)/(C/Y)=[C/Y−(C/Y)0]/(C/Y)0. To calculate the signal-to-noise ratio (SNR), the maximum d(C/Y)/(C/Y) was divided by the standard deviation of the d(C/Y)/(C/Y) during the 2-minute period (HeLa) right before adding the extracellular stimulation.
For puncta size analysis and the distance to the nearest punctum analysis, StarDist was used to identify and segment the boundaries of puncta followed by geometrical analysis with a custom MATLAB script. For the analysis of puncta spatial separation, a punctum with less than 5% overlap with the puncta of other sensors after immunostaining was counted as a spatially separate punctum. For the analysis of the motion of puncta in recorded and time-lapse movies (1 minute per frame; 1 hour total), puncta were automatically identified and tracked by the TrackMate plugin in ImageJ, then mean squared displacement (MSD) and diffusion coefficient were calculated in MATLAB (MathWorks, Natick, Mass.) with the script msdanalyzer [Tarantino, et al., 2014] based on the tracking results from TrackMate.
All statistical analysis was performed using the built-in statistical analysis tools in Prism (GraphPad, San Diego, Calif.), JMP (SAS), or MATLAB. The statistical details of each statistical analysis can be found in the Brief Description of the Drawings section, the Experiments, Results, and Discussion section, and Tables 3-78.
RNA Scaffolds can Assemble Fluorescent Sensors into Subcellular Puncta in Mammalian Cells
The MS2 system (Bertrand et al., 1998) and the PP7 system (Lim, Downey and Peabody, 2001), each of which comprises a specific viral RNA sequence and a viral coat protein that binds tightly to that sequence, were tested to determine whether either could be used to assemble fluorescent sensors fused to the protein component of each system onto the RNA component of the same system, in essence using the RNA components as scaffolds to assemble reporters fused to the protein components. Prior work (Bertrand et al., 1998; Wu, Chao and Singer, 2012) showed that fluorescent proteins fused to the RNA binding protein components from each of these systems could be used to fluorescently image the locations of mRNAs engineered to contain the RNA components. In studies described herein, fluorescent proteins previously described were replaced with a fluorescent reporter of calcium, the GFP-based sensor GCaMP6f (Chen et al., 2013), and a fluorescent CFP/YFP FRET reporter of cAMP, ICUE3 (DiPilato and Zhang, 2009) (
For the scaffold vector, a tandem array encoding 48 MS2 binding sites (“48×MBS”) and a tandem array encoding 72 PP7 binding sites (“72×PBS”) were cloned from 24-component arrays of each kind previously developed (Wu, Chao and Singer, 2012) (
Transfecting the MS2-based sensor vector with its cognate scaffold vector into HeLa cells resulted in GCaMP6f puncta formation, but the MS2-based sensor vector was unable to form GCaMP6f puncta on the orthogonal PP7 scaffold vector, or when no scaffold vector was provided (
As an additional confirmation that the fluorescent puncta were indeed scaffolded by the RNAs, RNA FISH was performed against the MS2 and PP7 RNA sequences, on HeLa cells transfected as described above. Almost all the sensor puncta also bore appropriate RNA FISH puncta (
The puncta assembled by RNA scaffolding of fluorescent reporters were tested to determine whether they were functional. MS2-scaffolded GCaMP6f and PP7-scaffolded ICUE3 were expressed in HeLa cells and the resultant signals elicited by extracellular stimuli (10 mM Ca2+ and 20 μM forskolin, respectively, as previously used (Williams et al., 2001; Miedlich, Gama and Breitwieser, 2002; Vay et al., 2007; Dipilato and Zhang, 2009)) were compared to those reported by the unscaffolded versions of the corresponding sensors. Both unscaffolded (
Next, the scaffolded reporters were examined to determine whether they were stationary in the cell, or whether they moved over time. In order for the post hoc identification of the sensor via immunostaining (or other highly multiplexed fixed cell imaging methods, such as serial FISH) to usefully explain the live cell imaging (
The density of puncta was also examined. Puncta should be separated by distances that are greater than the resolution limit of the microscope, or they will not be distinguishable. However, if they are too far apart, they may undersample any spatial heterogeneity of a biological signaling pathway within the cell that is desired to be measured. If obtaining a single measure from an entire cell is the goal, then this issue may not be a concern. But in cases where subcellular mapping of a biological signal is desired, having puncta spaced closer together than the characteristic length scales of the relevant biology will be helpful. For each punctum within a cell expressing either the MS2-scaffolded GCaMP6f or the PP7-scaffolded ICUE3, the distance to the nearest punctum was calculated (
Multiplexed Physiological Imaging of Ca2+ and cAMP in Single Cells Via Clustered Reporters of Overlapping Fluorescence Spectrum
The MS2-scaffolded GCaMP6f system and the PP7-scaffolded ICUE3 system, were co-expressed in single cells and assessed, using post hoc antibody staining against the HA and V5 epitopes respectively, to determine whether the orthogonality of clustering that was observed when the clusters were formed separately (
Having established the independence of these puncta, MS2 scaffolded GCaMP6f and PP7 scaffolded ICUE3 were then used to measure Ca2+ and cAMP simultaneously in the same cells, under 10 mM Ca2+ challenge. Interestingly, multiple kinds of responses emerged (
