[go: up one dir, main page]

WO2025071367A1 - Calcium-modulating fusion protein that can be expressed by adeno-associated virus - Google Patents

Calcium-modulating fusion protein that can be expressed by adeno-associated virus Download PDF

Info

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
Application number
PCT/KR2024/014799
Other languages
French (fr)
Korean (ko)
Inventor
이상규
국연희
이효인
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Institute for Basic Science
Original Assignee
Institute for Basic Science
Priority date (The priority date 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 date listed.)
Filing date
Publication date
Application filed by Institute for Basic Science filed Critical Institute for Basic Science
Publication of WO2025071367A1 publication Critical patent/WO2025071367A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/415Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from plants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/40Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation
    • C07K2319/42Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation containing a HA(hemagglutinin)-tag
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/40Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation
    • C07K2319/43Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation containing a FLAG-tag
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use 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

A calcium-modulating fusion protein that can be expressed by an adeno-associated virus, according to one aspect of the present invention, has monSTIM1 of which the coding sequence size is reduced compared to before so as to fit the packaging capacity of AAV, thereby establishing an AAV-based system that can be expressed in neurons and glial cells of a 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 an optical fiber inserted for a long period and has an effect of modulating neural activity in a non-invasive way, and thus is a promising tool that enables studying the spatiotemporal roles of calcium-mediated cellular activities in various brain functions and also has potentials to be applied in treating brain diseases related to abnormal calcium signaling.

Description

아데노 부속 바이러스로 발현 가능한 칼슘 조절 융합단백질Calcium-regulated fusion protein expressible by adeno-associated virus

아데노 부속 바이러스로 발현 가능한 칼슘 조절 융합단백질에 관련된 것이다.It relates to a calcium-regulated fusion protein that can be expressed by adeno-associated virus.

칼슘 이온(Ca2+)은 인지, 감정, 운동, 학습 및 기억을 포함한 광범위한 뇌 기능을 조절하는데 중요한 역할을 하는 유비쿼터스 제2의 메신저 역할을 한다. 세포 내 Ca2+ 신호는 공간과 시간에 따라 엄격하게 조절되며 중추 신경계(central nervous system)에서의 비정상적인 조절은 간질(epilepsy), 만성통증(chronic pain), 정신질환(psychiatric diseases) 및 신경퇴행(neurodegeneration)과 같은 다양한 신경 장애와 관련이 있다. 뇌 세포는 원형질막과 세포 소기관의 막에 위치한 Ca2+ 신호를 조절하는 채널과 펌프 등의 분자 기계를 보유하고 있다. Ca2+ 신호는 시냅스 가소성, 신경전달물질 방출, 유전자 발현 등 신경세포의 기능을 조절하며, 신호의 진폭, 지속 시간, 위치 등 다양한 매개변수에 따라 그 기능이 달라진다. 또한, Ca2+ 신호는 신경교세포(glial cells)에서도 신경전달물질 방출, 항상성 유지, 면역 반응 같은 중요한 과정을 조절하는 역할을 한다.Calcium ion (Ca2 + ) is a ubiquitous second messenger that plays a critical role in regulating a wide range of brain functions, including cognition, emotion, movement, learning, and memory. 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. In addition, Ca2 + signals also play a role in regulating important processes, such as neurotransmitter release, homeostasis, and immune responses, in glial cells.

광유전학 기술 분야(optogenetic technologies)에서는 빛 자극을 통해 세포 내 Ca2+ 수준을 제어하여 Ca2+ 신호와 시공간에서의 세포 기능 사이의 인과 관계를 직접 조사할 수 있는 다양한 기술이 활발히 연구되고 있다. 특히, Ca2+에 대해 높은 선택성을 지닌 CRAC(Ca2+-release-activated Ca2+) 채널이 주요 표적으로 주목받고 있다. 이러한 기술에서 SOCE(store-operated Ca2+ entry) 과정의 핵심 조절인자인 STIM1(Stromal interaction molecule 1)이 핵심적인 역할을 하며, CRAC 채널의 활성화를 위해 서로 다른 광수용체를 기반으로 하는 두 가지 유형의 광유전학적 액추에이터(optogenetic actuator)가 개발되었다. In the optogenetic technology field, various technologies are being actively studied to directly investigate the causal relationship between Ca 2+ signals and cellular functions in space and time by controlling intracellular Ca 2+ levels through light stimulation. In particular, CRAC (Ca 2+ -release-activated Ca 2+ ) channels with high selectivity for Ca 2+ are attracting attention as a major target. In these technologies, STIM1 (Stromal interaction molecule 1), a key regulator of the store-operated Ca 2+ entry (SOCE) process, plays a key role, and two types of optogenetic actuators based on different photoreceptors have been developed to activate CRAC channels.

첫 번째 유형의 광유전학적 액추에이터는 귀리(Avena Sativa)에서 추출한 광트로핀(phototropin)의 LOV2(Light-Oxygen-Voltage-sensing domain 2) 도메인을 활용한다. LOV2 도메인을 활성 STIM1 단편에 접합하면 입체 장애가 발생하여 빛이 없을 때 STIM1 활성이 억제된다. 청색광 자극은 LOV2 도메인 내의 형태 변화를 유도하여 C-말단 Jα 나선으로부터 해리를 유도한다. 이러한 해리는 STIM1의 억제를 완화하여 CRAC 채널의 활성화를 가능하게 하고 세포 내 Ca2+ 수준을 증가시킨다. The first type of optogenetic actuator utilizes the light-oxygen-voltage-sensing domain 2 (LOV2) domain of the phototropin isolated from oat ( Avena Sativa) . 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 Jα helix. This dissociation relieves STIM1 inhibition, allowing activation of the CRAC channel and increasing intracellular Ca2 + levels.

두 번째 유형의 광유전적 액추에이터는 애기장대(Arabidopsis thaliana)에서 추출한 크립토크롬 2(cryptochrome 2; CRY2)를 활용한다. CRY2의 빛을 매개로 한 동종 상호작용(light-mediated homo-interaction) 특성을 활용하여 STIM1의 세포질 단편은 CRY2와 결합하면 빛 자극 시 호모-올리고머화(homo-oligomerization) 과정을 거친다. 호모 올리고머화된 STIM1은 세포막으로 이동하여 CRAC 채널을 활성화하여 STIM1의 자연적인 작용 메커니즘을 모방한다(도 1a). 상기 두 번쨰 유형의 크립토크롬 2(cryptochrome 2; CRY2)를 이용한 광유전적 엑추에이터는 OptoSTIM1로 명명된다.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.

상기 OptoSTIM1은 다양한 세포 유형에서 Ca2+ 유입을 활성화하는 데 효과적이며 표적화된 뇌 기능을 성공적으로 제어하는 것으로 입증되었다(Nat Biotechnol. 2015;33:1092-6). LOV2 도메인 기반 액추에이터(actuator)는 (탈)활성화에 대한 더 빠른 동역학 및 OptoSTIM1에 비해 작은 크기인 점이 장점이지만, OptoSTIM1은 세포 내 Ca2+ 수준을 조절하는 데 있어 더 큰 폴드 변화를 유도하는 것으로 밝혀졌다(Cell Calcium. 2017;64:36-46). 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).

LOV2 도메인의 설계적 이점에도 불구하고, OptoSTIM1이 칼슘 유입을 조절하는 데 있어 더 뛰어난 성능을 보임에 따라 OptoSTIM1의 CRY2의 설계 개선을 위한 추가 연구가 진행되었다. 특히, CRY2 설계의 개선 과정에서 CRY2(E281A) 돌연변이가 어둠 속에서 OptoSTIM1의 기본 활동을 감소시키고, 청색광에 대한 민감도를 증가시키는 것으로 밝혀졌다. 또한 CRY2(E281A)와 A9 접합체를 결합하면 빛 의존적 올리고머화가 크게 증가한다는 사실도 확인되었다. 이를 바탕으로 2세대 OptoSTIM1인 monSTIM1이 개발되었으며, 이는 기존보다 청색광에 대한 감도가 약 55배 높아졌다. 이러한 개선으로 인해 광섬유 삽입 없이 비침습적으로 마우스 뇌에서 칼슘 신호를 활성화할 수 있게 되었다. Despite the design advantage of the LOV2 domain, further studies were conducted to improve the design of CRY2 of OptoSTIM1 because OptoSTIM1 showed a superior performance in regulating calcium influx. In particular, during the process of improving the CRY2 design, it was found that the CRY2(E281A) mutation decreased the basal activity of OptoSTIM1 in the dark and increased its sensitivity to blue light. In addition, it was confirmed that combining CRY2(E281A) and A9 conjugate significantly increased light-dependent oligomerization. Based on this, the second-generation OptoSTIM1, monSTIM1, was developed, which showed approximately 55-fold higher sensitivity to blue light than the previous one. These improvements enabled the activation of calcium signals in the mouse brain noninvasively without optical fiber insertion.

한편, 아데노 부속 바이러스(Adeno-Associated Virus, AAV)는 높은 유전자 전달 효율과 낮은 숙주 게놈 통합 확률, 낮은 면역원성으로 인해 신경과학 연구와 유전자 치료에서 널리 사용된다. 더욱이 최근 AAV 캡시드 엔지니어링의 발전으로 특정 조직과 세포에서 표적 발현을 가능하게 하는 전신 전달 기술이 개발되었다. 그러나 monSTIM1의 코딩서열과 ITR(inverted terminal repeats), WPRE(woodchuck hepatitis virus post-transcriptional regulatory element) 및 hGH(human growth hormone) 폴리아데닐화 신호를 포함한 AAV 카세트 구성 요소를 결합하면 프로모터를 제외하고 총 서열 크기가 ~ 5.1 kb로, 아데노 부속 바이러스(AAV)의 포장 용량(~ 5.0 kb)을 초과하게 된다(도 1b). Meanwhile, adeno-associated virus (AAV) is widely used in neuroscience research and gene therapy due to its high gene transfer efficiency, low host genome integration probability, and low immunogenicity. Moreover, recent advances in AAV capsid engineering have led to the development of systemic delivery technologies that enable targeted expression in specific tissues and cells. However, when combining 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).

기존 연구에서는 monSTIM1의 크기 제약으로 인해 마우스 뇌에서 광유전학적 Ca2+ 조절제(Ca2+ modulators)를 발현하기 위해 렌티바이러스(Lentivirus)를 사용했다. 렌티바이러스는 더 큰 유전자를 운반할 수 있지만, 숙주 게놈에 통합되는 특성이 있어 유전자 치료에 안전성 문제가 발생할 수 있으며, 뇌 조직 내 확산이 제한되어 형질전환되는 세포의 수가 줄어드는 한계가 있다.Previous studies have used lentiviruses to express optogenetic Ca2+ modulators in mouse brain due to the size constraints of monSTIM1. 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.

이에, 본 발명자는 AAV의 잠재적 이점을 활용하여 monSTIM1의 다양성을 확장하기 위해, 여러 전략을 통해 monSTIM1 코딩 서열을 축소하여 AAV의 패키징 용량에 맞는 AAV 호환 monSTIM1 변이체(monSTIM1 variants)를 개발하였다. 본 발명의 monSTIM1 변이체는 신경세포(neurons), 성상세포(astrocytes), 미세아교세포(microglia) 등 다양한 뇌 세포에서 세포 내 Ca2+ 수준을 효율적으로 증가시키고, 기존 monSTIM1의 초고광도(ultra-high light sensitivity) 특성을 유지하여 비침습적 빛 자극(non-invasive light illumination)에 반응하여 마우스 뇌의 신경세포와 성상세포에서 Ca2+ 매개 유전자 발현을 효과적으로 유도할 수 있다. Herein, in order to expand the diversity of monSTIM1 by utilizing the potential advantages of AAV, the present inventors developed AAV-compatible monSTIM1 variants (monSTIM1 variants) by shortening the monSTIM1 coding sequence through several strategies to fit the packaging capacity of AAV. 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.

일 양상은 태그(Tag) 단백질; CRY2(cryptochrome 2) 단백질 또는 그의 변이체; 및 STIMI1(Stromal interaction molecule 1) 단백질을 포함하는 융합단백질을 제공하는 것이다.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.

다른 양상은 상기 융합단백질을 암호화하는 폴리뉴클레오티드를 제공하는 것이다.Another aspect is to provide a polynucleotide encoding the above fusion protein.

또 다른 양상은 상기 융합단백질을 암호화하는 폴리뉴클레오티드를 포함하는 벡터를 제공하는 것이다.Another aspect is to provide a vector comprising a polynucleotide encoding the fusion protein.

또 다른 양상은 CIB1(cryptochromeinteracting basic-helix-loop-helix 1) 단백질 또는 그의 단편이 부착된 STIMI1(Stromal interaction molecule 1) 단백질; 및 CRY2(cryptochrome 2) 단백질 또는 그의 변이체를 포함하는 Ca2+ 모듈레이터를 제공하는 것이다.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.

또 다른 양상은 태그 단백질이 부착된 STIMI1 단백질; 및 상기 태그 단백질에 결합능이 있는 나노바디 항체가 부착된 CRY2단백질 또는 그의 변이체를 포함하는 Ca2+ 모듈레이터를 제공하는 것이다.Another aspect provides 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.

또 다른 양상은 STIMI1 단백질 또는 그의 단편을 암호화하는 폴리뉴클레오티드를 포함하는 제1 벡터; 및 CRY2단백질 또는 그의 변이체를 암호화하는 폴리뉴클레오티드를 포함하는 제2 벡터를 포함하는 발현 시스템을 제공하는 것이다.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.

일 양상은 태그(Tag) 단백질; CRY2(cryptochrome 2) 단백질 또는 그의 변이체; 및 STIMI1 단백질을 포함하는 융합단백질을 제공한다.One aspect provides a fusion protein comprising a Tag protein; CRY2 (cryptochrome 2) protein or a variant thereof; and STIMI1 protein.

본 명세서의 용어, 태그(Tag) 단백질 이란, 특정 단백질에 부착하여 단백질의 위치를 추적하거나 정제하는데 사용되는 단백질이다. 상기 태그는 단백질의 C-말단이나 N-말단에 연결되며, 단백질의 발현 수준을 모니터링하거나, 쉽게 검출할 수 있게 한다. The term "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.

일 구체예에 있어서, 상기 태그 단백질은 형광단백질일 수 있다. 예를 들어, 상기 형광단백질은 녹색형광단백질(green fluorescent protein, GFP), 황색형광단백질(yellow fluorescent protein, YFP), 적색형광단백질(red fluorescent protein, RFP), 주황형광단백질(orange fluorescent protein, OFP), 청록색형광단백질(cyan fluorescent protein, CFP), 청색형광단백질(blue fluorescent protein, BFP), 원적색형광단백질(far-red fluorescent protein) 또는 테트라시스테인 모티프(tetracystein motif)일 수 있다. 여기서, 상기 녹색형광단백질은 EGFP(enhanced green fluorescent protein), Emerald(Tsien, Annu. Rev. Biochem., 67: 509-544, 1998), Superfolder(Pedelacq et al., Nat. Biotech., 24: 79-88, 2006), GFP(Prendergast et al., Biochem., 17(17): 3448-3453, 1978), Azami Green(Karasawa, et al., J. Biol. Chem., 278: 34167-34171, 2003), TagGFP(Evrogen, Russia), TurboGFP(Shagin et al., Mol. Biol. Evol., 21(5): 841-850, 2004), ZsGreen(Matz et al., Nat. Biotechnol., 17: 969-973, 1999) 또는 T-Sapphire(Zapata-Hommer et al., BMC Biotechnol., 3:5, 2003)일 수 있다. 일 구체예에 있어서 상기 녹색형광단백질은 서열번호 41의 아미노산 서열로 이루어진 것일 수 있다.In one specific example, the tag protein can be a fluorescent protein. For example, 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. Here, the green fluorescent protein is EGFP (enhanced green fluorescent protein), Emerald (Tsien, Annu. Rev. Biochem., 67: 509-544, 1998), Superfolder (Pedelacq et al., Nat. Biotech., 24: 79-88, 2006), and GFP (Prendergast et al., Biochem., 17(17): 3448-3453, 1978), Azami Green (Karasawa, et al., J. Biol. Chem., 278: 34167-34171, 2003), TagGFP (Evrogen, Russia), TurboGFP (Shagin et al., Mol. Biol. Evol., 21(5): 841-850, 2004), ZsGreen (Matz et al., Nat. Biotechnol., 17: 969-973, 1999) or T-Sapphire (Zapata-Hommer et al., BMC Biotechnol., 3:5, 2003). In one specific example, the green fluorescent protein may be composed of an amino acid sequence of SEQ ID NO: 41.

