WO2022055320A1 - Adeno-associated virus vector for targeted gene delivery - Google Patents

Adeno-associated virus vector for targeted gene delivery Download PDF

Info

Publication number
WO2022055320A1
WO2022055320A1 PCT/KR2021/012420 KR2021012420W WO2022055320A1 WO 2022055320 A1 WO2022055320 A1 WO 2022055320A1 KR 2021012420 W KR2021012420 W KR 2021012420W WO 2022055320 A1 WO2022055320 A1 WO 2022055320A1
Authority
WO
WIPO (PCT)
Prior art keywords
sequence
vector
promoter
aimp2
disease
Prior art date
Application number
PCT/KR2021/012420
Other languages
French (fr)
Korean (ko)
Inventor
백경화
변미란
Original Assignee
주식회사 제너로스
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 주식회사 제너로스 filed Critical 주식회사 제너로스
Publication of WO2022055320A1 publication Critical patent/WO2022055320A1/en

Links

Images

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • C07K14/4703Inhibitors; Suppressors
    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • C12N2310/141MicroRNAs, miRNAs
    • 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

  • the present invention relates to an adeno-associated virus vector capable of delivering a gene by targeting the central nervous system and various uses thereof.
  • the mammalian brain can perform complex functions by generating a systematic neural network through a series of processes such as division and differentiation of neural stem cells, survival and death, and synapse formation. Even in the adult period, animal nerve cells produce many substances necessary for nerve growth, and axons and dendrites grow. remodeling), so it can be said that differentiation continues. In the process of cell differentiation and synapse formation, nerve cells die if they do not receive target-derived survival factors such as nerve growth factors. become a major cause of disease. Unlike the central nervous system, when the peripheral nervous system of animals is damaged, the axon regenerates over a long period of time.
  • the posterior axon at the site of nerve injury is degenerated by a process known as Wallerian degeneration, the nerve cell body resumes axonal regrowth, and Schwann cells divide and determine the target nerve by survival and death. It regenerates through the process of development, such as re-differentiation.
  • GDNF glial derived neuronal factor
  • AIMP2-DX2 is known to inhibit apoptosis by inhibiting the function of AIMP2 as an alternative splicing variant of AIMP2, a tumor suppressor factor involved in apoptosis in many ways.
  • AIMP2-DX2 inhibits the pro-apoptotic interaction of AIMP2 by competing with AIMP2 in the presence of TGF- ⁇ , TNF- ⁇ and DNA damage signals.
  • AIMP2 released from MSC (Multi-Synthetase Complex) by TGF- ⁇ and TNF- ⁇ stimulation binds to FBP and TRAF2, respectively, and is known to induce cell death by inducing ubiquitin-mediated degradation of FBP and TRAF2,
  • the presence of AIMP2-DX2 induces cell survival by competitively inhibiting the binding of AIMP2 to FBP and TRAF2.
  • a DNA damage signal such as UV irradiation
  • AIMP2 interacts with p53 to protect p53 from MDM2-mediated degradation to induce apoptosis, but AIMP2-DX2 inhibits the binding of p53 to AIMP2, leading to cell survival. do.
  • the present inventors have developed an adeno-associated viral vector comprising an ITR from which the TRS (terminal resolution site) region has been removed, a sequence targeting miR-142-3p, and a sequence encoding an AIMP2 mutant (AIMP2-DX2) gene with a deletion of exon 2 was constructed, and the present invention was completed by confirming that the vector can be selectively expressed only in the central nervous system, such as the brain or spinal cord.
  • an object of the present invention is an adeno-containing sequence encoding the ITR, miR-142-3p targeting sequence and exon 2 deleted AIMP2 variant (AIMP2-DX2) gene from which the TRS (terminal resolution site) region has been removed.
  • AIMP2-DX2 exon 2 deleted AIMP2 variant
  • the present invention provides a sequence encoding a first inverted terminal repeat (ITR); promoter sequence; a sequence encoding AIMP2-DX2 operably linked to the promoter sequence; a sequence targeting mir142-3p linked to the sequence encoding AIMP2-DX2; And it provides an adeno-associated viral vector comprising a sequence encoding a second ITR, comprising the sequence sequentially in a 5' to 3' direction.
  • ITR inverted terminal repeat
  • the present invention provides a composition for preventing or treating neurodegenerative diseases comprising the vector of the present invention.
  • the present invention provides a method for preventing or treating a neurodegenerative disease comprising administering the recombinant vector of the present invention to an individual in need of treatment.
  • the present invention provides a method for treating a neurodegenerative disease comprising administering the vector of the present invention to a subject suffering from a neurodegenerative disease in a pharmaceutically effective amount.
  • the present invention provides a use of the vector of the present invention for use in the manufacture of a pharmaceutical composition for the prevention or treatment of neurodegenerative diseases.
  • the adeno-associated virus vector of the present invention can be selectively expressed only in the central nervous system among various tissues, and thus can be usefully used in related industries.
  • FIG. 1 is a diagram showing a vector map of the adeno-associated viral vector of the present invention.
  • FIG. 2 is a diagram showing a vector map of the adeno-associated viral vector of the present invention.
  • Figure 3 is a schematic diagram showing the production process of the adeno-associated viral vector of the present invention.
  • FIG. 4 is a diagram showing that DX2 competitively inhibits the binding of TRAF2 and AIMP2, thereby reducing the pro-apoptotic activity of AIMP2.
  • the upper panel shows that DX2 inhibits AIMP2 binding to TRAF2 in a dose-dependent manner.
  • Lower panel shows NF- ⁇ B activation and cell viability.
  • FIG. 5 is a diagram showing that DX2 reduces TNF ⁇ -induced neuronal death.
  • A SH-SY5Y cells were transfected with EV (empty vector), DX2, and SOD1 G93A, and the level of ubiquitinated TRAF2 was confirmed by immunoblotting.
  • B SH-SY5Y cells were transfected with EV, DX2, SOD1 WT, G93A, and G85R and treated with TNF- ⁇ and CHX for 6 h. Cell viability was confirmed by MTT assay.
  • C Primary neuronal cells of WT and mutant SOD1 mice were transfected with ith EV and DX2, and viability was confirmed by performing MTT analysis.
  • FIG. 6 is a diagram showing the TRAF2 expression level in the ALS mouse model.
  • A Immunohistochemical staining results showing the expression of SOD1 (upper panel) and TRAF2 (lower panel) in each mouse.
  • B Immunoblot analysis results showing the expression of TRAF2, EPRS, KARS1, and SOD1 in each mouse.
  • CD Expression of GFP and DX2 in the spinal cord of mice:
  • C Expression of GFP after injection of AAV-GFP and
  • Br brain cortex, SC; spinal cord, scAAV-GFP; 2.4 x 10 10 vg, scAAV-DX2; 2.4x10 10 vg.
  • FIG. 7 is a view confirming the behavior, disease onset time and lifespan of ALS model mice by administration of DX2.
  • A Behavior was improved in the group administered with AAV-DX2.
  • B The onset of disease was delayed in the AAV-DX2 group.
  • TRAF2 which is known to inhibit apoptosis among inflammatory signaling factors caused by TNF- ⁇ , significantly decreased in mice injected with AAV-GFP (Con) into AAV-GFP (Con) mice injected with AAV-GFP (Con), whereas in mice injected with AAV-DX2 It was confirmed that the expression of TRAF2 was restored to a level similar to that of WT.
  • FIG. 9 is a schematic diagram showing the mechanism by which DX2 inhibits cell death by blocking ubiquitination of TRAF2.
  • FIG. 10 is a diagram showing that 67 laminin receptor stability is reduced in ALS.
  • A SK-N-SH cells were transfected with SOD1 WT and G93A. Cells were harvested and then immunoblotted for 67 laminin receptor (LR).
  • B Neuronal cells were transfected with SOD1 WT and G93A and then cultured on 22x22 coverslips. After fixing the cells, they were treated with KARS1 or 67LR antibody, and images were obtained by confocal microscopy.
  • FIG. 11 is a diagram showing the effect of DX2 on 67LR expression and Anoikis.
  • A Neuroblastoma cells were transfected with strep-DX2, SOD1 WT, G93A and G85R for 24 h. After harvesting the cells and performing ultracentrifugation, the samples were subjected to immunoblotting.
  • B SK-N-SH cells were transfected with SOD1 WT, G93A, KARS1, AIMP2 and DX2. After harvesting the cells, Western blot analysis was performed on the 67LR.
  • C Neuronal cells were transfected with SOD1 G93A and then either EV or DX2.
  • FIG. 12 is a diagram comparing scAAV and ssAAV.
  • A AAV-GFP transformation efficiency test result.
  • SK-SY5Y (neuroblastoma) cells were infected with scAAV-GFP or ssAAV-GFP (single-stranded AAV-GFP) and 48 hours later, GFP expression of (B) ssAAV-GFP and (C) scAAV-GFP was examined by fluorescence microscopy. observed.
  • AIMP2-DX2 is an alternative splicing variant of AIMP2, an apoptosis factor involved in apoptosis in many ways, and is known to inhibit apoptosis of tumors by inhibiting the function of AIMP2.
  • AIMP2 splicing variant refers to a variant resulting from partial or total loss of the region of exon 2 among exons 1 to 4, wherein the variant forms a heterodimer with the AIMP2 protein to interfere with the normal function of AIMP2. means that In addition, when the AIMP2 mutant is overexpressed in cells or tissues, cancer may be induced. Therefore, it is necessary to induce tissue-specific expression in order to suppress the induction of cancer.
  • AIMP2-DX2 can treat inflammatory diseases.
  • AIMP2/p38 promotes ubiquitination of TRAF2 to regulate TNF-alpha-induced apoptosis
  • AIMP2/p38 AIMP2-DX2 a splice variant of p38, acts as a competitive inhibitor of AIMP2 and inhibits ubiquitin of TRAF2, thereby inhibiting TNF-alpha-induced apoptosis, thereby promoting tumor progression and inflammatory marker Cox-2 It has been shown to inhibit the expression of
  • AIMP2-DX2 has been previously reported to have increased expression in lung cancer, AIMP2-DX2 itself does not have the ability to form tumors to transform normal cells.
  • AIMP2-DX2 can treat neurological diseases (US2019/0298858 A1).
  • the term "recombinant vector” refers to a vector capable of expressing a target protein or target RNA in an appropriate host cell, and refers to a genetic construct comprising essential regulatory elements operably linked to express a gene insert.
  • operably linked refers to a functional linkage between a nucleic acid expression control sequence and a nucleic acid sequence encoding a desired protein or RNA to perform a general function.
  • a promoter and a nucleic acid sequence encoding a protein or RNA may be operably linked to affect expression of the encoding nucleic acid sequence.
  • the operative linkage with the recombinant vector can be prepared using a genetic recombination technique well known in the art, and site-specific DNA cleavage and ligation are performed using enzymes generally known in the art.
  • the present invention provides a sequence encoding a first inverted terminal repeat (ITR); promoter sequence; a sequence encoding AIMP2-DX2 operably linked to the promoter sequence; a sequence targeting mir142-3p linked to the sequence encoding AIMP2-DX2; And it provides an adeno-associated viral vector comprising a sequence encoding a second ITR, wherein the sequence is sequentially included in a 5' to 3' direction.
  • ITR inverted terminal repeat
  • the adeno-associated viral vector of the present invention may have a vector structure as shown in FIGS. 1 and 2, but is not limited thereto.
  • the adeno-associated viral vector may include the sequence represented by SEQ ID NO: 7, but is not limited thereto.
  • the vector of the present invention can express DX2 specifically in the central nervous system, but is not expressed in hematopoietic cells such as leukocytes and lymphoid cells. Accordingly, the vectors of the present invention may be useful for specifically targeting neurons to treat neurological diseases.
  • AIMP2-DX2 polypeptide is a splicing variant of AIMP2, in which the second exon of AIMP2 is deleted.
  • the AIMP2-DX2 gene includes the nucleotide sequence shown in SEQ ID NO: 3, and the AIMP2-DX2 polypeptide includes the amino acid sequence shown in SEQ ID NO: 4.
  • the AIMP2-DX2 gene is at least 90%, at least 93%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% homologous to the sequence represented by SEQ ID NO: 4 It may comprise a nucleotide sequence encoding an amino acid.
  • the AIMP2-DX2 gene may include a nucleotide sequence encoding the amino acid sequence of SEQ ID NO: 4.
  • the AIMP2-DX2 gene may comprise a nucleotide sequence that is at least 90%, at least 93%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% homologous to the nucleotide sequence of SEQ ID NO:3.
  • the AIMP2-DX2 gene may include a nucleotide sequence represented by SEQ ID NO: 3.
  • micro RNA refers to a noncoding RNA consisting of about 20, 21, 22, 23 or 24 nucleotides MicroRNA (abbreviated miRNA) serves to regulate gene expression.
  • the microRNA acts in the post-transcription stage after gene transcription, and in mammals, it is known that about 60% of the gene expression is regulated by microRNA.
  • MicroRNAs play an important role in a variety of in vivo processes and have been implicated in cancer, heart disease, and neurological diseases.
  • miRNA is single-stranded RNA, a target sequence of either side of single-stranded RNA can be used as long as expression of the target gene among premature two-stranded RNAs can be suppressed.
  • miR-142 includes miR-142-3p and miR-142-5p.
  • any target sequence may be used, and miR-142 includes both miR-142-3p and miR-142-5p. is included, and may preferably be miR-142-3p.
  • miR-142-3p refers to its gene being present at the site of translocation in B-cell leukemia, and is expressed in hematopoietic tissues (bone marrow, spleen, thymus, etc.). is known In addition, expression of miR-142-3p has been confirmed in mouse fetal liver (fetal hematopoietic tissue), and is known to be involved in the differentiation of the hematopoietic system.
  • the nucleic acid targeting miR-142-3p may include a nucleotide sequence comprising ACACTA.
  • the miR-142 target nucleic acid may comprise a nucleotide sequence comprising ACACTA and 1-17 additional contiguous nucleotides of SEQ ID NO: 5.
  • the miR-142 target nucleic acid may include a nucleotide sequence comprising ACACTA and 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17 additional nucleotides.
  • the nucleic acid targeting miR-142-3p has at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% of the nucleotide sequence of SEQ ID NO: 5 (TCCATAAAGTAGGAAACACTACA; miR-142-3p). % homologous nucleotide sequences.
  • the miR-142 target nucleic acid may comprise the nucleotide sequence of SEQ ID NO:5.
  • the sequence targeting miR-142-3p may be repeated 2-10 times, for example, it may be repeated at least 2-8 times, at least 2-6 times, at least 4 times.
  • the sequence targeting the miR-142-3p may include the nucleotide sequence of SEQ ID NO: 5.
  • the sequence encoding the first ITR or the sequence encoding the second ITR may be a sequence in which a terminal resolution site (TRS) site has been removed, for example, the sequence encoding the first ITR or the second ITR. Either or both of the sequences may have the TRS region removed, but is not limited thereto.
  • the sequence encoding the first ITR (left ITR) is the sequence shown in SEQ ID NO: 1
  • the sequence coding the second ITR (right ITR) is the sequence shown in SEQ ID NO: 2.
  • the adeno-associated virus is a single-stranded provirus, and requires an auxiliary virus to replicate, and the AAV genome is 4,680 bp, which can be inserted into a specific site on chromosome 19 of an infected cell. Do.
  • the trans-gene is inserted into plasmid DNA linked by two inverted terminal repeat (ITR) sequence portions of 145 bp each and a signal sequence portion.
  • ITR inverted terminal repeat
  • Transgene-expressing DNA, AAV rep part, plasmid DNA expressing cap part, and helper DNA are transfected into 293 cells to generate an adeno-associated virus.
  • AAV has the advantages of a wide range of host cells to deliver genes, fewer immune side effects during repeated administration, and a long gene expression period.
  • the AAV genome is safe to integrate into the host cell's chromosomes and does not alter or rearrange the host's gene expression.
  • the adeno-associated virus has various serotypes.
  • Adeno-associated virus serotype 2 cystic fibrosis, hemophilia, and Canavan It is currently used for clinical gene transfer of disease.
  • rAAV recombinant adeno-associated virus
  • the adeno-associated virus may be a self complementary adeno-associated virus (scAAV), but is not limited thereto.
  • scAAV self complementary adeno-associated virus
  • EagI-NotI, Eco53KI, SacI, EcoRI, acc65I, KpnI and SexAI between the promoter and the sequence encoding AIMP2-DX2 may include any one or more restriction enzyme sites from the group consisting of, but is not limited thereto.
  • the first stop codon, the poly A coding sequence, and the second stop codon may be sequentially positioned toward the 3' end of the mir142-3P-targeting sequence, but the present invention is not limited thereto.
  • the stop codon may be repeated three or more times, but is not limited thereto.
  • One or more restriction enzyme sites from the group consisting of BspDI-ClaI, BamHI, and StuI may be included between the first stop codon and the second stop codon.
  • the adeno-associated viral recombinant vector of the present invention consists of a composition comprising ITR, miR-142-3p, and AIMP2-DX2, and regulates the inhibition of AIMP2-DX2 expression in cd45-derived cells, particularly lymphocytes and leukocytes, thereby reducing lymph nodes (lymph nodes). node) can control the side effect of overexpression of AIMP2-DX2 mutant.