Several cells were exposed to sequential stimuli, first to 20 μM forskolin and then to 10 mM Ca2+. Cells again exhibited a variety of relationships between the Ca2+ and cAMP responses (
Neurons exhibit complex signaling cascades important for learning, memory, and development, and that are compromised in complex ways in disease states. Primary cultures of rodent hippocampal and cortical neurons have been critical for the study of calcium and cAMP signaling cascades (Sheng, Thompson and Greenberg, 1991; Bito, Deisseroth and Tsien, 1996; Hardingham, Arnold and Bading, 2001; Redmond, Kashani and Ghosh, 2002), and may be an ideal testbed for the deployment of spatially multiplexed indicators, as long as they are distributed closely enough to sample the relevant spatial scales of neurons (e.g., at different points on dendrites). Expression of the RNA scaffolded Ca2+ and cAMP indicators that successfully worked in HeLa cells did not result in puncta in neurons (
Live neuron imaging was performed on a spinning disk confocal microscope (a Yokogawa CSU-W1 Confocal Scanner Unit on a Nikon Eclipse Ti microscope) equipped with a 40×1.15 NA water immersion objective (Nikon MRD77410) and a Zyla PLUS 4.2 Megapixel camera controlled by NIS-Elements AR software. The filter set for GFP was used for imaging GFP intensity-based reporters (GCaMP6f and cAMPr), and the filter set for GFP and the 405 nm excitation filter was used for imaging 488 nm/405 nm excitation ratiometric reporters (ExRaiAKAR and ExRaiCKAR). The filter set for RFP was used for imaging RFP intensity-based reporters (RAB_EKARev). For electrophysiological characterizations of GCaMP6f and S1-GCaMP6f (
In the experiments in
After recording, z stacks were taken at 40× in the GFP and miRFP channels, and then a tiled image in the GFP channel at 10× covering a 4×4 grid and at 4× covering the entire glass coverslip was taken for registration of live images with the images from downstream immunostaining. Neurons were then fixed for 15 minutes in TissuePrep buffered 10% formalin, followed by washing with 100 mM glycine (Millipore Sigma, Burlington, Mass.) in 1×PBS for 15 minutes, and then washing with 1×PBS three times, 5 minutes each. Cells were stored in 1× PBS at 4° C.
Whole cell patch clamp recordings in were made using Axopatch 200B or Multiclamp 700B amplifiers, a Digidata 1440 digitizer, and a PC running pClamp (Molecular Devices, San Jose, Calif.). For in vitro current-clamp recordings, neurons were patched at 14-18 DIV (7-11 days after AAV transduction) to allow for sodium channel maturation. Neurons were bathed in room temperature Tyrode containing 125 mM NaCl, 2 mM KCl, 3 mM CaCl2, 1 mM MgCl2, 10 mM HEPES, 30 mM glucose and the synaptic blockers 0.01 mM NBQX and 0.01 mM GABAzine. The Tyrode pH was adjusted to 7.3 with NaOH and the osmolarity was adjusted to 300 mOsm with sucrose. For in vitro voltage-clamp recordings, neurons were patched at 19-21 DIV (17-20 days post-transfection) and were done under similar conditions as current-clamp recordings, except the Tyrode also contained 1 μM tetrodotoxin (TTX, Tocris Bioscience, Bristol, UK). For recordings, borosilicate glass pipettes (Warner Instruments, Holliston, Mass.) with an outer diameter of 1.2 mm and a wall thickness of 0.255 mm were pulled to a resistance of 5-10 MΩ with a P-97 Flaming/Brown micropipette puller (Sutter Instruments, Novato, Calif.) and filled with a solution containing 155 mM K-gluconate, 8 mM NaCl, 0.1 mM CaCl2, 0.6 mM MgCl2, 10 mM HEPES, 4 mM Mg-ATP, and 0.4 mM Na-GTP. The pipette solution pH was adjusted to 7.3 with KOH and the osmolarity was adjusted to 298 mOsm with sucrose.
For additional methods, see methods described in Example 1, and additional details provided herein.
Protein Scaffolds can Assemble Reporters into Functional Clusters in Neurons
De novo-designed short peptides of coiled-coil forming hetero-dimers (Tripet et al., 1996; Oakley and Kim, 1998; Moll et al., 2001; Shekhawat et al., 2009; Boyle et al., 2012; Gradišar et al., 2013) or homo-oligomers (Grigoryan et al., 2011; Zaccai et al., 2011; Fletcher et al., 2012; Huang et al., 2014; Negron and Keating, 2014; Thomson et al., 2014; Zhang et al., 2018) were tested to determine whether they could, when fused to GCaMP6f, help assemble GCaMP6f into puncta. Two to five of these motifs were fused to the N-terminus or C-terminus of GCaMP6f, with glycine and serine rich flexible linkers adapted from a previous study (Boyle et al., 2012; Indelicato et al., 2016) (Tables 1-2). None of these constructs produced puncta (Dc=1), perhaps because they formed small oligomers instead of larger protein assemblies, as had been observed in certain conditions (Boyle et al., 2012).
Next, de novo-designed self-assembling peptides were tested that were not coiled-coil forming proteins, but that had been shown in solution to form large protein assemblies beyond oligomers (such as polyhedrons). These peptides included the 13-01 peptide that forms a 60-subunit 25 nm sized 1.33 MDa protein dodecahedron (Hsia et al., 2016); other supramolecular assembly forming peptides were also tested (Gonen et al., 2015; Garcia-Seisdedos et al., 2017) (Tables 1-2). The 13-01 peptide was able to form identifiable puncta of GCaMP6f in neurons that were modestly brighter than the non-puncta cytosol (Dc=2); none of the other designs produced any puncta in neurons (Dc=1). Thus, 13-01 served as a prototype for a general strategy for creating such puncta, to which improvements could be made, including improved brightness, and as a baseline for identifying other peptides with similar properties.