일 구체예에 있어서, 상기 태그 단백질은 에피토프 태그(epitope tags)일 수 있다. 상기 에프토프 태그는 짧은 아미노산 서열로, 외래 단백질에 추가되어 특정 항체에 의해 인식될 수 있는 표지 역할을 하는 것으로, 예를 들어, Myc, V5, T7, AU1/AU5, VSV-G(Vesicular Stomatitis Virus Glycoprotein)일 수 있고, 바람직하게는 FLAG 또는 HA인 것일 수 있으나 이에 제한되는 것은 아니다. 구체적으로, 상기 FLAG는 8개의 아미노산 서열(DYKDDDDK)로 구성된 에피토프 태그이며, HA는 Hemagglutinin epitope tag의 약자로, 인플루엔자 A 바이러스의 헤마글루티닌(hemagglutinin) 단백질에서 유래한 9개 아미노산 서열(YPYDVPDYA)로 구성된 에페토프 태그이다.In one specific example, 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. Specifically, 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.

본 명세서의 용어, CRY2(cryptochrome 2) 단백질이란, 주로 파란빛을 감지하고 일주기 리듬(circadian rhythm)을 조절하는 단백질로서 식물, 곰팡이, 동물에서 발견된다. 일 구체예에 있어서, CRY2(cryptochrome 2) 단백질은 애기장대(Arabidopsis thaliana)에서 추출한 것일 수 있으나, 이에 제한되는 것은 아니다.The term 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. In one specific example, the CRY2 (cryptochrome 2) protein may be extracted from Arabidopsis thaliana , but is not limited thereto.

본 명세서의 용어, 단편은 온전한 폴리펩타이드 서열 및 구조의 일부로만 구성되는 폴리펩타이드로 의도되며, 변이체의 C-말단 결손 또는 N-말단 결손이 존재할 수 있다. 예를 들어, 상기 CRY2 단편은 CRY2 활성을 가진 CRY2 폴리펩타이드의 기능성 단편을 의미한다. The term fragment, as used herein, 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. For example, the CRY2 fragment means a functional fragment of a CRY2 polypeptide having CRY2 activity.

본 명세서의 용어, 변이체(variant)란, 유전자 또는 단백질의 기본적인 구조나 서열에 변화가 생긴 것을 의미할 수 있다.The term variant, as used herein, may mean a change in the basic structure or sequence of a gene or protein.

일 구체예에 있어서, CRY2 변이체는 CRY2(E281A) 돌연변이를 의미하는 것 일 수 있다. 상기 CRY2(E281A) 돌연변이는 어둠 속에서 OptoSTIM1의 기본 활동을 감소시키고, 청색광에 대한 민감도를 증가시킨다.In one specific example, 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.

일 구체예에 있어서 CRY2 변이체는 CRY2(E281A)와 A9 접합체를 결합한 것 (CRY2(E281A, A9))일 수 있다. 상기 CRY2(E281A, A9)는 빛 의존적 올리고머화 및 청색광에 대한 민감도가 크게 증가한다.In one specific example, 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.

일 구체예에 있어서, 상기 CRY2(E281A, A9) 단백질은 서열번호 31의 아미노산 서열로 이루어진 것일 수 있다.In one specific example, the CRY2 (E281A, A9) protein may be composed of an amino acid sequence of SEQ ID NO: 31.

본 명세서의 용어, STIMI1(Stromal interaction molecule 1) 단백질은 세포의 소포체(endoplasmic reticulum; ER) 막에 존재하는 막투과 단백질로서, SOCE(store-operated Ca2+ entry) 과정의 핵심 조절인자이다. 소포체 내강의 Ca2+ 수준을 감지하는 역할을 하며, 소포체 Ca2+ 가 고갈되면 STIM1은 올리고머화되고 C-말단 도메인에서 형태적 변화를 겪는다. 이후 원형질막으로 이동하여 CRAC(Ca2+-release-activated Ca2+) 채널에 결합하고 활성화하여 세포 내 Ca2+ 수준을 증가시킨다.The term 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.

일 구체예에 있어서, 상기 STIMI1 단백질은 세포질 STIM1 단편 (cytosolic STIM1 fragment)인 것일 수 있다. 상기 세포질 STIM1 단편 (cytosolic STIM1 fragment)은 STIM1 단백질의 세포질 내 카르복실 말단(cytosolic carboxyl terminus, STIM1ct)을 의미할 수 있으며, 구체적으로, STIM1 단백질의 아미노산 서열 238-685 부분을 의미할 수 있다. 예를 들어, 상기 STIM1ct는 서열번호 30의 아미노산 서열로 이루어진 것일 수 있다.In one specific example, 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. For example, the STIM1ct may consist of an amino acid sequence of SEQ ID NO: 30.

일 구체예에 있어서, 상기 세포질 STIM1 단편은 Ca2+ 채널에 결합하고 개방하는 역할을 하는 STIM1 CRAC 활성화 도메인(CRAC-activation domain, CAD; 342-448)을 포함하는 것 일 수 있다. 예를 들어, 상기 STIM1 CRAC 활성화 도메인은 STIM1 단백질의 342-448 부분일 수 있고, 서열번호 34의 아미노산 서열로 이루어진 것일 수 있다.In one specific example, 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. For example, 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.

예를 들어, 상기 세포질 STIM1 단편은 STIMI1 단백질의 아미노산 서열 238-685(서열번호 30), 238-448(서열번호 35), 248-448(서열번호 36), 336-448(서열번호 37), 318-463(서열번호 38), 318-450(서열번호 39) 또는 342-483(서열번호 40) 부분일 수 있으나, 이에 제한되는 것은 아니다.For example, 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.

다른 양상은 상기 융합단백질을 암호화하는 폴리뉴클레오티드를 제공한다.Another aspect provides a polynucleotide encoding the fusion protein.

다른 양상은 상기 폴리뉴클레오티드를 포함하는 벡터를 제공한다. 상기 벡터는 상기 융합단백질을 발현할 수 있는 재조합 발현벡터일 수 있다.Another aspect provides a vector comprising the polynucleotide. The vector may be a recombinant expression vector capable of expressing the fusion protein.

일 구체예에 있어서, 상기 벡터는 아데노 부속 바이러스(Adeno-Associated Virus, AAV) 벡터인 것일 수 있다. In one specific example, the vector may be an adeno-associated virus (AAV) vector.

본 명세서의 용어, monSTIM1은 GFP(Green Fluorescent Protein), 크립토크롬 2(cryptochrome 2; CRY2) 및 STIM1 단백질을 융합한 구조를 포함하는 것을 의미할 수 있다. 구체적으로, 상기 STIM1 단백질은 STIM1 단편을 의미하는 것일 수 있고, 상기 CRY2는 CRY2(E281A)와 A9 접합체를 결합한 변이체(CRY2(E281A, A9)일 수 있다.The term monSTIM1 in this specification may mean a structure comprising a fusion of GFP (Green Fluorescent Protein), cryptochrome 2 (CRY2), and STIM1 protein. Specifically, the STIM1 protein may mean a STIM1 fragment, and the CRY2 may be a mutant (CRY2 (E281A, A9)) combining CRY2 (E281A) and A9.

일 구체예에 있어서 상기 CRY2 변이체(E281A, A9)는 서열번호 31의 아미노산 서열로 이루어진 것일 수 있다.In one specific example, the CRY2 variant (E281A, A9) may be composed of the amino acid sequence of SEQ ID NO: 31.

본 명세서의 용어, monSTIM1 변이체(monSTIM1 variants)는 상기 monSTIM1의 구조에 변형을 가한 것을 의미한다.The term monSTIM1 variants in this specification means a variant of the structure of monSTIM1.

일 구체예에 있어서, 상기 monSTIM1 변이체는 상기 monSTIM1에서 GFP보다 다른 작은 태그 단백질을 대체한 것을 의미할 수 있다.In one specific example, the monSTIM1 variant may mean that a small tag protein other than GFP is replaced in the monSTIM1.

일 구체예에 있어서, 상기 태그 단백질은 FLAG 또는 HA를 의미하는 것일 수 있다. 구체적으로, 상기 태그 단백질은 서열번호 32 또는 서열번호 33의 아미노산 서열로 이루어진 것일 수 있다.In one specific example, the tag protein may refer to FLAG or HA. Specifically, the tag protein may be composed of an amino acid sequence of SEQ ID NO: 32 or SEQ ID NO: 33.

일 구체예에 있어서, 상기 monSTIM1 변이체는 상기 monSTIM1에서 STIM1 단백질의 일부를 절단한 것을 의미할 수 있다. 예를 들어, 상기 절단된 STIM1 단백질은 세포질 STIM1 단편일 수 있으며, 상기 세포질 STIM1 단편은 STIMI1 단백질의 아미노산 서열 238-685(서열번호 30), 238-448(서열번호 35), 248-448(서열번호 36), 336-448(서열번호 37), 318-463(서열번호 38), 318-450(서열번호 39) 또는 342-483(서열번호 40) 부분일 수 있으나, 이에 제한되는 것은 아니다.In one specific example, the monSTIM1 variant may mean a STIM1 protein that is truncated from a portion of the STIM1 protein in the monSTIM1. For example, 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.

일 구체예에 있어서, 상기 monSTIM1 변이체는 상기 monSTIM1을 두 개의 구조체로 분리한 것을 의미할 수 있다. 예를 들어, 상기 두 개의 구조체는 STIMI1 단백질 또는 그의 단편을 암호화하는 폴리뉴클레오티드를 포함하는 제1 벡터; 및 CRY2단백질 또는 그의 변이체를 암호화하는 폴리뉴클레오티드를 포함하는 제2 벡터를 포함하는 것 일 수 있다.In one specific example, the monSTIM1 variant may mean that the monSTIM1 is separated into two structures. For example, 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.

다른 양상은 CIB1(cryptochromeinteracting basic-helix-loop-helix 1) 단백질 또는 그의 단편이 부착된 STIMI1 단백질; 및 CRY2단백질 또는 그의 변이체를 포함하는 Ca2+ 모듈레이터를 제공한다.Another aspect provides 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.

본 명세서의 용어, CIB1(cryptochromeinteracting basic-helix-loop-helix 1)단백질이란, 주로 식물에서 발견되는 bHLH (basic-helix-loop-helix) 전사 인자로, CRY2(Cryptochrome 2)와 상호작용하며, 파란빛에 의해 활성화된다. The term 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.

일 구체예에 있어서, 상기 CIB1 단백질의 단편은 CIB1 단백질의 N-말단 단편(N-terminal fragment of the CIB1, CIBN)을 의미하는 것일 수 있다.In one specific example, the fragment of the CIB1 protein may mean an N-terminal fragment of the CIB1 protein (CIBN).

일 구체예에 있어서, 상기 CIB1 단백질의 단편은 서열번호 42의 아미노산 서열로 이루어진 것일 수 있다.In one specific example, the fragment of the CIB1 protein may consist of an amino acid sequence of SEQ ID NO: 42.

본 명세서의 용어, Ca2+ 모듈레이터(modulator)란, 칼슘의 농도, 유입, 방출을 제어하여 세포 기능을 조절하는 것으로, 주로 칼슘 채널이나 칼슘 펌프를 조절하여 세포 내로 칼슘을 유입시키거나 방출하고, 신호 전달 과정을 통해 세포 활동을 조절한다.The term 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.

상기 "태그 단백질", "CRY2 단백질", "STIMI1 단백질","단편", "변이체" 등의 정의는 전술한 바와 같다.The definitions of the above “tag protein”, “CRY2 protein”, “STIMI1 protein”, “fragment”, “variant”, etc. are as described above.

다른 양상은 태그 단백질이 부착된 STIMI1 단백질; 및 상기 태그 단백질에 결합능이 있는 나노바디 항체가 부착된 CRY2 단백질 또는 그의 변이체를 포함하는 Ca2+ 모듈레이터를 제공한다.Another aspect provides 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.

본 명세서의 용어, 나노바디(nanobody) 항체란, 낙타과(camalids) 또는 연골어류(cartilaginous fish)의 중쇄 항체로부터 유래된 단일 도메인 항체(single-domain antibody, sdAb)의 일종으로, 경쇄(light chain)가 없고 중쇄(heavy chain)의 VHH 도메인만을 포함하는 것을 의미한다. The term 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.

일 구체예에 있어서, 상기 나노바디는 상기 태그 단백질에 특이적으로 결합할 수 있는 것을 의미한다. 예를 들어, GFP 나노바디는 높은 친화도(Kd = 0.23 nM)로 GFP에 특이적으로 결합할 수 있어, GFP-GFP 나노바디(vhhGFP) 상호작용을 통해 STIM1 올리고머화에 필요한 CRY2의 동종 상호작용을 효율적으로 전달할 수 있다.In one specific example, the nanobody is meant to be capable of specifically binding to the tag protein. For example, a GFP nanobody can specifically bind to GFP with high affinity (Kd = 0.23 nM), thereby efficiently delivering the homologous interaction of CRY2 required for STIM1 oligomerization through GFP-GFP nanobody (vhhGFP) interaction.

일 구체예에 있어서. GFP 나노바디(vhhGFP)는 서열번호 43의 아미노산 서열로 이루어진 것일 수 있다.In one specific example, the GFP nanobody (vhhGFP) may be composed of the amino acid sequence of SEQ ID NO: 43.

일 구체예에 있어서, 상기 태그 단백질에 결합능이 있는 나노바디 항체는 CRY2 단백질 또는 그의 변이체의 N- 말단 또는 C- 말단에 부착될 수 있다.In one specific example, 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.

일 구체예에 있어서, 상기 태그 단백질에 결합능이 있는 나노바디 항체는 CRY2 단백질 또는 그의 변이체의 N- 말단 또는 C- 말단에 하나 이상 부착될 수 있다. 예를 들어, CRY2 단백질 또는 그의 변이체의 N- 말단 또는 C- 말단에 하나, 둘 또는 세개의 상기 태그 단백질에 결합능이 있는 나노바디 항체의 카피를 부착할 수 있으나 이에 제한되는 것은 아니다. 구체적으로, 하나, 둘 또는 세 개의 GFP 나노바디(vhhGFP) 카피를 CRY2 단백질 또는 이의 변이체의 N- 또는 C- 말단에 융합할 수 있다.In one specific embodiment, 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. For example, but not limited to, 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. Specifically, 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.

다른 양상은 STIMI1 단백질 또는 그의 단편을 암호화하는 폴리뉴클레오티드를 포함하는 제1 벡터; 및 CRY2단백질 또는 그의 변이체를 암호화하는 폴리뉴클레오티드를 포함하는 제2 벡터를 포함하는 발현 시스템을 제공한다.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.

일 구체예에 있어서, 상기 제1 벡터는 CIB1 단백질 또는 그의 단편; 또는 태그 단백질을 암호화하는 폴리뉴클레오티드를 추가적으로 포함하는 것일 수 있다.In one specific embodiment, the first vector may additionally comprise a polynucleotide encoding a CIB1 protein or a fragment thereof; or a tag protein.

일 구체예에 있어서, 상기 제2 벡터는 태그 단백질에 결합능이 있는 나노바디 항체를 암호화하는 폴리뉴클레오티드를 포함하는 것일 수 있다.In one specific embodiment, the second vector may comprise a polynucleotide encoding a nanobody antibody capable of binding to a tag protein.

상기 "태그 단백질", "CRY2 단백질", "STIMI1 단백질", "CIB1 단백질", "단편", "변이체" 등의 정의는 전술한 바와 같다.The definitions of the above “tag protein”, “CRY2 protein”, “STIMI1 protein”, “CIB1 protein”, “fragment”, “variant”, etc. are as described above.

상기 발명에 대해 기술한 용어 및 방법 등은 각 발명들 간에 동일하게 적용된다.The terms and methods described for the above inventions are equally applicable to each invention.