  • the AIMP2-DX2 variant can be expressed only in the central nervous system or can be selectively expressed only in brain or spinal cord tissues and not other types of cells or tissues.
  • AIMP2-DX2 in cells expressing miR-142-3p, the expression or activity of AIMP2-DX2 can be inhibited, and in cells in which miR-142-3p is not expressed, the expression or activity of AIMP2-DX2 can be maintained or increased.
  • AIMP2-DX2 when the recombinant vector is injected in vivo, AIMP2-DX2 can be specifically expressed only in the central nervous system.
  • the promoter is a retrovirus (LTR) promoter, a cytomegalovirus (CMV) promoter, a Rous sarcoma virus (RSV) promoter, an MMT promoter, an EF-1 alpha promoter, a UB6 promoter, a chicken beta-actin promoter, a CAG promoter, a RPE65 promoter, It may be a promoter selected from the group consisting of an opsin promoter and combinations thereof, for example, a cytomegalovirus (CMV) promoter.
  • % sequence homology “% homology” or “% homology” to a nucleotide or amino acid sequence is identified by comparing two optimally aligned sequences with a comparison region, wherein the nucleotide sequence in the comparison region is A portion of may contain additions or deletions (ie, gaps) compared to a reference sequence (not including additions or deletions) for the optimal alignment of the two sequences.
  • the protein of the present invention includes not only a protein having its native amino acid sequence but also an amino acid sequence variant thereof within the scope of the present invention.
  • a variant of the protein of the present invention refers to a protein having a sequence different from the native amino acid sequence by deletion, insertion, non-conservative or conservative substitution of one or more amino acid residues, or a combination thereof.
  • Amino acid exchanges in proteins and peptides that do not entirely alter the activity of the molecule are known in the art (H.Neurath, R.L.Hill, The Proteins, Academic Press, New York, 1979).
  • the protein or variant thereof can be extracted from nature, synthesized (Merrifleld, J. Amer. chem. Soc. 85:2149-2156, 1963), or prepared by a genetic recombination method based on a DNA sequence (Sambrook et al. , Molecular Cloning, Cold Spring Harbor Laboratory Press, New York, USA, 2nd ed., 1989).
  • the amino acid mutation is made based on the relative similarity of the amino acid side chain substituents, such as hydrophobicity, hydrophilicity, charge, size, and the like.
  • arginine, lysine and histidine are all positively charged residues; alanine, glycine and serine have similar sizes; It can be seen that phenylalanine, tryptophan and tyrosine have similar shapes. Therefore, based on these considerations, arginine, lysine and histidine; alanine, glycine and serine; And phenylalanine, tryptophan and tyrosine can be said to be biologically functional equivalents.
  • the hydropathic index of the amino acid may be considered.
  • Each amino acid is assigned a hydrophobicity index according to its hydrophobicity and charge: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cysteine (+2.5); methionine (+1.9); alanine (+1.8); glycine (-0.4); threonine (-0.7); serine (-0.8); tryptophan (-0.9); tyrosine (-1.3); proline (-1.6); histidine (-3.2); glutamate (-3.5); glutamine (-3.5); aspartate (-3.5); asparagine (-3.5); lysine (-3.9); and arginine (-4.5).
  • the hydrophobic amino acid index is very important in conferring an interactive biological function of a protein. It is a known fact that amino acids having a similar hydrophobicity index must be substituted to retain similar biological activity. When introducing a mutation with reference to the hydrophobicity index, the substitution is made between amino acids showing a difference in the hydrophobicity index, preferably within ⁇ 2, more preferably within ⁇ 1, and even more preferably within ⁇ 0.5.
  • the substitution is made between amino acids exhibiting a difference in the hydrophilicity value within preferably ⁇ 2, more preferably within ⁇ 1, and even more preferably within ⁇ 0.5.
  • Amino acid exchanges in proteins that do not entirely alter the activity of the molecule are known in the art (H. Neurath, R.L. Hill, The Proteins, Academic Press, New York, 1979).
  • the most common exchanges are amino acid residues Ala/Ser, Val/Ile, Asp/Glu, Thr/Ser, Ala/Gly, Ala/Thr, Ser/Asn, Ala/Val, Ser/Gly, Thy/Phe, Ala/ It is an exchange between Pro, Lys/Arg, Asp/Asn, Leu/Ile, Leu/Val, Ala/Glu, Asp/Gly.
  • the vector of the present invention may be fused with other sequences as necessary to facilitate protein purification, and the fused sequences include, for example, glutathione S-transferase (Pharmacia, USA), maltose binding protein (NEB, USA). ), FLAG (IBI, USA) and 6x His (hexahistidine; Quiagen, USA) may be used, but is not limited thereto.
  • the expression vector of the present invention may contain an antibiotic resistance gene commonly used in the art as a selection marker, for example, ampicillin, gentamicin, carbenicillin, chloramphenicol, streptomycin, kanamycin, geneticin , resistance genes to neomycin and tetracycline.
  • the present invention also provides a method for delivering and expressing a heterologous gene such as AIMP2-DX2 to neurons, comprising administering the adeno-associated viral vector to an individual in need of treatment.
  • a heterologous gene such as AIMP2-DX2
  • the expression of the heterologous gene is increased in the central nervous system (the spinal canal and brain tissue), and the expression of the heterologous gene can be controlled in other tissues.
  • the present invention also provides a composition for preventing or treating neurodegenerative diseases comprising the adeno-associated viral vector as an active ingredient.
  • the present invention provides a method for preventing or treating a neurodegenerative disease comprising administering the recombinant vector of the present invention to an individual in need of treatment.
  • the neurological disease is Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, retinal degeneration, mild cognitive impairment, multiple-infarct dementia (Multi -infarct dementia, frontotemporal dementia, dementia with Lewy bodies, Huntington's disease, neurodegenerative disease, metabolic brain disease, depression, epilepsy, multiple sclerosis, cortex Corticobasal degeneration, multiple system atrophy, progressive supranuclear palsy, dentatorubropallidoluysian atrophy, spinocerebella ataxia It may be one or more selected from the group consisting of lateral sclerosis (primary lateral sclerosis), spinal muscular atrophy and stroke (stroke), but is not limited thereto.
  • lateral sclerosis primary lateral sclerosis
  • spinal muscular atrophy and stroke stroke
  • the neurological disease is amyotrophic lateral sclerosis; ALS).
  • the treatment may be to improve memory, motor impairment, motor ability or prolong the lifespan of an individual suffering from neurological disease such as ALS, Alzheimer's disease or Parkinson's disease.
  • the treatment may be to improve motor performance or prolong the lifespan of an individual suffering from a neurological disease such as ALS.
  • treatment includes partial cure, improvement and alleviation of symptoms according to the neurodegenerative disease as well as the complete cure of the neurodegenerative disease as a result of applying the pharmaceutical composition of the present invention to an individual having a neurodegenerative disease.
  • prevention refers to suppressing or blocking symptoms or phenomena such as cognitive impairment, behavioral disorder, and nerve destruction by applying the pharmaceutical composition of the present invention to an individual having a neurodegenerative disease, thereby causing various symptoms according to the neurodegenerative disease. This means that it does not happen in advance.
  • the pharmaceutical composition of the present invention may further include an adjuvant in addition to the active ingredient.
  • the adjuvant may be used without limitation as long as it is known in the art, for example, Freund's complete adjuvant or incomplete adjuvant may be further included to increase the immunity thereof.
  • the pharmaceutical composition according to the present invention may be prepared in a form in which the active ingredient is incorporated into a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier includes carriers, excipients and diluents commonly used in the pharmaceutical field.
  • Pharmaceutically acceptable carriers that can be used in the pharmaceutical composition of the present invention include, but are not limited to, lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia gum, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, polyvinyl pyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil.
  • the pharmaceutical composition of the present invention may be formulated in the form of oral dosage forms such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols, etc., external preparations, suppositories, or sterile injection solutions according to conventional methods, respectively. .
  • Solid preparations for oral administration include tablets, pills, powders, granules, capsules, etc., and such solid preparations include at least one excipient in the active ingredient, for example, starch, calcium carbonate, sucrose, lactose, gelatin. It can be prepared by mixing and the like. In addition to simple excipients, lubricants such as magnesium stearate and talc may also be used.
  • Liquid formulations for oral administration include suspensions, solutions, emulsions, syrups, etc.
  • compositions for parenteral administration include sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, lyophilized formulations and suppositories.
  • non-aqueous solvent and suspending agent propylene glycol, polyethylene glycol, vegetable oil such as olive oil, and injectable ester such as ethyl oleate may be used.
  • base of the suppository witepsol, tween 61, cacao butter, laurin, glycerogelatin, and the like can be used.
  • the pharmaceutical composition according to the present invention may be administered to an individual by various routes. Any mode of administration can be envisaged, for example, it can be administered by oral, intravenous, intramuscular, subcutaneous, intraperitoneal injection, brain striatal injection, intrathecal injection, and the like.
  • Preferred dosage of the therapeutic composition according to the present invention depends on factors such as formulation method, administration method, age, weight, sex, degree of disease symptoms, food, administration time, administration route, excretion rate, and response sensitivity of the patient. Although different, it may be appropriately selected by those skilled in the art.
  • the therapeutic agent includes intravenous injection, subcutaneous fat injection, intramuscular injection, and direct injection into the ventricle or spinal cord using a micro-injector.
  • the effective dose is 0.05 to 15 mg/kg of vector per 1 kg of body weight, and 5 ⁇ 10 11 to 3.3 in the case of recombinant virus.
  • ⁇ 10 14 virus particles (2.5 ⁇ 10 12 to 1.5 ⁇ 10 16 IU)/kg for cells 5 ⁇ 10 2 to 5 ⁇ 10 7 cells/kg, preferably 0.1 to 10 mg/kg for vectors kg, 5 ⁇ 10 12 to 3.3 ⁇ 10 13 particles (2.5 ⁇ 10 13 to 1.5 ⁇ 10 15 IU)/kg for recombinant virus, 5 ⁇ 10 3 to 5 ⁇ 10 6 cells/kg for cells, per week It may be administered 2-3 times.
  • the composition as described above is not necessarily limited thereto, and may vary depending on the patient's condition and the degree of onset of a neurological disease.
  • compositions as described above is not necessarily limited thereto, and may vary depending on the patient's condition and the degree of onset of a neurological disease. More specifically, the pharmaceutical composition of the present invention contains 1 ⁇ 10 10 to 1 ⁇ 10 12 vg (virus genome)/mL of recombinant adeno-associated virus, and typically 1 ⁇ 10 12 vg is injected every other day for 2 weeks. good to do Administration may be administered once a day, or may be administered in several divided doses.
  • the pharmaceutical composition may be formulated in various oral or parenteral dosage forms.
  • Formulations for oral administration include, for example, tablets, pills, hard, soft capsules, solutions, suspensions, emulsifiers, syrups, granules, and the like. crose, mannitol, sorbitol, cellulose and/or glycine), lubricants (eg silica, talc, stearic acid and its magnesium or calcium salts and/or polyethylene glycol).
  • the tablet may contain a binder such as magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidine, and optionally starch, agar, alginic acid. or a disintegrant such as its sodium salt or a boiling mixture and/or absorbent, coloring, flavoring and sweetening agent.
  • the formulation may be prepared by conventional mixing, granulating or coating methods.
  • a representative formulation for parenteral administration is an injection formulation
  • examples of the solvent for the injection formulation include water, Ringer's solution, isotonic saline, or suspension.
  • the sterile, fixed oil of the injectable preparation may be used as a solvent or suspending medium, and any non-irritating fixed oil including mono- and di-glycerides may be used for this purpose.
  • the injection preparation may use a fatty acid such as oleic acid.
  • the present invention provides a method for treating a neurodegenerative disease comprising administering the vector of the present invention to a subject suffering from a neurodegenerative disease in a pharmaceutically effective amount.
  • the present invention provides a use of the vector of the present invention for use in the manufacture of a pharmaceutical composition for the prevention or treatment of neurodegenerative diseases.
  • AIMP2 is one of the proteins involved in the formation of aminoacyl-tRNA synthetase (ARSs), and acts as an apoptosis inhibitor.
  • ARSs aminoacyl-tRNA synthetase
  • AIMP2 mutant of the present invention is represented by the nucleotide sequence of SEQ ID NO: 3, and is represented by the amino acid of SEQ ID NO: 4.
  • the AIMP2 variant of the present invention is overexpressed in tumors, and while it is safely expressed in nerve cells, which are target cells, to suppress the expression of the AIMP2 variant in leukocyte-related cells and lymphocyte-related cells, the expression of the AIMP2 variant can be controlled. miRNAs and their targets were selected.
  • miR-142-3p which is specifically expressed only in hematopoietic cells that produce leukocyte-related cells and lymphocyte-related cells, was selected as a target.
  • microarray data of mouse B cells and computer programming (mirSVR score) of the gene targeted by miR-142-3p were used.
  • the sequence targeting miR-142-3p of the present invention is represented by the nucleotide sequence shown in SEQ ID NO: 5.
  • the sequence targeting miR-142-3p of the present invention includes a restriction enzyme (NheI, Hind III, BmtI) site sequence (ccagaagcttgctagc) and a restriction enzyme (Hind III) site sequence (aagcttgtag) for cloning. It includes a nucleotide sequence represented by SEQ ID NO: 5 that is repeated 4 times together with linkers (tcac and gatatc) connecting them.
  • the sequence encoding the first ITR (SEQ ID NO: 1), the CMV promoter, the AIMP2 mutant of the present invention (SEQ ID NO: 3), miR-142-3p targeting sequence (SEQ ID NO: 5) and the second ITR (SEQ ID NO: 2) were inserted sequentially in the 5' to 3' direction.
  • the binding of the AIMP-2 mutant and the miR-142-3p target sequence was shown as the nucleotide sequence of SEQ ID NO: 6, and specifically, it was cut using NheI and Hind III sites.
  • the adeno-associated viral vector of the present invention is shown in Figures 1 and 2.
  • the vector prepared as described above was produced through the process shown in FIG. 3 .
  • DX2 TNF- ⁇ -induced neuroinflammation inhibitory effect of AIMP2-DX2
  • TNF- ⁇ tumor necrosis factor receptor-associated factor
  • TNF- ⁇ signaling TNF receptor-associated factor
  • TRADD TNF receptor-associated death domain
  • TRAF2 bind to TNFR1, a TNF- ⁇ receptor, and the activated complex mediates IKK activity.
  • caspase-8 caspase-8 is inactivated, TRAF2 is released from the TRADD complex, and the released TRAF2 is ubiquitinated by cIAP, an E3 ubiquitin ligase, to promote cell death.
  • AIMP2 a pro-apoptotic protein, promotes ubiquitination of TRAF2 by cIAP and induces cell death. Therefore, it was confirmed whether DX2, a competitor of AIMP2, affects the binding of TRAF2.
  • Neuroblastoma cells were transfected with mock, DX2 (AIMP2-DX2; AIMP2 from which exo 2 was removed) or AIMP2 expression plasmid and subjected to immunoprecipitation, and the results are shown in FIG. 4 .
  • the upper panel of FIG. 4 shows that DX2 inhibits AIMP2 binding to TRAF2 in a dose-dependent manner.
  • Lower panel shows NF- ⁇ B activation and cell viability.
  • DX2 overexpression plasmids were transfected into primary neurons extracted from WT or SOD1 transgenic mice. The transfected cells were treated with CHX/TNF- ⁇ and the cell death rate was analyzed.
  • CHX/TNF- ⁇ induced apoptosis in both WT or G93A primary neurons, but showed an approximately 13% higher rate of cell death in G93A primary neurons.
  • DX2 significantly reduced apoptosis in both CHX/TNF- ⁇ -treated WT or G93A primary neurons ( FIG. 5C ).
  • TRAF2 is an important protein mediating cell death by the TNF- ⁇ signaling pathway. Therefore, to confirm the importance of TRAF2 expression in ALS disease, TRAF2 levels were measured in an ALS mouse model (SOD1 G93A transgenic mice). Interestingly, the amount of KARS1 binding to AIMP2, which regulates TNF- ⁇ -induced ubiquitination of TRAF2, did not change; However, TRAF2 expression was significantly inhibited in ALS mice ( FIGS. 6A and 6B ). Accordingly, an AAV vector was designed (Example 1) to confirm the potential efficacy of DX2 as a therapeutic agent showing efficient expression of DX2 or GFP in ALS-induced mice.
  • AAV-GFP or AAV-DX2 was injected into the spinal cord of mice, and after 3 weeks, GFP and DX2 expression was analyzed by immunofluorescence and mRNA expression. GFP and DX2 expression was observed at the injection site of the spinal cord, indicating that either AAV-GFP or DX2 could be expressed only at the specific injection site ( FIGS. 6C and 6D ).
  • FIG. 7A it was confirmed that the behavior of the mouse model was improved ( FIG. 7A ) and the onset of the disease was delayed ( FIG. 7B ) due to the intrathecal injection of AAV-DX2.
  • FIG. 7C it was confirmed that the lifespan of the AAV-DX2 injected mouse group was extended compared to the AAV-GFP mouse group ( FIG. 7C ).
  • TNF- ⁇ signal is one of the inflammatory signal transduction systems, and has an important effect on the progression of neuroinflammation and ALS. .