It was hypothesized that self-assembling peptides could generate meaningful puncta if they were scalably assembled beyond the sizes achievable with a single self-assembling peptide: for example, if a fluorescent reporter was fused to two self-assembling peptides, it could form more scalable assemblies because each kind of reporter could enable new bridges to form (
A first goal of studies carried out was to increase the brightness of the puncta. To increase the number of GCaMP6f molecules per punctum, a short coiled-coil-forming homo-tetramer HexCoil-Ala [(Grigoryan et al., 2011); 30 amino acids long, abbreviated Coi11 in this study; four Coi11 peptides will assemble into a 12.8 kDa coiled-coil assembly] was added to the I3-01-fused GCaMP6f with the goal of allowing the I3-01-assembled puncta further assemble into even larger structures. The resulting dual self-assembling motif was named S1, and the reporter construct (including the immunoepitope Xpress for later antibody staining identification) was named S1-GCaMP6f (
S1-GCaMP6f had an extremely high Dc of >103 in the soma and >104 in the neurites (compare
The same strategy was further tested to determine whether it could be generalized to make many such clustering reagents. If the design were modular, and an existing fluorescent indicator could be adapted into a multiplexable form, compatible with simultaneous use with other similarly adapted reagents, any desired set of existing fluorescent reporters could in principle be enabled to become a multiplexable set for simultaneous imaging of multiple parts of a signal transduction network, without the laborious trial-and-error optimization common in protein engineering. Due to broad interest in the interplay between Ca2+, cAMP, and PKA dynamics, where feedforward and feedback connections between these signals has been shown to result in complex temporal dynamics of importance for driving cellular functions and states (Cooper, Mons and Karpen, 1995; Borodinsky and Spitzer, 2006; Mehta and Zhang, 2011; Sassone-Corsi, 2012), a high-performance cAMP reporter, cAMPr (Hackley, Mazzoni and Blau, 2018), and a high-performance protein kinase A (PKA) activity reporter, ExRaiAKAR (Mehta et al., 2018) were chosen for these experiments. The GFP-based, non-FRET-based cAMP sensor cAMPr (Hackley, Mazzoni and Blau, 2018) was chosen because a non-FRET-based sensor might require simpler optics than a FRET-based sensor, and might therefore facilitate the higher-speed simultaneous imaging of Ca2+ and cAMP in neurons by eliminating the need for filter switching for the single-camera microscopes commonly used in biology. Additionally, because cAMPr and GCaMP6f have identical fluorophores and therefore identical fluorescence spectra, one of skill in the art would expect that they would be indistinguishable to traditional fluorescence imaging strategies based upon spectral information. Both cAMPr and ExRaiAKAR are GFP-based (and thus cannot be used simultaneously with a conventional microscope).
cAMPr-based designs
cAMPr fusions to a variety of de novo-designed coiled-coil-forming peptides and polyhedron-forming peptides designed by analogy to the Coil1/I3-01 combination were screened. A short coiled-coil-forming homo-oligomer previously tested in GCaMP6f fusions was paired with each of the following self-assembling polyhedron-forming peptides: O3-33 (forms a 24-subunit 14 nm sized 480 kDa protein octahedron; King et al., 2012), T3-10 (forms a 12-subunit 11 nm sized 263 kDa protein tetrahedron; King et al., 2012), 3VDX (forms a 12-subunit 16 nm 593 kDa sized protein tetrahedron; Lai, Cascio and Yeates, 2012; Lai et al., 2016), ATC-HL3 (forms a 12-subunit 10 nm sized 365 kDa protein tetrahedron or a 24-subunit 20 nm sized 729 kDa protein octahedron; Lai et al., 2014), and T32-28 (forms a 24-subunit 15 nm sized 440 kDa protein tetrahedron; King et al., 2014), as well as with a group of fiber/foci-forming dihedral homomers (Garcia-Seisdedos et al., 2017) (Tables 1-2). In support of the generality of this approach, all five of these designs formed cAMPr puncta in cultured mouse hippocampal neurons (DC>1), although the expression level of the design with T32-28 was significantly lower than the other ones (
The construct with the largest Dc (Dc 102; see
To further explore the generality of this design strategy, a second successful punctum-forming cAMPr reporter, S2a-cAMPr, was thoroughly characterized using a different set of scaffolding peptides and showed excellent performance (
Movements of the puncta of S1-GCaMP6f (
Experimental results showed that the dual peptide-scaffolded SiRIs indeed were modularly assemblable: almost all the polyhedron/coil hybrid scaffolds that were tested provided DC>100, and a given scaffold could be used successfully with different sensors, showing the generality of the strategy (although of course, testing in cells is required for validation, as with any protein engineering project) (Table 2).
