WO2025071367A1 - Protéine de fusion modulant le calcium pouvant être exprimée par un virus adéno-associé - Google Patents
Protéine de fusion modulant le calcium pouvant être exprimée par un virus adéno-associé Download PDFInfo
- Publication number
- WO2025071367A1 WO2025071367A1 PCT/KR2024/014799 KR2024014799W WO2025071367A1 WO 2025071367 A1 WO2025071367 A1 WO 2025071367A1 KR 2024014799 W KR2024014799 W KR 2024014799W WO 2025071367 A1 WO2025071367 A1 WO 2025071367A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- monstim1
- protein
- stim1
- cry2
- gfp
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/415—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from plants
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
- C07K14/4701—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
- C07K14/4702—Regulators; Modulating activity
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
- C12N15/86—Viral vectors
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/56—Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
- C07K2317/569—Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/40—Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation
- C07K2319/42—Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation containing a HA(hemagglutinin)-tag
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/40—Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation
- C07K2319/43—Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation containing a FLAG-tag
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2750/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
- C12N2750/00011—Details
- C12N2750/14011—Parvoviridae
- C12N2750/14111—Dependovirus, e.g. adenoassociated viruses
- C12N2750/14141—Use of virus, viral particle or viral elements as a vector
- C12N2750/14143—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
Definitions
- It relates to a calcium-regulated fusion protein that can be expressed by adeno-associated virus.
- Ca2 + Calcium ion
- Intracellular Ca2 + signals are tightly regulated in space and time, and abnormal regulation in the central nervous system has been associated with various neurological disorders, such as epilepsy, chronic pain, psychiatric diseases, and neurodegeneration.
- Brain cells possess molecular machinery, such as channels and pumps located in the plasma membrane and membranes of cell organelles, that regulate Ca2 + signals.
- Ca2 + signals regulate neuronal functions, such as synaptic plasticity, neurotransmitter release, and gene expression, and their functions vary depending on various parameters, such as signal amplitude, duration, and location.
- Ca2 + signals also play a role in regulating important processes, such as neurotransmitter release, homeostasis, and immune responses, in glial cells.
- CRAC Ca 2+ -release-activated Ca 2+
- STIM1 Short interaction molecule 1
- SOCE store-operated Ca 2+ entry
- the first type of optogenetic actuator utilizes the light-oxygen-voltage-sensing domain 2 (LOV2) domain of the phototropin isolated from oat ( Avena Sativa) .
- LOV2 domain light-oxygen-voltage-sensing domain 2
- Conjugation of the LOV2 domain to an active STIM1 fragment causes steric hindrance, which inhibits STIM1 activity in the absence of light.
- Blue light stimulation induces a conformational change within the LOV2 domain that dissociates it from the C-terminal J ⁇ helix. This dissociation relieves STIM1 inhibition, allowing activation of the CRAC channel and increasing intracellular Ca2 + levels.
- the second type of optogenetic actuator utilizes cryptochrome 2 (CRY2) isolated from Arabidopsis thaliana . Taking advantage of the light-mediated homo-interaction property of CRY2, the cytoplasmic fragment of STIM1 binds to CRY2 and undergoes homo-oligomerization upon light stimulation. Homo-oligomerized STIM1 translocates to the cell membrane and activates CRAC channels, mimicking the natural action mechanism of STIM1 (Fig. 1a).
- This second type of cryptochrome 2 (CRY2)-based optogenetic actuator is named OptoSTIM1.
- the OptoSTIM1 has been demonstrated to be effective in activating Ca2 + influx in various cell types and successfully controls targeted brain functions (Nat Biotechnol. 2015;33:1092-6). Although LOV2 domain-based actuators have the advantage of faster kinetics for (de)activation and smaller size compared to OptoSTIM1, OptoSTIM1 was found to induce larger fold changes in regulating intracellular Ca2 + levels (Cell Calcium. 2017;64:36-46).
- adeno-associated virus is widely used in neuroscience research and gene therapy due to its high gene transfer efficiency, low host genome integration probability, and low immunogenicity.
- AAV capsid engineering have led to the development of systemic delivery technologies that enable targeted expression in specific tissues and cells.
- AAV cassette components including the coding sequence of monSTIM1 and inverted terminal repeats (ITRs), woodchuck hepatitis virus post-transcriptional regulatory element (WPRE), and human growth hormone (hGH) polyadenylation signal
- the total sequence size excluding the promoter is ⁇ 5.1 kb, which exceeds the packaging capacity of adeno-associated virus (AAV) ( ⁇ 5.0 kb) (Fig. 1b).
- Lentiviruses can carry larger genes, but they have the property of integrating into the host genome, which may raise safety issues for gene therapy, and their limited diffusion within brain tissue reduces the number of transduced cells.
- monSTIM1 variants AAV-compatible monSTIM1 variants
- the monSTIM1 variants of the present invention efficiently increase intracellular Ca2 + levels in various brain cells such as neurons, astrocytes, and microglia, and maintain the ultra-high light sensitivity property of the existing monSTIM1, so that they can effectively induce Ca2 + -mediated gene expression in neurons and astrocytes of the mouse brain in response to non-invasive light illumination.
- One aspect is to provide a fusion protein comprising a Tag protein; CRY2 (cryptochrome 2) protein or a variant thereof; and STIMI1 (Stromal interaction molecule 1) protein.
- Another aspect is to provide a polynucleotide encoding the above fusion protein.
- Another aspect is to provide a vector comprising a polynucleotide encoding the fusion protein.
- Another aspect is to provide a Ca2+ modulator comprising STIMI1 (Stromal interaction molecule 1) protein to which CIB1 (cryptochromeinteracting basic-helix-loop-helix 1) protein or a fragment thereof is attached; and CRY2 (cryptochrome 2) protein or a variant thereof.
- a Ca2 + modulator comprising a STIMI1 protein to which a tag protein is attached; and a CRY2 protein or a variant thereof to which a nanobody antibody capable of binding to the tag protein is attached.
- Another aspect provides an expression system comprising a first vector comprising a polynucleotide encoding a STIMI1 protein or a fragment thereof; and a second vector comprising a polynucleotide encoding a CRY2 protein or a variant thereof.
- One aspect provides a fusion protein comprising a Tag protein; CRY2 (cryptochrome 2) protein or a variant thereof; and STIMI1 protein.
- tag protein in this specification refers to a protein that is attached to a specific protein and used to track or purify the location of the protein.
- the tag is connected to the C-terminus or N-terminus of the protein, and allows the expression level of the protein to be monitored or easily detected.
- the tag protein can be a fluorescent protein.
- the fluorescent protein can be green fluorescent protein (GFP), yellow fluorescent protein (YFP), red fluorescent protein (RFP), orange fluorescent protein (OFP), cyan fluorescent protein (CFP), blue fluorescent protein (BFP), far-red fluorescent protein, or a tetracysteine motif.