  • FIG. 6 it was confirmed in FIG. 6 that the expression level of TRAF2 significantly decreased in the ALS model mouse.
  • DX2 regulates the expression level of TRAF2 in vitro in FIGS. 4 and 5, thereby suppressing cell death. Based on this, it was confirmed that the expression of DX2 in the actual mouse model regulates the expression of TRAF2.
  • the significantly reduced expression of TRAF2 in the mouse model of ALS is restored to the normal mouse (WT) level by injection of DX2 (AAV-DX2). was confirmed (FIG. 8C).
  • the neuroinflammatory marker factors FIGS. 8A, 8B
  • apoptosis factors FIGGS. 8D, 8E
  • Freeform AIMP2 binds to TRAF2 and promotes ubiquitination of TRAF2 protein, and ultimately TRAF2-binding AIMP2 induces cell death.
  • ubiquitination of TRAF2 by AIMP2 was significantly reduced in the presence of DX2, and by this decrease, DX2 inhibits AIMP2-stimulated apoptosis in the TNF- ⁇ -active condition ( FIG. 9 ).
  • 67kDa laminin receptor a dimer form of p40/37LRP
  • 67LR has been reported as an important factor in cell migration and cell death, and KARS1 enables cell migration through stabilization of 67LR.
  • KARS1 phosphorylated at threonine 52 by p38MAPK is separated from the multi-tRNA synthetase complex (MSC) and migrated to the plasma membrane.
  • MSC multi-tRNA synthetase complex
  • KARS1 binds to 67LR and inhibits the interaction of 67LR with Nedd4, an E3 ubiquitin ligase, thereby inhibiting ubiquitination-mediated degradation of 67LR. Accordingly, it was confirmed whether 67LR protein stability was related to neuronal cell death in ALS conditions.
  • SK-N-SH cells a human neuroblastoma cell
  • SOD1-G93A SOD1-G93A
  • 67 LR levels were analyzed.
  • 67LR protein in the plasma membrane was significantly reduced under ALS-like conditions.
  • MAPK/ERK Extracellular Signal-regulated Kinase
  • Anoikis is a type of apoptosis induced by reduced contact between the extracellular matrix (ECM) and cell membrane proteins, and the resistance of anoikis plays an important role in cell survival.
  • ECM extracellular matrix
  • Cells were inoculated on poly HEMA-coated plates to observe whether KARS1 and laminin treatment affect cell viability induced by anoikis. As a result, it was confirmed that the cell death of 93A-transfected cells was significantly lower than that of WT cells (FIG. 10E). And, KARS1 expression level did not affect the anoikis-induced cell death (FIG. 10F).
  • the AAV-GFP transformation efficiency test was performed.
  • SK-SY5Y (neuroblastoma) cells were infected with scAAV-GFP or ssAAV-GFP (single-stranded AAV-GFP), and 48 hours later, GFP expression of ssAAV-GFP and scAAV-GFP was observed under a fluorescence microscope.
  • FIG. 12 it was confirmed that the scAAV of the present invention and the conventional ssAAV showed a better GFP expression level. That is, it was indirectly confirmed that the AAV vector of the present invention was an optimized vector in order to increase the expression of AIMP2-DX2 in the central nervous system, restrict expression in blood or other organs, and express it specifically only in the central nervous system.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biochemistry (AREA)
  • Neurology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biophysics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Neurosurgery (AREA)
  • General Chemical & Material Sciences (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Physics & Mathematics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Epidemiology (AREA)
  • Hospice & Palliative Care (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Psychiatry (AREA)
  • Virology (AREA)
  • Toxicology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to an adeno-associated virus vector capable of delivering genes by targeting the central nervous system, and to various uses of same. The vector of the present invention comprises: a sequence encoding a first inverted terminal repeat (ITR); a promoter sequence; a sequence encoding AIMP2-DX2 operably linked to the promoter sequence; a sequence targeting mir142-3p linked to the sequence encoding the AIMP2-DX2; and a sequence encoding a second ITR, wherein the adeno-associated virus vector can be selectively expressed only in the central nervous system among various tissues, and thus can be useful in related industries.

Description

표적화된 유전자 전달을 위한 아데노-부속 바이러스 벡터Adeno-associated viral vectors for targeted gene delivery
본 발명은 본 발명은 중추신경계를 표적으로 하여 유전자를 전달할 수 있는 아데노 부속 바이러스 벡터 및 이의 다양한 용도에 관한 것이다.The present invention relates to an adeno-associated virus vector capable of delivering a gene by targeting the central nervous system and various uses thereof.
포유동물의 뇌는 신경줄기세포(neuronal stem cell)의 분열과 분화 및 생존과 사멸, 시냅스 형성 등 일련의 과정을 거쳐 체계적인 신경회로망(neural network)을 발생시킴으로써 복잡한 기능을 수행할 수 있게 된다. 성체시기에도 동물의 신경세포에서는 신경성장에 필요한 많은 물질을 생산하여 축색돌기(axon)과 수상돌기(dendrite)가 성장하게 되고 새로운 학습과 기억을 할 때마다 시냅스 연결과 신경회로망을 끊임없이 재구성(synaptic remodeling)하므로 분화가 계속된다고 할 수 있다. 신경세포는 세포분화하고 시냅스를 형성하는 과정에서 신경성장인자와 같은 표적유래 생존인자(target-derived survival factor)를 받지 못하면 세포사멸하며 스트레스와 세포독성물질(cytotoxic agent)에 의한 세포사멸은 퇴행성 뇌질환의 주요원인이 된다. 동물의 말초신경계는 손상되었을 때는 중추신경계와 달리 축색이 오랜 시간에 걸쳐 재생한다. 신경 상해 부위의 뒤쪽 축색은 월러변성(Wallerian degeneration)으로 알려진 과정에 의해 퇴화되고 신경의 세포체는 축색의 성장(axonal regrowth)을 다시 시작하며 슈반세포는 분열한 후 생존과 사멸에 의해 표적신경을 결정하고 다시 분화하는 등, 발생과정을 다시 거쳐 재생하게 된다.The mammalian brain can perform complex functions by generating a systematic neural network through a series of processes such as division and differentiation of neural stem cells, survival and death, and synapse formation. Even in the adult period, animal nerve cells produce many substances necessary for nerve growth, and axons and dendrites grow. remodeling), so it can be said that differentiation continues. In the process of cell differentiation and synapse formation, nerve cells die if they do not receive target-derived survival factors such as nerve growth factors. become a major cause of disease. Unlike the central nervous system, when the peripheral nervous system of animals is damaged, the axon regenerates over a long period of time. The posterior axon at the site of nerve injury is degenerated by a process known as Wallerian degeneration, the nerve cell body resumes axonal regrowth, and Schwann cells divide and determine the target nerve by survival and death. It regenerates through the process of development, such as re-differentiation.
전세계적으로 고령화 인구가 급증함에 따라 퇴행성 뇌질환의 발병 추세도 매년 증가하는 추세에 있으며, 아직까지 그 예방법과 치료법이 명확하지 않아, 이러한 질환을 치료하기 위한 효능이 뛰어난 약제를 발견하지 못하고 있는 실정이다. 또한 이러한 질환에 사용되고 있는 치료제와 치료법은 장기 복용에 따른 부작용 및 독성을 나타내는 경우가 많으며, 완벽한 질환 치료보다는 증상의 경과를 늦추거나 증상의 정도를 일시적으로 경감시키는 효과만 있기 때문에 부작용 및 독성을 감소시키고 결정적인 치료효과가 있는 소재의 발굴 및 개발이 시급한 실정이다.As the aging population is rapidly increasing worldwide, the incidence of degenerative brain disease is also increasing every year. am. In addition, the therapeutic agents and treatments used for these diseases often exhibit side effects and toxicity due to long-term use, and they only have the effect of delaying the course of symptoms or temporarily alleviating the severity of symptoms rather than a complete disease treatment, so side effects and toxicity are reduced. There is an urgent need to discover and develop materials with decisive therapeutic effects.
유전자치료법은 인간을 대상으로 1990년 처음 임상시험이 시작된 이래 2002년까지 약 600여건의 임상시험이 3500명의 환자를 대상으로 진행 중이다. 2003년 인간 유전체서열 분석의 완료를 계기로 다양한 유전자 발굴에 따른 새로운 유전자 치료법의 개발은 가속화 될 것이다. 그러나 지금까지 승인된 유전자치료법 중 75%가 암이나 낭포성 섬유증 등의 단일유전 질환의 치료를 목표로 하고 있고 신경질환이나 신경재생을 위한 유전자치료제의 개발은 활발하지 않다 (미국 NIH 재조합 DNA 자문보고서(2002); Gene therapy clinical trials (2002) J Gene Med. www. wiley.co.uk/genmed). 그러나 이미 파킨슨 병의 치료, 감각신경의 재생등에 NT-3, GDNF(glial derived neuronal factor)등의 신경성장인자를 이용한 유전자치료법의 개발이 시도되고 있다(GDNF family ligands activate multiple events during axonal growth in mature sensory neurons (2004) Mol. cell. Neurosci. 25, 4453-4459). 신경 계통의 질환은 뇌기능 연구에 대한 신경과학의 전반적인 연구가 아직 부진한 상태이므로 각종 만성 신경계 질환의 치료 약제개발 또한 난관에 처해 있다. Since the first clinical trial in humans for gene therapy started in 1990, about 600 clinical trials have been conducted on 3,500 patients until 2002. With the completion of human genome sequencing in 2003, the development of new gene therapy according to the discovery of various genes will be accelerated. However, 75% of the gene therapies approved so far target the treatment of monogenetic diseases such as cancer or cystic fibrosis, and the development of gene therapies for neurological diseases or nerve regeneration is not active (NIH Recombinant DNA Advisory Report, USA). (2002); Gene therapy clinical trials (2002) J Gene Med. www.wiley.co.uk/genmed). However, the development of gene therapy using nerve growth factors such as NT-3 and GDNF (glial derived neuronal factor) for the treatment of Parkinson's disease and regeneration of sensory nerves has already been attempted (GDNF family ligands activate multiple events during axonal growth in mature). sensory neurons (2004) Mol. cell. Neurosci. 25, 4453-4459). As for neurological diseases, overall neuroscience research on brain function research is still in a sluggish state, so the development of therapeutic agents for various chronic nervous system diseases is also difficult.
한편 AIMP2-DX2는 세포 사멸에 다방면으로 관련된 종양억제인자 AIMP2의 얼터너티브 스플라이싱 변이체(alternative splicing variant)로서 AIMP2의 기능을 저해함으로써 세포사멸을 억제하는 것으로 알려져 있다. AIMP2-DX2는 TGF-β, TNF-α 및 DNA 손상 신호가 주어지면 AIMP2와 경쟁하여 AIMP2의 친-세포사멸(pro-apoptotic) 상호 작용을 억제한다. TGF-β 및 TNF-α 자극에 의해 MSC(Multi-Synthetase Complex)에서 방출된 AIMP2는 각각 FBP 및 TRAF2에 결합하고, 유비퀴틴 매개인 FBP 및 TRAF2의 분해를 유도하여 세포 사멸을 유도하는 것으로 알려져 있으나, AIMP2-DX2가 존재하면 FBP 및 TRAF2에 AIMP2가 결합하는 것을 경쟁적으로 저해하여 세포 생존을 유도한다. 또한 UV 조사와 같은 DNA 손상 신호가 주어질 때, AIMP2는 p53과 상호 작용하여 p53을 MDM2 매개 분해로부터 보호하여 세포 사멸을 유도하지만, AIMP2-DX2에 의해 p53과 AIMP2의 결합이 저해되어 세포 생존이 유도된다.Meanwhile, AIMP2-DX2 is known to inhibit apoptosis by inhibiting the function of AIMP2 as an alternative splicing variant of AIMP2, a tumor suppressor factor involved in apoptosis in many ways. AIMP2-DX2 inhibits the pro-apoptotic interaction of AIMP2 by competing with AIMP2 in the presence of TGF-β, TNF-α and DNA damage signals. AIMP2 released from MSC (Multi-Synthetase Complex) by TGF-β and TNF-α stimulation binds to FBP and TRAF2, respectively, and is known to induce cell death by inducing ubiquitin-mediated degradation of FBP and TRAF2, The presence of AIMP2-DX2 induces cell survival by competitively inhibiting the binding of AIMP2 to FBP and TRAF2. In addition, when a DNA damage signal such as UV irradiation is given, AIMP2 interacts with p53 to protect p53 from MDM2-mediated degradation to induce apoptosis, but AIMP2-DX2 inhibits the binding of p53 to AIMP2, leading to cell survival. do.
본 발명자들은 TRS(terminal resolution site) 부위가 제거된 ITR, miR-142-3p를 표적하는 서열 및 엑손 2가 결실된 AIMP2 변이체(AIMP2-DX2) 유전자를 코딩하는 서열을 포함하는 아데노-부속 바이러스 벡터를 제작하였고, 상기 벡터가 뇌 또는 척수와 같은 중추신경계에서만 선택적으로 발현할 수 있음을 확인하여 본 발명을 완성하였다.The present inventors have developed an adeno-associated viral vector comprising an ITR from which the TRS (terminal resolution site) region has been removed, a sequence targeting miR-142-3p, and a sequence encoding an AIMP2 mutant (AIMP2-DX2) gene with a deletion of exon 2 was constructed, and the present invention was completed by confirming that the vector can be selectively expressed only in the central nervous system, such as the brain or spinal cord.
따라서, 본 발명의 목적은 TRS(terminal resolution site) 부위가 제거된 ITR, miR-142-3p를 표적하는 서열 및 엑손 2가 결실된 AIMP2 변이체(AIMP2-DX2) 유전자를 코딩하는 서열을 포함하는 아데노-부속 바이러스 벡터 및 이의 퇴행성 신경 질환 예방 및 치료 용도를 제공하는 것이다. Therefore, an object of the present invention is an adeno-containing sequence encoding the ITR, miR-142-3p targeting sequence and exon 2 deleted AIMP2 variant (AIMP2-DX2) gene from which the TRS (terminal resolution site) region has been removed. - To provide an accessory viral vector and its use for preventing and treating neurodegenerative diseases.
상기 목적의 달성을 위해, 본 발명은 제 1 ITR(inverted terminal repeat)을 코딩하는 서열; 프로모터 서열; 상기 프로모터 서열과 작동가능하게 연결된 AIMP2-DX2를 코딩하는 서열; 상기 AIMP2-DX2를 코딩하는 서열과 연결된 mir142-3p를 표적하는 서열; 및 제 2 ITR을 코딩하는 서열을 포함하되, 상기 서열을 5'에서 3' 방향으로 순차적으로 포함하는, 아데노 부속 바이러스 벡터를 제공한다.In order to achieve the above object, the present invention provides a sequence encoding a first inverted terminal repeat (ITR); promoter sequence; a sequence encoding AIMP2-DX2 operably linked to the promoter sequence; a sequence targeting mir142-3p linked to the sequence encoding AIMP2-DX2; And it provides an adeno-associated viral vector comprising a sequence encoding a second ITR, comprising the sequence sequentially in a 5' to 3' direction.
또한, 본 발명은 본 발명의 벡터를 포함하는 퇴행성 신경질환의 예방 또는 치료용 조성물을 제공한다.In addition, the present invention provides a composition for preventing or treating neurodegenerative diseases comprising the vector of the present invention.
또한, 본 발명은 본 발명의 재조합 벡터를 치료가 필요한 개체에 투여하는 단계를 포함하는 퇴행성 신경질환의 예방 또는 치료 방법을 제공한다.In addition, the present invention provides a method for preventing or treating a neurodegenerative disease comprising administering the recombinant vector of the present invention to an individual in need of treatment.
또한, 본 발명은 본 발명의 벡터를 약학적으로 유효한 양으로 퇴행성 신경질환에 걸린 개체에 투여하는 단계를 포함하는 퇴행성 신경질환의 치료 방법을 제공한다.In addition, the present invention provides a method for treating a neurodegenerative disease comprising administering the vector of the present invention to a subject suffering from a neurodegenerative disease in a pharmaceutically effective amount.
아울러, 본 발명은 퇴행성 신경질환 예방 또는 치료용 약학 조성물의 제조에 사용하기 위한, 본 발명의 벡터의 용도를 제공한다.In addition, the present invention provides a use of the vector of the present invention for use in the manufacture of a pharmaceutical composition for the prevention or treatment of neurodegenerative diseases.
본 발명의 아데노 부속 바이러스 벡터는 여러 조직 중 중추신경계에서만 선택적으로 발현될 수 있는 바, 관련 산업에 유용하게 이용될 수 있다. The adeno-associated virus vector of the present invention can be selectively expressed only in the central nervous system among various tissues, and thus can be usefully used in related industries.
도 1은 본 발명의 아데노-부속 바이러스 벡터의 벡터맵을 나타낸 도이다.1 is a diagram showing a vector map of the adeno-associated viral vector of the present invention.
도 2는 본 발명의 아데노-부속 바이러스 벡터의 벡터맵을 나타낸 도이다.2 is a diagram showing a vector map of the adeno-associated viral vector of the present invention.
도 3은 본 발명의 아데노-부속 바이러스 벡터의 제조 공정을 도시한 개략도이다.Figure 3 is a schematic diagram showing the production process of the adeno-associated viral vector of the present invention.