Additional constructs were made using ExRaiAKAR and were similarly tested to further demonstrate this strategy (
To further test the strategy and investigate Akt (Protein Kinase B (PKB)) activity, an S3-ExRaiAktAR reporter was prepared by replacing the ExRaiAKAR sequence in the S3-ExRaiAKAR polypeptide with ExRaiAktAR, a fluorescent sensor for Akt/PKB (Mehta et al., 2018). S3-ExRaiAktAR was expressed in cultured mouse hippocampal neurons, and as with S3-ExRaiAKAR, green puncta were observed at the soma and neurites in neurons via fluorescent imaging. Following stimulation of neurons expressing S3-ExRaiAktAR with N-Methyl-d-aspartic acid (NMDA) which is known to increase Akt activity, the S3-ExRaiAktAR green puncta reported increased Akt activity (data not shown).
Thus, the results of these experiments showed that the combination of polyhedron-assembling motifs and coiled-coil forming motifs enables multiple kinds of fluorescent reporter to self-assemble into clusters, effectively, robustly, and safely.
These experiments were performed to investigate whether the SiRI toolbox for Ca2+, cAMP, and PKA could be used simultaneously to map out relationships between signals within this complex network, in response to a biologically meaningful stimulus. Simultaneous measurements of Ca2+, cAMP, and PKA within a single cell is important because of the many ways that these signals may interact with one another, with both feedforward pathways and feedback loops potentially connecting them, in their coupling of cellular input to cellular output. For example, the time dynamics of one signal may influence the amplitude of another signal within this network, important because such relationships determine whether this signal transduction network may effectively drive a cellular output function or state in response to a given cellular input stimulus (Abrams, Karl and Kandel, 1991; Cooper, Mons and Karpen, 1995; Borodinsky and Spitzer, 2006; Mehta and Zhang, 2011; Sassone-Corsi, 2012).
See methods described in Examples 1-2 and described in Experiments, Results, and Discussion section.
First, S1-GCaMP6f, S2a-cAMPr, and the cell morphology marker miRFP were co-expressed in cultured mouse hippocampal neurons. Live cell imaging was performed at 20 frames per second in the GFP channel with extracellular stimulation by 5 μM forskolin, and two-color immunostaining was performed against the Xpress epitope (which was added as part of the S1-GCaMP6f protein;
As with HeLa cells, Ca2+, and cAMP responses in neurons exhibited different relationships that were apparent when simultaneously measured, but which would be lost if measured in separate populations of neurons and then compared in the aggregate. For example, when measured at the soma (
Spatially multiplexed imaging was further tested by co-expressing three (S1-GCaMP6f, S2a-cAMPr, and S3-ExRaiAKAR;
Simultaneous imaging of three fluorescent reporters of Ca2+, cAMP, and PKA that are based on different fluorescent proteins (Mehta et al., 2018) would require either filter switching and the reuse of the camera for serial imaging of the three channels, which may slow down the frame rate of imaging, or multiple light paths and cameras, increasing instrument complexity and expense. In addition, the use of multiple fluorophores would typically involve the risk of bleedthrough of light from one sensor into a separate spectral channel, requiring extensive characterization of how sets of sensors overlap before they are used in a given experiment, and computational correction to unmix the signals could add additional complexity or error due to potential sample-to-sample variation of the bleedthrough. These experiments therefore investigated whether the use of SiRIs with fluorophores all of the same color, made possible a recording rate of 5 seconds per volume, using just one filter set, and whether SiRIs avoided bleedthrough concerns due to their spatially multiplexed nature.
After performing live imaging, the neurons were fixed and three-color immunostaining was performed against the epitopes associated with each scaffold (
Ca2+, cAMP, and PKA activity was imaged in 24 neurons during and after 3 min of forskolin stimulation (
As referred to elsewhere herein, forskolin-driven cAMP has been shown to increase the frequency of spontaneous Ca2+ oscillations, in some, but not all, neurons thus treated (Gorbunova and Spitzer, 2002). It was observed that some cultured hippocampal neurons exhibited transient, often oscillatory, somatic Ca2+ responses, with each increase in Ca2+ concentration lasting 1 minute or less (
Because Ca2+ and cAMP signals could influence each other bidirectionally, experiments were then performed to investigate whether such variations in Ca2+ dynamics were associated with any variations in cAMP dynamics. Neurons that exhibited sustained, shorter-latency Ca2+ responses at the soma had faster rises in cAMP at the soma than did those with transient, longer-latency Ca2+ responses (
Thus, the simultaneous nature of SiRI-based imaging of multiple components of a signal transduction network allowed the derivation of relationships between the different components, and, ultimately, the analysis of how dynamics within the network related to network output. Importantly, such relationships could not be derived by inspecting
Spatially Multiplexed Imaging of More than 3 Fluorescent Reporters in Neurons, and Combined Spatial and Spectral Multiplexing
Spatially multiplexed imaging was further tested by co-expressing multiple (S1-GCaMP6f, S2a-cAMPr, S3-ExRaiAKAR, and S4-ExRaiCKAR;
When S1-GCaMP6f, S2-cAMPr, S3-ExRaiAKAR, and S4-ExRaiCKAR were co-expressed in neurons, all 4 kinds of puncta were observed (
To show functionality of all 4 sensors, neurons were challenged with two drugs, 50 μM forskolin (which as described elsewhere herein modulates cAMP, Ca2+, and PKA) for 3 minutes, followed after a break of 3 minutes by 100 ng/mL phorbol 12-myristate 13-acetate (PMA), a drug known to drive PKC activation, for 3 minutes, and observed responses of S1-GCaMP6f, S2-cAMPr, S3-ExRaiAKAR, and S4-ExRaiCKAR to these interventions (
Spatial multiplexing does not replace spectral multiplexing, but instead is complementary. To investigate the compatibility of spatial and spectral multiplexing techniques for even greater multiplexing capabilities, spatial and spectral multiplexing were combined by sharing one protein scaffold among multiple reporters that emitted different colors. The S2 scaffold, used in Example 1 for cAMPr, was adapted for a red fluorescent protein (RFP)-based reporter for extracellular signal-regulated kinase (ERK), RAB-EKARev (Ding et al., 2015; Mehta et al., 2018), resulting in S2-RAB_EKARev (
This combination of spatial and spectral multiplexing could enable the readout of SiRI reporter identity without requiring immunostaining: for example, in cells co-expressing S1-GCaMP6f, S3-ExRaiAKAR, and S3-RAB_EKARev (generated by replacing ExRaiAKAR in S3-ExRaiAKAR with RAB_EKARev), S3-ExRaiAKAR and S3-RAB_EKARev co-clustered into puncta because they shared the same protein scaffold (
Spatially multiplexed imaging of Ca2+ and PKA activities via two GFP-based sensors was performed during long-term potentiation to examine whether the methods described in the preceding Examples could also be used over longer time periods, and whether signals within the Ca2+/cAMP/PKA signal transduction network could be measured in an intact biological preparation.