- the green fluorescent protein is EGFP (enhanced green fluorescent protein), Emerald (Tsien, Annu. Rev. Biochem., 67: 509-544, 1998), Superfolder (Pedelacq et al., Nat.
- the green fluorescent protein may be composed of an amino acid sequence of SEQ ID NO: 41.
- the tag protein may be an epitope tag.
- the epitope tag is a short amino acid sequence that is added to a foreign protein and acts as a label that can be recognized by a specific antibody, and may be, for example, Myc, V5, T7, AU1/AU5, VSV-G (Vesicular Stomatitis Virus Glycoprotein), and preferably, FLAG or HA, but is not limited thereto.
- the FLAG is an epitope tag composed of an eight amino acid sequence (DYKDDDDK), and HA is an abbreviation for Hemagglutinin epitope tag, and is an epitope tag composed of a nine amino acid sequence (YPYDVPDYA) derived from the hemagglutinin protein of influenza A virus.
- CRY2 (cryptochrome 2) protein used in this specification refers to a protein that mainly detects blue light and regulates circadian rhythm, and is found in plants, fungi, and animals.
- the CRY2 (cryptochrome 2) protein may be extracted from Arabidopsis thaliana , but is not limited thereto.
- fragment is intended to mean a polypeptide consisting only of a portion of the complete polypeptide sequence and structure, wherein a C-terminal deletion or an N-terminal deletion of the variant may be present.
- the CRY2 fragment means a functional fragment of a CRY2 polypeptide having CRY2 activity.
- variant may mean a change in the basic structure or sequence of a gene or protein.
- the CRY2 variant may refer to a CRY2(E281A) mutation.
- the CRY2(E281A) mutation reduces the basal activity of OptoSTIM1 in the dark and increases sensitivity to blue light.
- the CRY2 variant may be a combination of CRY2(E281A) and A9 conjugate (CRY2(E281A, A9)).
- the CRY2(E281A, A9) has significantly increased light-dependent oligomerization and sensitivity to blue light.
- the CRY2 (E281A, A9) protein may be composed of an amino acid sequence of SEQ ID NO: 31.
- STIMI1 (Stromal interaction molecule 1) protein in this specification is a transmembrane protein present in the endoplasmic reticulum (ER) membrane of cells, and is a key regulator of the SOCE (store-operated Ca2+ entry) process. It plays a role in sensing the Ca2 + level in the lumen of the ER, and when ER Ca2 + is depleted, STIM1 oligomerizes and undergoes a conformational change in the C-terminal domain. It then translocates to the plasma membrane, binds to and activates the CRAC (Ca2+-release-activated Ca2+) channel, thereby increasing the intracellular Ca2 + level.
- CRAC Ca2+-release-activated Ca2+
- the STIMI1 protein may be a cytosolic STIM1 fragment.
- the cytosolic STIM1 fragment may refer to the cytosolic carboxyl terminus (STIM1ct) of the STIM1 protein, and specifically, may refer to a portion of the amino acid sequence 238-685 of the STIM1 protein.
- the STIM1ct may consist of an amino acid sequence of SEQ ID NO: 30.
- the cytoplasmic STIM1 fragment may include a STIM1 CRAC activation domain (CRAC-activation domain, CAD; 342-448), which plays a role in binding to and opening a Ca2+ channel.
- CRAC-activation domain CAD; 342-448
- the STIM1 CRAC activation domain may be a portion of 342-448 of the STIM1 protein and may consist of an amino acid sequence of SEQ ID NO: 34.
- the cytoplasmic STIM1 fragment may be, but is not limited to, a portion of the amino acid sequence 238-685 (SEQ ID NO: 30), 238-448 (SEQ ID NO: 35), 248-448 (SEQ ID NO: 36), 336-448 (SEQ ID NO: 37), 318-463 (SEQ ID NO: 38), 318-450 (SEQ ID NO: 39), or 342-483 (SEQ ID NO: 40) of the STIMI1 protein.
- Another aspect provides a polynucleotide encoding the fusion protein.
- the vector may be a recombinant expression vector capable of expressing the fusion protein.
- the vector may be an adeno-associated virus (AAV) vector.
- AAV adeno-associated virus
- monSTIM1 in this specification may mean a structure comprising a fusion of GFP (Green Fluorescent Protein), cryptochrome 2 (CRY2), and STIM1 protein.
- the STIM1 protein may mean a STIM1 fragment
- the CRY2 may be a mutant (CRY2 (E281A, A9)) combining CRY2 (E281A) and A9.
- the CRY2 variant (E281A, A9) may be composed of the amino acid sequence of SEQ ID NO: 31.
- monSTIM1 variants in this specification means a variant of the structure of monSTIM1.
- the monSTIM1 variant may mean that a small tag protein other than GFP is replaced in the monSTIM1.
- the tag protein may refer to FLAG or HA.
- the tag protein may be composed of an amino acid sequence of SEQ ID NO: 32 or SEQ ID NO: 33.
- the monSTIM1 variant may mean a STIM1 protein that is truncated from a portion of the STIM1 protein in the monSTIM1.
- the truncated STIM1 protein may be a cytoplasmic STIM1 fragment, and the cytoplasmic STIM1 fragment may be a portion of the amino acid sequence 238-685 (SEQ ID NO: 30), 238-448 (SEQ ID NO: 35), 248-448 (SEQ ID NO: 36), 336-448 (SEQ ID NO: 37), 318-463 (SEQ ID NO: 38), 318-450 (SEQ ID NO: 39), or 342-483 (SEQ ID NO: 40) of the STIMI1 protein, but is not limited thereto.
- the monSTIM1 variant may mean that the monSTIM1 is separated into two structures.
- the two structures may include a first vector comprising a polynucleotide encoding a STIMI1 protein or a fragment thereof; and a second vector comprising a polynucleotide encoding a CRY2 protein or a variant thereof.
- a Ca2+ modulator comprising a STIMI1 protein to which a CIB1 (cryptochromeinteracting basic-helix-loop-helix 1) protein or a fragment thereof is attached; and a CRY2 protein or a variant thereof.
- CIB1 cryptoochromeinteracting basic-helix-loop-helix 1
- CIB1 (cryptochromeinteracting basic-helix-loop-helix 1) protein in this specification refers to a bHLH (basic-helix-loop-helix) transcription factor found mainly in plants, which interacts with CRY2 (Cryptochrome 2) and is activated by blue light.
- the fragment of the CIB1 protein may mean an N-terminal fragment of the CIB1 protein (CIBN).
- the fragment of the CIB1 protein may consist of an amino acid sequence of SEQ ID NO: 42.
- Ca2+ modulator used in this specification refers to a substance that regulates cell function by controlling the concentration, influx, and release of calcium, and mainly regulates calcium channels or calcium pumps to influx or release calcium into cells and regulates cell activity through signal transduction processes.
- a Ca2+ modulator comprising a STIMI1 protein to which a tagged protein is attached; and a CRY2 protein or a variant thereof to which a nanobody antibody capable of binding to the tagged protein is attached.