도 4는 DX2가 TRAF2 및 AIMP2의 결합을 경쟁적으로 저해하여 AIMP2의 친-세포사멸(pro-apoptotic) 활성을 감소시킴을 나타낸 도이다. 상부 패널은 DX2가 투여량 의존적으로 AIMP2가 TRAF2에 결합하는 것을 저해함을 나타낸다. 하부 패널은 NF-κB 활성화 및 세포 생존율을 나타낸다. 4 is a diagram showing that DX2 competitively inhibits the binding of TRAF2 and AIMP2, thereby reducing the pro-apoptotic activity of AIMP2. The upper panel shows that DX2 inhibits AIMP2 binding to TRAF2 in a dose-dependent manner. Lower panel shows NF-κB activation and cell viability.
도 5는 DX2가 TNFα-유도 신경세포사를 감소시킴을 나타낸 도이다. (A) SH-SY5Y 세포를 EV (empty vector), DX2, 및 SOD1 G93A로 트랜스펙션시키고 면역블롯팅하여 유비퀴틴화된 TRAF2의 수준을 확인하였다. (B) SH-SY5Y 세포를 EV, DX2, SOD1 WT, G93A, 및 G85R로 트랜스펙션시키고, TNF-α 및 CHX로 6시간 동안 처리하였다. 세포 생존율을 MTT 검정으로 확인하였다. (C) WT 및 돌연변이 SOD1 마우스의 일차 신경 세포를 ith EV 및 DX2로 트랜스펙션시키고, MTT 분석을 실시하여 생존율을 확인하였다.5 is a diagram showing that DX2 reduces TNFα-induced neuronal death. (A) SH-SY5Y cells were transfected with EV (empty vector), DX2, and SOD1 G93A, and the level of ubiquitinated TRAF2 was confirmed by immunoblotting. (B) SH-SY5Y cells were transfected with EV, DX2, SOD1 WT, G93A, and G85R and treated with TNF-α and CHX for 6 h. Cell viability was confirmed by MTT assay. (C) Primary neuronal cells of WT and mutant SOD1 mice were transfected with ith EV and DX2, and viability was confirmed by performing MTT analysis.
도 6은 ALS 마우스 모델에서의 TRAF2 발현 수준을 나타낸 도이다. (A) 각 마우스에서 SOD1 (상부 패널) 및 TRAF2 (하부 패널)의 발현을 나타내는 면역조직화학 염색 결과. (B) 각 마우스에서 TRAF2, EPRS, KARS1, 및 SOD1의 발현을 나타내는 면역블롯 분석 결과. (C-D) 마우스의 척수에서 GFP 및 DX2의 발현: (C) AAV-GFP의 주입 후 GFP의 발현 및 (D) 척수(n = 2 per group)에서 DX2 mRNA 발현의 정량. Br; brain cortex, S.C; spinal cord, scAAV-GFP; 2.4 x 1010vg, scAAV-DX2; 2.4x1010vg. 6 is a diagram showing the TRAF2 expression level in the ALS mouse model. (A) Immunohistochemical staining results showing the expression of SOD1 (upper panel) and TRAF2 (lower panel) in each mouse. (B) Immunoblot analysis results showing the expression of TRAF2, EPRS, KARS1, and SOD1 in each mouse. (CD) Expression of GFP and DX2 in the spinal cord of mice: (C) Expression of GFP after injection of AAV-GFP and (D) Quantification of DX2 mRNA expression in the spinal cord (n = 2 per group). Br; brain cortex, SC; spinal cord, scAAV-GFP; 2.4 x 10 10 vg, scAAV-DX2; 2.4x10 10 vg.
도 7은 DX2의 투여에 의한 루게릭병 모델 마우스의 행동, 질환 발병 시기 및 수명을 확인한 도이다. (A) AAV-DX2를 투여한 군에서 행동이 개선되었다. (B) 질환의 발병 시기는 AAV-DX2 군에서 지연되었다. (C) 마우스의 수명은 AAV-GFP 군에 비해 AAV-DX2 군에서 연장되었다. Animals; WT n=5, SOD1G93A n=6, SOD1G93A AAV2-Con n=9 및 SOD1G93A AAV2-DX2 n=9.7 is a view confirming the behavior, disease onset time and lifespan of ALS model mice by administration of DX2. (A) Behavior was improved in the group administered with AAV-DX2. (B) The onset of disease was delayed in the AAV-DX2 group. (C) Lifespan of mice was prolonged in the AAV-DX2 group compared to the AAV-GFP group. Animals; WT n=5, SOD1 G93A n=6, SOD1 G93A AAV2-Con n=9 and SOD1 G93A AAV2-DX2 n=9.
도 8은 DX2가 루게릭 모델 마우스에서 신경세포 염증 및 세포 사멸을 억제하는지를 단백질 수준에서 확인한 도이다. 루게릭 모델 마우스에 AAV-GFP (Con)를 주입한 마우스에서 TNF-α에 의한 염증신호전달인자 중 세포 사멸을 억제하는 것으로 알려진 TRAF2가 현저하게 감소하는 것을 확인한 반면, AAV-DX2가 주입된 마우스에서 WT과 비슷한 수준으로 TRAF2의 발현이 회복되는것을 확인하였다. 8 is a diagram confirming at the protein level whether DX2 inhibits neuronal inflammation and apoptosis in ALS model mice. While it was confirmed that TRAF2, which is known to inhibit apoptosis among inflammatory signaling factors caused by TNF-α, significantly decreased in mice injected with AAV-GFP (Con) into AAV-GFP (Con) mice injected with AAV-GFP (Con), whereas in mice injected with AAV-DX2 It was confirmed that the expression of TRAF2 was restored to a level similar to that of WT.
도 9는 DX2가 TRAF2의 유비퀴틴화를 차단하여 세포사를 저해하는 기작을 나타낸 개략도이다.9 is a schematic diagram showing the mechanism by which DX2 inhibits cell death by blocking ubiquitination of TRAF2.
도 10은 ALS에서 67 라미닌 수용체 안정성이 감소됨을 나타낸 도이다. (A) SK-N-SH 세포를 SOD1 WT 및 G93A로 트랜스펙션시켰다. 세포를 수확한 후 67 라미닌 수용체 (LR)에 대해 면역블롯팅하였다. (B) 신경 세포를 SOD1 WT 및 G93A로 트랜스펙션한 후 22x22 커버 슬립 상에 배양하였다. 세포를 고정한 후 KARS1 또는 67LR 항체로 처리하고 공초점 현미경으로 이미지를 얻었다. (C) 세포 이동에서 SOD1 WT 및 G93A의 효과를 확인하기 위해, 신경 세포를 8.0 μm 포어 크기의 맴브레인으로 분리된 트랜스웰 플레이트의 상부 챔버에 로딩하고 WT 및 G93A를 하부 챔버에 로딩하였다. 그런 다음 맴브레인을 잘라내고 염색하였다. (D) 신경 세포를 각 플라스미드로 트랜스펙션시키고 라미닌-1 (LN1)을 0, 15, 30 및 60 분 동안 처리하였다. pERK 및 ERK 수준을 웨스턴 블롯으로 측정하였다. (E) SH-SY5Y 세포를 각 플라스미드 및 siRNA KARS1로 트랜스펙션시키고 트랜스펙션된 세포를 poly HEMA-코팅 플레이트에 접종한 후 TNF-α, CHX 및 라미닌 1 (LN1)으로 6시간 동안 처리하였다. 그런 다음 세포를 MTT 분석하여 세포 생존율을 확인하였다. (F) SH-SY5Y 세포를 각 플라스미드로 24시간 동안 트랜스펙션시켰다. 이후 세포를 poly HEMA-코팅 플레이트에 접종하고 TNF-α 및 cycloheximide (CHX)를 6시간 동안 처리하였다. MTT 검정을 실시하여 세포 생존율을 확인하였다.10 is a diagram showing that 67 laminin receptor stability is reduced in ALS. (A) SK-N-SH cells were transfected with SOD1 WT and G93A. Cells were harvested and then immunoblotted for 67 laminin receptor (LR). (B) Neuronal cells were transfected with SOD1 WT and G93A and then cultured on 22x22 coverslips. After fixing the cells, they were treated with KARS1 or 67LR antibody, and images were obtained by confocal microscopy. (C) To confirm the effect of SOD1 WT and G93A on cell migration, neurons were loaded into the upper chamber of a transwell plate separated by a membrane with an 8.0 μm pore size, and WT and G93A were loaded into the lower chamber. The membrane was then cut and stained. (D) Neuronal cells were transfected with each plasmid and treated with laminin-1 (LN1) for 0, 15, 30 and 60 min. pERK and ERK levels were determined by Western blot. (E) SH-SY5Y cells were transfected with each plasmid and siRNA KARS1, and the transfected cells were inoculated on poly HEMA-coated plates and then treated with TNF-α, CHX and laminin 1 (LN1) for 6 hours. . Then, the cells were analyzed by MTT to determine the cell viability. (F) SH-SY5Y cells were transfected with each plasmid for 24 h. Thereafter, the cells were inoculated on poly HEMA-coated plates and treated with TNF-α and cycloheximide (CHX) for 6 hours. Cell viability was confirmed by performing an MTT assay.
도 11은 67LR 발현 및 아노이키스에서의 DX2의 효과를 나타낸 도이다. (A) 신경모세포종 세포를 strep-DX2, SOD1 WT, G93A 및 G85R로 24시간 동안 트랜스펙션시켰다. 세포를 수확하고 초원심세포분획법을 실시한 후 샘플을 면역블롯팅하였다. (B) SK-N-SH 세포를 SOD1 WT, G93A, KARS1, AIMP2 및 DX2로 트랜스펙션시켰다. 세포를 수확한 후 67LR에 대해 웨스턴블롯 분석을 실시하였다. (C) 신경 세포를 SOD1 G93A로 트랜스펙션한 후 EV 또는 DX2를 트랜스펙션하였다. 세포를 라미닌 1 (LN1)으로 0, 15, 30 및 60 분간 처리한 후 세포용해시키고 p-ERK 및 ERK 수준에 대해 면역블롯팅하였다. (D) SH-SY5Y 세포를 SOD1 G93A로 트랜스펙션시킨 후 TNF-α (30 ng/mL)로 24시간 동안 처리하였다. 세포의 부착을 iCelligence로 측정하였다.11 is a diagram showing the effect of DX2 on 67LR expression and Anoikis. (A) Neuroblastoma cells were transfected with strep-DX2, SOD1 WT, G93A and G85R for 24 h. After harvesting the cells and performing ultracentrifugation, the samples were subjected to immunoblotting. (B) SK-N-SH cells were transfected with SOD1 WT, G93A, KARS1, AIMP2 and DX2. After harvesting the cells, Western blot analysis was performed on the 67LR. (C) Neuronal cells were transfected with SOD1 G93A and then either EV or DX2. Cells were treated with laminin 1 (LN1) for 0, 15, 30 and 60 min before lysis and immunoblotting for p-ERK and ERK levels. (D) SH-SY5Y cells were transfected with SOD1 G93A and then treated with TNF-α (30 ng/mL) for 24 hours. Cell adhesion was measured by iCelligence.
도 12는 scAAV와 ssAAV를 비교한 도이다. (A) AAV-GFP의 형질전환 효율 테스트 결과이다. SK-SY5Y (neuroblastoma) 세포를 scAAV-GFP 또는 ssAAV-GFP (single-stranded AAV-GFP)로 감염시키고 48시간 후에, (B) ssAAV-GFP 및 (C) scAAV-GFP의 GFP 발현을 형광 현미경으로 관찰하였다.12 is a diagram comparing scAAV and ssAAV. (A) AAV-GFP transformation efficiency test result. SK-SY5Y (neuroblastoma) cells were infected with scAAV-GFP or ssAAV-GFP (single-stranded AAV-GFP) and 48 hours later, GFP expression of (B) ssAAV-GFP and (C) scAAV-GFP was examined by fluorescence microscopy. observed.
AIMP2-DX2는 세포 사멸에 다방면으로 관련된 세포사멸인자 AIMP2의 얼터너티브 스플라이싱 변이체(alternative splicing variant)로서 AIMP2의 기능을 저해함으로써 종양의 세포사멸을 억제하는 것으로 알려져 있다.AIMP2-DX2 is an alternative splicing variant of AIMP2, an apoptosis factor involved in apoptosis in many ways, and is known to inhibit apoptosis of tumors by inhibiting the function of AIMP2.
본 발명에서 용어, "AIMP2 스플라이싱 변이체"는 엑손 1 내지 4 중 엑손 2의 영역이 부분적으로나 전체적으로 상실되어 생긴 변이체로서, 상기 변이체는 AIMP2 단백질과 헤테로다이머를 형성하여 AIMP2의 정상적인 기능을 방해하는 것을 의미한다. 또한, 상기 AIMP2 변이체가 세포 또는 조직에서 과발현되는 경우, 암이 유발될 수 있다. 따라서, 암의 유발을 억제하기 위하여 조직 특이적으로 발현을 유도할 필요가 있다.As used herein, the term "AIMP2 splicing variant" refers to a variant resulting from partial or total loss of the region of exon 2 among exons 1 to 4, wherein the variant forms a heterodimer with the AIMP2 protein to interfere with the normal function of AIMP2. means that In addition, when the AIMP2 mutant is overexpressed in cells or tissues, cancer may be induced. Therefore, it is necessary to induce tissue-specific expression in order to suppress the induction of cancer.
KR 10-1067816 (2011)에서 AIMP2-DX2의 저해제가 염증 질환을 치료할 수 있음을 기재하고 있으며, 또한, AIMP2/p38이 TRAF2의 유비퀴틴화를 촉진하여 TNF-알파-유도 세포사멸을 조절하고 AIMP2/p38의 스플라이스 변이체인 AIMP2-DX2이 AIMP2의 경쟁적 저해제로서 작용하여 TRAF2의 유비퀴틴을 저해함으로써 TNF-알파-유도 세포사멸을 저해할 수 있고, 이에 따라 종양의 진행을 촉진하고 염증 마커인 Cox-2의 발현을 저해함을 기재하고 있다.In KR 10-1067816 (2011), it is described that an inhibitor of AIMP2-DX2 can treat inflammatory diseases. Also, AIMP2/p38 promotes ubiquitination of TRAF2 to regulate TNF-alpha-induced apoptosis and AIMP2/p38 AIMP2-DX2, a splice variant of p38, acts as a competitive inhibitor of AIMP2 and inhibits ubiquitin of TRAF2, thereby inhibiting TNF-alpha-induced apoptosis, thereby promoting tumor progression and inflammatory marker Cox-2 It has been shown to inhibit the expression of
또한, AIMP2-DX2는 기존에 폐암에서 발현이 증가되어 있는 것이 보고된 바 있으나, AIMP2-DX2 자체는 정상세포를 변형시키는 종양 형성 능력을 갖지 않는다.In addition, although AIMP2-DX2 has been previously reported to have increased expression in lung cancer, AIMP2-DX2 itself does not have the ability to form tumors to transform normal cells.
또한, AIMP2-DX2가 신경 질환을 치료할 수 있다는 보고도 있었다 (US2019/0298858 A1).There was also a report that AIMP2-DX2 can treat neurological diseases (US2019/0298858 A1).
본 발명에서 용어, "재조합 벡터"란 적당한 숙주세포에서 목적 단백질 또는 목적 RNA을 발현할 수 있는 벡터로서, 유전자 삽입물이 발현되도록 작동가능하게 연결된 필수적인 조절 요소를 포함하는 유전자 작제물을 말한다.As used herein, the term "recombinant vector" refers to a vector capable of expressing a target protein or target RNA in an appropriate host cell, and refers to a genetic construct comprising essential regulatory elements operably linked to express a gene insert.
본 발명에서 용어, "작동가능하게 연결된(operably linked)"은 일반적 기능을 수행하도록 핵산 발현조절 서열과 목적하는 단백질 또는 RNA를 코딩하는 핵산 서열이 기능적으로 연결(functional linkage)되어 있는 것을 말한다. 예를 들어 프로모터와 단백질 또는 RNA를 코딩하는 핵산 서열이 작동가능하게 연결되어 코딩하는 핵산서열의 발현에 영향을 미칠 수 있다. 재조합 벡터와의 작동적 연결은 당해 기술분야에서 잘 알려진 유전자 재조합 기술을 이용하여 제조할 수 있으며, 부위-특이적 DNA 절단 및 연결은 당해 기술 분야에서 일반적으로 알려진 효소 등을 사용한다.As used herein, the term “operably linked” refers to a functional linkage between a nucleic acid expression control sequence and a nucleic acid sequence encoding a desired protein or RNA to perform a general function. For example, a promoter and a nucleic acid sequence encoding a protein or RNA may be operably linked to affect expression of the encoding nucleic acid sequence. The operative linkage with the recombinant vector can be prepared using a genetic recombination technique well known in the art, and site-specific DNA cleavage and ligation are performed using enzymes generally known in the art.