Embryonic day (E) 15.5 timed-pregnant female Swiss Webster (Taconic Biosciences, Rensselaer, N.Y.) mice were deeply anaesthetized with 2% isoflurane. Uterine horns were exposed and periodically rinsed with warm sterile PBS. Plasmid DNA was injected into the lateral ventricle of one cerebral hemisphere of an embryo. Final plasmid DNA concentration was 4.5 μg/μl in water (DNA mass ratio of pAAV-CAG-S1-GCaMP6f, pAAV-CAG-S3-ExRaiAKAR, and pAAV-CAG-mRuby3-6×FLAG at 1:2:2). Fast Green FCF dye (Millipore Sigma, Burlington, Mass.) was added to the DNA mixture to visualize the mixture during DNA injection. Five voltage pulses (50 V, 50 ms duration, 1 Hz) were delivered two times using 5-mm round plate electrodes (Harvard Apparatus, Holliston, Mass.), with the cathode placed on top of the skull to target the hippocampus. Electroporated embryos were placed back into the dam, and allowed to mature for delivery.
3 to 4 week-old mice expressing SiRIs and miRFP (from in utero electroporation) were anesthetized with isoflurane and decapitated; the brains were taken out and placed in ice-cold choline-based cutting solution containing the following: 110 mM choline chloride, 7 mM MgCl2, 2.5 mM KCl, 0.5 mM CaCl2, 1.25 mM NaHPO4, 25 mM NaHCO3, 25 mM D-glucose, 11.6 mM ascorbic acid, and 3.1 mM pyruvic acid to a final pH of 7.75. The sagittal brain slices containing the hippocampus (300 μm) were cut in cold choline-based cutting solution with a Compresstome VF-300 (Precisionary Instruments, Natick, Mass.), and transferred to a holding chamber containing ACSF: 124 mM NaCl, 2 mM MgSO4, 2.5 mM KCl, 2 mM CaCl2, 1.2 mM NaHPO4, 24 mM NaHCO3, 5 mM HEPES, and 12.5 mM D-glucose to a final pH of 7.35, and recovered for 20 minutes at 30-32° C. Slices were subsequently maintained at room temperature until use. The cutting solution, ACSF, and ACSF with 50 μM forskolin added (or ‘forskolin-ACSF’) were constantly bubbled with 95% 02, 5% CO2. Slices were screened for positive green expression with a hand-held LED and filter.
Individual slices were transferred to a submersion perfusion chamber and continuously perfused with room temperature (23-26° C.) ACSF, constantly bubbled with 95% O2, 5% CO2, at a 4-5 ml/minute rate. Recording pipettes (4-6MΩ resistance) were pulled from borosilicate glass (Warner Instruments, Holliston, Mass.) and filled with ACSF. fEPSP recordings were made with a microelectrode amplifier (Multiclamp 700B, Molecular Devices, San Jose, Calif.). Signals were low-pass-filtered at 3 kHz and sampled at 20 kHz with a Digidata 1550B plus Humsilencer (Molecular Devices, San Jose, Calif.), and data were stored on a computer for subsequent offline analysis. In fEPSP recordings, recording pipettes were placed in the stratum radiatum of CA1. All evoked responses were elicited by delivering constant current pulses (duration 0.2 millisecond, 0.05 Hz) through a bipolar tungsten stimulating electrode placed into the mid stratum radiatum to activate Schaffer collateral/commissural fibers. In LTP experiments, stimulation intensity was adjusted to about 50% of the threshold for maximum fEPSPs and maintained at that level throughout all the recordings. Chemical LTP was induced by perfusing forskolin-ACSF, instead of ACSF, to the slice for 15 minutes, after which the perfusion was switched back to ACSF. fEPSP was recorded every 3 minutes, from 12 minutes before the onset of forskolin-ACSF perfusion to 57 minutes after the onset of forskolin-ACSF perfusion, in 24 fEPSP recordings. fEPSP slopes were measured by calculating the slopes between 10-60% of the fEPSP rising phase.