본 발명의 일 양상에 따른 아데노 부속 바이러스로 발현 가능한 칼슘 조절 융합단백질은 기존 monSTIM1 코딩 서열의 크기를 줄여 AAV 패키징 용량에 맞춤으로써 마우스 뇌의 신경세포와 아교세포에서 발현할 수 있는 AAV 기반 시스템을 구축하였다. 일 양상에 따른 AAV에 의해 발현된 monSTIM1 변이체는 장기간의 광섬유 삽입으로 인해 발생하는 조직 손상, 신경교 흉터 형성, 염증 및 조직 가열 등의 문제점을 최소화할 수 있으며, 비침습적인 방식으로 신경 활동을 조절할 수 있는 효과가 있다.According to one aspect of the present invention, 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.

도 1은 일 양상의 monSTIM1 변이체 또는 OptoCRAC의 광유전학적 활성화 양상을 나타낸 것이다: Figure 1 illustrates the optogenetic activation pattern of monSTIM1 mutants or OptoCRAC in one aspect:

도 1a는 GFP-monSTIM1의 작동 메커니즘을 나타낸 모식도이고; 도 1b는 monSTIM1의 코딩 서열 및 AAV 카세트의 구성 요소의 크기를 나타낸 구조도이며; 도 1c는 R-GECO1과 일 양상의 monSTIM1 변이체 또는 OptoCRAC을 공동 발현하는 세포의 형광 이미지를 나타낸 사진이고; 도 1d는 일 양상의 monSTIM1 변이체와 OptoCRAC의 일시적 활성화에 따른 시간에 따른 R-GECO1의 정규화된 강도의 변화를 보여주는 그래프이며; 도 1e는 청색광 자극 시 일 양상의 monSTIM1 변이체 및 OptoCRAC에 대한 R-GECO1 강도의 최대 폴드(fold) 변화(△F/F0)를 보여주는 그래프이고; 도 1f는 일 양상의 monSTIM1 변이체의 활성화 동역학을 정량화한 그래프이며; 도 1g는 일 양상의 monSTIM1 변이체의 비활성화 동역학을 정량화한 그래프이고; 도 1h는 각 광유전학 모듈(EGFP-monSTIM1, FLAG-monSTIM1, HA-monSTIM1, OptoCRAC)의 광 감도를 보여주는 그래프이다.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. 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).

도 2는 STIM1 CRAC 활성화 도메인(CAD; 342-448)을 포함하는 CRY2 융합 STIM1 단편의 6가지의 융합 단백질의 구조 및 이들 단백질의 발현 수준과 기저 R-GECO1 강도 사이의 상관관계를 나타낸 것이다: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:

도 2a는 CRY2 융합 STIM1 단편 구조를 나타낸 모식도이고; 도 2b는 STIM1 CRAC 활성화 도메인(CRAC-activation domain, CAD; 342-448)을 포함하는 6개의 CRY2 융합 STIM1 단편(monSTIM1 변이체)의 발현 수준을 보여주는 그래프이며; 도 2c는 각 CRY2 융합 STIM1 단편(monSTIM1 변이체 세트 1-6)을 공동 발현하는 HeLa 세포에서 R-GECO1의 형광 이미지를 청색광으로 조명한 결과를 나타낸 사진이고; 도 2d는 각 CRY2 융합 STIM1 단편(monSTIM1 변이체 세트 1-6) 활성화 시 R-GECO1 강도의 최대 폴드(fold) 변화((△F/F0)를 보여주는 그래프이며; 도 2e는 청색광이 없을 때 R-GECO1의 형광 강도를 나타낸 그래프이고, 도 2f는 CRY2-융합 STIM1 단편의 발현 수준과 기저 R-GECO1 강도 사이의 상관 관계를 보여주는 그래프이고; 도2g는 IRES2에 의해 발현이 유도되는 CRY2 융합 STIM1 단편 구조(세트 1-7)를 나타낸 모식도이고; 도 2h는 청색광으로 조명시, 각 IRES2에 의해 발현이 유도되는 CRY2 융합 STIM1 단편(세트 1-7)을 공동 발현하는 HeLa 세포에서 R-GECO1의 형광 이미지를 나타낸 사진이며; 도 2i는 각 IRES2에 의해 발현이 유도되는 CRY2 융합 STIM1 단편(세트 1-7) 활성화 시 R-GECO1 강도의 최대 폴드(fold) 변화((△F/F0)를 보여주는 그래프이고; 도 2j는 청색광이 없을 때, 각 IRES2에 의해 발현이 유도되는 CRY2 융합 STIM1 단편(세트 1-7)에서 R-GECO1의 형광 강도를 나타낸 그래프이다.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, and Figure 2f is a graph showing the correlation between the expression level of CRY2-fused STIM1 fragments and the basal R-GECO1 intensity; Figure 2g is a schematic diagram showing the structure of CRY2-fused STIM1 fragments (set 1-7) whose expression is induced by IRES2; Figure 2h is a photograph showing the fluorescence image of R-GECO1 in HeLa cells co-expressing each IRES2-induced CRY2-fused STIM1 fragment (set 1-7) when illuminated with blue light; Figure 2i is a graph showing the fluorescence image of R-GECO1 upon activation of each IRES2-induced CRY2-fused STIM1 fragment (set 1-7). Graph showing the maximum fold change in R-GECO1 intensity ((△F/F0); 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.

도 3은 monSTIM1 단백질을 두 가지 구성 요소로 분리한 시스템 및 이들 단백질의 발현 수준과 기저 R-GECO1 강도 사이의 상관관계를 나타낸 것이다:Figure 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:

도 3a는 monSTIM1 단백질을 두 가지 구성 요소 시스템의 작동 메커니즘을 나타낸 모식도이고; 도 3b는 monSTIM1 단백질을 두 가지 구성 요소로 분리한 시스템의 융합 구조를 나타낸 모식도이며; 도 3c는 청색광으로 조명시, 각 단백질 쌍(세트 1-5)을 공동 발현하는 HeLa 세포에서 R-GECO1의 형광 이미지를 나타낸 사진이고; 도 3d는 monSTIM1 변이체 활성화 시 R-GECO1 강도의 최대 폴드(fold) 변화((△F/F0)를 보여주는 그래프이며; 도 3e는 청색광이 없을 때 R-GECO1의 형광 강도를 나타낸 그래프이다.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; and Figure 3e is a graph showing the fluorescence intensity of R-GECO1 in the absence of blue light.

도 4는 각 monSTIM1 변이체(EGFP-monSTIM1; FLAG-monSTIM1; GFP-CRY2-STIM1(318-450); GFP-IRES2-CRY2-STIM1(238-448); GFP-STIM1+vhhGFP-CRY2; CIBN-STIM1+CRY2)를 발현하는 세포에서 Fura-2 이미징을 통한 상대적 Ca2+ 수준 측정한 그래프이다(왼쪽: 어두운 조건에서 측정한 Fura-2 비율(방출 340 nm/380 nm); 오른쪽: 청색광 조사 후 측정한 Fura-2 비율).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).

도 5는 배양된 해마 신경세포(hippocampal neurons)에서 일 양상의 monSTIM1 변이체의 활성화를 통한 Ca2+ 증가를 나타낸 것이다: Figure 5 shows the Ca2 + increase induced by activation of one monSTIM1 mutant in cultured hippocampal neurons:

도 5a는 빛 조사(light illumination) 프로토콜을 설명하는 모식도이고; 도 5b는 각 monSTIM1 변이체(EGFP-monSTIM1; FLAG-monSTIM1; GFP-CRY2-STIM1(318-450); GFP-IRES2-CRY2-STIM1(238-448); GFP-STIM1+vhhGFP-CRY2; CIBN-STIM1+CRY2)를 발현하는 뉴런에서 푸른 빛으로 조명한 R-GECO1의 형광 이미지(위)를 나타낸 사진 및 반복적인 빛 조명에 따른 시간 경과에 따른 R-GECO1 강도의 상대적 변화를 보여주는 그래프(아래)를 나타낸 것이며; 도 5c는 monSTIM1 변이체 활성화 시 R-GECO1 강도의 최대 폴드(fold) 변화((△F/F0)를 나타내는 그래프이다.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.

도 6은 배양된 성상세포 및 미세아교세포에서 일 양상의 monSTIM1 변이체의 활성화를 통해 Ca2+가 증가됨을 확인한 것이다: Figure 6 shows that activation of one monSTIM1 mutant in cultured astrocytes and microglia increases Ca2 + :

도 6a는 청색광으로 조명한 성상세포(astrocyte,왼쪽)와 각 monSTIM1 변이체(EGFP-monSTIM1; FLAG-monSTIM1; GFP-CRY2-STIM1(318-450); GFP-IRES2-CRY2-STIM1(238-448); GFP-STIM1+vhhGFP-CRY2; CIBN-STIM1+CRY2)를 공동 발현하는 미세아교세포(BV2 세포, 오른쪽)에서의 R-GECO1 형광 이미지를 나타낸 사진이고; 도 6b는 조명 프로토콜을 설명하는 개략도를 나타낸 것이며; 도 6c는 조명에 따른 R-GECO1 강도의 시간 경과에 따른 상대적 변화를 보여주는 그래프이고; 도 6d는 monSTIM1 변이체 활성화 시 R-GECO1 강도의 최대 폴드(fold) 변화((△F/F0)를 보여주는 그래프이고; 도 6e는 백색 선이 있는 원으로 표시된 세포하부 영역(subcellular region)에 빛을 비추었을 때 성상세포에서 R-GECO1의 형광 이미지를 나타낸 사진이며; 도 6f는 도 6e의 a-b 및 c-d 라인에 해당하는 R-GECO1의 키모그래프(Kymograph)를 나타낸 것이다.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; Figure 6f is a kymograph of R-GECO1 corresponding to lines a-b and c-d of Figure 6e.

도 7은 신경세포에서 일 양상의 monSTIM1 변이체의 적용 결과를 나타낸 것이다: Figure 7 shows the results of application of one aspect of monSTIM1 mutants in neurons:

도 7a는 일 양상의 AAV-호환성 monSTIM1 변이체가 해마 CA1 영역을 표적으로 하는 CaMKIIα 프로모터의 제어하에 발현되는 실험의 모식도(위) 및 맞춤형 경두개(transcranial) 광 조사 시스템을 통해 청색 LED 빛을 마우스에 조사하고, 면역조직화학을 위해 체류시킨 후 분석(아래)을 나타낸 것이고; 도 7b는 CA1 영역에서 cFos와 monSTIM1 변이체를 발현하는 활성화된 신경세포의 정량화에 사용된 영역을 표시하는 도식이며(두 개의 AAV-호환 monSTIM1변이체(AAV-CaMKIIα-FLAG-monSTIM1 및 AAV-CaMKIIα-EGFP-STIM1(318-450))가 사용됨. 청색, DAPI; 녹색, FLAG; 분홍색, NeuN. 스케일 바, 50 μm); 도 7c는 각 monSTIM1 변이체를 발현하는 cFos 양성 세포를 나타내는 대표이미지로, 비침습적 광 전달 유무 및 EGFP(대조군)를 발현하는 경우를 나타내는 이미지이며(스케일 바, 50 μm); 도 7d는 FLAG-monSTIM1을 발현하는 cFos 양성 세포를 정량화한 그래프이고, 도 7e는 GFP-CRY2-STIM1(318-450)을 발현하는 cFos 양성 세포를 정량화한 그래프이다.Figure 7a is a schematic representation of the experiment in which one AAV-compatible monSTIM1 variant was expressed under the control of the CaMKIIα 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-CaMKIIα-FLAG-monSTIM1 and AAV-CaMKIIα-EGFP-STIM1(318-450). Blue, DAPI; green, FLAG; pink, NeuN. Scale bar, 50 μm); 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 μm); Figure 7d is a graph quantifying cFos-positive cells expressing FLAG-monSTIM1, and Figure 7e is a graph quantifying cFos-positive cells expressing GFP-CRY2-STIM1(318-450).

도 8은 성상세포에 일 양상의 monSTIM1 변이체를 적용한 결과를 나타낸 것이다: Figure 8 shows the results of applying one type of monSTIM1 mutant to astrocytes:

도 8a는 해마 CA1 영역을 표적으로 한 GfaABC1D 프로모터의 제어 하에 발현된 FLAG-monSTIM1을 보여주는 사진이고(녹색, FLAG; 빨간색, c-Fos; 파란색, DAPI. 스케일 바, 100 μm); 도 8b는 비침습적 광 전달 유무에 관계없이 FLAG-monSTIM1을 발현하는 cFos 양성 세포를 보여주는 사진이며(녹색, FLAG; 청색, DAPI; 분홍색, GFAP; 적색, c-Fos. 스케일 바, 50 μm); 도 8c는 CA1 성상세포에서 FLAG-monSTIM1을 발현하는 cFos 양성 세포를 정량화한 그래프이다.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 μm); 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 μm); Figure 8c is a graph showing the quantification of cFos-positive cells expressing FLAG-monSTIM1 in CA1 astrocytes.

도9는 SST-Cre 마우스 계통에서 FLAG-monSTIM1을 발현하는 SST 양성 세포를 보여주는 이미지를 나타낸 사진이다(파란색은 DAPI, 녹색은 FLAG, 빨간색은 tdTomato).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).

이하 실험예를 통하여 보다 상세하게 설명한다. 그러나, 이들 실험예는 하나 이상의 구체예를 예시적으로 설명하기 위한 것으로 본 발명의 범위가 이들 실험예에 한정되는 것은 아니다.The following experimental examples will be described in more detail. However, these experimental examples are intended to exemplify one or more specific examples, and the scope of the present invention is not limited to these experimental examples.

실시예 1. 플라스미드 준비Example 1. Preparation of plasmids

R-GECO1용 발현 플라스미드(Addgene 플라스미드 #32444)는 Addgene에서 얻었다. HA-monSTIM1 및 FLAG-monSTIM1의 발현 플라스미드 제작을 위해 GFP-monSTIM1의 monSTIM1 서열을 HA-F, FLAG-F 및 HA-R 프라이머를 사용하여 중합효소 연쇄 반응(PCR)으로 증폭하였다. 증폭된 서열은 EGFP를 절제한 후 NheI 및 SalI 제한효소 자리를 이용해 GFP-monSTIM1에 연결하였다. The expression plasmid for R-GECO1 (Addgene plasmid #32444) was obtained from Addgene. To construct expression plasmids of HA-monSTIM1 and FLAG-monSTIM1, 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.

OptoCRAC의 LOV2(404-546) 서열은 LOV2-F 및 LOV2-R 프라이머를 사용하여 PCR 증폭하고 BsrGI 및 HindIII 부위에서 EGFP-C1 벡터에 결합하여 EGFP-LOV2 벡터를 생성하였다. 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.

GFP-monSTIM1의 STIM1(336-486) 서열은 STIM1(336-486)-F 및 STIM1(336-486)-R 프라이머를 사용하여 PCR 증폭한 후 HindIII 및 BamHI 부위의 EGFP-LOV2 벡터에 결합하여 OptoCRAC 발현 플라스미드를 생성했다. 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) 및 GFP-CRY2-STIM1(342-448) 발현 플라스미드의 구축을 위해, STIM1(238-448), STIM1(248-448)을 암호화하는 서열을 생성하였다, STIM1(336-448) 및 STIM1(342-448) 단편을 STIM1(238-448)-F, STIM1(248-448)-F, STIM1(336-448)-F, STIM1(342-448)-F 및 STIM1(238-448)-R 프라이머를 사용하여 PCR 증폭하고 BspEI 및 BamHI 제한효소 자리를 이용해 monSTIM1에 결합시켰다. For the construction of 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) 및 GFP-CRY2-STIM1(318-450) 발현 플라스미드 구축을 위해 STIM1(318-463) 및 STIM1(318-450) 단편을 암호화하는 서열을 STIM1(318-463)-F를 사용하여 PCR 증폭하였다. STIM1(318-463)-R 및 STIM1(318-450)-R 프라이머를 사용하여 증폭하고 BspEI 및 BamHI 부위에서 monSTIM1에 연결하였다. To construct 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.