- nanobody antibody as used herein means a type of single-domain antibody (sdAb) derived from a heavy chain antibody of camelids or cartilaginous fish, which has no light chain and contains only the VHH domain of the heavy chain.
- sdAb single-domain antibody
- the nanobody is meant to be capable of specifically binding to the tag protein.
- the GFP nanobody (vhhGFP) may be composed of the amino acid sequence of SEQ ID NO: 43.
- the nanobody antibody capable of binding to the tag protein can be attached to the N-terminus or C-terminus of the CRY2 protein or a variant thereof.
- one or more copies of the nanobody antibody capable of binding to the tag protein can be attached to the N-terminus or the C-terminus of the CRY2 protein or a variant thereof.
- one, two or three copies of the nanobody antibody capable of binding to the tag protein can be attached to the N-terminus or the C-terminus of the CRY2 protein or a variant thereof.
- one, two or three copies of a GFP nanobody (vhhGFP) can be fused to the N- or C-terminus of the CRY2 protein or a variant thereof.
- Another aspect provides an expression system comprising a first vector comprising a polynucleotide encoding a STIMI1 protein or a fragment thereof; and a second vector comprising a polynucleotide encoding a CRY2 protein or a variant thereof.
- the second vector may comprise a polynucleotide encoding a nanobody antibody capable of binding to a tag protein.
- a calcium-regulatory fusion protein expressible by adeno-associated virus was constructed by reducing the size of the existing monSTIM1 coding sequence to fit the AAV packaging capacity, thereby establishing an AAV-based system that can be expressed in neurons and glial cells of the mouse brain.
- the monSTIM1 variant expressed by AAV according to one aspect can minimize problems such as tissue damage, glial scar formation, inflammation, and tissue heating caused by long-term optical fiber insertion, and has the effect of regulating neural activity in a noninvasive manner.
- Figure 1 illustrates the optogenetic activation pattern of monSTIM1 mutants or OptoCRAC in one aspect:
- FIG. 1a is a schematic diagram showing the mechanism of action of GFP-monSTIM1;
- FIG. 1b is a structural diagram showing the coding sequence of monSTIM1 and the sizes of components of the AAV cassette;
- FIG. 1c is a photograph showing a fluorescent image of a cell co-expressing R-GECO1 and one aspect of monSTIM1 variants or OptoCRAC;
- FIG. 1d is a graph showing the change in normalized intensity of R-GECO1 over time according to the transient activation of one aspect of monSTIM1 variants and OptoCRAC;
- FIG. 1e is a graph showing the maximum fold change ( ⁇ F/F0) of R-GECO1 intensity for one aspect of monSTIM1 variants and OptoCRAC upon blue light stimulation;
- FIG. 1a is a schematic diagram showing the mechanism of action of GFP-monSTIM1
- FIG. 1b is a structural diagram showing the coding sequence of monSTIM1 and the sizes of components of the AAV cassette
- FIG. 1f is a graph quantifying the activation kinetics of one aspect of monSTIM1 variants
- FIG. 1g is a graph quantifying the deactivation kinetics of one aspect of monSTIM1 variants
- Figure 1h is a graph showing the light sensitivity of each optogenetic module (EGFP-monSTIM1, FLAG-monSTIM1, HA-monSTIM1, OptoCRAC).
- Figure 2 shows the structures of six fusion proteins of CRY2-fused STIM1 fragments containing the STIM1 CRAC activation domain (CAD; 342-448) and the correlation between the expression levels of these proteins and basal R-GECO1 intensity:
- Figure 2a is a schematic diagram showing the structure of CRY2 fused STIM1 fragments
- Figure 2b is a graph showing the expression levels of six CRY2 fused STIM1 fragments (monSTIM1 variants) containing the STIM1 CRAC activation domain (CRAC-activation domain, CAD; 342-448);
- Figure 2c is a photograph showing the result of fluorescence images of R-GECO1 in HeLa cells co-expressing each CRY2 fused STIM1 fragment (monSTIM1 variant sets 1-6) under blue light illumination
- Figure 2d is a graph showing the maximum fold change (( ⁇ F/F0) of R-GECO1 intensity upon activation of each CRY2-fused STIM1 fragment (monSTIM1 mutant set 1-6)
- Figure 2e is a graph showing the fluorescence intensity of R-GECO1 in the absence of blue light
- Figure 2f is a graph showing the correlation between the expression level of CRY2-fused STIM1 fragments and the basal R-GECO1 intensity
- Fig. 2j is a graph showing the fluorescence intensity of R-GECO1 in CRY2 fused STIM1 fragments (set 1-7) whose expression is induced by each IRES2 in the absence of blue light.
- FIG. 3 shows the system that separates the monSTIM1 protein into two components and the correlation between the expression levels of these proteins and the basal R-GECO1 intensity:
- Figure 3a is a schematic diagram showing the working mechanism of the two-component system of monSTIM1 protein
- Figure 3b is a schematic diagram showing the fusion structure of the system in which monSTIM1 protein is separated into two components
- Figure 3c is a photograph showing the fluorescence image of R-GECO1 in HeLa cells co-expressing each protein pair (set 1-5) when illuminated with blue light
- Figure 3d is a graph showing the maximum fold change (( ⁇ F/F0)) of R-GECO1 intensity upon activation of monSTIM1 mutants
- Figure 3e is a graph showing the fluorescence intensity of R-GECO1 in the absence of blue light.
- Figure 4 is a graph showing the relative Ca2+ levels measured by Fura-2 imaging in cells expressing each monSTIM1 mutant (EGFP-monSTIM1; FLAG-monSTIM1; GFP-CRY2-STIM1(318-450); GFP-IRES2-CRY2-STIM1(238-448); GFP-STIM1+vhhGFP-CRY2; CIBN-STIM1+CRY2) (left: Fura-2 ratio measured under dark conditions (emission 340 nm/380 nm); right: Fura-2 ratio measured after blue light irradiation).
- Figure 5 shows the Ca2 + increase induced by activation of one monSTIM1 mutant in cultured hippocampal neurons:
- Figure 5a is a schematic diagram illustrating the light illumination protocol
- Figure 5b is a photograph (top) showing fluorescence images of R-GECO1 illuminated with blue light in neurons expressing each monSTIM1 variant (EGFP-monSTIM1; FLAG-monSTIM1; GFP-CRY2-STIM1(318-450); GFP-IRES2-CRY2-STIM1(238-448); GFP-STIM1+vhhGFP-CRY2; CIBN-STIM1+CRY2) and a graph (bottom) showing the relative change in R-GECO1 intensity over time upon repetitive light illumination;
- Figure 5c is a graph showing the maximum fold change (( ⁇ F/F0)) in R-GECO1 intensity upon activation of monSTIM1 variants.