Acute brain slice neuron imaging was performed on a spinning disk confocal microscope (a Yokogawa CSU-W1 Confocal Scanner Unit on a Nikon Eclipse Ti microscope) equipped with a 40×1.15 NA water immersion objective (Nikon MRD77410) and a Zyla PLUS 4.2 Megapixel camera controlled by NIS-Elements AR software. Individual slices were transferred to a submersion perfusion chamber and continuously perfused with room temperature (23-26° C.) ACSF, constantly bubbled with 95% O2, 5% CO2, at a 4-5 ml/minute rate.
A volume containing the soma, apical dendrites, and basal dendrites of CA1 pyramidal neurons in the slice were imaged under the GFP (for SiRIs) and RFP (for the morphological marker mRuby3-FLAG) channels, and then volumetric time-lapse imaging (1.5-2.0 μm per Z step) was performed in the GFP emission channel (30 seconds per volume; each X-Y plane in the volume was captured twice, once under 405 nm excitation and the other under 488 nm excitation). Chemical stimulation was by perfusing forskolin-ACSF, instead of ACSF, to the slice for 15 minutes, after which the perfusion was switched back to ACSF. Time-lapse imaging started 5 minutes before the onset of forskolin-ACSF perfusion and ended 60 minutes after the onset of forskolin-ACSF perfusion, in total 65 minutes.
For the CA1 pyramidal neurons recorded from acute mouse brain slice, apical dendrites and basal dendrites were identified by cell morphology. For ExRaiAKAR or ExRaiCKAR, the time courses of the fluorescence intensity in the GFP emission channel under 488 nm and 405 nm excitations were measured as F_488 and F_405, respectively.
Significant photobleaching was observed in the fluorescence time courses measured from cAMPr and RAB_EKARev in recordings longer than 5 minutes in cultured neurons, as well as those measured from GCaMP6f and ExRaiAKAR in the 65-minute-long recordings in slice. Photobleaching correction was performed on these fluorescence time courses as described herein. Fluorescence time courses during the pre-stimulation baseline periods measured from cAMPr or RAB_EKARev were fitted to exponential decay functions, and the exponential decay components were then removed from the full-length time courses. Fluorescence time courses during the pre-stimulation baseline periods measured from GCaMP6f in the 65-minute-long recordings in slice were fitted to a bi-exponential decay function, and the bi-exponential decay components were then removed from the full-length time courses. The fluorescence time courses during the pre-stimulation baseline periods measured from ExRaiAKAR in the 65-minute-long recordings in slice under 488 nm excitation, i.e. F_488_baseline, were fitted to a bi-exponential decay function, and the bi-exponential decay components were then removed from the full-length time courses. The fluorescence time courses during the pre-stimulation baseline periods measured from ExRaiAKAR in slice under 405 nm excitation, i.e. F_405_baseline, were fitted to a bi-exponential decay function with the time constants fixed to those obtained from the curve fitting of the corresponding F_488_baseline time courses (so that the resultant time constants from the bi-exponential fitting were identical between each F_488_baseline/F_405_baseline pair), and the resulted bi-exponential decay components were then removed from the full-length time courses.
To calculate the dF/F0 for GCaMP6f, cAMPr, or RAB_EKARev expressing HeLa cells or neurons, the baseline fluorescence, F0, was first calculated as the average net fluorescence during the pre-stimulation baseline period. dF/F0 was then calculated as dF/F0=(F−F0)/F0. To calculate the signal-to-noise ratio (SNR) the maximum dF/F0 was divided by the standard deviation of the net fluorescence during the pre-stimulation baseline period.
To calculate the dR/R0 for ExRaiAKAR or ExRaiCKAR expressing HeLa cells or neurons, the baseline fluorescence, F0_488 and F0_405, was first calculated as the average net fluorescence during the pre-stimulation baseline period in the GFP emission channel under 488 nm and 405 nm excitations, respectively. dR/R0 was then calculated as dR/R0=(F_488/F_405)/(F0_488/F0_405)−1. To calculate the signal-to-noise ratio (SNR) the maximum dF/F0 was divided by the standard deviation of the net fluorescence during the pre-stimulation baseline period.
The slice was then fixed in TissuePrep buffered 10% formalin for 15 minutes at room temperature (RT) followed by incubation in 100 mM glycine in 1×PBS for 15-30 minutes at RT and three washes in 1×PBS. The slice was then stained against the epitope tags and morphological markers.
Additional materials and methods were as described in Examples 1-3 and in Experiments, Results, and Discussion section.