본 발명은 제 1 ITR(inverted terminal repeat)을 코딩하는 서열; 프로모터 서열; 상기 프로모터 서열과 작동가능하게 연결된 AIMP2-DX2를 코딩하는 서열; 상기 AIMP2-DX2를 코딩하는 서열과 연결된 mir142-3p를 표적하는 서열; 및 제 2 ITR을 코딩하는 서열을 포함하되, 상기 서열을 5`에서 3` 방향으로 순차적으로 포함하는, 아데노-부속 바이러스 벡터를 제공한다. The present invention provides a sequence encoding a first inverted terminal repeat (ITR); promoter sequence; a sequence encoding AIMP2-DX2 operably linked to the promoter sequence; a sequence targeting mir142-3p linked to the sequence encoding AIMP2-DX2; And it provides an adeno-associated viral vector comprising a sequence encoding a second ITR, wherein the sequence is sequentially included in a 5' to 3' direction.
본 발명의 아데노-부속 바이러스 벡터는 도 1 및 2에 도시된 바와 같은 벡터구조를 가질 수 있으나, 이에 제한되지 않는다. 상기 아데노 부속 바이러스 벡터는 서열번호 7로 표시되는 서열을 포함할 수 있으나, 이에 제한되지 않는다. The adeno-associated viral vector of the present invention may have a vector structure as shown in FIGS. 1 and 2, but is not limited thereto. The adeno-associated viral vector may include the sequence represented by SEQ ID NO: 7, but is not limited thereto.
본 발명의 벡터는 중추신경계에서 특이적으로 DX2를 발현할 수 있으나, 백혈구 및 림프계 세포와 같은 조혈 세포에서는 발현되지 않는다. 따라서, 본 발명의 벡터는 신경 질환을 치료하기 위해 신경세포를 특이적으로 표적화하는데 유용할 수 있다.The vector of the present invention can express DX2 specifically in the central nervous system, but is not expressed in hematopoietic cells such as leukocytes and lymphoid cells. Accordingly, the vectors of the present invention may be useful for specifically targeting neurons to treat neurological diseases.
AIMP2-DX2 폴리펩티드는 AIMP2의 스플라이싱 변이체로, AIMP2의 두 번째 엑손이 결실되었다. 구체적으로, 상기 AIMP2-DX2 유전자는 서열번호 3으로 표시되는 염기서열을 포함하며, AIMP2-DX2 폴리펩티드는 서열번호 4로 표시되는 아미노산 서열을 포함한다.AIMP2-DX2 polypeptide is a splicing variant of AIMP2, in which the second exon of AIMP2 is deleted. Specifically, the AIMP2-DX2 gene includes the nucleotide sequence shown in SEQ ID NO: 3, and the AIMP2-DX2 polypeptide includes the amino acid sequence shown in SEQ ID NO: 4.
본 발명의 일 구현예에서, AIMP2-DX2 유전자는 서열번호 4로 표시되는 서열과 적어도 90%, 적어도 93%, 적어도 95%, 적어도 96%, 적어도 97%, 적어도 98%, 적어도 99% 상동인 아미노산을 코딩하는 뉴클레오티드 서열을 포함할 수 있다. AIMP2-DX2 유전자는 서열번호 4의 아미노산 서열을 코딩하는 뉴클레오티드 서열을 포함할 수 있다.In one embodiment of the present invention, the AIMP2-DX2 gene is at least 90%, at least 93%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% homologous to the sequence represented by SEQ ID NO: 4 It may comprise a nucleotide sequence encoding an amino acid. The AIMP2-DX2 gene may include a nucleotide sequence encoding the amino acid sequence of SEQ ID NO: 4.
AIMP2-DX2 유전자는 서열번호 3의 뉴클레오티드 서열과 적어도 90%, 적어도 93%, 적어도 95%, 적어도 96%, 적어도 97%, 적어도 98%, 또는 적어도 99% 상동인 뉴클레오티드 서열을 포함할 수 있다. AIMP2-DX2 유전자는 서열번호 3으로 표시되는 뉴클레오티드 서열을 포함할 수 있다.The AIMP2-DX2 gene may comprise a nucleotide sequence that is at least 90%, at least 93%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% homologous to the nucleotide sequence of SEQ ID NO:3. The AIMP2-DX2 gene may include a nucleotide sequence represented by SEQ ID NO: 3.
본 발명에서 용어, "마이크로 RNA(miRNA)"는 MicroRNA(약칭 miRNA)는 약 20, 21, 22, 23 또는 24개 nucleotide로 이루어진 noncoding RNA로 유전자 발현을 조절하는 역할을 한다. 상기 microRNA는 유전자의 전사 후 post-transcription 단계에서 작용하며, 포유류의 경우 유전자의 60% 정도가 microRNA에 의해 발현이 조절되는 것으로 알려져 있다. MicroRNA는 다양한 생체 내 프로세스에서 중요한 역할을 하며 암, 심장 질환, 신경 관련 질병에 연관이 있는 것으로 밝혀져 있다. miRNA 는 1 개 사슬 RNA 이지만, premature 의 2 개 사슬 RNA 중 목적 유전자의 발현을 억제할 수 있는 한, 어느 측의 1 개 사슬 RNA 의 표적 배열을 사용할 수 있다. 예컨대, miR-142에는 miR-142-3p 및 miR-142-5p가 존재하며, 본 발명에 있어서는 어느 표적 배열을 사용해도 되며, miR-142에는 miR-142-3p 및 miR-142-5p의 양자가 포함되고, 바람직하게는 miR-142-3p일 수 있다.As used herein, the term "micro RNA (miRNA)" refers to a noncoding RNA consisting of about 20, 21, 22, 23 or 24 nucleotides MicroRNA (abbreviated miRNA) serves to regulate gene expression. The microRNA acts in the post-transcription stage after gene transcription, and in mammals, it is known that about 60% of the gene expression is regulated by microRNA. MicroRNAs play an important role in a variety of in vivo processes and have been implicated in cancer, heart disease, and neurological diseases. Although miRNA is single-stranded RNA, a target sequence of either side of single-stranded RNA can be used as long as expression of the target gene among premature two-stranded RNAs can be suppressed. For example, miR-142 includes miR-142-3p and miR-142-5p. In the present invention, any target sequence may be used, and miR-142 includes both miR-142-3p and miR-142-5p. is included, and may preferably be miR-142-3p.
본 발명에서 용어, "miR-142-3p"는 이의 유전자가 B 세포 백혈병(aggressive B cell leukemia)에 있어서 전좌가 일어나는 부위에 존재하고 있으며, 조혈 조직(골수, 비장, 흉선 등)에서 발현하고 있다고 알려져 있다. 또한, miR-142-3p 는 마우스 태아 간장(태아의 조혈 조직)에 있어서 발현이 확인되어, 조혈계의 분화에 관여하고 있는 것으로 알려져 있다.As used herein, the term "miR-142-3p" refers to its gene being present at the site of translocation in B-cell leukemia, and is expressed in hematopoietic tissues (bone marrow, spleen, thymus, etc.). is known In addition, expression of miR-142-3p has been confirmed in mouse fetal liver (fetal hematopoietic tissue), and is known to be involved in the differentiation of the hematopoietic system.
상기 miR-142-3p를 표적하는 핵산은 ACACTA를 포함하는 뉴클레오티드 서열을 포함할 수 있다. 상기 miR-142 표적 핵산은 ACACTA를 포함하는 뉴클레오티드 서열 및 서열번호 5의 1-17개의 추가 인접 뉴클레오티드를 포함할 수 있다. 예를 들어, miR-142 표적 핵산은 ACACTA를 포함하는 뉴클레오티드 서열 및 서열번호 5에 표시되는 ACACTA의 인접한 5’ 또는 3’인 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 또는 17개의 추가 뉴클레오티드를 포함할 수 있다 .The nucleic acid targeting miR-142-3p may include a nucleotide sequence comprising ACACTA. The miR-142 target nucleic acid may comprise a nucleotide sequence comprising ACACTA and 1-17 additional contiguous nucleotides of SEQ ID NO: 5. For example, the miR-142 target nucleic acid may include a nucleotide sequence comprising ACACTA and 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17 additional nucleotides.
상기 miR-142-3p를 표적하는 핵산은 서열번호5(TCCATAAAGTAGGAAACACTACA; miR-142-3p)의 뉴클레오티드 서열과 적어도 50%, 적어도 60%, 적어도 70%, 적어도 80%, 적어도 90%, 또는 적어도 95% 상동인 뉴클레오티드 서열을 포함할 수 있다. miR-142 표적 핵산은 서열번호 5의 뉴클레오티드 서열을 포함할 수 있다.The nucleic acid targeting miR-142-3p has at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% of the nucleotide sequence of SEQ ID NO: 5 (TCCATAAAGTAGGAAACACTACA; miR-142-3p). % homologous nucleotide sequences. The miR-142 target nucleic acid may comprise the nucleotide sequence of SEQ ID NO:5.
상기 miR-142-3p를 표적하는 서열은 2-10번 반복될 수 있으며, 예를 들어 적어도 2-8번, 적어도 2-6번, 적어도 4번 반복될 수 있다. 상기 miR-142-3p를 표적하는 서열은 서열번호 5의 뉴클레오티드 서열을 포함할 수 있다.The sequence targeting miR-142-3p may be repeated 2-10 times, for example, it may be repeated at least 2-8 times, at least 2-6 times, at least 4 times. The sequence targeting the miR-142-3p may include the nucleotide sequence of SEQ ID NO: 5.
상기 제 1 ITR을 코딩하는 서열 또는 제 2 ITR을 코딩하는 서열은 TRS(terminal resolution site) 부위가 제거된 서열일 수 있으며, 예를 들어, 제 1 ITR을 코딩하는 서열 또는 제 2 ITR을 코딩하는 서열 중 어느 하나 또는 둘 다 TRS 부위가 제거된 것일 수 있으나, 이에 제한되지 않는다. 상기 제 1 ITR을 코딩하는 서열(left ITR)은 서열번호 1로 표시되는 서열이고, 제 2 ITR을 코딩하는 서열(right ITR)은 서열번호 2로 표시되는 서열이다.The sequence encoding the first ITR or the sequence encoding the second ITR may be a sequence in which a terminal resolution site (TRS) site has been removed, for example, the sequence encoding the first ITR or the second ITR. Either or both of the sequences may have the TRS region removed, but is not limited thereto. The sequence encoding the first ITR (left ITR) is the sequence shown in SEQ ID NO: 1, and the sequence coding the second ITR (right ITR) is the sequence shown in SEQ ID NO: 2.
상기 아데노-부속 바이러스(AAV)는 단일가닥의 원바이러스(Provirus)로서, 복제하기 위하여 보조바이러스를 필요로 하고, AAV 게놈(genome)은 4,680 bp로서 감염세포의 염색체 19번 특정부위에 삽입이 가능하다. 트랜스 유전자(trans-gene)는 각각 145bp의 두 개의 역위말단반복(inverted terminal repeat, ITR) 서열부분과 시그날 서열(signal sequence)부분에 의해 연결된 플라즈미드 DNA(plasmid DNA)에 삽입된다. 트랜스 유전자(Transgene)가 발현되는 DNA, AAV rep부분, cap 부분을 발현시키는 플라스미드 DNA 및 helper DNA를 293세포에 트랜스펙션(transfection)시켜 아데노-부속 바이러스를 생성한다. AAV는 유전자를 전달하는 숙주세포의 범위가 넓고, 반복투여 시 면역 부작용이 적으며, 유전자 발현 기간이 긴 장점을 지니고 있다. 더구나, AAV 게놈이 숙주세포의 염색체에 통합되어도 안전하고, 숙주의 유전자발현을 변형시키거나 재배열시키지 않는다.The adeno-associated virus (AAV) is a single-stranded provirus, and requires an auxiliary virus to replicate, and the AAV genome is 4,680 bp, which can be inserted into a specific site on chromosome 19 of an infected cell. Do. The trans-gene is inserted into plasmid DNA linked by two inverted terminal repeat (ITR) sequence portions of 145 bp each and a signal sequence portion. Transgene-expressing DNA, AAV rep part, plasmid DNA expressing cap part, and helper DNA are transfected into 293 cells to generate an adeno-associated virus. AAV has the advantages of a wide range of host cells to deliver genes, fewer immune side effects during repeated administration, and a long gene expression period. Moreover, the AAV genome is safe to integrate into the host cell's chromosomes and does not alter or rearrange the host's gene expression.
상기 아데노-부속 바이러스는 다양한 혈청형이 있는 것으로 알려져 있다. 목적유전자의 전달에 사용될 수 있는 많은 아데노-부속 바이러스의 혈청형들 중에서, 가장 널리 연구된 벡터는 아데노-부속 바이러스 혈청형 2(Adeno-associated virus serotype 2)로, 낭포성섬유증, 혈우병 및 카나반 병의 임상적 유전자 전달에 현재 사용되고 있다. 또한, 최근에는 암 유전자 치료 분야 (cancer gene therapy)에서 rAAV(recombinant adeno-associated virus)의 잠재성이 증가하고 있다. It is known that the adeno-associated virus has various serotypes. Among the many adeno-associated virus serotypes that can be used for target gene delivery, the most widely studied vector is Adeno-associated virus serotype 2, cystic fibrosis, hemophilia, and Canavan It is currently used for clinical gene transfer of disease. In addition, recently, the potential of rAAV (recombinant adeno-associated virus) in cancer gene therapy is increasing.
상기 아데노 부속 바이러스는 scAAV(self complementary adeno-associated virus)일 수 있으나, 이에 제한되지 않는다.The adeno-associated virus may be a self complementary adeno-associated virus (scAAV), but is not limited thereto.
상기 프로모터와 AIMP2-DX2를 코딩하는 서열 사이에 EagI-NotI, Eco53KI, SacI, EcoRI, acc65I, KpnI 및 SexAI로 이루어진 군 중 어느 하나 이상의 제한효소 사이트를 포함할 수 있으나, 이에 제한되지 않는다.EagI-NotI, Eco53KI, SacI, EcoRI, acc65I, KpnI and SexAI between the promoter and the sequence encoding AIMP2-DX2 may include any one or more restriction enzyme sites from the group consisting of, but is not limited thereto.
상기 mir142-3P를 표적하는 서열의 3` 말단 쪽으로 제 1 종결코돈, 폴리 A를 코딩하는 서열 및 제 2 종결코돈이 순차적으로 위치할 수 있으나, 이에 제한되지 않는다. 상기 제 1 종결코돈 또는 제 2 종결코돈은 종결코돈이 3회 이상 반복될 수 있으나, 이에 제한되지 않는다. 상기 제 1 종결코돈과 제 2 종결코돈 사이에 BspDI-ClaI, BamHI 및 StuI로 이루어진 군 중 어느 하나 이상의 제한효소 사이트를 포함할 수 있다.The first stop codon, the poly A coding sequence, and the second stop codon may be sequentially positioned toward the 3' end of the mir142-3P-targeting sequence, but the present invention is not limited thereto. In the first stop codon or the second stop codon, the stop codon may be repeated three or more times, but is not limited thereto. One or more restriction enzyme sites from the group consisting of BspDI-ClaI, BamHI, and StuI may be included between the first stop codon and the second stop codon.
본 발명의 아데노-부속 바이러스 재조합 벡터는 ITR, miR-142-3p, AIMP2-DX2를 포함하는 구성으로 이루어져 있어, cd45-유래 세포, 특히 림프계 및 백혈구에서 AIMP2-DX2 발현 억제를 조절하여 림프절(lymph node)에서 AIMP2-DX2 변이체가 과발현되는 부작용을 제어할 수 있다. 따라서, AIMP2-DX2 변이체는 중추신경계에서만 발현될 수 있거나 다른 유형의 세포 또는 조직이 아닌 뇌 또는 척수 조직에서만 선택적으로 발현될 수 있다. 즉, miR-142-3p가 발현되는 세포에서는 AIMP2-DX2의 발현 또는 활성을 저해할 수 있고, miR-142-3p가 발현되지 않는 세포에서는 AIMP2-DX2의 발현 또는 활성을 유지 또는 증가시킬 수 있다. 또한, 생체 내에서 재조합 벡터를 주입하는 경우, AIMP2-DX2가 중추신경계에만 특이적으로 발현될 수 있다. The adeno-associated viral recombinant vector of the present invention consists of a composition comprising ITR, miR-142-3p, and AIMP2-DX2, and regulates the inhibition of AIMP2-DX2 expression in cd45-derived cells, particularly lymphocytes and leukocytes, thereby reducing lymph nodes (lymph nodes). node) can control the side effect of overexpression of AIMP2-DX2 mutant. Thus, the AIMP2-DX2 variant can be expressed only in the central nervous system or can be selectively expressed only in brain or spinal cord tissues and not other types of cells or tissues. That is, in cells expressing miR-142-3p, the expression or activity of AIMP2-DX2 can be inhibited, and in cells in which miR-142-3p is not expressed, the expression or activity of AIMP2-DX2 can be maintained or increased. . In addition, when the recombinant vector is injected in vivo, AIMP2-DX2 can be specifically expressed only in the central nervous system.
상기 프로모터는 레트로바이러스(LTR) 프로모터, 사이토메갈로바이러스(CMV) 프로모터, 라우스 육종 바이러스(RSV) 프로모터, MMT 프로모터, EF-1 알파 프로모터, UB6 프로모터, 치킨 베타-액틴 프로모터, CAG 프로모터, RPE65 프로모터, 옵신 프로모터 및 이들의 조합으로 이루어진 군으로부터 선택된 프로모터일 수 있으며, 예를 들어 사이토메갈로바이러스(CMV) 프로모터일 수 있다.The promoter is a retrovirus (LTR) promoter, a cytomegalovirus (CMV) promoter, a Rous sarcoma virus (RSV) promoter, an MMT promoter, an EF-1 alpha promoter, a UB6 promoter, a chicken beta-actin promoter, a CAG promoter, a RPE65 promoter, It may be a promoter selected from the group consisting of an opsin promoter and combinations thereof, for example, a cytomegalovirus (CMV) promoter.