- Figure 6 shows that activation of one monSTIM1 mutant in cultured astrocytes and microglia increases Ca2 + :
- Figure 6a is a photograph showing R-GECO1 fluorescence images in astrocytes (left) illuminated with blue light and microglia (BV2 cells, right) co-expressing each monSTIM1 mutant (EGFP-monSTIM1; FLAG-monSTIM1; GFP-CRY2-STIM1(318-450); GFP-IRES2-CRY2-STIM1(238-448); GFP-STIM1+vhhGFP-CRY2; CIBN-STIM1+CRY2);
- Figure 6b is a schematic illustrating the illumination protocol;
- Figure 6c is a graph showing the relative change in R-GECO1 intensity over time upon illumination;
- Figure 6d is a graph showing the maximum fold change (( ⁇ F/F0) of R-GECO1 intensity upon activation of the monSTIM1 mutant;
- Figure 6e is a photograph showing a fluorescence image of R-GECO1 in astrocytes when light was shined on the subcellular region indicated by the circle with the white line
- FIG. 7 shows the results of application of one aspect of monSTIM1 mutants in neurons:
- Figure 7a is a schematic representation of the experiment in which one AAV-compatible monSTIM1 variant was expressed under the control of the CaMKII ⁇ promoter targeting the CA1 region of the hippocampus (top) and mice were irradiated with blue LED light via a custom transcranial light illumination system, followed by retention for immunohistochemistry and analysis (bottom);
- Figure 7b is a schematic representation indicating the region used for quantification of activated neurons expressing cFos and monSTIM1 variants in the CA1 region (two AAV-compatible monSTIM1 variants were used: AAV-CaMKII ⁇ -FLAG-monSTIM1 and AAV-CaMKII ⁇ -EGFP-STIM1(318-450).
- Figure 7c is a representative image showing cFos-positive cells expressing each monSTIM1 mutant, with or without non-invasive light delivery and when expressing EGFP (control) (scale bar, 50 ⁇ m);
- Figure 7d is a graph quantifying cFos-positive cells expressing FLAG-monSTIM1
- Figure 7e is a graph quantifying cFos-positive cells expressing GFP-CRY2-STIM1(318-450).
- Figure 8 shows the results of applying one type of monSTIM1 mutant to astrocytes:
- Figure 8a is a photograph showing FLAG-monSTIM1 expressed under the control of the GfaABC1D promoter targeting the CA1 region of the hippocampus (green, FLAG; red, c-Fos; blue, DAPI. Scale bar, 100 ⁇ m);
- Figure 8b is a photograph showing cFos-positive cells expressing FLAG-monSTIM1 with or without noninvasive light delivery (green, FLAG; blue, DAPI; pink, GFAP; red, c-Fos. Scale bar, 50 ⁇ m);
- Figure 8c is a graph showing the quantification of cFos-positive cells expressing FLAG-monSTIM1 in CA1 astrocytes.
- Figure 9 is a photograph showing an image showing SST-positive cells expressing FLAG-monSTIM1 in the SST-Cre mouse line (blue is DAPI, green is FLAG, and red is tdTomato).
- the expression plasmid for R-GECO1 (Addgene plasmid #32444) was obtained from Addgene.
- the monSTIM1 sequence of GFP-monSTIM1 was amplified by polymerase chain reaction (PCR) using HA-F, FLAG-F, and HA-R primers. The amplified sequence was ligated to GFP-monSTIM1 using NheI and SalI restriction sites after excising EGFP.
- the LOV2 (404-546) sequence of OptoCRAC was PCR amplified using LOV2-F and LOV2-R primers and ligated into the EGFP-C1 vector at the BsrGI and HindIII sites to generate the EGFP-LOV2 vector.
- the STIM1(336-486) sequence of GFP-monSTIM1 was PCR amplified using STIM1(336-486)-F and STIM1(336-486)-R primers, and then ligated into the EGFP-LOV2 vector at the HindIII and BamHI sites to generate the OptoCRAC expression plasmid.
- GFP-CRY2-STIM1(238-448), GFP-CRY2-STIM1(248-448), GFP-CRY2-STIM1(336-448), and GFP-CRY2-STIM1(342-448) expression plasmids sequences encoding STIM1(238-448), STIM1(248-448) were generated, and the STIM1(336-448) and STIM1(342-448) fragments were PCR amplified using STIM1(238-448)-F, STIM1(248-448)-F, STIM1(336-448)-F, STIM1(342-448)-F, and STIM1(238-448)-R primers and ligated into monSTIM1 using BspEI and BamHI restriction enzyme sites. Combined.
- GFP-CRY2-STIM1(318-463) and GFP-CRY2-STIM1(318-450) expression plasmids sequences encoding STIM1(318-463) and STIM1(318-450) fragments were PCR amplified using STIM1(318-463)-F primers. The sequences were amplified using STIM1(318-463)-R and STIM1(318-450)-R primers and ligated into monSTIM1 at the BspEI and BamHI sites.
- the IRES2 sequence was PCR amplified using the IRES2-F and IRES2-R primers, excised EGFP, and ligated into GFP-CRY2-STIM1(238-448) at the NheI and BsrGI sites. Then, the sequence encoding EGFP was inserted into the NheI and NotI sites to generate the EGFP-IRES2-CRY2-STIM1(238-448) expression plasmid. The STIM1(238-448) fragment was replaced with other STIM1 mutants at the BspEI and BamHI sites.
- the CRY2 (E281A, A9) sequence of monSTIM1 was PCR amplified using CRY2-F and CRY2-R primers, excised EGFP to generate the CRY2-N1 vector, and then ligated into EGFP-N1 at AgeI and BsrGI sites.
- the vhhGFP sequence of mCherry-CRY2-vhhGFP was PCR amplified using vhhGFP-F1 and vhhGFP-R1 primers and ligated into CRY2-N1 at NheI and AgeI sites to generate the vhhGFP-CRY2 expression plasmid.
- the CRY2 (E281A, A9) sequence of CRY2-N1 was digested with AgeI and BsrGI, EGFP was excised, and ligated into EGFP-C1 (Clontech) to generate the CRY2-C1 vector.
- vhhGFP sequence of mCherry-CRY2-vhhGFP was PCR amplified using primers vhhGFP-F2 and vhhGFP-R2 and ligated to CRY2-C1 at the BsrGI and XhoI sites to generate the CRY2-vhhGFP vector.
- an exchange enzyme site was designed using NheI-BamHI oligomers and inserted into pAAV-CamKIIa-EGFP (addgene #50469).
- GFP-monSTIM1 was ligated to pAAV-CamKIIa-EGFP at the NheI and BamHI sites after excising EGFP.
- the W3SL sequence of pAAV-CW3SL-EGFP was PCR amplified using W3SL-F and W3SL-R primers, then ligated into pAAV-CamKIIa-GFP-monSTIM1 vector at BamHI and RsrII sites after excising WPRE and hGH poly(A) signals, generating pAAV-CamKIIa-GFP-monSTIM1-W3SL vector.