S1-GCaMP6f, S3-ExRaiAKAR, and the cell morphological marker mRuby3 were expressed in pyramidal neurons in mouse hippocampal area CA1 by targeted in utero electroporation, to examine the relationship between these signals over timescales of ˜1 hour. S1-GCaMP6f (green) and S3-ExRaiAKAR (green) SiRIs formed puncta (
Volumetric imaging of SiRIs was performed in in 14 hippocampal CA1 pyramidal neurons in acute brain slices before, during, and after a 15 minute administration of 50 forskolin (
At a population level, neurons exhibited a Ca2+ increase during the forskolin administration period, which decayed after forskolin removal, accompanied by a ramp up in PKA activity that was more sustained similar to the cultured neuron experiments of
Next, experiments were performed to determine whether neurons with immediate versus delayed Ca2+ signals had different PKA outputs from this signaling network. Neurons with immediate Ca2+ increases had PKA responses that were faster, and that achieved higher ultimate magnitudes, than those of neurons with delayed Ca2+ responses (
Responses in apical and basal dendrites (identified based on morphology) were then analyzed under forskolin stimulation. As at the soma, some dendrites exhibited immediate Ca2+ responses, while others exhibited delayed responses. Some also exhibited spontaneous Ca2+ activities before forskolin stimulation. However, analysis of PKA rise slope, ultimate amplitude, and duration of signaling showed no clear relationship to the timing of the Ca2+ signals (
Results of experiments and studies that have been performed, some of which are described herein, demonstrated that it is possible to localize, via engineered RNA and protein scaffolds that are bio-orthogonal to mammalian cells, spectrally similar or even spectrally identical fluorescent reporters of different biological signals at different points in space, safely clustered into bright, stationary puncta (with brightness 100-1000× brighter than the surrounding background) within living cells. This protein architecture, termed a signaling reporter island (SiRI), is very modular—that is, given a desired set of signals to be imaged at once within a single living cell, a set of existing fluorescent indicators may be reliably adapted by fusing each to a different pair of self-assembling peptides, so that each will cluster, stochastically, at a different set of points in space. These puncta end up several microns from each other, so that many different signals may be dynamically imaged within a living cell at the same time, even if they all possess the same fluorescent spectrum. After live imaging of the cell, post hoc reconstruction of sensor identity in fixed cells may proceed via epitope immunostaining, RNA FISH, or other highly multiplexed fixed cell imaging methods. In this way, multiple fluorescent signals may be recorded at different points in space, with minimal crosstalk, in a fashion where the identity of the signal may be clearly defined. SiRIs allow the high multiplexing capacity of fixed cell imaging to be translated to help the live cell imaging case, using the spatial dimension as an asset. Results indicated methods and compositions if the invention could be used for spatially multiplexed imaging to sample physiological signals with 1-2 micron resolution and that such imaging was able to capture the relevant cell biology; see, for example,
SiRIs are easy to design, with a highly modular architecture—most of the SiRIs designed and tested herein worked well upon initial validation testing, with minimal trial-and-error refinement required. In this way, SiRIs may offer a highly complementary strategy to traditional fluorescent reporter engineering: rather than having to engineer a set of fluorescent reporters with different fluorophores in order to use them simultaneously within the same cell, a nonmodular strategy that often requires significant post-design optimization and sometimes even directed evolution of each reporter (Piatkevich et al., 2018), one could simply create reporters all with a single fluorophore, or utilize existing fluorescent reporters without regard to the fluorophores utilized, and then to modularly attach them to different SiRI peptide pairs, without the need for significant post-design iterative work.
Unlike live cell imaging, where distinguishing different fluorescent reporters on a conventional microscope almost exclusively relies on having different excitation and/or emission spectra for the fluorescent reporters, fixed cell imaging may be highly multiplexed. For example, fluorescent probes that bind to different targets may be washed in and out over many cycles, with some procedures (e.g., serial antibody staining against immunoepitopes (Micheva and Smith, 2007; Murray et al., 2015), serial FISH against RNA sequences (Moffitt et al., 2016; Shah et al., 2017)) enable dozens to even hundreds of distinct biomolecules to be identified. In addition, combinations of immunoepitopes as protein barcodes (Wroblewska et al., 2018) may be used in methods of the invention to tag individual kinds of fluorescent reporters, expanding the number of fluorescent reporters that may be identified in a single round of antibody staining. Here, the high degree of multiplexing of fixed cell imaging technologies has been used and demonstrated to boost the multiplexing capability of live cell imaging technologies.
Important components of the success of this strategy are the findings that the scaffolded reporters of the invention are stationary over timescales appropriate for live cell imaging (else the identity of a given punctum could not be reconstructed), and that the puncta be located distant enough to be resolved by a microscope but close enough to spatially sample the relevant biology. It was observed that the puncta engineered in these studies were spaced typically a few microns from each other, and thus appropriate for cellular imaging and also for subcellular imaging where length scales of a few microns do not matter. In the future, creating a wide diversity of such scaffolds with different kinds of spacing may be used to allow tradeoffs between the number of signals that may be simultaneously observed, and the spatial sampling that is permitted. For a cell with 10,000 resolvable spots, for example, one might have 100 kinds of sensors distributed at each of 100 points, or 10 sensors distributed at each of 1000 points. Reported herein are >10 candidate scaffolds that showed at least some degree of clustering (both RNA-based and protein-based), which enable many different kinds of signals to be observed simultaneously in a single cell. Programmable RNA scaffolds such as the Pumby system (Adamala, Martin-Alarcon and Boyden, 2016), new kinds of engineered protein polyhedra, and the simultaneous use of RNA scaffolds and protein polyhedral systems together in cells that may permit the utility of both, rapidly allow the expansion of this system. Certain embodiments of methods of the invention also include de novo/computationally designed, bio-orthogonal protein motifs (King et al., 2012; Hsia et al., 2016; Huang, Boyken and Baker, 2016; Lai et al., 2016; Garcia-Seisdedos et al., 2017) and RNA motifs (Delebecque et al., 2011). Methods of the invention may also be used in vivo in multiple model organisms of interest in biology, such as C. elegans, zebrafish, Drosophila, and mice, to monitor simultaneous pathways in the cells of an intact organism.