뉴클레오티드 또는 아미노산 서열에 대한 "서열 상동성의 %", “%의 상동성” 또는 “% 상동인”의 용어는 두 개의 최적으로 배열된 서열과 비교 영역을 비교함으로써 확인되며, 비교 영역에서의 염기 서열의 일부는 두 서열의 최적 배열에 대한 참고 서열(추가 또는 삭제를 포함하지 않음)에 비해 추가 또는 삭제(즉, 갭)를 포함할 수 있다.The term “% sequence homology”, “% homology” or “% homology” to a nucleotide or amino acid sequence is identified by comparing two optimally aligned sequences with a comparison region, wherein the nucleotide sequence in the comparison region is A portion of may contain additions or deletions (ie, gaps) compared to a reference sequence (not including additions or deletions) for the optimal alignment of the two sequences.
본 발명의 단백질은 이의 천연형 아미노산 서열을 갖는 단백질뿐만 아니라 이의 아미노산 서열 변이체가 또한 본 발명의 범위에 포함된다.The protein of the present invention includes not only a protein having its native amino acid sequence but also an amino acid sequence variant thereof within the scope of the present invention.
본 발명의 단백질의 변이체란 천연 아미노산 서열과 하나 이상의 아미노산 잔기가 결실, 삽입, 비보전적 또는 보전적 치환 또는 이들의 조합에 의하여 상이한 서열을 가지는 단백질을 의미한다. 분자의 활성을 전체적으로 변경시키지 않는 단백질 및 펩티드에서의 아미노산 교환은 당해 분야에 공지되어 있다 (H.Neurath, R.L.Hill, The Proteins, Academic Press, New York, 1979).A variant of the protein of the present invention refers to a protein having a sequence different from the native amino acid sequence by deletion, insertion, non-conservative or conservative substitution of one or more amino acid residues, or a combination thereof. Amino acid exchanges in proteins and peptides that do not entirely alter the activity of the molecule are known in the art (H.Neurath, R.L.Hill, The Proteins, Academic Press, New York, 1979).
상기 단백질 또는 이의 변이체는 천연에서 추출하거나 합성 (Merrifleld, J. Amer. chem. Soc. 85:2149-2156, 1963) 또는 DNA 서열을 기본으로 하는 유전자 재조합 방법에 의해 제조될 수 있다 (Sambrook et al, Molecular Cloning, Cold Spring Harbour Laboratory Press, New York, USA, 2판, 1989).The protein or variant thereof can be extracted from nature, synthesized (Merrifleld, J. Amer. chem. Soc. 85:2149-2156, 1963), or prepared by a genetic recombination method based on a DNA sequence (Sambrook et al. , Molecular Cloning, Cold Spring Harbor Laboratory Press, New York, USA, 2nd ed., 1989).
상기 아미노산 변이는 아미노산 곁사슬 치환체의 상대적 유사성, 예컨대, 소수성, 친수성, 전하, 크기 등에 기초하여 이루어진다. 아미노산 곁사슬 치환체의 크기, 모양 및 종류에 대한 분석에 의하여, 아르기닌, 라이신과 히스티딘은 모두 양전하를 띤 잔기이고; 알라닌, 글라이신과 세린은 유사한 크기를 갖으며; 페닐알라닌, 트립토판과 타이로신은 유사한 모양을 갖는다는 것을 알 수 있다. 따라서, 이러한 고려 사항에 기초하여, 아르기닌, 라이신과 히스티딘; 알라닌, 글라이신과 세린; 그리고 페닐알라닌, 트립토판과 타이로신은 생물학적으로 기능 균등물이라 할 수 있다.The amino acid mutation is made based on the relative similarity of the amino acid side chain substituents, such as hydrophobicity, hydrophilicity, charge, size, and the like. According to the analysis of the size, shape and type of amino acid side chain substituents, arginine, lysine and histidine are all positively charged residues; alanine, glycine and serine have similar sizes; It can be seen that phenylalanine, tryptophan and tyrosine have similar shapes. Therefore, based on these considerations, arginine, lysine and histidine; alanine, glycine and serine; And phenylalanine, tryptophan and tyrosine can be said to be biologically functional equivalents.
변이를 도입하는 데 있어서, 아미노산의 소수성 인덱스 (hydropathic index)가 고려될 수 있다. 각각의 아미노산은 소수성과 전하에 따라 소수성 인덱스가 부여되어 있다: 아이소루이신 (+4.5); 발린 (+4.2); 루이신(+3.8); 페닐알라닌(+2.8); 시스테인/시스타인 (+2.5); 메티오닌 (+1.9); 알라닌 (+1.8); 글라이신 (-0.4); 쓰레오닌 (-0.7); 세린 (-0.8); 트립토판 (-0.9); 타이로신 (-1.3); 프롤린 (-1.6); 히스티딘 (-3.2); 글루타메이트 (-3.5); 글루타민 (-3.5); 아스파르테이트 (-3.5); 아스파라긴 (-3.5); 라이신 (-3.9); 및 아르기닌 (-4.5).In introducing the mutation, the hydropathic index of the amino acid may be considered. Each amino acid is assigned a hydrophobicity index according to its hydrophobicity and charge: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cysteine (+2.5); methionine (+1.9); alanine (+1.8); glycine (-0.4); threonine (-0.7); serine (-0.8); tryptophan (-0.9); tyrosine (-1.3); proline (-1.6); histidine (-3.2); glutamate (-3.5); glutamine (-3.5); aspartate (-3.5); asparagine (-3.5); lysine (-3.9); and arginine (-4.5).
단백질의 상호적인 생물학적 기능(interactive biological function)을 부여하는 데 있어서 소수성 아미노산 인덱스는 매우 중요하다. 유사한 소수성 인덱스를 가지는 아미노산으로 치환하여야 유사한 생물학적 활성을 보유할 수 있다는 것은 공지된 사실이다. 소수성 인덱스를 참조하여 변이를 도입시키는 경우, 바람직하게는 ± 2이내, 보다 바람직하게는 ± 1 이내, 보다 더 바람직하게는 ± 0.5 이내의 소수성 인덱스 차이를 나타내는 아미노산 사이에 치환을 한다.The hydrophobic amino acid index is very important in conferring an interactive biological function of a protein. It is a known fact that amino acids having a similar hydrophobicity index must be substituted to retain similar biological activity. When introducing a mutation with reference to the hydrophobicity index, the substitution is made between amino acids showing a difference in the hydrophobicity index, preferably within ±2, more preferably within ±1, and even more preferably within ±0.5.
한편, 유사한 친수성 값(hydrophilicity value)을 가지는 아미노산 사이의 치환이 균등한 생물학적 활성을 갖는 단백질을 초래한다는 것도 잘 알려져 있다. 미국 특허 제4,554,101호에 개시된 바와 같이, 다음의 친수성 값이 각각의 아미노산 잔기에 부여되어 있다: 아르기닌 (+3.0); 라이신 (+3.0); 아스팔테이트(+3.0± 1); 글루타메이트 (+3.0± 1); 세린 (+0.3); 아스파라긴 (+0.2); 글루타민 (+0.2); 글라이신 (0); 쓰레오닌 (-0.4); 프롤린 (-0.5 ± 1); 알라닌 (-0.5); 히스티딘 (-0.5); 시스테인 (-1.0); 메티오닌 (-1.3); 발린 (-1.5); 루이신(-1.8); 아이소루이신 (-1.8); 타이로신 (-2.3); 페닐알라닌 (-2.5); 트립토판 (-3.4).On the other hand, it is also well known that substitution between amino acids having similar hydrophilicity values results in proteins having equivalent biological activity. As disclosed in US Pat. No. 4,554,101, the following hydrophilicity values are assigned to each amino acid residue: arginine (+3.0); lysine (+3.0); Aspartate (+3.0± 1); glutamate (+3.0± 1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine (-0.4); proline (-0.5 ± 1); alanine (-0.5); histidine (-0.5); cysteine (-1.0); methionine (-1.3); valine (-1.5); leucine (-1.8); isoleucine (-1.8); tyrosine (-2.3); phenylalanine (-2.5); Tryptophan (-3.4).
친수성 값을 참조하여 변이를 도입시키는 경우, 바람직하게는 ± 2 이내, 보다 바람직하게는 ± 1 이내, 보다 더 바람직하게는 ± 0.5 이내의 친수성 값 차이를 나타내는 아미노산 사이에 치환을 한다.When the mutation is introduced with reference to the hydrophilicity value, the substitution is made between amino acids exhibiting a difference in the hydrophilicity value within preferably ±2, more preferably within ±1, and even more preferably within ±0.5.
분자의 활성을 전체적으로 변경시키지 않는 단백질에서의 아미노산 교환은 당해 분야에 공지되어 있다(H. Neurath, R.L.Hill, The Proteins, Academic Press, New York, 1979). 가장 통상적으로 일어나는 교환은 아미노산 잔기 Ala/Ser, Val/Ile, Asp/Glu, Thr/Ser, Ala/Gly, Ala/Thr, Ser/Asn, Ala/Val, Ser/Gly, Thy/Phe, Ala/Pro, Lys/Arg, Asp/Asn, Leu/Ile, Leu/Val, Ala/Glu, Asp/Gly 간의 교환이다.Amino acid exchanges in proteins that do not entirely alter the activity of the molecule are known in the art (H. Neurath, R.L. Hill, The Proteins, Academic Press, New York, 1979). The most common exchanges are amino acid residues Ala/Ser, Val/Ile, Asp/Glu, Thr/Ser, Ala/Gly, Ala/Thr, Ser/Asn, Ala/Val, Ser/Gly, Thy/Phe, Ala/ It is an exchange between Pro, Lys/Arg, Asp/Asn, Leu/Ile, Leu/Val, Ala/Glu, Asp/Gly.
본 발명의 벡터는 단백질의 정제를 용이하게 하기 위하여, 필요에 따라 다른 서열과 융합될 수도 있으며, 융합되는 서열은 예컨대, 글루타티온 S-트랜스퍼라제(Pharmacia, USA), 말토스 결합 단백질(NEB, USA), FLAG (IBI, USA) 및 6x His(hexahistidine; Quiagen, USA) 등이 이용될 수 있으나, 이에 제한되지는 않는다. 또한, 본 발명의 발현 벡터는 선택표지로서, 당업계에서 통상적으로 이용되는 항생제 내성 유전자를 포함할 수 있으며, 예를 들어 암피실린, 겐타마이신, 카베니실린, 클로람페니콜, 스트렙토마이신, 카나마이신, 게네티신, 네오마이신 및 테트라사이클린에 대한 내성 유전자가 있다.The vector of the present invention may be fused with other sequences as necessary to facilitate protein purification, and the fused sequences include, for example, glutathione S-transferase (Pharmacia, USA), maltose binding protein (NEB, USA). ), FLAG (IBI, USA) and 6x His (hexahistidine; Quiagen, USA) may be used, but is not limited thereto. In addition, the expression vector of the present invention may contain an antibiotic resistance gene commonly used in the art as a selection marker, for example, ampicillin, gentamicin, carbenicillin, chloramphenicol, streptomycin, kanamycin, geneticin , resistance genes to neomycin and tetracycline.
또한 본 발명은 상기 아데노-부속 바이러스 벡터를 치료가 필요한 개체에 투여하는 단계;를 포함하는 AIMP2-DX2와 같은 이종 유전자를 신경세포에 전달 및 발현시키는 방법을 제공한다.The present invention also provides a method for delivering and expressing a heterologous gene such as AIMP2-DX2 to neurons, comprising administering the adeno-associated viral vector to an individual in need of treatment.
상기 방법은, 중추신경계 (척수강 및 뇌조직)에서는 이종 유전자의 발현이 증가하고, 다른 조직에서는 이종 유전자 발현이 제어시킬 수 있다. In this method, the expression of the heterologous gene is increased in the central nervous system (the spinal canal and brain tissue), and the expression of the heterologous gene can be controlled in other tissues.
또한 본 발명은 상기 아데노-부속 바이러스 벡터를 유효성분으로 포함하는 퇴행성 신경질환 예방 또는 치료용 조성물을 제공한다. The present invention also provides a composition for preventing or treating neurodegenerative diseases comprising the adeno-associated viral vector as an active ingredient.
이에 따라, 본 발명은 본 발명의 재조합 벡터를 치료가 필요한 개체에 투여하는 단계를 포함하는 퇴행성 신경 질환의 예방 또는 치료 방법을 제공한다. 상기 신경 질환은 알츠하이머 질환(alzheimer's disease), 파킨슨 병(Parkinson’s disease), 루게릭 병(amyotrophic lateral sclerosis), 망막 변성증(retinal degeneration), 경도인지 장애(mild cognitive impairment), 다발-경색성 치매((Multi-infarct dementia), 전두측두치매(frontotemporal dementia), 루이소체 치매(dementia with Lewy bodies), 헌팅턴 질환(Huntington's disease), 신경퇴행질환, 대사성 뇌질환, 우울증, 간질, 다발성 경화증(Multiple sclerosis), 피질기저퇴행증(corticobasal degeneration), 다계통위축병(multiple system atrophy), 진행성핵상마비(progressive supranuclear palsy), 치상핵적핵담창구시상하핵위축증(dentatorubropallidoluysian atrophy), 척수소뇌실조병(spinocerebella ataxia), 원발성 측삭 경화증(primary lateral sclerosis), 척수근육위축병(spinal muscular atrophy) 및 뇌졸중(stroke) 구성된 군으로부터 선택된 1종 이상일 수 있으나, 이에 제한되지 않는다. 일 구현예에서, 상기 신경 질환은 루게릭 병(amyotrophic lateral sclerosis; ALS)일 수 있다. 상기 치료는 ALS, 알츠하이머 질환 또는 파킨슨 병과 같은 신경 질환에 걸린 개체의 기억, 운동장애, 운동 능력을 개선시키거나 수명을 연장시키는 것일 수 있다. 일 구현예에서, 상기 치료는 ALS와 같은 신경 질환에 걸린 개체의 운동 능력을 개선시키거나 수명을 연장시키는 것일 수 있다.Accordingly, the present invention provides a method for preventing or treating a neurodegenerative disease comprising administering the recombinant vector of the present invention to an individual in need of treatment. The neurological disease is Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, retinal degeneration, mild cognitive impairment, multiple-infarct dementia (Multi -infarct dementia, frontotemporal dementia, dementia with Lewy bodies, Huntington's disease, neurodegenerative disease, metabolic brain disease, depression, epilepsy, multiple sclerosis, cortex Corticobasal degeneration, multiple system atrophy, progressive supranuclear palsy, dentatorubropallidoluysian atrophy, spinocerebella ataxia It may be one or more selected from the group consisting of lateral sclerosis (primary lateral sclerosis), spinal muscular atrophy and stroke (stroke), but is not limited thereto. In one embodiment, the neurological disease is amyotrophic lateral sclerosis; ALS).The treatment may be to improve memory, motor impairment, motor ability or prolong the lifespan of an individual suffering from neurological disease such as ALS, Alzheimer's disease or Parkinson's disease. The treatment may be to improve motor performance or prolong the lifespan of an individual suffering from a neurological disease such as ALS.
본 발명에서 용어, "치료"는 본 발명의 약학 조성물을 퇴행성 신경질환을 갖는 개체에 적용한 결과로서 퇴행성 신경질환의 완치는 물론 퇴행성 신경질환에 따른 제증상의 부분적 완치, 호전 및 경감을 포함한다.As used herein, the term "treatment" includes partial cure, improvement and alleviation of symptoms according to the neurodegenerative disease as well as the complete cure of the neurodegenerative disease as a result of applying the pharmaceutical composition of the present invention to an individual having a neurodegenerative disease.
본 발명에서 용어, "예방"은 본 발명의 약학 조성물을 퇴행성 신경질환을 갖는 개체에 적용하여 인지장애, 행동장애, 신경 파괴 등의 증세 내지 현상을 억제 또는 차단함으로써, 퇴행성 신경질환에 따른 제증상이 사전에 발생되지 않도록 하는 것을 의미한다.In the present invention, the term "prevention" refers to suppressing or blocking symptoms or phenomena such as cognitive impairment, behavioral disorder, and nerve destruction by applying the pharmaceutical composition of the present invention to an individual having a neurodegenerative disease, thereby causing various symptoms according to the neurodegenerative disease. This means that it does not happen in advance.
본 발명의 약학 조성물에는 유효성분 이외에 보조제(adjuvant)를 추가로 포함할 수 있다. 상기 보조제는 당해 기술분야에 알려진 것이라면 어느 것이나 제한 없이 사용할 수 있으나, 예를 들어 프로인트(Freund)의 완전 보조제 또는 불완전 보조제를 더 포함하여 그 면역성을 증가시킬 수 있다. The pharmaceutical composition of the present invention may further include an adjuvant in addition to the active ingredient. The adjuvant may be used without limitation as long as it is known in the art, for example, Freund's complete adjuvant or incomplete adjuvant may be further included to increase the immunity thereof.