- GFP-monSTIM1 was excised, and FLAG-monSTIM1 or GFP-STIM1(318-450) was inserted into the pAAV-CamKIIa-GFP-monSTIM1-W3SL vector using NheI and BamHI sites.
- the GfaABC1D promoter sequence was PCR amplified using primers GfaABC1D-F and GfaABC1D-R, and the CaMKII ⁇ promoter sequence was excised, followed by ligation into pAAV-CaMKII ⁇ -FLAG-monSTIM1 at the MluI and XbaI sites to generate pAAV-GfaABC1D-FLAG-monSTIM1.
- the sequences of the primers used for plasmid construction are shown in Table 1.
- HeLa cells ATCC and BV2 cells (ATCC) were maintained in Dulbecco's Modified Eagle's Medium (DMEM; Gibco, Cat# 11965092, Massachusetts, USA) supplemented with 10% fetal bovine serum (FBS; Gibco, Cat# 16,000-044) at 37°C in a humidified 5% CO2 atmosphere.
- DMEM Dulbecco's Modified Eagle's Medium
- FBS fetal bovine serum
- Hippocampal neurons were prepared from embryonic day 15–16 mice, and the collected embryonic hippocampi were dissected in Hank's balanced salt solution (HBSS; Gibco, Cat# 14175-095). The collected hippocampi were dissociated by incubation with 0.05% trypsin for 5 min at 37 °C, filtered through a 0.4- ⁇ m filter, and seeded onto coated 24-well polymer-coverslip-bottom plates (ibiTreat; ibidi, Cat# 82426, Gräfelfing, Germany) coated with 50 ⁇ g/mL poly-D-lysine (Millipore, Cat# A003-E, MA, USA).
- HBSS Hank's balanced salt solution
- the collected hippocampi were dissociated by incubation with 0.05% trypsin for 5 min at 37 °C, filtered through a 0.4- ⁇ m filter, and seeded onto coated 24-well polymer-coverslip-bottom plates (ibiTreat; ibidi
- Neurons were cultured in Neurobasal medium (Cat# 21103-049) supplemented with 2% B-27, 2% N-2 supplement, 2 mM GlutaMAX (Gibco, Cat# 35050061), 1000 units/mL penicillin-streptomycin and maintained at 37°C in a humidified 5% CO2 atmosphere.
- Astrocytes were dissected from P0-P1 C57BL/6 mouse pups by removing attached meninges and dissociated into single cell suspensions by osmotic pressure through a Pasteur pipette. Dissociated cells were plated on 60 mm dishes coated with 50 ⁇ g/mL poly-D-lysine.
- Cells were cultured in high-glucose DMEM (Gibco) containing L-glutamine and supplemented with 10% horse serum, 10% FBS, and 1000 units/mL penicillin-streptomycin and maintained at 37°C in a humidified atmosphere with 5% CO2.
- high-glucose DMEM Gibco
- FBS penicillin-streptomycin
- a Nikon A1R confocal microscope (Nikon Instruments) mounted on a Nikon Eclipse Ti body and equipped with a CFI Plan Apochromat VC objective ( ⁇ 60/1.4 numerical aperture (NA)) and digital zoom Nikon imaging software (NIS Element AR 64-bit version 3.21; Laboratory Imaging) was used.
- Example 4-1 Reduction of monSTIM1 size by replacing GFP with a small tag
- GFP of monSTIM1 was replaced with a smaller tagging component (HA (SEQ ID NO: 33) or FLAG tag (SEQ ID NO: 32)) by the method of Examples 1 and 2, and each mutant was expressed in HeLa cells together with R-GECO1, a red fluorescent Ca 2+ indicator.
- Figure 1c is a fluorescence image of cells co-expressing R-GECO1 and a monSTIM1 mutant or OptoCRAC (LOV2-STIM1). Blue light was delivered for 2 min at 30-min intervals, and changes in intracellular Ca2 + levels were monitored by imaging R-GECO1 (left, pink (magenta) image), while expression of monSTIM1 mutant and OptoCRAC is shown in green images (right, green image).
- Figure 1d is a graph showing the change in normalized intensity of R-GECO1 over time upon transient activation of one aspect of monSTIM1 mutants and OptoCRAC
- Figure 1f is a graph quantifying the activation kinetics of one aspect of monSTIM1 mutants
- Figure 1g is a graph quantifying the deactivation kinetics of one aspect of monSTIM1 mutants
- Figure 1h is a graph showing the light sensitivity of each optogenetic module (EGFP-monSTIM1, FLAG-monSTIM1, HA-monSTIM1, OptoCRAC)
- Data are presented as mean ⁇ SEM. (*p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001, ****p ⁇ 0.0001; Student two-tailed t test); ns, not significant (p>0.05)).
- HA-tagged monSTIM1 exhibited slower activation and faster deactivation kinetics than GFP or FLAG-tagged monSTIM1, suggesting that the properties of monSTIM1 may be influenced to some extent by the labeling components.
- FLAG-monSTIM1 coupled with the light-insensitive CRY2(D387A) mutant did not induce an increase in Ca2+ levels.
- Both FLAG- and HA-monSTIM1 mutants coupled with CRY2 showed similar sensitivity to blue light (Fig. 1h), indicating that the light sensitivity is mainly determined by the CRY2 photoreceptor.
- the LOV2-based method, OptoCRAC confirmed that the light sensitivity was reduced and induced a significantly smaller increase in Ca2+ compared to the monSTIM1 mutant (Figs. 1c to 1e and 1h).
- CAD STIM1 CRAC-activation domain
- Each truncated STIM1 construct is surrounded by different domain regions responsible for CAD autoinhibition.
- the expression levels of the six CRY2-fused STIM1 fragments (monSTIM1 mutants) were measured, and the results are shown in Fig. 2b.
- Figure 2b is a graph showing the expression levels of six CRY2-fused STIM1 fragments (monSTIM1 mutants) containing the STIM1 CRAC-activation domain (CAD; 342-448) (expressed as mean ⁇ SEM (one way ANOVA followed by multiple comparison test); ns, not significant (p>0.05)).
- Figure 2c is a photograph showing the result of fluorescence imaging of R-GECO1 in HeLa cells co-expressing each CRY2-fused STIM1 fragment (monSTIM1 mutant set 1-6) under blue light illumination;
- Figure 2d is a graph showing the maximum fold change (( ⁇ F/F0)) of R-GECO1 intensity upon activation of each CRY2-fused STIM1 fragment (monSTIM1 mutant set 1-6);
- Figure 2e is a graph showing the fluorescence intensity of R-GECO1 in the absence of blue light, and
- Figure 2f is a graph showing the correlation between the expression level of CRY2-fused STIM1 fragment and the basal R-GECO1 intensity.
- the STIM1 construct of set 5 (aa 318-450 fused EGFP-CRY2) was confirmed to exhibit similar levels of Ca2 + influx upon light stimulation and basal R-GECO1 intensity in the absence of light stimulation (dark) to monSTIM1.