IRES2-CRY2-STIM1(238-448) 발현 플라스미드 생성을 위해 IRES2 서열을 IRES2-F 및 IRES2-R 프라이머를 사용하여 PCR 증폭하고 EGFP를 절제한 후, NheI 및 BsrGI 부위에서 GFP-CRY2-STIM1(238-448)에 결합시켰다. 그 후, EGFP를 코딩하는 서열을 NheI 및 NotI 부위에 삽입하여 EGFP-IRES2-CRY2-STIM1(238-448) 발현 플라스미드를 생성하였다. STIM1(238-448) 단편은 BspEI 및 BamHI 부위에서 다른 STIM1 변이체로 대체되었다. To generate the IRES2-CRY2-STIM1(238-448) expression plasmid, 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.

monSTIM1의 CRY2(E281A, A9) 서열은 CRY2-F 및 CRY2-R 프라이머를 사용하여 PCR 증폭하고 EGFP를 절제하여 CRY2-N1 벡터를 생성한 후 AgeI 및 BsrGI 부위에서 EGFP-N1에 결합시켰다. 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.

mCherry-CRY2-vhhGFP의 vhhGFP 서열을 vhhGFP-F1 및 vhhGFP-R1 프라이머를 사용하여 PCR 증폭하고 NheI 및 AgeI 부위에서 CRY2-N1에 결합하여 vhhGFP-CRY2 발현 플라스미드를 생성하였다. CRY2-N1의 CRY2(E281A, A9) 서열을 AgeI 및 BsrGI로 분해하고 EGFP를 절제한 후 EGFP-C1(클론텍)에 결합하여 CRY2-C1 벡터를 생성하였다.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.

mCherry-CRY2-vhhGFP의 vhhGFP 염기서열을 vhhGFP-F2 및 vhhGFP-R2 프라이머를 사용하여 PCR 증폭하고 BsrGI 및 XhoI 부위에서 CRY2-C1에 결합하여 CRY2-vhhGFP 벡터를 생성하였다. The 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.

pAAV-CamKIIa-GFP-monSTIM1용 바이러스 벡터를 제작하기 위해 NheI-BamHI 올리고머를 사용하여 교환 효소 부위를 설계하고 pAAV-CamKIIa-EGFP(addgene #50469)에 삽입하였다. GFP-monSTIM1은 EGFP를 절제한 후 NheI 및 BamHI 부위에서 pAAV-CamKIIa-EGFP에 연결되었다. To construct the viral vector for pAAV-CamKIIa-GFP-monSTIM1, 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.

pAAV-CW3SL-EGFP(Addgene 플라스미드 #61463)의 W3SL 서열을 W3SL-F 및 W3SL-R 프라이머를 사용하여 PCR 증폭한 다음 WPRE 및 hGH 폴리(A) 신호를 절제한 후 BamHI 및 RsrII 부위에서 pAAV-CamKIIa-GFP-monSTIM1 벡터에 결합하여 pAAV-CamKIIa-GFP-monSTIM1-W3SL 벡터를 생성하였다. The W3SL sequence of pAAV-CW3SL-EGFP (Addgene plasmid #61463) 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.

pAAV-CamKIIa-FLAG-monSTIM1-W3SL 및 pAAV-CamKIIa-GFP-STIM1(318-450)-W3SL의 바이러스 벡터를 만들기 위해, GFP-monSTIM1을 절제한 후 NheI 및 BamHI 부위를 사용하여 FLAG-monSTIM1 또는 GFP-STIM1(318-450)을 pAAV-CamKIIa-GFP-monSTIM1-W3SL 벡터에 삽입하였다. To generate viral vectors of pAAV-CamKIIa-FLAG-monSTIM1-W3SL and pAAV-CamKIIa-GFP-STIM1(318-450)-W3SL, 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.

GfaABC1D 프로모터 서열은 GfaABC1D-F 및 GfaABC1D-R 프라이머를 사용하여 PCR 증폭하고 CaMKIIα 프로모터 서열을 절제한 후 MluI 및 XbaI 부위에서 pAAV-CaMKIIα-FLAG-monSTIM1에 결합하여 pAAV-GfaABC1D-FLAG-monSTIM1을 생성하였다. 플라스미드 제작을 위해 사용된 프라이머의 서열은 표 1에 나타내었다.The GfaABC1D promoter sequence was PCR amplified using primers GfaABC1D-F and GfaABC1D-R, and the CaMKIIα promoter sequence was excised, followed by ligation into pAAV-CaMKIIα-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.

PrimerPrimer 서열order 서열번호Sequence number HA-FHA-F 5'-GACTGCTAGCGCCACCATGGGATACCCATACGACGTGCCTGACTACGCCCCACCGGTCATGAAGATGGAC-3'5'-GACTGCTAGCGCCACCATGGGATACCCATACGACGTGCCTGACTACGCCCCACCGGTCATGAAGATGGAC-3' 11 HA-RHA-R 5'-GCATCCACGAGTGGGTACC-3'5'-GCATCCACGAGTGGGTACC-3' 22 FLAG-RFLAG-R 5'-GACTGCTAGCGCCACCATGGGAGACTACAAGGATGACGACGATAAGCCACCGGTCATGAAGATG-3'5'-GACTGCTAGCGCCACCATGGGAGACTACAAGGATGACGACGATAAGCCACCGGTCATGAAGATG-3' 33 LOV2-FLOV2-F 5'-GACTTGTACAAGGGCAGCCTGGCCACCACTCTAGAGCG-3'5'-GACTTGTACAAGGGCAGCCTGGCCACCACTCTAGAGCG-3' 44 LOV2-RLOV2-R 5'-GACTAAGCTTCAGCTCCTTGGCGGCCTC-3'5'-GACTAAGCTTCAGCTCCTTGGCGGCCTC-3' 55 STIM1(336-486)-FSTIM1(336-486)-F 5'-GACTAAGCTTGAATCTCACAGCTCATGGTATGCTC-3'5'-GACTAAGCTTGAATCTCACAGCTCATGGTATGCTC-3' 66 STIM1(336-486)-RSTIM1(336-486)-R 5'-GACTGGATCCTTAAGACACAATCTCCTCATCCATGTCATC-3'5'-GACTGGATCCTTAAGACACAATCTCCTCATCCATGTCATC-3' 77 STIM1(238-448)-FSTIM1(238-448)-F 5'-GGAGGCTCCGGACTCAGATC-3'5'-GGAGGCTCCGGACTCAGATC-3' 88 STIM1(238-448)-RSTIM1(238-448)-R 5'-GACTGGATCCCTAGTGGATGCCAGGGTTGTTG-3'5'-GACTGGATCCCTAGTGGATGCCAGGGTTTGTTG-3' 99 STIM1(248-448)-FSTIM1(248-448)-F 5'-GACTTCCGGATTGGAGGGGTTACACCGAGC-3'5'-GACTTCCGGATTGGAGGGGTTACACCGAGC-3' 1010 STIM1(336-448)-FSTIM1(336-448)-F 5'-GACTTCCGGAGAATCTCACAGCTCATGGTATGC-3'5'-GACTTCCGGAGAATCTCACAGCTCATGGTATGC-3' 1111 STIM1(342-448)-FSTIM1(342-448)-F 5'-GACTTCCGGATATGCTCCAGAGGCCC-3'5'-GACTTCCGGATATGCTCCAGAGGCCC-3' 1212 STIM1(318-463)-FSTIM1(318-463)-F 5 -GACTTCCGGAGAGGAGGAGTTGG-3'5 -GACTTCCGGAGAGGAGGAGTTGG-3' 1313 STIM1(318-463)-RSTIM1(318-463)-R 5'-GACTGGATCCCTAACTGCCCATCCA-3'5'-GACTGGATCCCTAACTGCCCATCCA-3' 1414 STIM1(318-450)-RSTIM1(318-450)-R 5'-GACTGGATCCCTACAGTGAGTGGATGC-3'5'-GACTGGATCCCTACAGTGAGTGGATGC-3' 1515 IRES2-FIRES2-F 5'-GATCGCTAGCGATCGATCGCGGCCGCATCCGCCCCTCTCCCTCC-3'5'-GATCGCTAGCGATCGATCGCGGCCGCATCCGCCCCTCTCCCTCC-3' 1616 IRES2-RIRES2-R 5'-GATCTGTACACCATGGTTGTGGCCATATTATCATC-3'5'-GATCTGTACACCATGGTTGTGGCCATATTATCATC-3' 1717 CRY2-FCRY2-F 5'-GACTACCGGTCGCCACCATGAAGATGGACAAAAAGACCATCG-3'5'-GACTACCGGTCGCCACCATGAAGATGGACAAAAAGACCATCG-3' 1818 CRY2-RCRY2-R 5'-GACTTGTACAATTCGTTGTCGAGGTCGGG-3'5'-GACTTGTACAATTCGTTGTCGAGGTCGGG-3' 1919 vhhGFP-F1vhhGFP-F1 5'-GACTGCTAGCGCCACCATGGTCCAACTGGTGGAGTCTGG-3'5'-GACTGCTAGCGCCACCATGGTCCAACTGGTGGAGTCTGG-3' 2020 vhhGFP-R1vhhGFP-R1 5'-GACTACCGGTGGGCTTCCGCCGCTGGAGACGGTGACCTG-3'5'-GACTACCGGTGGGCTTCCGCCGCTGGAGACGGTGACCTG-3' 2121 vhhGFP-F2vhhGFP-F2 5'-GACTTGTACAGCCACCATGGTCCAACTGGTGGAGTCTGG-3'5'-GACTTGTACAGCCACCATGGTCCAACTGGTGGAGTCTGG-3' 2222 vhhGFP-R2vhhGFP-R2 5'-GACTCTCGAGGGGCTTCCGCCGCTGGAGACGGTGACCTG-3'5'-GACTCTCGAGGGGCTTCCGCCGCTGGAGACGGTGACCTG-3' 2323 Agel-Nhel-BamHⅠ-HindⅢ oligomer-FAgel-Nhel-BamHⅠ-HindⅢ oligomer-F 5'-CCGGGCTAGCGGTTCATCAGGTTCATCAGGATCC-3'5'-CCGGGCTAGCGGTTCATCAGGTTCATCAGGATCC-3' 2424 Agel-Nhel-BamHⅠ-HindⅢ oligomer-RAgel-Nhel-BamHⅠ-HindⅢ oligomer-R 5-AGCTGGATCCTGATGAACCTGATGAACCGCTAGC-3'5-AGCTGGATCCTGATGAACCTGATGAACCGCTAGC-3' 2525 W3SL-FW3SL-F 5'-TGGATGGGCAGTTAGGGATCCCTCGAGATAATCAACCTCTGGATTACAAAATTTG-3'5'-TGGATGGGCAGTTAGGGATCCCTCGAGATAATCAACCTCTGGATTACAAAATTTG-3' 2626 W3SL-RW3SL-R 5'-TCCTGCGGCCGCTCGGTCCGTCCTGCGGCCGCTTTAAAAAAC-3'5'-TCCTGCGGCCGCTCGGTCCGTCCTGCGGCCGCTTTTAAAAAAC-3' 2727 GfaABC1D-FGfaABC1D-F 5'-ATCGACGCGTAACATATCCTGGTGTGGAGTAGGGG-3'5'-ATCGACGCGTAACATATCCTGGTGTGGAGTAGGGG-3' 2828 GfaABC1D-RGfaABC1D-R 5'-ATCGTCTAGAGCGAGCAGCGGA-3'5'-ATCGTCTAGAGCGAGCAGCGGA-3' 2929

실시예 2. 세포 배양 및 감염Example 2. Cell culture and infection

HeLa 세포(ATCC)와 BV2 세포(ATCC)는 5% CO2 가습된 37°C에서 10% 태아 소 혈청(FBS; Gibco, Cat# 16,000-044)을 보충한 Dulbecco's Modified Eagle's Medium (DMEM; Gibco, Cat# 11965092, 미국 매사추세츠주)에서 유지(maintain)하였다. 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.

해마 신경세포는 배아일(embryonic day) 15~16 마우스로부터 준비되었고, 수집된 배아의 해마는 Hank's balanced salt solution(HBSS; Gibco, Cat# 14175-095)에서 해부되었다. 수집된 해마는 37 °C에서 5분간 0.05% 트립신과 함께 배양하여 분리하고, 0.4-μm 필터로 여과한 다음, 50 μg/mL 폴리-D-리신(Millipore, Cat# A003-E, MA, USA)으로 코팅된 코팅된 24웰-폴리머-커버슬립-바닥-플레이트(24-well polymer-coverslip-bottom plate)(ibiTreat; ibidi, Cat# 82426, 독일 그라펠핑)에 접종(seed)하였다. 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-μm filter, and seeded onto coated 24-well polymer-coverslip-bottom plates (ibiTreat; ibidi, Cat# 82426, Gräfelfing, Germany) coated with 50 μg/mL poly-D-lysine (Millipore, Cat# A003-E, MA, USA).

신경세포는 2% B-27, 2% N-2 보충제, 2mM GlutaMAX(Gibco, Cat# 35050061), 1000 units/mL 페니실린-스트렙토마이신을 보충한 Neurobasal 배지(Cat# 21103-049)에서 배양하고 5% CO2 습도에서 37°C로 유지하였다. P0-P1 C57BL/6 마우스 새끼에서 부착된 수막을 제거하여 원피질 성상세포를 해부한 다음, 파스퇴르 피펫을 통해 삼투압 처리하여 단일 세포 현탁액으로 해리하였다. 해리된 세포를 50μg/mL 폴리-D-라이신으로 코팅된 60mm 접시에 도말하였다. 세포는 L-글루타민을 함유하고 10% 말 혈청, 10% FBS 및 1000 units/mL 페니실린-스트렙토마이신을 보충한 고포도당 DMEM(Gibco)에서 배양하고 5% CO2 및 습한 공기조건에서 37°C로 유지하였다.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 μg/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.

제조업체의 지침에 따라 Lipofectamine LTX(Invitrogen, Cat# 15338-100, 미국 캘리포니아주)를 사용하여 세포를 감염시켰다.Cells were transfected using Lipofectamine LTX (Invitrogen, Cat# 15338-100, CA, USA) according to the manufacturer's instructions.

실시예 3. 실시간 세포 이미닝(Live-cell imaging) 확인Example 3. Confirmation of live-cell imaging

실시간 세포 이미징을 위해, Nikon Eclipse Ti 본체에 장착되고 CFI Plan Apochromat VC 대물렌즈(Х60/1.4-개구수(NA))와 디지털 줌 Nikon 이미징 소프트웨어(NIS Element AR 64비트 버전 3.21; Laboratory Imaging)가 장착된 Nikon A1R 공초점 현미경(Nikon Instruments)을 사용하였다. For live cell imaging, a Nikon A1R confocal microscope (Nikon Instruments) mounted on a Nikon Eclipse Ti body and equipped with a CFI Plan Apochromat VC objective (Х60/1.4 numerical aperture (NA)) and digital zoom Nikon imaging software (NIS Element AR 64-bit version 3.21; Laboratory Imaging) was used.

현미경에 장착된 Chamlide TC 시스템(Live Cell Instruments, Inc., 한국)으로 이미징하는 동안 세포는 5% CO2 및 37°C에서 유지되었다. R-GECO1을 공동 발현하는 세포의 Ca2+ 유입은 각 monSTIM1 변이체와 함께 유전적으로 인코딩된 적색 Ca2+ 지표를 사용하여 측정했으며, 488 및 561nm 레이저 소스를 사용하여 30초 간격으로 세포를 여기(excite)시켰다. 캡처된 이미지는 NIS-Elements AR 현미경 이미징 소프트웨어(NIS-element AR 64비트 버전 3.21, Nikon)를 사용하여 분석하였다. Ca2+ 유입의 타임랩스 이미지를 세포의 정의된 관심 영역(ROI)에서 분석하고, R-GECO1 강도의 변화를 정량화하였다.Cells were maintained at 5% CO 2 and 37°C during imaging with a Chamlide TC system (Live Cell Instruments, Inc., Korea) mounted on the microscope. Ca 2+ influx in cells co-expressing R-GECO1 was measured using a red Ca 2+ indicator genetically encoded with each monSTIM1 mutant, exciting cells at 30 s intervals using 488 and 561 nm laser sources. Captured images were analyzed using NIS-Elements AR microscope imaging software (NIS-element AR 64-bit version 3.21, Nikon). Time-lapse images of Ca 2+ influx were analyzed in defined regions of interest (ROIs) of the cells, and changes in R-GECO1 intensity were quantified.