본 발명에 따른 약학 조성물은 유효성분을 약학적으로 허용된 담체에 혼입시킨 형태로 제조될 수 있다. 여기서, 약학적으로 허용된 담체는 제약 분야에서 통상 사용되는 담체, 부형제 및 희석제를 포함한다. 본 발명의 약학 조성물에 이용할 수 있는 약학적으로 허용된 담체는 이들로 제한되는 것은 아니지만, 락토스, 덱스트로스, 수크로스, 솔비톨, 만니톨, 자일리톨, 에리스리톨, 말티톨, 전분, 아카시아 고무, 알지네이트, 젤라틴, 칼슘 포스페이트, 칼슘 실리케이트, 셀룰로스, 메틸 셀룰로스, 폴리비닐 피롤리돈, 물, 메틸히드록시벤조에이트, 프로필히드록시벤조에이트, 탈크, 마그네슘 스테아레이트 및 광물유를 들 수 있다.The pharmaceutical composition according to the present invention may be prepared in a form in which the active ingredient is incorporated into a pharmaceutically acceptable carrier. Here, the pharmaceutically acceptable carrier includes carriers, excipients and diluents commonly used in the pharmaceutical field. Pharmaceutically acceptable carriers that can be used in the pharmaceutical composition of the present invention include, but are not limited to, lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia gum, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, polyvinyl pyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil.
본 발명의 약학 조성물은 각각 통상의 방법에 따라 산제, 과립제, 정제, 캡슐제, 현탁액, 에멀전, 시럽, 에어로졸 등의 경구형 제형, 외용제, 좌제 또는 멸균 주사용액의 형태로 제형화하여 사용될 수 있다.The pharmaceutical composition of the present invention may be formulated in the form of oral dosage forms such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols, etc., external preparations, suppositories, or sterile injection solutions according to conventional methods, respectively. .
제제화할 경우에는 통상 사용하는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 사용하여 조제될 수 있다. 경구투여를 위한 고형제제에는 정제, 환제, 산제, 과립제, 캡슐제 등이 포함되며, 그러한 고형 제제는 유효성분에 적어도 하나 이상의 부형제, 예를 들면 전분, 칼슘 카르보네이트, 수크로스, 락토오스, 젤라틴 등을 섞어 조제될 수 있다. 또한, 단순한 부형제 이외에 마그네슘 스테아레이트, 탈크 같은 윤활제들도 사용될 수 있다. 경구투여를 위한 액상 제제로는 현탁제, 내용액제, 유제, 시럽제 등이 해당되는데, 일반적으로 사용되는 희석제인 물, 리퀴드 파라핀 이외에 여러 가지 부형제, 예를 들면 습윤제, 감미제, 방향제, 보존제 등이 포함될 수 있다. 비경구 투여를 위한 제제에는 멸균된 수용액, 비수용성용제, 현탁제, 유제, 동결건조 제제 및 좌제가 포함된다. 비수용성용제, 현탁제로는 프로필렌 글리콜, 폴리에틸렌 글리콜, 올리브유와 같은 식물성 기름, 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용될 수 있다. 좌제의 기제로는 위텝솔(witepsol), 트윈(tween) 61, 카카오지, 라우린지, 글리세로젤라틴 등이 사용될 수 있다.In the case of formulation, it can be prepared using a diluent or excipient such as a filler, extender, binder, wetting agent, disintegrant, surfactant, etc. commonly used. Solid preparations for oral administration include tablets, pills, powders, granules, capsules, etc., and such solid preparations include at least one excipient in the active ingredient, for example, starch, calcium carbonate, sucrose, lactose, gelatin. It can be prepared by mixing and the like. In addition to simple excipients, lubricants such as magnesium stearate and talc may also be used. Liquid formulations for oral administration include suspensions, solutions, emulsions, syrups, etc. In addition to water and liquid paraffin, which are commonly used diluents, various excipients such as wetting agents, sweeteners, fragrances, and preservatives may be included. can Formulations for parenteral administration include sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, lyophilized formulations and suppositories. As the non-aqueous solvent and suspending agent, propylene glycol, polyethylene glycol, vegetable oil such as olive oil, and injectable ester such as ethyl oleate may be used. As the base of the suppository, witepsol, tween 61, cacao butter, laurin, glycerogelatin, and the like can be used.
본 발명에 따른 약학 조성물은 개체에 다양한 경로로 투여될 수 있다. 투여의 모든 방식이 예상될 수 있는데, 예를 들면 경구, 정맥, 근육, 피하, 복강내 주사, 뇌 선조체 주사, 척수강 내 주사 등에 의해 투여될 수 있다.The pharmaceutical composition according to the present invention may be administered to an individual by various routes. Any mode of administration can be envisaged, for example, it can be administered by oral, intravenous, intramuscular, subcutaneous, intraperitoneal injection, brain striatal injection, intrathecal injection, and the like.
본 발명에 따른 치료용 조성물의 바람직한 투여량은 제제화 방법, 투여 방식, 환자의 연령, 체중, 성, 질병 증상의 정도, 음식, 투여 시간, 투여 경로, 배설 속도 및 반응 감응성과 같은 요인들에 따라 다르지만, 당업자에 의해 적절하게 선택될 수 있다.Preferred dosage of the therapeutic composition according to the present invention depends on factors such as formulation method, administration method, age, weight, sex, degree of disease symptoms, food, administration time, administration route, excretion rate, and response sensitivity of the patient. Although different, it may be appropriately selected by those skilled in the art.
그러나 치료효과를 위해서, 보통으로 숙련된 의사는 목적하는 치료에 효과적인 투여량을 용이하게 결정 및 처방할 수 있다. 예를 들어 상기 치료제는 혈관주사, 피하지방, 근육주사 및 미세주사기를 이용한 뇌실 또는 척수에 직접주사를 포함한다. 이때, 단회 투여, 다회 투여(multiple injection) 및 반복 투여가 가능하며, 예를 들어 유효 용량은 체중 1 kg당 벡터의 경우에는 0.05 내지 15 mg/kg, 재조합 바이러스의 경우에는 5 Х 1011 내지 3.3 Х 1014 바이러스 입자(2.5 Х 1012 내지 1.5 Х 1016 IU)/kg, 세포의 경우에는 5 Х 102 내지 5 Х 107세포/kg이고, 바람직하게는 벡터의 경우에는 0.1 내지 10 mg/kg, 재조합 바이러스의 경우에는 5 Х 1012 내지 3.3 Х 1013 입자(2.5 Х 1013 내지 1.5 Х1015 IU)/kg, 세포의 경우에는 5 Х103 내지 5 Х 106 세포/kg이며, 일주일에 2 내지 3회 투여될 수 있다. 상기와 같은 조성은 반드시 이에 한정되는 것은 아니고, 환자의 상태 및 신경 질환의 발병 정도에 따라 변할 수 있다. 기타 피하지방, 근육주사, 환부 직접 투여에 대한 유효 용량은 10cm 의 간격으로 9 Х 1010 내지 3.3 Х 1014의 재조합 바이러스 입자가 투여되며, 일주일에 2 ~ 3회 투여될 수 있다. 상기와 같은 조성은 반드시 이에 한정되는 것은 아니고, 환자의 상태 및 신경 질환의 발병 정도에 따라 변할 수 있다. 보다 구체적으로, 본 발명의 약학 조성물은 1 Х 1010 내지 1 Х 1012 vg(virus genome)/mL의 재조합 아데노조력 바이러스를 포함하며, 통상적으로 1 Х 1012 vg를 이틀에 한번씩 2주 동안 주사하는 것이 좋다. 투여는 하루에 한번 투여할 수도 있고, 수회 나누어 투여할 수도 있다. However, for therapeutic effect, an ordinary skilled physician can easily determine and prescribe an effective dosage for the desired treatment. For example, the therapeutic agent includes intravenous injection, subcutaneous fat injection, intramuscular injection, and direct injection into the ventricle or spinal cord using a micro-injector. In this case, single administration, multiple injection and repeated administration are possible. For example, the effective dose is 0.05 to 15 mg/kg of vector per 1 kg of body weight, and 5 Х 10 11 to 3.3 in the case of recombinant virus. Х 10 14 virus particles (2.5 Х 10 12 to 1.5 Х 10 16 IU)/kg for cells 5 Х 10 2 to 5 Х 10 7 cells/kg, preferably 0.1 to 10 mg/kg for vectors kg, 5 Х 10 12 to 3.3 Х 10 13 particles (2.5 Х 10 13 to 1.5 Х10 15 IU)/kg for recombinant virus, 5 Х10 3 to 5 Х 10 6 cells/kg for cells, per week It may be administered 2-3 times. The composition as described above is not necessarily limited thereto, and may vary depending on the patient's condition and the degree of onset of a neurological disease. Other effective doses for subcutaneous fat, intramuscular injection, and direct administration to the affected area are 9 Х 10 10 to 3.3 Х 10 14 recombinant virus particles at an interval of 10 cm, and can be administered 2-3 times a week. The composition as described above is not necessarily limited thereto, and may vary depending on the patient's condition and the degree of onset of a neurological disease. More specifically, the pharmaceutical composition of the present invention contains 1 Х 10 10 to 1 Х 10 12 vg (virus genome)/mL of recombinant adeno-associated virus, and typically 1 Х 10 12 vg is injected every other day for 2 weeks. good to do Administration may be administered once a day, or may be administered in several divided doses.
상기 약학 조성물은 다양한 경구 또는 비경구 투여 형태로 제형화될 수 있다.The pharmaceutical composition may be formulated in various oral or parenteral dosage forms.
경구 투여용 제형으로는 예를 들면 정제, 환제, 경질, 연질 캅셀제, 액제, 현탁제, 유화제, 시럽제, 과립제 등이 있는데, 이들 제형은 유효성분 이외에 희석제(예: 락토즈, 덱스트로즈, 수크로즈, 만니톨, 솔비톨, 셀룰로즈 및/또는 글리신), 활택제(예: 실리카, 탈크, 스테아르산 및 그의 마그네슘 또는 칼슘염 및/ 또는 폴리에틸렌 글리콜)를 추가로 포함할 수 있다. 또한, 상기 정제는 마그네슘 알루미늄 실리케이트, 전분 페이스트, 젤라틴, 트라가칸스, 메틸셀룰로즈, 나트륨 카복시메틸셀룰로즈 및/또는 폴리비닐피롤리딘과 같은 결합제를 함유할 수 있으며, 경우에 따라 전분, 한천, 알긴산 또는 그의 나트륨 염과 같은 붕해제 또는 비등 혼합물 및/또는 흡수제, 착색제, 향미제 및 감미제를 함유할 수 있다. 상기 제형은 통상적인 혼합, 과립화 또는 코팅 방법에 의해 제조될 수 있다.Formulations for oral administration include, for example, tablets, pills, hard, soft capsules, solutions, suspensions, emulsifiers, syrups, granules, and the like. crose, mannitol, sorbitol, cellulose and/or glycine), lubricants (eg silica, talc, stearic acid and its magnesium or calcium salts and/or polyethylene glycol). In addition, the tablet may contain a binder such as magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidine, and optionally starch, agar, alginic acid. or a disintegrant such as its sodium salt or a boiling mixture and/or absorbent, coloring, flavoring and sweetening agent. The formulation may be prepared by conventional mixing, granulating or coating methods.
또한, 비경구 투여용 제형의 대표적인 것은 주사용 제제이며, 주사용 제제의 용매로서 물, 링거액, 등장성 생리식염수 또는 현탁액을 들 수 있다. 상기 주사용 제제의 멸균 고정 오일은 용매 또는 현탁 매질로서 사용할 수있으며 모노-, 디-글리세라이드를 포함하여 어떠한 무자극성 고정오일도 이러한 목적으로 사용될 수 있다. 또한, 상기 주사용 제제는 올레산과 같은 지방산을 사용할 수 있다.In addition, a representative formulation for parenteral administration is an injection formulation, and examples of the solvent for the injection formulation include water, Ringer's solution, isotonic saline, or suspension. The sterile, fixed oil of the injectable preparation may be used as a solvent or suspending medium, and any non-irritating fixed oil including mono- and di-glycerides may be used for this purpose. In addition, the injection preparation may use a fatty acid such as oleic acid.
또한, 본 발명은 본 발명의 벡터를 약학적으로 유효한 양으로 퇴행성 신경질환에 걸린 개체에 투여하는 단계를 포함하는 퇴행성 신경질환의 치료 방법을 제공한다.In addition, the present invention provides a method for treating a neurodegenerative disease comprising administering the vector of the present invention to a subject suffering from a neurodegenerative disease in a pharmaceutically effective amount.
아울러, 본 발명은 퇴행성 신경질환 예방 또는 치료용 약학 조성물의 제조에 사용하기 위한, 본 발명의 벡터의 용도를 제공한다.In addition, the present invention provides a use of the vector of the present invention for use in the manufacture of a pharmaceutical composition for the prevention or treatment of neurodegenerative diseases.
하기의 실시예를 통하여 본 발명을 보다 상세하게 설명한다. 그러나 하기 실시예는 본 발명의 내용을 구체화하기 위한 것일 뿐 이에 의해 본 발명이 한정되는 것은 아니다.The present invention will be described in more detail through the following examples. However, the following examples are only intended to embody the contents of the present invention, and the present invention is not limited thereto.
<실시예 1> 본 발명의 재조합 벡터 제작<Example 1> Construction of the recombinant vector of the present invention
1-1. AIMP2 변이체 제작1-1. Creation of AIMP2 variants
AIMP2는 아미노아실-tRNA 합성효소 복합체(Aminoacyl-tRNA synthetase: ARSs)의 형성에 관련된 단백질 중의 하나로, 세포사멸억제제 (apoptosis inhibitor)로서 작용한다. 상기 AIMP2의 엑손 2가 결실된 변이체를 발현하는 플라스미드를 구축하기 위하여, AIMP2 변이체의 cDNA를 pcDNA3.1-myc으로 클로닝하였다. 이는 AIMP2 변이체를 H322 cDNA에서 EcoR1 및 Xho I linker가 달린 primer를 이용하여 증폭시킨 후 EcoR1 과 Xho1을 이용하여 pcDNA3.1-myc에 클로닝하였다.AIMP2 is one of the proteins involved in the formation of aminoacyl-tRNA synthetase (ARSs), and acts as an apoptosis inhibitor. To construct a plasmid expressing a mutant in which exon 2 of AIMP2 is deleted, cDNA of the AIMP2 mutant was cloned into pcDNA3.1-myc. This AIMP2 mutant was amplified from H322 cDNA using EcoR1 and Xho I linker primers, and then cloned into pcDNA3.1-myc using EcoR1 and Xho1.
본 발명의 AIMP2 변이체는 서열번호 3의 염기서열로 표시되고, 서열번호 4의 아미노산으로 표시된다.AIMP2 mutant of the present invention is represented by the nucleotide sequence of SEQ ID NO: 3, and is represented by the amino acid of SEQ ID NO: 4.
1-2. miRNA 선별 및 이의 표적 서열 선발1-2. miRNA selection and its target sequence selection
본 발명의 AIMP2 변이체는 종양에서 과발현되는 것이 확인되었는 바, 이를 표적 세포인 신경 세포에서 안전하게 발현되면서도 백혈구 관련 세포 및 림프구 관련 세포에서는 상기 AIMP2 변이체의 발현을 억제하기 위하여, 상기 AIMP2 변이체 발현을 조절할 수 있는 miRNA 및 이의 표적을 선발하였다. It was confirmed that the AIMP2 variant of the present invention is overexpressed in tumors, and while it is safely expressed in nerve cells, which are target cells, to suppress the expression of the AIMP2 variant in leukocyte-related cells and lymphocyte-related cells, the expression of the AIMP2 variant can be controlled. miRNAs and their targets were selected.
이를 위하여, 백혈구 관련 세포 및 림프구 관련 세포를 만들어 내는 조혈모세포(hematopoietic cell)에서만 특이적으로 발현하는 miR-142-3p를 타겟으로 선발하였다. 상기 miR-142-3p를 타겟하는 서열을 제작하기 위하여, 마우스 B 세포의 마이크로어레이 데이터와 miR-142-3p에 의해 타겟되는 유전자의 Computer programing (mirSVR score)를 이용하였다. 본 발명의 miR-142-3p를 표적하는 서열은 서열번호 5로 표시되는 염기서열로 나타내었다.To this end, miR-142-3p, which is specifically expressed only in hematopoietic cells that produce leukocyte-related cells and lymphocyte-related cells, was selected as a target. In order to prepare the sequence targeting the miR-142-3p, microarray data of mouse B cells and computer programming (mirSVR score) of the gene targeted by miR-142-3p were used. The sequence targeting miR-142-3p of the present invention is represented by the nucleotide sequence shown in SEQ ID NO: 5.
상기 본 발명의 miR-142-3p를 표적하는 서열은, 클로닝을 위한 제한효소(NheⅠ, Hind Ⅲ, BmtⅠ) 사이트 서열(ccagaagcttgctagc), 제한효소(Hind Ⅲ) 사이트 서열(aagcttgtag)을 포함한다. 이는 이를 연결시켜주는 링커(tcac and gatatc)와 함께 4번 반복되는 서열번호 5로 표시되는 뉴클레오티드 서열을 포함한다.The sequence targeting miR-142-3p of the present invention includes a restriction enzyme (NheI, Hind III, BmtI) site sequence (ccagaagcttgctagc) and a restriction enzyme (Hind III) site sequence (aagcttgtag) for cloning. It includes a nucleotide sequence represented by SEQ ID NO: 5 that is repeated 4 times together with linkers (tcac and gatatc) connecting them.