- Figure 2g is a schematic diagram showing the structures of CRY2 fusion STIM1 fragments (sets 1-7) whose expression is induced by IRES2;
- Figure 2h is a photograph showing a fluorescence image of R-GECO1 in HeLa cells co-expressing each IRES2-induced CRY2 fusion STIM1 fragment (sets 1-7) when illuminated with blue light;
- Figure 2i is a graph showing the maximum fold change (( ⁇ F/F0) of R-GECO1 intensity upon activation of CRY2 fusion STIM1 fragments (Sets 1-7) whose expression is induced by each IRES2;
- the method using internal ribosome entry sequence 2 resulted in a marked decrease in basal R-GECO1 fluorescence, and in particular, the set 1 construct showed a much lower level of basal R-GECO1 fluorescence than monSTIM1.
- set 7 showed a much higher basal R-GECO1 intensity because it contains the constitutively activated domain of STIM1.
- the set 5 construct expressed by IRES2 did not efficiently induce Ca2 + influx under light stimulation, and only the set 1 construct expressed by IRES2 induced a Ca2 + increase at a level similar to monSTIM1.
- CRY2-fused STIM1 protein can affect both the basal Ca2 + level in the dark and the maximal Ca2 + level under light illumination.
- STIM1 (238-685) (SEQ ID NO: 30) was conjugated to CIBN (SEQ ID NO: 42), an N-terminal fragment of the cryptochrome-interacting basic-helix-loop-helix 1 (CIB1) protein that binds to oligomerized CRY2 in a light-dependent manner.
- CIBN SEQ ID NO: 42
- CIB1 cryptochrome-interacting basic-helix-loop-helix 1
- GFP-tagged STIM1 and GFP nanobody (vhhGFP)-conjugated CRY2 were utilized.
- vhhGFP GFP nanobody
- SEQ ID NO: 43 the copy number of vhhGFP
- E281A, A9 the copy number of CRY2
- Figure 3a is a schematic diagram showing the working mechanism of the two-component system of monSTIM1 protein
- Figure 3b is a schematic diagram showing the fusion structure of the system in which monSTIM1 protein is separated into two components
- Figure 3c is a photograph showing the fluorescence image of R-GECO1 in HeLa cells co-expressing each protein pair (set 1-5) when illuminated with blue light
- Figure 3d is a graph showing the maximum fold change (( ⁇ F/F0) of R-GECO1 intensity upon activation of monSTIM1 mutant
- Figure 3e is a graph showing the fluorescence intensity of R-GECO1 in the absence of blue light
- Data are presented as means ⁇ SEM (*p ⁇ 0.05, **p ⁇ 0.01, ****p ⁇ 0.0001; Student two-tailed t test); ns,
- proteins fused to the N-terminus of CRY2 with vhhGFP (sets 4 and 5) effectively induced Ca 2+ influx, indicating that the N-terminal fusion of vhhGFP maintained the GFP-binding properties and the light-dependent oligomerization of CRY2.
- the C-terminal fusion proteins (sets 2 and 3) showed significantly lower levels of Ca 2+ elevation, suggesting that the C-terminal fusion interfered with GFP binding and the light-dependent oligomerization of CRY2.
- all sets did not show an increase in basal R-GECO1 fluorescence in the dark, indicating that the STIM1 fragment (aa 238-685) sufficiently inhibited CAD activity in the absence of light stimulation (Fig. 3e).
- the five monSTIM1 variants that are potentially compatible with AAV are as follows:
- FLAG-CRY2-STIM1(238-685) FLAG-monSTIM1 (SEQ ID NO: 45); GFP-CRY2-STIM1(318-450)(SEQ ID NO: 46); GFP-IRES2-CRY2-STIM1(238-448); CIBN-STIM1(238-685)(SEQ ID NO: 47) + CRY2 (SEQ ID NO: 31) and GFP-STIM1(238-685)(SEQ ID NO: 48) + vhhGFP-CRY2 (SEQ ID NO: 49).
- R-GECO1 signals were utilized as surrogate measurements of relative Ca2+ levels.
- Fura-2 imaging was performed on cells expressing each monSTIM1 mutant (EGFP-monSTIM1; FLAG-monSTIM1; GFP-CRY2-STIM1(318-450); GFP-IRES2-CRY2-STIM1(238-448); GFP-STIM1+vhhGFP-CRY2; CIBN-STIM1+CRY2).
- HeLa cells expressing each of the above monSTIM1 mutants (EGFP-monSTIM1; FLAG-monSTIM1; GFP-CRY2-STIM1(318-450); GFP-IRES2-CRY2-STIM1(238-448); GFP-STIM1+vhhGFP-CRY2; CIBN-STIM1+CRY2) were loaded with Fura-2 AM (Invitrogen, Cat# F6774) diluted to 2 ⁇ M in DMEM and incubated at room temperature for 30 min, after which the cells were washed three times for 5 min each.
- Fura-2 AM Invitrogen, Cat# F6774
- Fura-2 imaging was performed using a LAMBDA DG-4 lamp (Sutter Instrument Company) and a ⁇ 40/0.75 NA CFI Plan Fluor objective with intermittent excitation using 340 and 380 nm filtered fluorescent lamps.
- the emitted light was collected with a Nikon DS-Qi1 black-and-white digital camera after passing through a 510 nm emission filter, and the results are shown in Fig. 4.
- the proteins (EGFP-monSTIM1; FLAG-monSTIM1; EGFP-CRY2-STIM1(318-450); EGFP-IRES2-CRY2-STIM1(238-448); EGFP-STIM1+vhhGFP-CRY2; CIBN-STIM1+CRY2) were introduced into cultured neurons, astrocytes, and microglia using the method of Example 2.
- Neurons expressing the above monSTIM1 mutant were treated with ACSF for 30 minutes, and then stimulated with blue light eight times for 0.5 seconds at 2-minute intervals. Astrocytes and microglia were given repetitive light stimulation (five times for 2 minutes at 30-second intervals), and changes in Ca2 + levels were confirmed using the method of Example 3.
- Figure 5a is a schematic diagram illustrating the light illumination protocol
- Figure 5b is a photograph (top) showing fluorescence images of R-GECO1 illuminated with blue light in neurons expressing each monSTIM1 mutant and a graph (bottom) showing relative changes in R-GECO1 intensity over time upon repeated light illumination
- Figure 3c is a graph showing the maximum fold change (( ⁇ F/F0) of R-GECO1 intensity upon activation of monSTIM1 mutants
- EGFP-monSTIM1: n 6
- Data are expressed as mean ⁇ SEM (one way ANOVA followed by multiple comparison test); n
- Figure 6 confirms that activation of monSTIM1 mutants increases Ca2+ in cultured astrocytes and microglia:
- Figure 6a is a photograph showing R-GECO1 fluorescence images in astrocytes (left) illuminated with blue light and microglia (BV2 cells, right) co-expressing each monSTIM1 mutant;
- Figure 6b is a schematic illustrating the illumination protocol;
- Figure 6c is a graph showing the relative change in R-GECO1 intensity over time upon illumination;
- Figure 6d is a graph showing the maximum fold change (( ⁇ F/F0)) of R-GECO1 intensity upon activation of monSTIM1 mutants.