실시예 4. monSTIM1 크기 감소를 위한 변형 및 최적화Example 4. Modification and optimization for monSTIM1 size reduction

실시예 4-1. GFP를 작은 태그로의 대체를 통한 monSTIM1 크기 감소Example 4-1. Reduction of monSTIM1 size by replacing GFP with a small tag

녹색 형광 단백질(green fluorescent protein; GFP), CRY2(E281A, A9)(서열번호 31), 세포질 STIM1 단편(a.a. 238-685)(서열번호 30)의 세 가지 구성 요소로 이루어진 기존의 monSTIM1(그림 1b)의 AAV의 포장 용량을 초과하는 문제점을 해결하기 위해, 실시예 1 및 실시예 2의 방법으로 monSTIM1의 GFP를 더 작은 표지 성분(HA(서열번호 33) 또는 FLAG 태그(서열번호 32))으로 대체하고, 각 변이체를 적색 형광 Ca2+ 지표(red fuorescence Ca2+ indicator)인 R-GECO1과 함께 HeLa 세포에서 발현하였다. To address the issue of exceeding the packaging capacity of AAV of the existing monSTIM1 (Figure 1b), which consists of three components: green fluorescent protein (GFP), CRY2 (E281A, A9) (SEQ ID NO: 31), and cytoplasmic STIM1 fragment (aa 238-685) (SEQ ID NO: 30), 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.

FLAG-monSTIM1 및 HA-monSTIM1의 Ca2+ 유입 촉진 효율을 확인하기 위해 실시예 3의 방법으로 실시간 세포 이미징(Live-cell imaging)을 확인 및 분석하였고, 그 결과를 도1c 내지 도 1h에 나타내었다.To confirm the Ca2 + influx promoting efficiency of FLAG-monSTIM1 and HA-monSTIM1, live-cell imaging was confirmed and analyzed using the method of Example 3, and the results are shown in Figures 1c to 1h.

도 1c는 R-GECO1과 일 양상의 monSTIM1 변이체 또는 OptoCRAC(LOV2-STIM1)을 공동 발현하는 세포의 형광 이미지를 나타낸 사진이다. 청색광을 30분 간격으로 2분 동안 전달하고, R-GECO1을 영상화하여 세포 내 Ca2+ 수준의 변화를 모니터링하였고(왼쪽, 분홍(마젠타) 이미지), monSTIM1 변이체와 OptoCRAC의 발현은 녹색 이미지로 나타내었다(오른쪽, 녹색 이미지).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).

도 1d는 일 양상의 monSTIM1 변이체와 OptoCRAC의 일시적 활성화에 따른 시간에 따른 R-GECO1의 정규화된 강도의 변화를 보여주는 그래프이고; 도 1e는 청색광 자극 시 일 양상의 monSTIM1 변이체 및 OptoCRAC에 대한 R-GECO1 강도의 최대 폴드(fold) 변화(△F/F0)를 보여주는 그래프이다(EGFP-monSTIM1: n=18; FLAG-monSTIM1: n=23; HA-monSTIM1: n=24; OptoCRAC: n=31; FLAG-monSTIM1(CRY2D387A): n=38 cells).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 1e is a graph showing the maximum fold change (△F/F0) in R-GECO1 intensity for one aspect of monSTIM1 mutants and OptoCRAC upon blue light stimulation (EGFP-monSTIM1: n=18; FLAG-monSTIM1: n=23; HA-monSTIM1: n=24; OptoCRAC: n=31; FLAG-monSTIM1(CRY2 D387A ): n=38 cells).

도 1d 내지 도 1e에 나타난 바와 같이, 청색광 자극 시 FLAG-monSTIM1은 원래 GFP-monSTIM1과 비슷한 효율로 Ca2+ 유입을 촉진한 반면, HA-monSTIM1은 원래에 비해 Ca2+ 증가가 감소한 것으로 나타났다.As shown in Figures 1d to 1e, upon blue light stimulation, FLAG-monSTIM1 promoted Ca2 + influx with an efficiency similar to that of the original GFP-monSTIM1, whereas HA-monSTIM1 showed a decreased increase in Ca2 + compared to the original.

한편, 일 양상의 monSTIM1 변이체의 활성화, 비활성화 동역학 및 청색광에 대한 민감도를 측정하였다.Meanwhile, the activation and deactivation kinetics and sensitivity to blue light of monSTIM1 mutants of different types were measured.

도 1f는 일 양상의 monSTIM1 변이체의 활성화 동역학을 정량화한 그래프이며; 도 1g는 일 양상의 monSTIM1 변이체의 비활성화 동역학을 정량화한 그래프이고(EGFP-monSTIM1: n=30-38; FLAG-monSTIM1: n=42-50; HA-monSTIM1: n=45-47 cells); 도 1h는 각 광유전학 모듈(EGFP-monSTIM1, FLAG-monSTIM1, HA-monSTIM1, OptoCRAC)의 광 감도를 보여주는 그래프이다(데이터는 평균 ± SEM으로 표시됨. (*p<0.05, **p<0.01, ***p<0.001, ****p<0.0001; Student two-tailed t test); ns, not significant (p>0.05)).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 (EGFP-monSTIM1: n=30-38; FLAG-monSTIM1: n=42-50; HA-monSTIM1: n=45-47 cells); 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)).

도 1f 및 도1g에 나타난 바와 같이, HA-태그가 부착된 monSTIM1은 GFP 또는 FLAG-태그가 부착된 monSTIM1보다 느린 활성화 및 빠른 비활성화 동역학(kinetics)을 나타냄을 확인하였다. 이는 monSTIM1의 특성이 표지 성분(labeling components)에 의해 어느 정도 영향을 받을 수 있음을 시사한다.As shown in Figures 1f and 1g, 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.

또한, 음성 대조군 구조인 빛에 둔감한 CRY2(D387A) 돌연변이와 결합된 FLAG-monSTIM1은 Ca2+ 수준의 증가를 일으키지 않았다. CRY2와 결합된 FLAG- 및 HA-monSTIM1 변이체는 모두 청색광에 대해 유사한 민감도를 보였으며(도 1h), 이는 빛에 대한 민감도가 주로 CRY2 광수용체에 의해 결정된다는 것을 나타낸다. 반면, LOV2 기반 방법인 OptoCRAC은 monSTIM1 변종에 비해 빛 감도가 감소하고, Ca2+ 의 현저히 작은 증가를 유도하는 것을 확인하였다(도 1c 내지 도1e 및 도 1h). In addition, FLAG-monSTIM1 coupled with the light-insensitive CRY2(D387A) mutant, a negative control construct, 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. In contrast, 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).

이러한 결과는 GFP를 작은 태그로 대체하면 기존 monSTIM1의 기능을 유지하면서 monSTIM1의 크기를 크게 줄일 수 있음을 나타낸다.These results indicate that replacing GFP with a small tag can significantly reduce the size of monSTIM1 while maintaining the original monSTIM1 function.

실시예 4-2. STIM1절단을 통한 monSTIM1의 크기 감소Example 4-2. Size reduction of monSTIM1 through STIM1 truncation

어둠 속에서 칼슘 유입과 기저 칼슘 수준의 효율성을 유지하면서도 STIM1의 절단(truncation)이 monSTIM1의 크기를 더욱 줄일 수 있는지 테스트하기 위해, 실시예 1의 방법으로 일련의 융합 단백질을 생성하였다. To test whether truncation of STIM1 could further reduce the size of monSTIM1 while maintaining the efficiency of calcium influx and basal calcium levels in the dark, a series of fusion proteins were generated using the method of Example 1.

구체적으로, CRY2와 융합된 STIM1 단편 중, Ca2+ 채널에 결합하고 개방하는 역할을 하는 STIM1 CRAC 활성화 도메인(CRAC-activation domain, CAD; 342-448)을 포함하는 6가지의 융합 단백질(monSTIM1 변이체)을 설계하였다(도 2a, 표 2). Specifically, among the STIM1 fragments fused to CRY2, six fusion proteins (monSTIM1 mutants) containing the STIM1 CRAC-activation domain (CAD; 342-448), which plays a role in binding to and opening Ca2 + channels, were designed (Fig. 2a, Table 2).

단백질protein 아미노산 서열Amino acid sequence 서열번호Sequence number STIM1(342-448) (CRAC-activation domain, CAD)STIM1(342-448) (CRAC-activation domain, CAD) YAPEALQKWLQLTHEVEVQYYNIKKQNAEKQLLVAKEGAEKIKKKRNTLFGTFHVAHSSSLDDVDHKILTAKQALSEVTAALRERLHRWQQIEILCGFQIVNNPGIHYAPEALQKWLQLTHEVEVQYYNIKKQNAEKQLLVAKEGAEKIKKKRNTLFGTFHVAHSSSLDDVDHKILTAKQALSEVTAALRERLHRWQQIEILCGFQIVNNPGIH 3434 STIM1(238-448)STIM1(238-448) KEHMKKMMKDLEGLHRAEQSLHDLQERLHKAQEEHRTVEVEKVHLEKKLRDEINLAKQEAQRLKELREGTENERSRQKYAEEELEQVREALRKAEKELESHSSWYAPEALQKWLQLTHEVEVQYYNIKKQNAEKQLLVAKEGAEKIKKKRNTLFGTFHVAHSSSLDDVDHKILTAKQALSEVTAALRERLHRWQQIEILCGFQIVNNPGIHKEHMKKMMKDLEGLHRAEQSLHDLQERLHKAQEEHRTVEVEKVHLEKKLRDEINLAKQEAQRLKELREGTENERSRQKYAEEELEQVREALRKAEKEKELESHSSWY APEALQKWLQLTHEVEVQYYNIKKQNAEKQLLVAKEGAEKIKKKRNTLFGTFHVAHSSSLDDVDHKILTAKQALSEVTAALRERLHRWQQIEILCGFQIVNNPGIH 3535 STIM1(248-448)STIM1(248-448) LEGLHRAEQSLHDLQERLHKAQEEHRTVEVEKVHLEKKLRDEINLAKQEAQRLKELREGTENERSRQKYAEEELEQVREALRKAEKELESHSSWYAPEALQKWLQLTHEVEVQYYNIKKQNAEKQLLVAKEGAEKIKKKRNTLFGTFHVAHSSSLDDVDHKILTAKQALSEVTAALRERLHRWQQIEILCGFQIVNNPGIHLEGLHRAEQSLHDLQERLHKAQEEHRTVEVEKVHLEKKLRDEINLAKQEAQRLKELREGTENERSRQKYAEEELEQVREALRKAEKELESHSSWYAPEAL QKWLQLTHEVEVQYYNIKKQNAEKQLLVAKEGAEKIKKKRNTLFGTFHVAHSSSLDDVDHKILTAKQALSEVTAALRERLHRWQQIEILCGFQIVNNPGIH 3636 STIM1(336-448)STIM1(336-448) ESHSSWYAPEALQKWLQLTHEVEVQYYNIKKQNAEKQLLVAKEGAEKIKKKRNTLFGTFHVAHSSSLDDVDHKILTAKQALSEVTAALRERLHRWQQIEILCGFQIVNNPGIHESHSSWYAPEALQKWLQLTHEVEVQYYNIKKQNAEKQLLVAKEGAEKIKKKRNTLFGTFHVAHSSSLDDVDHKILTAKQALSEVTAALRERLHRWQQIEILCGFQIVNNPGIH 3737 STIM1(318-463)STIM1(318-463) EEELEQVREALRKAEKELESHSSWYAPEALQKWLQLTHEVEVQYYNIKKQNAEKQLLVAKEGAEKIKKKRNTLFGTFHVAHSSSLDDVDHKILTAKQALSEVTAALRERLHRWQQIEILCGFQIVNNPGIHSLVAALNIDPSWMGSEEELEQVREALRKAEKELESHSSWYAPEALQKWLQLTHEVEVQYYNIKKQNAEKQLLVAKEGAEKIKKKRNTLFGTFHVAHSSSLDDVDHKILTAKQALSEVTAALRERLHRWQQIEILCGFQIVNNPGIHSLVAALNIDPSWMGS 3838 STIM1(318-450)STIM1(318-450) EEELEQVREALRKAEKELESHSSWYAPEALQKWLQLTHEVEVQYYNIKKQNAEKQLLVAKEGAEKIKKKRNTLFGTFHVAHSSSLDDVDHKILTAKQALSEVTAALRERLHRWQQIEILCGFQIVNNPGIHSLEEELEQVREALRKAEKELESHSSWYAPEALQKWLQLTHEVEVQYYNIKKQNAEKQLLVAKEGAEKIKKKRNTLFGTFHVAHSSSLDDVDHKILTAKQALSEVTAALRERLHRWQQIEILCGFQIVNNPGIHSL 3939 STIM1(342-483)STIM1(342-483) YAPEALQKWLQLTHEVEVQYYNIKKQNAEKQLLVAKEGAEKIKKKRNTLFGTFHVAHSSSLDDVDHKILTAKQALSEVTAALRERLHRWQQIEILCGFQIVNNPGIHSLVAALNIDPSWMGSTRPNPAHFIMTDDVDDMDEEYAPEALQKWLQLTHEVEVQYYNIKKQNAEKQLLVAKEGAEKIKKKRNTLFGTFHVAHSSSLDDVDHKILTAKQALSEVTAALRERLHRWQQIEILCGFQIVNNPGIHSLVAALNIDPSWMGSTRPNPAHFIMTDDVDDMDEE 4040

각 절단된 STIM1 구조체는 CAD의 자가 억제를 담당하는 서로 다른 도메인 영역들에 의해 둘러싸여 있다. 상기 6개의 CRY2 융합 STIM1 단편(monSTIM1 변이체)의 발현수준을 측정하였고, 그 결과를 도 2b에 나타내었다.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.

도 2b는 STIM1 CRAC 활성화 도메인(CRAC-activation domain, CAD; 342-448)을 포함하는 6개의 CRY2 융합 STIM1 단편(monSTIM1 변이체)의 발현 수준을 보여주는 그래프이다(평균 ± SEM으로 표시됨 (일원배치 분산분석(one way ANOVA) 후 다중 비교 검정(multiple comparison test)); ns, not significant (p>0.05)).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)).

도 2b에 나타난 바와 같이, 발현된 CRY2 융합 STIM1 단백질 단편의 평균 발현 수준에는 유의미한 차이가 없음을 발견하였다.As shown in Fig. 2b, we found that there was no significant difference in the average expression levels of the expressed CRY2 fusion STIM1 protein fragments.

한편, 상기 6개의 CRY2 융합 STIM1 단편(monSTIM1 변이체 세트 1-6)의 Ca2+ 유입 촉진 효율을 확인하기 위해 실시예 3의 방법으로 실시간 세포 이미징(Live-cell imaging)을 확인 및 분석하였고, 그 결과를 도 2c 내지 도 2e에 나타내었다.Meanwhile, in order to confirm the Ca2 + influx promoting efficiency of the above six CRY2 fused STIM1 fragments (monSTIM1 mutant set 1-6), live-cell imaging was confirmed and analyzed using the method of Example 3, and the results are shown in Figs. 2c to 2e.

도 2c는 각 CRY2 융합 STIM1 단편(monSTIM1 변이체 세트 1-6)을 공동 발현하는 HeLa 세포에서 R-GECO1의 형광 이미지를 청색광으로 조명한 결과를 나타낸 사진이고; 도 2d는 각 CRY2 융합 STIM1 단편(monSTIM1 변이체 세트 1-6) 활성화 시 R-GECO1 강도의 최대 폴드(fold) 변화((△F/F0)를 보여주는 그래프이며; 도 2e는 청색광이 없을 때 R-GECO1의 형광 강도를 나타낸 그래프이고, 도 2f는 CRY2-융합 STIM1 단편의 발현 수준과 기저 R-GECO1 강도 사이의 상관 관계를 보여주는 그래프이다.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.

도 2c 내지 도 2e에 나타난 바와 같이, 6개의 CRY2 융합 STIM1 단편 구조체 중 세트 5의 STIM1 구조체(a.a. 318-450 융합 EGFP-CRY2)는 빛 자극시 Ca2+ 유입 수준과 빛 자극이 없는 상태(어둠)에서의 기저(basal) R-GECO1 강도가 monSTIM1과 유사하게 나타남을 확인하였다. As shown in Figures 2c to 2e, among the six CRY2-fused STIM1 fragment constructs, 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.

세트 1 내지 4도 Ca2+ 유입을 유도했지만, 암흑 상태에서 R-GECO1 형광의 강도가 증가 기저 Ca2+ 수준이 증가하는 것으로 나타났으며, 이는 STIM1 도메인 내의 자가억제 수준의 차이로 인한 것일 수 있다. 특히, 각 세포의 기저 R-GECO1 강도는 EGFP-CRY2-STIM1 발현 수준과 양의 상관관계를 보였으며, 이는 CRY2-STIM1 분자의 하위 집단(subpopulation)이 청색광이 없는 상태에서도 활성화되어 있음을 의미한다(도 2f).Although sets 1 to 4 induced Ca 2+ influx, the intensity of R-GECO1 fluorescence was increased in the dark state, indicating an increase in basal Ca 2+ levels, which may be due to differences in the level of autoinhibition within the STIM1 domain. Notably, the basal R-GECO1 intensity in each cell was positively correlated with the level of EGFP-CRY2-STIM1 expression, suggesting that a subpopulation of CRY2-STIM1 molecules is activated even in the absence of blue light (Fig. 2f).