Coupling between Ca2+, cAMP, and PKA signals, three signals of immense importance for a large number of normal biological functions, and that go awry in many pathological contexts, was examined herein in cultured hippocampal neurons and in acute mouse hippocampal brain slices. Understanding how these signals relate to each other is complex, because of the many feedforward and feedback connections between these signals. Using neurons of both cultured hippocampus and acute hippocampal slice, experiments described herein investigated how the timing of Ca2+ and cAMP signals relate to the amplitude of PKA signals. In summary, neurons that had shorter-latency cAMP and Ca2+ responses to forskolin, exhibited stronger PKA activation than neurons with longer-latency cAMP and Ca2+ responses, showing how an output signal like PKA may be governed by the properties of the second messengers upstream. In cultured neurons, the neurons with longer-latency Ca2+ responses also exhibited transient, even oscillatory, responses, indicated that the interactions within this signal transduction network might generate extremely complex, nonlinear, dynamics with computational consequences for how cells filter, integrate, and combine information towards the generation of precise cellular outputs. Importantly, such relationships between the different components of this signal transduction network could not be derived by observing these signals in separate cells, and then comparing them after averaging.
Spatial multiplexing may be particularly useful for imaging fast cellular dynamics, such as those in neurons, under inexpensive single-camera microscopes with fluorescent sensors that may all be imaged in one shared optical channel (e.g. using all GFP-based sensors), because using methods of the invention, the imaging speed is not limited by the number of cameras available or any mechanical filter switching required to record from multiple channels. By reducing the reliance on spectral multiplexing, methods of the invention may also avoid concerns related to bleedthrough caused by using multiple, spectrally similar fluorescent reporters within the same cell. Spatial multiplexing methods of the invention are also capable of freeing up optical channels for use for other purposes, such as cellular control. For example, the GFP channel may be used to observe cellular activity readout from multiple GFP-based sensors, and then the red channel of the microscope may be utilized to operate red-light driven optogenetic tools (Chuong et al., 2014; Klapoetke et al., 2014).
DiPilato, L. M. and Zhang, J. (2009) “The role of membrane microdomains in shaping beta2-adrenergic receptor-mediated cAMP dynamics.,” Molecular bioSystems, 5(8), pp. 832-7. doi: 10.1039/b823243a.
Ménard, C. and Quirion, R. (2012) “Group 1 metabotropic glutamate receptor function and its regulation of learning and memory in the aging brain,” Frontiers in Pharmacology, 3 OCT. doi: 10.3389/fphar.2012.00182.
Wu, B., Chao, J. A. and Singer, R. H. (2012) “Fluorescence Fluctuation Spectroscopy Enables Quantitative Imaging of Single mRNAs in Living Cells,” Biophysical Journal, 102(12), pp. 2936-2944. doi: 10.1016/j.bpj.2012.05.017.
Although several embodiments of the present invention have been described and illustrated herein, those of ordinary skill in the art will readily envision a variety of other means and/or structures for performing the functions and/or obtaining the results and/or one or more of the advantages described herein, and each of such variations and/or modifications is deemed to be within the scope of the present invention. More generally, those skilled in the art will readily appreciate that all parameters, dimensions, materials, and configurations described herein are meant to be exemplary and that the actual parameters, dimensions, materials, and/or configurations will depend upon the specific application or applications for which the teachings of the present invention is/are used. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. It is, therefore, to be understood that the foregoing embodiments are presented by way of example only and that, within the scope of the appended claims and equivalents thereto; the invention may be practiced otherwise than as specifically described and claimed. The present invention is directed to each individual feature, system, article, material, and/or method described herein. In addition, any combination of two or more such features, systems, articles, materials, and/or methods, if such features, systems, articles, materials, and/or methods are not mutually inconsistent, is included within the scope of the present invention.
All definitions, as defined and used herein, should be understood to control over dictionary definitions, definitions in documents incorporated by reference, and/or ordinary meanings of the defined terms.
The indefinite articles “a” and “an,” as used herein in the specification and in the claims, unless clearly indicated to the contrary, should be understood to mean “at least one.” The phrase “and/or,” as used herein in the specification and in the claims, should be understood to mean “either or both” of the elements so conjoined, i.e., elements that are conjunctively present in some cases and disjunctively present in other cases. Other elements may optionally be present other than the elements specifically identified by the “and/or” clause, whether related or unrelated to those elements specifically identified, unless clearly indicated to the contrary.
All references, patents and patent applications and publications that are cited or referred to in this application are incorporated by reference in their entirety herein.
This application claims benefit under 35 U.S.C. § 119(e) of U.S. Provisional application Ser. No. 62/899,416 filed Sep. 12, 2019, the disclosure of which is incorporated by reference herein in its entirety.
This invention was made with government support under W911NF1510548 6933228 awarded by the U. S. Army Research Laboratory and the U. S. Army Research Office; NIH 1R24MH106075 6930901, NIH R44EB021054 6935482, NIH 1R01DA045549 6937110, NIH 1R01MH114031 6937063, NIH 2R01DA029639 6932279, NIH 1R01EB024261 6936689, NIH Director's Pioneer Award 1DP1NS087724 6928706, and NIH 1R01GM104948 6926932 awarded by the National Institutes of Health; and NSF CBET 1344219 6928628 awarded by the National Science Foundation. The Government has certain rights in the invention.
Number | Date | Country | |
---|---|---|---|
62899416 | Sep 2019 | US |