- Figure 6e is a photograph showing a fluorescence image of R-GECO1 in astrocytes when light is shone on the subcellular region indicated by a circle with a white line
- Figure 6f is a kymograph of R-GECO1 corresponding to lines a-b and c-d of Figure 6e.
- the local stimulation induced a reversible, local increase in Ca2 + levels within the light-irradiated region, while no detectable increase in Ca2 + signals was observed in the contralateral, non-irradiated subcellular region.
- monSTIM1 mutants can effectively regulate intracellular Ca2 + levels in neurons and glial cells in a temporal and spatial manner.
- mice used in the experiment were male C57BL/6 mice aged 12 to 15 weeks.
- Male C57BL/6 mice aged 12 to 15 weeks were group-housed on a 12-hour light/dark cycle and had free access to food and water.
- the experimental protocol was approved by the Institutional Animal Care and Use Committee of IBS (Daejeon, Republic of Korea). Mice were randomly assigned to experimental groups.
- mice Surgical procedures for mice were performed according to the IBS IACUC guidelines. Mice were anaesthetized with 5% isoflurane and maintained with 1–2% isoflurane during stereotaxic surgery. After fixation of the skull in a stereotaxic device (RWD), the skin was shaved, the scalp was sterilized with povidone iodine, and a small craniotomy was performed. The following coordinates were used for microinjections into CA1: AP, 2.0 mm; ML, ⁇ 1.5 mm; DV, -1.4 mm.
- AAV vectors were generated using the method of Example 1 using two single-component mutants (FLAG-monSTIM1 and GFP-CRY2-STIM1(318-450)).
- WPRE and the hGH polyadenylation signal in the AAV cassette were replaced with a smaller component, W3SL, to enhance gene expression.
- monSTIM1 mutants were selectively expressed in excitatory neurons using the small CaMKII ⁇ promoter.
- Immunohistochemical staining was performed as follows. Mice were anesthetized with a mixture of alfaxan (40 mg/kg) and xylazine (10 mg/kg) for 90 min under dim or bright light conditions, and perfused transcardially first with phosphate-buffered saline (PBS) and then with PBS containing 4% paraformaldehyde (PFA). The brains were removed, fixed in 4% PFA overnight at 4°C, and then sectioned at 30 ⁇ m thickness using a vibratome (Leica).
- PBS phosphate-buffered saline
- PFA paraformaldehyde
- fluorescence images were acquired using a Leica Stellaris 8 confocal microscope equipped with ⁇ 20, ⁇ 40, and ⁇ 60 objectives.
- fluorescence images were acquired using a Zeiss Axio scan Z1 equipped with ⁇ 10 objective, and images were analyzed using ImageJ (NIH).
- NIR ImageJ
- FIG. 7 shows the results of application of one aspect of monSTIM1 mutants in neurons:
- Figure 7a is a schematic representation of the experiment in which AAV-compatible monSTIM1 variants were expressed under the control of the CaMKII ⁇ promoter targeting the CA1 region of the hippocampus (top) and mice were irradiated with blue LED light via a custom transcranial light illumination system, followed by retention for immunohistochemistry and analysis (bottom);
- Figure 7b is a schematic representation indicating the region used for quantification of activated neurons expressing cFos and monSTIM1 variants in the CA1 region (two AAV-compatible monSTIM1 variants were used: AAV-CaMKII ⁇ -FLAG-monSTIM1 and AAV-CaMKII ⁇ -EGFP-STIM1(318-450).
- Figure 7c is a representative image showing cFos-positive cells expressing each monSTIM1 mutant, with or without non-invasive light delivery and when expressing EGFP (control) (scale bar, 50 ⁇ m);
- Figure 7d is a graph quantifying cFos-positive cells expressing FLAG-monSTIM1
- Figure 7e is a graph quantifying cFos-positive cells expressing GFP-CRY2-STIM1(318-450).
- the monSTIM1 mutant showed broad and selective expression in neurons (NeuN-positive cells).
- Figure 8 shows the results of applying one aspect of monSTIM1 mutants to astrocytes:
- Figure 8a is a photograph showing FLAG-monSTIM1 expressed under the control of the GfaABC1D promoter targeting the hippocampal CA1 region (green, FLAG; red, c-Fos; blue, DAPI. Scale bar, 100 ⁇ m);
- Figure 8b is a photograph showing cFos-positive cells expressing FLAG-monSTIM1 with or without noninvasive light delivery (green, FLAG; blue, DAPI; pink, GFAP; red, c-Fos.
- the virus (AAV-nEF1 ⁇ -DIO-FLAG-CRY2(EA9)-STIM1(238-685)) was injected into the right anterior cingulate cortex (ACC) and CA1 region of the hippocampus of the brain of mice (SST-Cre; Ai-14), which were crossbred with Ai14 mice (Rosa26 locusCAG(promoter)-loxp-Stop-loxp-tdTomato) in which Cre recombinase is expressed in somatostatin (SST) interneurons, and immunohistochemical staining was performed using the method of Experimental Example 3.
- Figure 9 is a photograph showing the results of applying Cre-dependent FLAG-monSTIM1 expression control in the SST-Cre mouse strain, showing images of SST-positive cells expressing FLAG-monSTIM1 (blue is DAPI, green is FLAG, and red is tdTomato).