한편, CRY2-STIM1 변이체의 발현 수준을 낮춤으로써 R-GECO1의 기저 강도를 감소시킬 수 있는지 확인하기 위해, IRES2(internal ribosome entry sequence 2)(서열번호 44)를 사용하여 단백질 발현을 유도하였다. Meanwhile, to determine whether the basal strength of R-GECO1 could be reduced by lowering the expression level of the CRY2-STIM1 mutant, protein expression was induced using internal ribosome entry sequence 2 (IRES2) (SEQ ID NO: 44).

도2g는 IRES2에 의해 발현이 유도되는 CRY2 융합 STIM1 단편 구조(세트 1-7)를 나타낸 모식도이고; 도 2h는 청색광으로 조명시, 각 IRES2에 의해 발현이 유도되는 CRY2 융합 STIM1 단편(세트 1-7)을 공동 발현하는 HeLa 세포에서 R-GECO1의 형광 이미지를 나타낸 사진이며; 도 2i는 각 IRES2에 의해 발현이 유도되는 CRY2 융합 STIM1 단편(세트 1-7) 활성화 시 R-GECO1 강도의 최대 폴드(fold) 변화((△F/F0)를 보여주는 그래프이고; 도 2j는 청색광이 없을 때, 각 IRES2에 의해 발현이 유도되는 CRY2 융합 STIM1 단편(세트 1-7)에서 R-GECO1의 형광 강도를 나타낸 그래프이다(monSTIM1: n=42; Set 1: n=30; Set 2: n=32; Set 3: n=43; Set 4: n=27; Set 5: n=41; Set 6: n=26; Set 7: n=81 cells. Data are presented as means±SEM (*p<0.05, ***p<0.001, ****p<0.0001; Student two-tailed t test); ns, not significant (p>0.05)).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; Figure 2j is a graph showing the fluorescence intensity of R-GECO1 in CRY2 fusion STIM1 fragments (Sets 1-7) whose expression is induced by each IRES2 in the absence of blue light (monSTIM1: n=42; Set 1: n=30; Set 2: n=32; Set 3: n=43; Set 4: n=27; Set 5: n=41; Set 6: n=26; Set 7: n=81 cells. Data are presented as means±SEM (*p<0.05, ***p<0.001, ****p<0.0001; Student two-tailed t test); ns, not significant (p>0.05)).

도 2g 내지 도 2j에 나타난 바와 같이, IRES2(internal ribosome entry sequence 2)를 사용한 방법은 기저 R-GECO1 형광의 눈에 띄는 감소를 가져왔으며, 특히 세트 1 구조에서는 monSTIM1보다 훨씬 낮은 수준의 기저 R-GECO1 형광을 보였다. 그러나 세트 7은 STIM1의 구성적으로 활성화된 도메인을 포함하기 때문에 훨씬 더 높은 기저 R-GECO1 강도를 보였다. 특히, 도 2d와는 달리, IRES2에 의해 발현된 세트 5 구조는 빛 자극 하에서 Ca2+ 유입을 효율적으로 유도하지 못하였고, IRES2에 의해 발현된 세트 1 구조만이 monSTIM1과 비슷한 수준으로 Ca2+ 증가를 유도함을 확인하였다.As shown in Figs. 2g to 2j, the method using internal ribosome entry sequence 2 (IRES2) 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. However, set 7 showed a much higher basal R-GECO1 intensity because it contains the constitutively activated domain of STIM1. In particular, unlike Fig. 2d, 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.

이는 CRY2 융합 STIM1 단백질의 발현 수준이 어둠 속에서 기저 Ca2+ 수준과 빛 조명 아래에서 최대 Ca2+ 수준에 모두 영향을 미칠 수 있음을 의미한다.This suggests that the expression level of CRY2-fused STIM1 protein can affect both the basal Ca2 + level in the dark and the maximal Ca2 + level under light illumination.

실시예 4-3. monSTIM1의 분할을 통한 크기 감소Example 4-3. Size reduction through splitting of monSTIM1

기존 monSTIM1 코딩 서열의 크기를 줄여 AAV 패키징 용량에 맞추기 위해 monSTIM1 단백질을 두 가지 구성 요소로 분리하는 두 가지 접근법을 테스트하였다. To reduce the size of the original monSTIM1 coding sequence to fit AAV packaging capacity, we tested two approaches to split the monSTIM1 protein into two components.

구체적으로, 첫번째 접근법(세트 1)에서는 STIM1(238-685)(서열번호 30)을 빛에 의존하는 방식으로 올리고머화된 CRY2에 결합하는 CIB1(cryptochrome-interacting basic-helix-loop-helix 1) 단백질의 N-말단 단편인 CIBN(서열번호 42)에 접합시켰다. Specifically, in the first approach (set 1), 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.

두 번째 접근법(세트 2 내지 5)에서는 GFP로 표지된 STIM1과 GFP 나노바디(vhhGFP)-접합 CRY2를 활용하였다. 융합 단백질의 최적 구성과 vhhGFP(서열번호 43)의 카피 수를 결정하기 위해, 하나 또는 두 개의 vhhGFP 카피를 CRY2(E281A, A9)의 N- 또는 C- 말단에 융합하였다(도 3b). GFP 나노바디는 중쇄 항체의 가변 도메인에서 파생된 단일 도메인 항체이며 높은 친화도(Kd = 0.23 nM)로 GFP에 특이적으로 결합할 수 있다. 따라서 GFP-vhhGFP 상호작용은 STIM1 올리고머화에 필요한 CRY2의 동종 상호작용을 효율적으로 전달한다. In the second approach (sets 2–5), GFP-tagged STIM1 and GFP nanobody (vhhGFP)-conjugated CRY2 were utilized. To determine the optimal composition of the fusion protein and the copy number of vhhGFP (SEQ ID NO: 43), one or two copies of vhhGFP were fused to the N- or C-terminus of CRY2 (E281A, A9) (Fig. 3B). GFP nanobodies are single-domain antibodies derived from the variable domain of heavy-chain antibodies and can specifically bind GFP with high affinity (Kd = 0.23 nM). Therefore, the GFP-vhhGFP interaction efficiently transmits the homotypic interaction of CRY2 required for STIM1 oligomerization.

상기 monSTIM1 단백질을 두 가지 구성 요소로 분리한 시스템 및 이들 단백질의 발현 수준과 기저 R-GECO1 강도 사이의 상관관계를 도 3에 나타내었다.The system in which the above monSTIM1 protein was separated into two components and the correlation between the expression levels of these proteins and the basal R-GECO1 intensity are shown in Figure 3.

도 3a는 monSTIM1 단백질을 두 가지 구성 요소 시스템의 작동 메커니즘을 나타낸 모식도이고; 도 3b는 monSTIM1 단백질을 두 가지 구성 요소로 분리한 시스템의 융합 구조를 나타낸 모식도이며; 도 3c는 청색광으로 조명시, 각 단백질 쌍(세트 1-5)을 공동 발현하는 HeLa 세포에서 R-GECO1의 형광 이미지를 나타낸 사진이고; 도 3d는 monSTIM1 변이체 활성화 시 R-GECO1 강도의 최대 폴드(fold) 변화((△F/F0)를 보여주는 그래프이며; 도 3e는 청색광이 없을 때 R-GECO1의 형광 강도를 나타낸 그래프이다(monSTIM1: n=52; Set 1: n=27; Set 2: n=24; Set 3: n=14; Set 4: n=21; Set 5: n=51 cells. Data are presented as means±SEM (*p<0.05, **p<0.01, ****p<0.0001; Student two-tailed t test); ns, not significant (p>0.05)).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 (monSTIM1: n=52; Set 1: n=27; Set 2: n=24; Set 3: n=14; Set 4: n=21; Set 5: n=51 cells. Data are presented as means±SEM (*p<0.05, **p<0.01, ****p<0.0001; Student two-tailed t test); ns, not significant (p>0.05)).

도 3에 나타난 바와 같이, 각 단백질 쌍을 R-GECO1과 함께 공동 발현하는 세포를 청색광으로 자극한 결과, 세트 1에서 R-GECO1 형광이 가장 크게 증가하였다(도 3c 및 도 3d). As shown in Figure 3, when cells co-expressing each protein pair with R-GECO1 were stimulated with blue light, the fluorescence of R-GECO1 increased the most in Set 1 (Figures 3c and 3d).

또한, vhhGFP를 CRY2의 N-말단에 융합한 단백질(세트 4 및 5)은 Ca2+ 유입을 효과적으로 유도하였으며, 이는 vhhGFP의 N-말단 융합이 GFP 결합 특성과 CRY2의 빛 의존적 올리고머화를 유지했음을 나타낸다. 대조적으로, C-말단 융합 단백질(세트 2 및 3)은 현저히 낮은 수준의 Ca2+ 증가를 보였으며, 이는 C-말단 융합이 GFP 결합 및 CRY2의 빛 의존적 올리고머화에 방해가 되었음을 의마한다. 특히, 모든 세트들이 어두운 상태에서 기저 R-GECO1 형광의 증가를 나타내지 않았으며, 이는 STIM1 단편(a.a. 238-685)이 빛 자극이 없을 때 CAD 활성을 충분히 억제한다는 것을 의미한다(도 3e). In addition, 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. In contrast, 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. Notably, 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).

상기 실시예 4-1 내지 4-3의 결과를 종합하여, AAV와 잠재적으로 호환될 수 있는 5개의 monSTIM1 변이체를 선별하였다. AAV와 잠재적으로 호환될 수 있는 5개의 monSTIM1 변이체는 다음과 같다:Based on the results of Examples 4-1 to 4-3 above, five monSTIM1 variants that are potentially compatible with AAV were selected. The five monSTIM1 variants that are potentially compatible with AAV are as follows:

FLAG-CRY2-STIM1(238-685) (FLAG-monSTIM1) (서열번호 45); GFP-CRY2-STIM1(318-450)(서열번호 46); GFP-IRES2-CRY2-STIM1(238-448); CIBN-STIM1(238-685)(서열번호 47) + CRY2(서열번호 31) 및 GFP-STIM1(238-685)(서열번호 48) + vhhGFP-CRY2(서열번호 49). 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).

상기 각 monSTIM1변이체에 대한 코딩 서열의 크기에 대한 정보는 표 3에 나타내었다.Information on the size of the coding sequence for each of the above monSTIM1 variants is shown in Table 3.

사이즈(bp)Size (bp) 표유류 발현 백터Expression vector of the stray 단일 구성Single configuration GFP-CRY2-STIM1(238-685)GFP-CRY2-STIM1(238-685) 35553555 HA-CRY2-STIM1(238-685)HA-CRY2-STIM1(238-685) 28172817 FLAG-CRY2-STIM1(238-685)FLAG-CRY2-STIM1(238-685) 28682868 GFP- CRY2-STIM1(318-450)GFP- CRY2-STIM1(318-450) 26102610 GFP-IRES2-CRY2-STIM1(238-448)GFP-IRES2-CRY2-STIM1(238-448) 34323432 두가지 구성Two configurations VhhGFP-CRY2VhhGFP-CRY2 19201920 GFP-STIM1(238-685)GFP-STIM1(238-685) 20612061 CIBN-STIM1(238-685)CIBN-STIM1(238-685) 18541854 CRY2CRY2 14941494 AAV 발현 벡터AAV expression vector 삽입용량(~4.8 kb)Insert size (~4.8 kb) AAV- CaMKIIα-FLAG- STIM1(238-685)AAV-CaMKIIα-FLAG-STIM1(238-685) 41474147 AAV- CaMKIIα- EGFP-CRY2-STIM1(318-450)AAV-CaMKIIα-EGFP-CRY2-STIM1(318-450) 38643864 AAV-GfaABC1D-FLAG-STIM1(238-685)AAV-GfaABC1D-FLAG-STIM1(238-685) 44674467

실험예 1. Fura-2 이미징을 통한 상대적 Ca2+ 수준 측정Experimental Example 1. Measurement of relative Ca2+ levels using Fura-2 imaging

상대적인 Ca2+ 수준의 대리 측정(surrogate measurement)하기 위해 R-GECO1 신호를 활용하였다. 어두운 조건과 밝은 조건 모두에서 Ca2+ 수준을 보다 정밀하게 분석하기 위해 각 monSTIM1 변이체(EGFP-monSTIM1; FLAG-monSTIM1; GFP-CRY2-STIM1(318-450); GFP-IRES2-CRY2-STIM1(238-448); GFP-STIM1+vhhGFP-CRY2; CIBN-STIM1+CRY2)를 발현하는 세포에 대해 Fura-2 이미징을 수행하였다.R-GECO1 signals were utilized as surrogate measurements of relative Ca2+ levels. To more precisely characterize Ca2 + levels under both dark and light conditions, 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).

구체적으로, 상기 각 monSTIM1 변이체(EGFP-monSTIM1; FLAG-monSTIM1; GFP-CRY2-STIM1(318-450); GFP-IRES2-CRY2-STIM1(238-448); GFP-STIM1+vhhGFP-CRY2; CIBN-STIM1+CRY2)를 발현하는 HeLa세포를 DMEM에 2μM으로 희석한 Fura-2 AM(Invitrogen, Cat# F6774)으로 로드(load)하였으며 상온에서 30분 동안 인큐베이션 한 후 세포를 5분씩 3회 세척하였다. Fura-2 이미징은 340 및 380nm 필터 형광등을 사용한 간헐적 여기(excitation)와 함께 LAMBDA DG-4 램프(Sutter Instrument Company)와 × 40/0.75 NA CFI Plan Fluor 대물렌즈를 사용하여 수행하였다. 방출된 빛은 510nm 방출 필터를 통과한 후 Nikon DS-Qi1 흑백 디지털 카메라로 수집하였고 그 결과를 도 4에 나타내었다.Specifically, 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 μM in DMEM and incubated at room temperature for 30 min, after which the cells were washed three times for 5 min each. 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.

도 4는 각 monSTIM1 변이체(EGFP-monSTIM1; FLAG-monSTIM1; GFP-CRY2-STIM1(318-450); GFP-IRES2-CRY2-STIM1(238-448); GFP-STIM1+vhhGFP-CRY2; CIBN-STIM1+CRY2)를 발현하는 세포에서 Fura-2 이미징을 통한 상대적 Ca2+ 수준 측정한 그래프이다(왼쪽: 어두운 조건에서 측정한 Fura-2 비율(방출 340 nm/380 nm); 오른쪽: 청색광 조사 후 측정한 Fura-2 비율(EGFP-monSTIM1: n=120; FLAG-monSTIM1: n=150; EGFP-CRY2-STIM1(318-450): n=201; EGFP-IRES2-CRY2-STIM1(238-448): n=146; EGFP-STIM1+vhhGFP-CRY2: n=152; CIBN-STIM1+CRY2: n=144 cells. 데이터는 평균 ± SEM으로 표시됨 (****p<0.0001; Student two-tailed t test)).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 condition (emission 340 nm/380 nm); Right: Fura-2 ratio measured after blue light irradiation (EGFP-monSTIM1: n=120; FLAG-monSTIM1: n=150; EGFP-CRY2-STIM1(318-450): n=201; EGFP-IRES2-CRY2-STIM1(238-448): n=146; EGFP-STIM1+vhhGFP-CRY2: n=152; CIBN-STIM1+CRY2: n=144 cells. Data are presented as mean ± SEM (****p<0.0001; Student two-tailed t test)).