- FLAG-monSTIM1 is selectively expressed in SST interneurons, implying that FLAG-monSTIM1 is a platform that can be specifically expressed in cells expressing Cre recombinase as well as excitatory neurons and astrocytes.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Genetics & Genomics (AREA)
- Zoology (AREA)
- General Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Biophysics (AREA)
- Biochemistry (AREA)
- Molecular Biology (AREA)
- Biomedical Technology (AREA)
- Gastroenterology & Hepatology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Wood Science & Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Engineering & Computer Science (AREA)
- Biotechnology (AREA)
- Medicinal Chemistry (AREA)
- Plant Pathology (AREA)
- Toxicology (AREA)
- Virology (AREA)
- Physics & Mathematics (AREA)
- Microbiology (AREA)
- Botany (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Une protéine de fusion modulant le calcium qui peut être exprimée par un virus adéno-associé, selon un aspect de la présente invention, comprend monSTIM1 dont la taille de séquence codante est réduite par comparaison à avant de sorte à s'adapter à la capacité d'encapsulation du VAA, ce qui permet d'établir un système à base de VAA qui peut être exprimé dans les neurones et les cellules gliales d'un cerveau de souris. Le variant de monSTIM1 exprimé par le VAA selon un aspect peut réduire à un minimum les problèmes tels que les lésions tissulaires, la formation de cicatrices gliales, l'inflammation et l'échauffement des tissus, provoqués par une fibre optique insérée pendant une longue période et a pour effet de moduler l'activité neuronale d'une manière non invasive, et est ainsi un outil prometteur qui permet d'étudier les rôles spatiotemporels d'activités cellulaires à médiation par le calcium dans diverses fonctions cérébrales et présente également des potentiels à appliquer dans le traitement de maladies cérébrales associées à une signalisation calcique anormale.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| KR20230131100 | 2023-09-27 | ||
| KR10-2023-0131100 | 2023-09-27 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025071367A1 true WO2025071367A1 (fr) | 2025-04-03 |
Family
ID=95201944
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/KR2024/014799 Pending WO2025071367A1 (fr) | 2023-09-27 | 2024-09-27 | Protéine de fusion modulant le calcium pouvant être exprimée par un virus adéno-associé |
Country Status (2)
| Country | Link |
|---|---|
| KR (1) | KR20250047636A (fr) |
| WO (1) | WO2025071367A1 (fr) |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| KR20130088304A (ko) * | 2012-01-31 | 2013-08-08 | 한국과학기술원 | 빛에 의해 세포내 칼슘 이온 농도의 제어가 가능한 융합단백질 및 그의 용도 |
| KR20200055677A (ko) * | 2018-11-13 | 2020-05-21 | 기초과학연구원 | 광감도가 향상된 cry2 변이체 및 그 용도 |
| KR20210094950A (ko) * | 2020-01-22 | 2021-07-30 | (주)휴룩스 | 광감도가 향상된 cry2 변이체 융합 단백질 및 이를 이용한 치료학적 용도 |
-
2024
- 2024-09-27 WO PCT/KR2024/014799 patent/WO2025071367A1/fr active Pending
- 2024-09-27 KR KR1020240131827A patent/KR20250047636A/ko active Pending
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| KR20130088304A (ko) * | 2012-01-31 | 2013-08-08 | 한국과학기술원 | 빛에 의해 세포내 칼슘 이온 농도의 제어가 가능한 융합단백질 및 그의 용도 |
| KR20200055677A (ko) * | 2018-11-13 | 2020-05-21 | 기초과학연구원 | 광감도가 향상된 cry2 변이체 및 그 용도 |
| KR20210094950A (ko) * | 2020-01-22 | 2021-07-30 | (주)휴룩스 | 광감도가 향상된 cry2 변이체 융합 단백질 및 이를 이용한 치료학적 용도 |
Non-Patent Citations (2)
| Title |
|---|
| MA GUOLIN, HE LIAN, LIU SHUZHONG, XIE JIANSHENG, HUANG ZIXIAN, JING JI, LEE YI-TSANG, WANG RUI, LUO HESHENG, HAN WEIDONG, HUANG YU: "Optogenetic engineering to probe the molecular choreography of STIM1-mediated cell signaling", NATURE COMMUNICATIONS, NATURE PUBLISHING GROUP, UK, vol. 11, no. 1, 1 February 2020 (2020-02-01), UK, pages 1039 - 15, XP093299064, ISSN: 2041-1723, DOI: 10.1038/s41467-020-14841-9 * |
| TAN PENG, HE LIAN, HUANG YUN, ZHOU YUBIN: "Optophysiology: Illuminating cell physiology with optogenetics", PHYSIOLOGICAL REVIEWS, AMERICAN PHYSIOLOGICAL SOCIETY, US, vol. 102, no. 3, 1 July 2022 (2022-07-01), US , pages 1263 - 1325, XP093283202, ISSN: 0031-9333, DOI: 10.1152/physrev.00021.2021 * |
Also Published As
| Publication number | Publication date |
|---|---|
| KR20250047636A (ko) | 2025-04-04 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| WO2017034330A1 (fr) | Protéine recombinante (cp-bmp) de protéine morphogénétique osseuse perméable aux cellules et son utilisation | |
| US10561742B2 (en) | Methods and compositions for treatment of disease or injury of the nervous system | |
| JP2003511071A (ja) | Jnkシグナル導入経路の細胞透過性ペプチドインヒビター | |
| EA014330B1 (ru) | Пептидные ингибиторы пути трансдукции сигнала jnk, обладающие способностью проникать в клетку | |
| WO2012050402A2 (fr) | Protéine parkin recombinante à perméation cellulaire et composition pharmaceutique de traitement des maladies dégénératives du cerveau l'incluant | |
| WO2015156649A1 (fr) | Peptide présentant une activité inhibitrice contre la fibrose, et composition le contenant | |
| WO2014183491A1 (fr) | Polypeptide ayant une activité anti-tumorale et son utilisation | |
| WO2017018787A1 (fr) | Protéine recombinée de la parkine à perméabilité cellulaire améliorée (icp) et son utilisation | |
| WO2017026779A1 (fr) | Protéine recombinée cre à perméabilité cellulaire améliorée (icp-cre) et son utilisation | |
| CN110267673A (zh) | 利用chrimson来进行光遗传视觉恢复 | |
| US12139519B2 (en) | CRY2 variant having increased photosensitivity and use thereof | |
| EP3334755A1 (fr) | Protéine recombinante de facteur de reprogrammation à perméabilité cellulaire améliorée (icp-rf), et utilisation de ladite protéine | |
| US7033794B2 (en) | Basolateral sorting signal and inhibitors thereof | |
| WO2025071367A1 (fr) | Protéine de fusion modulant le calcium pouvant être exprimée par un virus adéno-associé | |
| WO2020009320A1 (fr) | Composition comprenant oct4 pour induire une transdifférenciation directe dans une cellule osseuse | |
| KR20210094952A (ko) | Cry2 변이체 융합 단백질을 이용한 엑소좀 분비 방법 | |
| KR20210094950A (ko) | 광감도가 향상된 cry2 변이체 융합 단백질 및 이를 이용한 치료학적 용도 | |
| WO2024054062A1 (fr) | Nouvelle composition polypeptidique pour transfection intracellulaire | |
| WO2018088733A1 (fr) | Nouveau domaine de transduction de protéine et utilisation associée | |
| US20070065904A1 (en) | Recombinant expression vectors for functional nav1.9 sodium channels | |
| WO2017209519A1 (fr) | Ligands sur mesure de la famille ab6 de la superfamille tgf-bêta | |
| WO2021162375A1 (fr) | Exosome comprenant une protéine photoclivable, et utilisation associée | |
| KR20210094951A (ko) | 광감도가 향상된 cry2 변이체 융합 단백질을 이용한 세포 대량 배양 방법 | |
| Magee | Cell adhesion molecules and intracellular signalling: from fly to man | |
| Alvarado | C-Terminal Properties of Teneurin, a Bacterial Toxin Homologue at the Synapse |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24873074 Country of ref document: EP Kind code of ref document: A1 |