도 4에 나타난 바와 같이, 모든 monSTIM1 변이체가 빛 자극에 따라 Ca2+ 농도를 크게 증가시키는 것을 확인하였다. 청색광이 없는 경우, GFP-CRY2-STIM1(318-450)을 제외한 모든 변이체는 형질전환 되지 않은 세포에서 관찰된 것과 비슷한 수준의 기저 Ca2+ 을 보였으며, 이는 R-GECO1을 사용하여 관찰된 경향과 일치한다. 한편, GFP-CRY2-STIM1(318-450)에서 관찰된 약간 상승된 Ca2+ 수준은 STIM1의 N-말단 자가 억제 도메인이 CAD의 활성을 완전히 억제하기에 불충분하다는 것을 의미한다.As shown in Fig. 4, we confirmed that all monSTIM1 mutants significantly increased Ca2 + concentration in response to light stimulation. In the absence of blue light, all mutants except GFP-CRY2-STIM1(318-450) showed basal Ca2 + levels similar to those observed in untransfected cells, which is consistent with the trend observed using R-GECO1. Meanwhile, the slightly elevated Ca2 + level observed in GFP-CRY2-STIM1(318-450) implies that the N-terminal autoinhibitory domain of STIM1 is insufficient to completely inhibit the activity of CAD.

실험예 2. 신경세포, 성상세포 및 미세아교세포에서 빛에 의해 유도된 Ca2+유입 확인Experimental Example 2. Confirmation of light-induced Ca2+ influx in neurons, astrocytes, and microglia

뇌에서 monSTIM1 변이체가 Ca2+ 신호를 조절하는 효능을 평가하기 위해, 배양된 신경세포(neurons), 성상세포(astrocyte) 및 미세아교세포(microglia)에 실시예 2의 방법으로, 상기 단백질들(EGFP-monSTIM1; FLAG-monSTIM1; EGFP-CRY2-STIM1(318-450); EGFP-IRES2-CRY2-STIM1(238-448); EGFP-STIM1+vhhGFP-CRY2; CIBN-STIM1+CRY2)을 도입하였다. 상기 monSTIM1 변이체를 발현한 신경세포(neurons)는 ACSF를 30분간 처리해 준 이후 2분 간격으로 0.5초 동안 8번의 청색광 자극을 주었고, 성상세포(astrocyte) 및 미세아교세포(microglia)는 반복적인 빛 자극(30초 간격으로 2분 동안 5회)을 주고, Ca2+ 수치의 변화를 실시예 3의 방법으로 확인하였다.To evaluate the efficacy of monSTIM1 mutants in modulating Ca2 + signals in the brain, 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.

2.1 신경세포에서 빛에 의해 유도된 Ca2+ 유입 확인2.1 Confirmation of light-induced Ca2+ influx in neurons

배양된 해마 신경세포(hippocampal neurons)에서 monSTIM1 변이체의 활성화를 통한 Ca2+ 수치 변화와 관련된 결과를 도 5에 나타내었다.Results related to changes in Ca2 + levels through activation of monSTIM1 mutants in cultured hippocampal neurons are shown in Figure 5.

도 5a는 빛 조사(light illumination) 프로토콜을 설명하는 모식도이고; 도 5b는 각 monSTIM1 변이체를 발현하는 뉴런에서 푸른 빛으로 조명한 R-GECO1의 형광 이미지(위)를 나타낸 사진 및 반복적인 빛 조명에 따른 시간 경과에 따른 R-GECO1 강도의 상대적 변화를 보여주는 그래프(아래)를 나타낸 것이며; 도 3c는 monSTIM1 변이체 활성화 시 R-GECO1 강도의 최대 폴드(fold) 변화((△F/F0)를 나타내는 그래프이다(EGFP-monSTIM1: n=6; FLAG-monSTIM1: n=5; EGFP-CRY2-STIM1(318-450): n=7; EGFP-IRES2-CRY2-STIM1(238-448): n=7; EGFP-STIM1+vhhGFP-CRY2: n=5; CIBN-STIM1+CRY2: n=6 cells. 데이터는 평균 ± SEM으로 표시됨(일원배치 분산분석(one way ANOVA) 후 다중 비교 검정(multiple comparison test)); ns, not significant (p>0.05)).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; FLAG-monSTIM1: n=5; EGFP-CRY2-STIM1(318-450): n=7; EGFP-IRES2-CRY2-STIM1(238-448): n=7; EGFP-STIM1+vhhGFP-CRY2: n=5; CIBN-STIM1+CRY2: n=6 cells. Data are expressed as mean ± SEM (one way ANOVA followed by multiple comparison test); ns, not significant (p>0.05)).

도 5a 내지 도 5c에 나타난 바와 같이, monSTIM1 변이체를 발현하는 신경세포에서 반복적인 빛 노출(2분 간격으로 5회)에 따라 세포 내 Ca2+ 수치가 빠르고 지속적으로 증가하는 것을 관찰하였다(도 5b). 한편, monSTIM1 변이체에서 Ca2+ 증가 수준에는 유의미한 차이가 없었다(도 5c). As shown in Figures 5a to 5c, we observed a rapid and sustained increase in intracellular Ca2 + levels following repeated light exposure (five times at 2-minute intervals) in neurons expressing monSTIM1 mutants (Figure 5b). Meanwhile, there was no significant difference in the level of Ca2 + increase in monSTIM1 mutants (Figure 5c).

이러한 결과는 monSTIM1 변이체가 신경세포의 세포 내 Ca2+ 수준을 효과적으로 조절할 수 있음을 의미하는 것이다.These results suggest that monSTIM1 mutants can effectively regulate intracellular Ca2 + levels in neurons.

2.2 성상세포 및 미세아교세포에서 빛에 의해 유도된 Ca2+ 유입 확인2.2 Confirmation of light-induced Ca2+ influx in astrocytes and microglia

배양된 성상세포(astrocyte) 및 미세아교세포(microglia)에서 일 양상에 따른 monSTIM1 변이체의 활성화를 통한 Ca2+ 수치 변화 결과를 도 6에 나타내었다.The results of changes in Ca2 + levels through activation of monSTIM1 mutants according to daily pattern in cultured astrocytes and microglia are shown in Figure 6.

도 6은 배양된 성상세포와 미세아교세포에서 monSTIM1 변이체의 활성화를 통해 Ca2+가 증가됨을 확인한 것이다: 도 6a는 청색광으로 조명한 성상세포(astrocyte,왼쪽)와 각 monSTIM1 변이체를 공동 발현하는 미세아교세포(BV2 세포, 오른쪽)에서의 R-GECO1 형광 이미지를 나타낸 사진이고; 도 6b는 조명 프로토콜을 설명하는 개략도를 나타낸 것이며; 도 6c는 조명에 따른 R-GECO1 강도의 시간 경과에 따른 상대적 변화를 보여주는 그래프이고; 도 6d는 monSTIM1 변이체 활성화 시 R-GECO1 강도의 최대 폴드(fold) 변화((△F/F0)를 보여주는 그래프이다.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.

도 6a 내지 도 6d에 나타난 바와 같이, 각 변이체 간 최대 폴드 변화의 차이는 있지만, 빛 자극 시 모든 변이체에서 성상세포와 미세아교세포에서 Ca2+ 수준이 현저하게 상승했다. 특히, monSTIM1 변이체에 의해 신경교 세포에서 유도된 Ca2+ 신호의 증가 정도는 신경세포에서 관찰된 것을 능가했으며, 이는 다양한 세포 유형에서 내인성 Ca2+ 채널의 발현 수준 또는 밀도의 차이에 기인할 수 있다. As shown in Figures 6a to 6d, although there were differences in the maximum fold changes among the mutants, all mutants significantly increased Ca 2+ levels in astrocytes and microglia upon light stimulation. Notably, the magnitude of the increase in Ca 2+ signals induced in glial cells by the monSTIM1 mutants exceeded that observed in neurons, which may be due to differences in the expression levels or densities of endogenous Ca 2+ channels in different cell types.

한편, monSTIM1에 의한 Ca2+ 수준의 국소적 조절을 확인하기 위해 성상세포에서 FLAG-monSTIM1을 발현하고 특정 세포 하부 영역에 1초 동안 빛을 조사하였고, 그 결과를 도 6e 및 도 6f에 나타내었다.Meanwhile, to confirm the local regulation of Ca2 + levels by monSTIM1, FLAG-monSTIM1 was expressed in astrocytes and light was irradiated to specific subcellular regions for 1 s, and the results are shown in Fig. 6e and Fig. 6f.

도 6e는 백색 선이 있는 원으로 표시된 세포하부 영역(subcellular region)에 빛을 비추었을 때 성상세포에서 R-GECO1의 형광 이미지를 나타낸 사진이며, 도 6f는 도 6e의 a-b 및 c-d 라인에 해당하는 R-GECO1의 키모그래프(Kymograph)를 나타낸 것이다.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, and Figure 6f is a kymograph of R-GECO1 corresponding to lines a-b and c-d of Figure 6e.

도 6e 및 도 6f에 나타난 바와 같이, 상기 국소 자극은 빛이 조사된 영역 내에서 Ca2+ 수치의 가역적이고 국소적인 증가를 일으켰으며, 빛이 조사되지 않은 반대쪽 세포하부 영역(subcellular region)에서는 Ca2+ 신호의 감지 가능한 증가가 나타나지 않았다.As shown in Figures 6e and 6f, 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.

이러한 결과는 monSTIM1 변이체가 신경세포 및 신경교세포의 세포 내 Ca2+ 수준을 시간적 및 공간적으로 효과적으로 조절할 수 있음을 의미하는 것이다.These results imply that monSTIM1 mutants can effectively regulate intracellular Ca2 + levels in neurons and glial cells in a temporal and spatial manner.

실험예 3. 마우스 뇌에서 생체 내 monSTIM1 변이체의 적용Experimental Example 3. In vivo application of monSTIM1 mutants in the mouse brain

3.1. 신경세포에서 monSTIM1 변이체의 적용3.1. Application of monSTIM1 mutants in neurons

monSTIM1 변이체가 마우스 뇌의 생체 내(in vivo)에서 Ca2+ 신호를 조절할 수 있는지 확인하였다. We investigated whether monSTIM1 mutants can modulate Ca2 + signals in the mouse brain in vivo .

실험에 사용한 마우스는 12~15주령의 수컷 C57BL/6를 사용하였다. 12~15주령의 수컷 C57BL/6 마우스를 12시간 명암 주기로 그룹 사육하고 음식과 물을 자유로이 이용할 수 있도록 하였다. 실험 프로토콜은 IBS(대전, 대한민국)의 기관 동물 관리 및 사용 위원회의 승인을 받았다. 마우스는 무작위로 실험군에 배정되었다.The 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.

마우스 수술 절차는 IBS IACUC 가이드라인에 따라 수행되었다. 마우스의 마취는 5% 이소플루란(isoflurane)으로 유도하고, 정위 수술 동안 1-2% 이소플루란으로 유지하였다. 정위 장치(RWD)로 두개골을 고정시킨 후 피부를 면도하고 두피를 포비돈 요오드로 멸균한 후 작은 개두술(craniotomy)을 시행하였다. CA1에 미세 주사할 때 다음 좌표가 사용되었다: AP, 2.0mm, ML, ± 1.5mm, DV, -1.4mm. 모든 미세 주사는 유리모세관 Picospritzer(Parker)를 사용하여 수행하였고, 각 주사 후 모세혈관은 확산을 위해 추가로 10분 동안 제자리에 유지한 다음 천천히 빼냈다. 생체 내 광자극 실험은 주사 3주 후, 고체 LED 여기(excitation) 시스템(Live Cell Instrument)을 통해 전달된 473nm 광을 사용하여 수행하였고, 빛의 조명 시간은 30분이었다.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. All microinjections were performed using a glass capillary Picospritzer (Parker), and after each injection, the capillaries were held in place for an additional 10 min to allow for diffusion and then slowly withdrawn. In vivo photostimulation experiments were performed 3 weeks after injection using 473 nm light delivered through a solid-state LED excitation system (Live Cell Instrument), with an illumination time of 30 min.

두 가지 단일구성요소 변이체(FLAG-monSTIM1및 GFP-CRY2-STIM1(318-450))를 사용하여 실시예 1의 방법으로 AAV 벡터를 생성하였다. 코딩 서열을 패키징하는 데 사용할 수 있는 공간을 더 늘리기 위해, AAV 카세트에서 WPRE와 hGH 폴리아데닐화 신호를 더 작은 구성 요소인 W3SL로 교체하여 유전자의 발현을 강화시켰다. 또한, 작은 CaMKIIα 프로모터를 사용하여 흥분성 신경세포에서 선택적으로 monSTIM1 변이체를 발현시켰다. 마우스 해마 CA1 영역에 AAV를 주입한 지 3주 후, 면역조직화학 염색법을 통해 확인하였다.AAV vectors were generated using the method of Example 1 using two single-component mutants (FLAG-monSTIM1 and GFP-CRY2-STIM1(318-450)). To increase the space available for packaging the coding sequence, WPRE and the hGH polyadenylation signal in the AAV cassette were replaced with a smaller component, W3SL, to enhance gene expression. In addition, monSTIM1 mutants were selectively expressed in excitatory neurons using the small CaMKIIα promoter. Three weeks after AAV injection into the mouse hippocampal CA1 region, immunohistochemical staining was performed to confirm this.

면역조직화학 염색은 다음과 같은 과정으로 진행하였다. 마우스를 어두운 조명 또는 밝은 조명 조건에서 90분 후 알팍산(40 mg/kg)과 자일라진(10 mg/kg)의 혼합물로 마취하고, 먼저 PBS로 경심관류(transcardial perfusion)한 다음 4% 파라포름알데히드(PFA)를 함유한 PBS로 경심관류하였다. 뇌를 적출하여 4% PFA에 4℃에서 하룻밤 동안 고정시킨 다음, 바이브라톰(vibratome)(Leica)을 사용하여 30μm 두께로 절편화 하였다. 면역염색을 위해 자유-부유 섹션(free-floating sections)을 PBS로 세척하고, 차단 용액(5% 정상염소혈청(normal goat serum)과 0.3% 트리톤 X-100을 함유한 PBS)에서 1시간 동안 배양한 후, 차단 용액에 1차 항체를 넣어 4℃에서 하룻밤 동안 배양하였다. 그런 다음 섹션을 PBS로 세척하고 PBS에 2차 항체를 넣고 실온에서 2시간 동안 배양하였다. 마지막으로 뇌 조직 섹션을 PBS로 세척하고 DAPI(4',6-diamidino-2-phenylindole)(Vector Laboratories, Cat# H-1200, CA, USA))가 포함된 Vectashield antifade 마운팅 배지로 슬라이드 장착하였다. CA1 이미징의 경우, 20배, 40배, 60배 대물렌즈를 장착한 Leica Stellaris 8 공초점 현미경을 사용하여 형광 이미지를 얻었다. 모든 뇌 영역의 경우, 형광 이미지는 10배 대물렌즈를 장착한 Zeiss Axio scan Z1을 사용하여 획득하였고, 이미지는 ImageJ(NIH)를 사용하여 분석하였다. 각 마우스 샘플에 대해 세 개 이상의 관상 뇌 섹션을 분석하여 cFos+ 및 FLAG+ 세포를 정량화하였다.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 μm thickness using a vibratome (Leica). For immunostaining, free-floating sections were washed with PBS, incubated in blocking solution (PBS containing 5% normal goat serum and 0.3% Triton X-100) for 1 h, and then primary antibodies were added to the blocking solution and incubated overnight at 4°C. Sections were then washed with PBS and incubated with secondary antibodies in PBS for 2 h at room temperature. Finally, brain tissue sections were washed with PBS and slide-mounted with Vectashield antifade mounting medium containing DAPI (4',6-diamidino-2-phenylindole) (Vector Laboratories, Cat# H-1200, CA, USA). For CA1 imaging, fluorescence images were acquired using a Leica Stellaris 8 confocal microscope equipped with ×20, ×40, and ×60 objectives. For all brain regions, fluorescence images were acquired using a Zeiss Axio scan Z1 equipped with ×10 objective, and images were analyzed using ImageJ (NIH). For each mouse sample, at least three coronal brain sections were analyzed to quantify cFos+ and FLAG+ cells.

도 7은 신경세포에서 일 양상의 monSTIM1 변이체의 적용에 대한 결과를 나타낸 것이다: Figure 7 shows the results of application of one aspect of monSTIM1 mutants in neurons:

도 7a는 AAV-호환성 monSTIM1 변이체가 해마 CA1 영역을 표적으로 하는 CaMKIIα 프로모터의 제어하에 발현되는 실험의 모식도(위) 및 맞춤형 경두개(transcranial) 광 조사 시스템을 통해 청색 LED 빛을 마우스에 조사하고, 면역조직화학을 위해 체류시킨 후 분석(아래)을 나타낸 것이고; 도 7b는 CA1 영역에서 cFos와 monSTIM1 변이체를 발현하는 활성화된 신경세포의 정량화에 사용된 영역을 표시하는 도식이며(두 개의 AAV-호환 monSTIM1변이체(AAV-CaMKIIα-FLAG-monSTIM1 및 AAV-CaMKIIα-EGFP-STIM1(318-450))가 사용됨. 청색, DAPI; 녹색, FLAG; 분홍색, NeuN. 스케일 바, 50 μm); 도 7c는 각 monSTIM1 변이체를 발현하는 cFos 양성 세포를 나타내는 대표이미지로, 비침습적 광 전달 유무 및 EGFP(대조군)를 발현하는 경우를 나타내는 이미지이며(스케일 바, 50 μm); 도 7d는 FLAG-monSTIM1을 발현하는 cFos 양성 세포를 정량화한 그래프이고, 도 7e는 GFP-CRY2-STIM1(318-450)을 발현하는 cFos 양성 세포를 정량화한 그래프이다.Figure 7a is a schematic representation of the experiment in which AAV-compatible monSTIM1 variants were expressed under the control of the CaMKIIα 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-CaMKIIα-FLAG-monSTIM1 and AAV-CaMKIIα-EGFP-STIM1(318-450). Blue, DAPI; green, FLAG; pink, NeuN. Scale bar, 50 μm); 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 μm); Figure 7d is a graph quantifying cFos-positive cells expressing FLAG-monSTIM1, and Figure 7e is a graph quantifying cFos-positive cells expressing GFP-CRY2-STIM1(318-450).

도 7a 및 도 7b에 나타난 바와 같이, AVV를 해마 CA1 영역에 주입한 후 3주가 지나, monSTIM1 변이체가 신경세포(NeuN 양성 세포)에서 넓고 선택적인 발현을 나타냈다. As shown in Figures 7a and 7b, 3 weeks after AVV injection into the hippocampal CA1 region, the monSTIM1 mutant showed broad and selective expression in neurons (NeuN-positive cells).

또한, 도 7c 내지 도 7e에 나타난 바와 같이, 청색광에 의한 monSTIM1 변이체의 활성화는 Ca2+-반응성 즉각 초기 유전자(Ca2+-responsive immediate-early gene)인 cFos의 유의미한 유도를 이끌어냈으며, STIM1 양성 신경세포 중 약 10%에서 이러한 반응이 관찰되었다. Additionally, as shown in Figures 7c to 7e, activation of the monSTIM1 mutant by blue light led to significant induction of cFos, a Ca 2+ -responsive immediate-early gene, and this response was observed in approximately 10% of STIM1-positive neurons.

반면, 어두운 조건의 마우스와 빛 자극 조건에서 GFP만을 발현하는 마우스는 cFos 발현의 유의미한 증가가 관찰되지 않았으며, 이는 cFos 발현이 빛 자극 자체가 아니라 monSTIM1 변이체의 활성에 의해 유도되었음을 의미한다.In contrast, no significant increase in cFos expression was observed in mice under dark conditions and in mice expressing only GFP under light stimulation conditions, suggesting that cFos expression was induced by the activity of the monSTIM1 mutant and not by the light stimulus itself.

이러한 결과는 유전자 발현이 비침습적인 빛 자극에 의해 성공적으로 유도되었으며, 상기 두 가지 변이체(FLAG-monSTIM1 및 GFP-CRY2-STIM1(318-450))가 여전히 기존 monSTIM1과 유사한 초고광도 감도를 가지고 있음을 의미한다.These results suggest that gene expression was successfully induced by noninvasive light stimulation, and that the two mutants (FLAG-monSTIM1 and GFP-CRY2-STIM1(318-450)) still possess ultrahigh light sensitivity similar to that of native monSTIM1.

3.2. 성상세포에서 monSTIM1 변이체의 적용3.2. Application of monSTIM1 mutants in astrocytes

성상세포에서 Ca2+ 신호를 조절하는 monSTIM1의 능력을 평가하기 위해, CA1 성상세포에서 GfaABC1D 프로모터에 의해 구동되는 FLAG-monSTIM1 발현 여부 및 GFAP 양성 세포에서 선택적 발현 여부를 확인하였고, 그 결과를 도 8에 나타내었다.To evaluate the ability of monSTIM1 to regulate Ca2+ signals in astrocytes, we examined whether FLAG-monSTIM1 expression driven by the GfaABC1D promoter was expressed in CA1 astrocytes and whether selective expression occurred in GFAP-positive cells, and the results are shown in Fig. 8.

도 8은 성상세포에 일 양상의 monSTIM1 변이체를 적용한 결과를 나타낸 것이다: 도 8a는 해마 CA1 영역을 표적으로 한 GfaABC1D 프로모터의 제어 하에 발현된 FLAG-monSTIM1을 보여주는 사진이고(녹색, FLAG; 빨간색, c-Fos; 파란색, DAPI. 스케일 바, 100 μm); 도 8b는 비침습적 광 전달 유무에 관계없이 FLAG-monSTIM1을 발현하는 cFos 양성 세포를 보여주는 사진이며(녹색, FLAG; 청색, DAPI; 분홍색, GFAP; 적색, c-Fos. 스케일 바, 50 μm); 도 8c는 CA1 성상세포에서 FLAG-monSTIM1을 발현하는 cFos 양성 세포를 정량화한 그래프이다(FLAG-monSTIM1 (-Light): n=5; FLAG-monSTIM1 (+Light): n=6 mice. 데이터는 평균 ± SEM으로 표시됨 (*p<0.05, **p<0.01; Unpaired student's t test); ns, not significant (p>0.05)).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 μm); 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 μm); Figure 8c is a graph quantifying cFos-positive cells expressing FLAG-monSTIM1 in CA1 astrocytes (FLAG-monSTIM1 (-Light): n=5; FLAG-monSTIM1 (+Light): n=6 mice. Data are presented as mean ± SEM (*p<0.05, **p<0.01; Unpaired student's t test); ns, not significant (p>0.05)).

도 8a 및 도 8b에 나타난 바와 같이, CA1 성상세포에서 GfaABC1D 프로모터에 의해 구동되는 FLAG-monSTIM1을 발현하고, GFAP 양성 세포에서 선택적으로 발현되는 것을 확인하였다. 또한, 빛 자극 시, monSTIM1 양성 성상세포의 약 50%에서 cFos 발현이 강력하게 증가한 반면, monSTIM1 음성 성상세포에서는 이러한 반응이 나타나지 않음을 확인하였다(도 8b, 도 8c). As shown in Fig. 8a and Fig. 8b, we confirmed that FLAG-monSTIM1, driven by the GfaABC1D promoter, was expressed in CA1 astrocytes and selectively expressed in GFAP-positive cells. In addition, when light stimulated, cFos expression was strongly increased in about 50% of monSTIM1-positive astrocytes, whereas this response was not observed in monSTIM1-negative astrocytes (Fig. 8b and Fig. 8c).

이러한 결과는 조사된 monSTIM1 변이체의 비침습적 빛 자극이 신경세포와 신경교세포에서 Ca2+ 신호를 효과적이고 선택적으로 활성화함을 의미하는 것이다.These results imply that noninvasive light stimulation of the investigated monSTIM1 mutant effectively and selectively activates Ca2 + signaling in neurons and glial cells.

실험예 4. SST-Cre 마우스 계통에서 Cre-의존적인 FLAG-monSTIM1 발현 제어Experimental Example 4. Cre-dependent FLAG-monSTIM1 expression control in the SST-Cre mouse strain

SST-Cre 마우스 계통에서 Cre-의존적인 FLAG-monSTIM1 발현 제어(Cre-dependent FLAG-monSTIM1 expression control) 여부를 확인하기 위해, Somatostatin (SST) interneuron 에 Cre recombinase 가 발현되어 있는 마우스와 Ai14 마우스 (Rosa26 locusCAG(promoter)-loxp-Stop-loxp-tdTomato)를 교배한 마우스(SST-Cre; Ai-14) 뇌의 오른쪽 전측 대상피질(Anterior Cingulate Cortex, ACC)과 해마의 CA1 영역에 바이러스(AAV-nEF1α-DIO-FLAG-CRY2(EA9)-STIM1(238-685))를 주입하고, 실험예 3의 방법으로 면역조직화학염색을 수행하였다. To determine whether Cre-dependent FLAG-monSTIM1 expression control exists in the SST-Cre mouse strain, The virus (AAV-nEF1α-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.

도9는 SST-Cre 마우스 계통에서 Cre-의존적인 FLAG-monSTIM1 발현 제어의 적용 결과를 나타낸 것으로, FLAG-monSTIM1을 발현하는 SST 양성 세포를 보여주는 이미지를 나타낸 사진이다(파란색은 DAPI, 녹색은 FLAG, 빨간색은 tdTomato).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).

도 9에 나타난 바와 같이, 붉은색 형광단백질 (tdTomato)로 확인되는 SST interneuron 에 특이적으로 녹색 형광단백질 (FLAG)이 발현됨을 확인하였다. 이는 SST interneuron에 FLAG-monSTIM1이 선택적으로 발현됨을 의미하는 것으로, FLAG-monSTIM1 가 excitatory neuron 과 astrocyte 뿐만 아니라 Cre recombinase 가 발현되어 있는 세포 특이적으로 발현될 수 있는 플랫폼임을 의미한다. As shown in Fig. 9, it was confirmed that green fluorescent protein (FLAG) was specifically expressed in SST interneurons identified by red fluorescent protein (tdTomato). This means that 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.

Claims (11)

태그(Tag) 단백질; CRY2(cryptochrome 2) 단백질 또는 그의 변이체; 및 STIMI1(Stromal interaction molecule 1) 단백질을 포함하는 융합단백질.A fusion protein comprising a Tag protein; CRY2 (cryptochrome 2) protein or a mutant thereof; and STIMI1 (Stromal interaction molecule 1) protein. 청구항 1에 있어서, 상기 태그 단백질은 FLAG 단백질 또는 HA 단백질인 것인 융합단백질.A fusion protein according to claim 1, wherein the tag protein is a FLAG protein or an HA protein. 청구항 1에 있어서, 상기 STIMI1 단백질은 세포질 STIM1 단편(cytosolic STIM1 fragment)인 것인 융합단백질.A fusion protein according to claim 1, wherein the STIMI1 protein is a cytosolic STIM1 fragment. 청구항 1 내지 3 중 어느 한 항의 융합단백질을 암호화하는 폴리뉴클레오티드.A polynucleotide encoding a fusion protein according to any one of claims 1 to 3. 청구항 4의 폴리뉴클레오티드를 포함하는 벡터.A vector comprising the polynucleotide of claim 4. 청구항 5에 있어서, 상기 벡터는 아데노 부속 바이러스(AAV) 벡터인 것인 벡터.A vector according to claim 5, wherein the vector is an adeno-associated virus (AAV) vector. CIB1(cryptochromeinteracting basic-helix-loop-helix 1) 단백질 또는 그의 단편이 부착된 STIMI1(Stromal interaction molecule 1) 단백질; 및 CRY2(cryptochrome 2) 단백질 또는 그의 변이체를 포함하는 Ca2+ 모듈레이터(modulator).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. 태그 단백질이 부착된 STIMI1 단백질; 및 상기 태그 단백질에 결합능이 있는 나노바디 항체가 부착된 CRY2단백질 또는 그의 변이체를 포함하는 Ca2+ 모듈레이터(modulator).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. STIMI1 단백질 또는 그의 단편을 암호화하는 폴리뉴클레오티드를 포함하는 제1 벡터; 및 A first vector comprising a polynucleotide encoding a STIMI1 protein or a fragment thereof; and CRY2단백질 또는 그의 변이체를 암호화하는 폴리뉴클레오티드를 포함하는 제2 벡터를 포함하는 발현 시스템.An expression system comprising a second vector comprising a polynucleotide encoding a CRY2 protein or a variant thereof. 청구항 9에 있어서, 상기 제1 벡터는 CIB1 단백질 또는 그의 단편; 또는 태그 단백질을 암호화하는 폴리뉴클레오티드를 추가적으로 포함하는 것인 발현 시스템.An expression system according to claim 9, wherein the first vector additionally comprises a polynucleotide encoding a CIB1 protein or a fragment thereof; or a tag protein. 청구항 9에 있어서, 상기 제2 벡터는 태그 단백질에 결합능이 있는 나노바디 항체를 암호화하는 폴리뉴클레오티드를 포함하는 것인 발현 시스템.An expression system according to claim 9, wherein the second vector comprises a polynucleotide encoding a nanobody antibody having binding ability to a tag protein.
PCT/KR2024/014799 2023-09-27 2024-09-27 Calcium-modulating fusion protein that can be expressed by adeno-associated virus Pending WO2025071367A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2023-0131100 2023-09-27
KR20230131100 2023-09-27

Publications (1)

Publication Number Publication Date
WO2025071367A1 true WO2025071367A1 (en) 2025-04-03

Family

ID=95201944

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2024/014799 Pending WO2025071367A1 (en) 2023-09-27 2024-09-27 Calcium-modulating fusion protein that can be expressed by adeno-associated virus

Country Status (2)

Country Link
KR (1) KR20250047636A (en)
WO (1) WO2025071367A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20130088304A (en) * 2012-01-31 2013-08-08 한국과학기술원 Fusion proteins capable of regulating intracellular calcium ion level by light and uses thereof
KR20200055677A (en) * 2018-11-13 2020-05-21 기초과학연구원 Modified CRY2 with high photosensitivity and use of the same
KR20210094950A (en) * 2020-01-22 2021-07-30 (주)휴룩스 Therapeutic use of fusion protein having modified cry2 and therapeutic use based the same

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20130088304A (en) * 2012-01-31 2013-08-08 한국과학기술원 Fusion proteins capable of regulating intracellular calcium ion level by light and uses thereof
KR20200055677A (en) * 2018-11-13 2020-05-21 기초과학연구원 Modified CRY2 with high photosensitivity and use of the same
KR20210094950A (en) * 2020-01-22 2021-07-30 (주)휴룩스 Therapeutic use of fusion protein having modified cry2 and therapeutic use based the same

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
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 (en) 2025-04-04

Similar Documents

Publication Publication Date Title
WO2017034330A1 (en) Cell-permeable bone morphogenetic protein (cp-bmp) recombinant protein and use thereof
US10561742B2 (en) Methods and compositions for treatment of disease or injury of the nervous system
JP2003511071A (en) Cell penetrating peptide inhibitor of JNK signal transduction pathway
EA014330B1 (en) Cell-permeable peptide inhibitors of the jnk signal transduction pathway
WO2012050402A2 (en) Cell-permeable recombinant parkin protein and a pharmaceutical composition for treating degenerative brain diseases containing the same
WO2015156649A1 (en) Peptide having fibrosis inhibitory activity and composition containing same
WO2014183491A1 (en) Polypeptide having anti-tumor activity and use thereof
WO2017018787A1 (en) Improved cell-permeable (icp) parkin recombinant protein and use thereof
WO2017026779A1 (en) Improved cell-permeable cre (icp-cre) recombinant protein and use thereof
CN110267673A (en) Optogenetic Vision Restoration Using CHRIMSON
US12139519B2 (en) CRY2 variant having increased photosensitivity and use thereof
EP3334755A1 (en) Improved cell-permeable reprogramming factor (icp-rf) recombinant protein and use thereof
US7033794B2 (en) Basolateral sorting signal and inhibitors thereof
WO2025071367A1 (en) Calcium-modulating fusion protein that can be expressed by adeno-associated virus
WO2020009320A1 (en) Composition comprising oct4 for inducing direct transdifferentiation into bone-related cell
JP7474512B2 (en) Photoactivatable Tet expression control system
KR20210094952A (en) Exosome secretion method using modified cry2 fusion protein
KR20210094950A (en) Therapeutic use of fusion protein having modified cry2 and therapeutic use based the same
WO2024054062A1 (en) Novel polypeptide composition for intracellular transfection
WO2018088733A1 (en) Novel protein transduction domain and use thereof
US20070065904A1 (en) Recombinant expression vectors for functional nav1.9 sodium channels
WO2017209519A1 (en) Ab6 family designer ligands of tgf-beta superfamily
WO2021162375A1 (en) Exosome comprising photocleavable protein, and use thereof
KR20210094951A (en) Mass cell culture method using modified cry2 fusion protein
Magee Cell adhesion molecules and intracellular signalling: from fly to man

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