1-3. 본 발명의 재조합 벡터 제작1-3. Construction of the recombinant vector of the present invention
본 발명의 재조합 벡터를 제작하기 위하여, 제 1 ITR을 코딩하는 서열(서열번호 1), CMV 프로모터, 상기 본 발명의 AIMP2 변이체(서열번호 3), miR-142-3p를 표적하는 서열(서열번호 5) 및 제 2 ITR(서열번호 2)를 5`에서 3` 방향으로 순차적으로 포함하도록 삽입하였다. AIMP-2 변이체와 miR-142-3p 표적 서열이 결합된 것은 서열번호 6의 염기서열로 나타내었으며, 구체적으로, NheI 및 Hind Ⅲ 사이트를 이용하여 잘라넣었다. 본 발명의 아데노-부속 바이러스 벡터는 도 1 및 2에 나타내었다. In order to construct the recombinant vector of the present invention, the sequence encoding the first ITR (SEQ ID NO: 1), the CMV promoter, the AIMP2 mutant of the present invention (SEQ ID NO: 3), miR-142-3p targeting sequence (SEQ ID NO: 5) and the second ITR (SEQ ID NO: 2) were inserted sequentially in the 5' to 3' direction. The binding of the AIMP-2 mutant and the miR-142-3p target sequence was shown as the nucleotide sequence of SEQ ID NO: 6, and specifically, it was cut using NheI and Hind Ⅲ sites. The adeno-associated viral vector of the present invention is shown in Figures 1 and 2.
상기와 같이 제조된 벡터는 도 3에 도시된 바와 같은 공정을 통해 생산하였다.The vector prepared as described above was produced through the process shown in FIG. 3 .
<실시예 2> AIMP2-DX2(이하 DX2라고 함)의 TNF-α-유도 신경염증 억제효과 확인<Example 2> Confirmation of TNF-α-induced neuroinflammation inhibitory effect of AIMP2-DX2 (hereinafter referred to as DX2)
2-1. DX2의 친-세포사멸(pro-apoptotic) 활성 억제효과 확인2-1. Confirmation of inhibitory effect on pro-apoptotic activity of DX2
신경염증은 ALS 환자의 신경계에서 일반적으로 관찰되는 특징이고, TNF-α는 염증을 유도하는 중요한 사이토카인으로 알려져 있다. TNF-α 시그널링에서, TRAF (Tumor necrosis factor receptor-associated factor)2는 세포사에 중요한 역할을 한다. TNF-α 시그널링이 활성화될 때, TRADD (TNF 수용체-associated death domain) 및 TRAF2가 TNF-α 수용체인 TNFR1에 결합하고, 활성화된 복합체가 IKK 활성을 매개한다. caspase-8이 불활성화됨에 따라, TRAF2가 TRADD 복합체로부터 방출되고, 방출된 TRAF2가 E3 유비퀴틴 리가아제인 cIAP에 의해 유비퀴틴화되어 세포사를 촉진한다. 친-세포사멸 단백질인 AIMP2는 cIAP에 의한 TRAF2의 유비퀴틴화를 촉진하고 세포사를 유도한다. 그러므로, AIMP2의 경쟁자인 DX2가 TRAF2의 결합에 영향을 미치는지를 확인하였다. 신경모세포종 세포를 mock, DX2(AIMP2-DX2; 엑소 2가 제거된 AIMP2 ) 또는 AIMP2 발현 플라스미드로 트랜스펙션시키고 면역침강법을 실시하고 그 결과를 도 4에 나타냈다. 도 4의 상부 패널은 DX2가 투여량 의존적으로 AIMP2가 TRAF2에 결합하는 것을 저해함을 나타낸다. 하부 패널은 NF-κB 활성화 및 세포 생존율을 나타낸다. 세포 생존율은 정상 조건에서는 대조, AIMP2 및 DX2 트랜스펙션 세포에서 차이가 없었으나, TNF-α 처리 후, DX2 과발현 세포에서 AIMP2-트랜스펙션 세포에 비해 증가된 세포 생존율 및 NF-κB 활성화가 관찰되었다.Neuroinflammation is a feature commonly observed in the nervous system of ALS patients, and TNF-α is known as an important cytokine inducing inflammation. In TNF-α signaling, tumor necrosis factor receptor-associated factor (TRAF)2 plays an important role in cell death. When TNF-α signaling is activated, TRADD (TNF receptor-associated death domain) and TRAF2 bind to TNFR1, a TNF-α receptor, and the activated complex mediates IKK activity. As caspase-8 is inactivated, TRAF2 is released from the TRADD complex, and the released TRAF2 is ubiquitinated by cIAP, an E3 ubiquitin ligase, to promote cell death. AIMP2, a pro-apoptotic protein, promotes ubiquitination of TRAF2 by cIAP and induces cell death. Therefore, it was confirmed whether DX2, a competitor of AIMP2, affects the binding of TRAF2. Neuroblastoma cells were transfected with mock, DX2 (AIMP2-DX2; AIMP2 from which exo 2 was removed) or AIMP2 expression plasmid and subjected to immunoprecipitation, and the results are shown in FIG. 4 . The upper panel of FIG. 4 shows that DX2 inhibits AIMP2 binding to TRAF2 in a dose-dependent manner. Lower panel shows NF-κB activation and cell viability. There was no difference in cell viability in control, AIMP2 and DX2 transfected cells under normal conditions, but after TNF-α treatment, increased cell viability and NF-κB activation were observed in DX2 overexpressing cells compared to AIMP2-transfected cells. became
2-2. DX2의 TNF-α 유도 신경세포사 억제 확인 2-2. Confirmation of inhibition of TNF-α-induced neuronal death by DX2
그런 다음, DX2가 TRAF2의 유비퀴틴화를 저해하는지를 확인하였다. in vitro ALS-유사 조건을 위한 모델로서 돌연변이 SOD1를 사용하였다. 신경모세포종 세포를 SOD1 G93A 및 DX2 발현 플라스미드로 트랜스펙션시키고, in vitro 유비퀴틴화 분석을 실시하였다. 도 5A에서 볼 수 있는 바와 같이, DX2가 투여량 의존적으로 돌연변이 SOD1-유도 TRAF2 유비퀴틴화를 효과적으로 저해함이 관찰되었다. Then, it was confirmed whether DX2 inhibits ubiquitination of TRAF2. Mutant SOD1 was used as a model for in vitro ALS-like conditions. Neuroblastoma cells were transfected with SOD1 G93A and DX2 expression plasmids and subjected to in vitro ubiquitination assay. 5A , it was observed that DX2 effectively inhibited mutant SOD1-induced TRAF2 ubiquitination in a dose-dependent manner.
그 다음, 신경세포사에서 DX2의 억제 효과를 확인하였다. CHX/TNF-α는 WT SOD1 또는 돌연변이 SOD1 (G85R 및 G93A) 과발현 세포 둘 다에서 세포사를 유도하나, 돌연변이 SOD1 과발현 세포에서 약 20% 정도 더 높은 세포사 비율을 보였다. DX2는 CHX/TNF-α-처리 WT 또는 돌연변이 SOD1 (G85R and G93A) 세포 둘 다에서 세포사멸 수준을 상당히 감소시켰다 (도 5B). Then, the inhibitory effect of DX2 on neuronal cell death was confirmed. CHX/TNF-α induced apoptosis in both WT SOD1 or mutant SOD1 (G85R and G93A) overexpressing cells, but showed an approximately 20% higher cell death rate in mutant SOD1 overexpressing cells. DX2 significantly reduced the level of apoptosis in both CHX/TNF-α-treated WT or mutant SOD1 (G85R and G93A) cells ( FIG. 5B ).
또한, DX2의 세포사 억제 효과가 일차 신경원에서도 관찰되었다. DX2 과발현 플라스미드를 WT 또는 SOD1 트랜스제닉 마우스로부터 추출한 일차 신경 세포로 트랜스펙션시켰다. 트랜스펙션된 세포를 CHX/TNF-α로 처리하고 세포사 비율을 분석하였다. In addition, the cell death inhibitory effect of DX2 was also observed in primary neurons. DX2 overexpression plasmids were transfected into primary neurons extracted from WT or SOD1 transgenic mice. The transfected cells were treated with CHX/TNF-α and the cell death rate was analyzed.
그 결과, CHX/TNF-α는 WT 또는 G93A 일차 신경 세포 둘 다에서 세포사를 유도하였지만, G93A 일차 신경 세포에서 약 13% 더 높은 세포사 비율을 보였다. DX2는 CHX/TNF-α-처리된 WT 또는 G93A 일차 신경 세포 둘 다에서 세포사멸을 상당히 감소시켰다 (도 5C). As a result, CHX/TNF-α induced apoptosis in both WT or G93A primary neurons, but showed an approximately 13% higher rate of cell death in G93A primary neurons. DX2 significantly reduced apoptosis in both CHX/TNF-α-treated WT or G93A primary neurons ( FIG. 5C ).
상기 결과는 DX2가 유전적 돌연변이와 관련없는 일반적인 신경염증 상황 또는 SOD1 돌연변이 관련 신경염증 상황에서 신경세포의 세포사멸을 억제함을 입증하는 것이다.The above results demonstrate that DX2 inhibits neuronal cell death in a normal neuroinflammatory situation unrelated to genetic mutation or in a neuroinflammatory situation related to SOD1 mutation.
2-3. TRAF2 발현 억제효과 확인2-3. Confirmation of TRAF2 expression inhibitory effect
TRAF2는 TNF-α 시그널링 경로에 의한 세포사를 매개하는 중요한 단백질이다. 따라서, ALS 질환에서 TRAF2 발현의 중요성을 확인하기 위해 ALS 마우스 모델(SOD1 G93A 트랜스제닉 마우스)에서 TRAF2 수준을 측정하였다. 흥미롭게도, TNF-α에 의해 유도되는 TRAF2의 유비퀴틴화를 조절하는 AIMP2로의 KARS1 결합량이 변하지 않았다; 그러나, TRAF2 발현은 ALS 마우스에서 상당히 저해되었다 (도 6A 및 6B). 이에 따라, ALS-유도 마우스에서 DX2 또는 GFP의 효율적인 발현을 보이는 치료제로서 DX2의 잠재적 효능을 확인하기 위해 AAV 벡터를 설계(실시예 1)하였다. AAV-DX2 발현의 in vivo 평가를 위해, AAV-GFP 또는 AAV-DX2를 마우스의 척수에 주입하고, 3주 후에 GFP 및 DX2 발현을 면역형광 및 mRNA 발현으로 분석하였다. GFP 및 DX2 발현이 척수의 주입 부위에서 관찰되었으며, 이는 AAV-GFP 또는 DX2가 특정 주입 부위에서만 발현할 수 있음을 나타내는 것이다 (도 6C 및 6D).TRAF2 is an important protein mediating cell death by the TNF-α signaling pathway. Therefore, to confirm the importance of TRAF2 expression in ALS disease, TRAF2 levels were measured in an ALS mouse model (SOD1 G93A transgenic mice). Interestingly, the amount of KARS1 binding to AIMP2, which regulates TNF-α-induced ubiquitination of TRAF2, did not change; However, TRAF2 expression was significantly inhibited in ALS mice ( FIGS. 6A and 6B ). Accordingly, an AAV vector was designed (Example 1) to confirm the potential efficacy of DX2 as a therapeutic agent showing efficient expression of DX2 or GFP in ALS-induced mice. For in vivo evaluation of AAV-DX2 expression, AAV-GFP or AAV-DX2 was injected into the spinal cord of mice, and after 3 weeks, GFP and DX2 expression was analyzed by immunofluorescence and mRNA expression. GFP and DX2 expression was observed at the injection site of the spinal cord, indicating that either AAV-GFP or DX2 could be expressed only at the specific injection site ( FIGS. 6C and 6D ).
2-4. DX2 유전자-주입 루게릭병 마우스 모델에서 마우스의 수명 연장 확인2-4. Confirmation of Lifespan Extension of Mice in DX2 Gene-Injected ALS Mouse Model
In vivo에서 DX2의 효과를 연구하기 위해 ALS 마우스 (SOD1 트랜스제닉 마우스)의 척추관에 AAV-GFP (GFP) 또는 AAV-DX2(실시예 1에서 제조)를 처리하였다. 도 7A에서 볼 수 있는 바와 같이, AAV-DX2의 척추강내 주입으로 인해 마우스 모델의 행동이 개선되고 (도 7A), 질환의 발병 시기가 지연되는 (도 7B) 것을 확인하였다. 뿐만 아니라, AAV-GFP 마우스 군에 비해 AAV-DX2를 주입한 마우스 군의 수명이 연장된 것을 확인하였다 (도 7C). In order to study the effect of DX2 in vivo , the spinal canal of ALS mice (SOD1 transgenic mice) was treated with AAV-GFP (GFP) or AAV-DX2 (prepared in Example 1). As can be seen in FIG. 7A , it was confirmed that the behavior of the mouse model was improved ( FIG. 7A ) and the onset of the disease was delayed ( FIG. 7B ) due to the intrathecal injection of AAV-DX2. In addition, it was confirmed that the lifespan of the AAV-DX2 injected mouse group was extended compared to the AAV-GFP mouse group ( FIG. 7C ).
2-5. ALS 마우스 모델에서 TRAF2 발현의 감소 및 DX2에 의한 TRAF2의 발현 증가 확인2-5. Confirmation of decrease in TRAF2 expression and increase in TRAF2 expression by DX2 in ALS mouse model
TNF-α 신호는 염증신호전달체계 중 하나로, 신경염증 및 루게릭병의 진행에 중요한 영향을 미치며, TNF-α 신호에서 TRAF2의 발현은 TNF-α에 의한 세포사멸을 조절하는 핵심적인 역할을 수행한다. 실제로, 도 6에서 루게릭병 모델 마우스에서 현저하게 감소하는 TRAF2의 발현 수준을 확인할 수 있었다. 또한, 도 4 및 도 5에서 in vitro에서 DX2가 TRAF2의 발현 수준을 조절하고, 이로 인해 세포 사멸이 억제되는 것을 이미 확인하였다. 이를 바탕으로 실제 마우스 모델에서 DX2의 발현이 TRAF2 발현을 조절하는지 확인한 결과, 루게릭 질환 모델 마우스에서 현저하게 감소된 TRAF2 발현이 DX2의 주입 (AAV-DX2)에 의해 정상 마우스 (WT) 수준으로 회복되는 것을 확인하였다 (도 8C). 또한, ALS 마우스 모델에서 확인되는 신경 염증 마커 인자 (도 8A, 8B) 및 세포 사멸 인자 (도 8D, 8E) 역시 DX2에 의해 감소되는 것을 확인하였다.TNF-α signal is one of the inflammatory signal transduction systems, and has an important effect on the progression of neuroinflammation and ALS. . In fact, it was confirmed in FIG. 6 that the expression level of TRAF2 significantly decreased in the ALS model mouse. In addition, it was confirmed that DX2 regulates the expression level of TRAF2 in vitro in FIGS. 4 and 5, thereby suppressing cell death. Based on this, it was confirmed that the expression of DX2 in the actual mouse model regulates the expression of TRAF2. As a result, the significantly reduced expression of TRAF2 in the mouse model of ALS is restored to the normal mouse (WT) level by injection of DX2 (AAV-DX2). was confirmed (FIG. 8C). In addition, it was confirmed that the neuroinflammatory marker factors (FIGS. 8A, 8B) and apoptosis factors (FIGS. 8D, 8E) confirmed in the ALS mouse model were also reduced by DX2.
2-6. 작용 메커니즘2-6. mechanism of action
자유형의 AIMP2은 TRAF2에 결합하여 TRAF2 단백질의 유비퀴틴화를 촉진하고, 궁극적으로는 TRAF2-결합 AIMP2는 세포사를 유도한다. 그러나, AIMP2에 의한 TRAF2의 유비퀴틴화는 DX2의 존재하에서는 상당히 감소하였으며, 이러한 감소에 의해, DX2는 TNF-α-활성 조건에서 AIMP2-자극 세포사멸을 저해한다 (도 9).Freeform AIMP2 binds to TRAF2 and promotes ubiquitination of TRAF2 protein, and ultimately TRAF2-binding AIMP2 induces cell death. However, ubiquitination of TRAF2 by AIMP2 was significantly reduced in the presence of DX2, and by this decrease, DX2 inhibits AIMP2-stimulated apoptosis in the TNF-α-active condition ( FIG. 9 ).
<실시예 3> ALS 질환에서 본 발명의 AAV 벡터의 아노이키스-유도 신경 세포사 감소 확인<Example 3> Confirmation of reduction in anoikis-induced neuronal cell death of the AAV vector of the present invention in ALS disease
3-1. ALS 질환에서 아노이키스를 유도, 맴브레인 67 라미닌 수용체 감소 확인3-1. Induction of anoikis in ALS disease, confirmation of membrane 67 laminin receptor reduction
p40/37LRP의 이량체 형태인 67kDa 라미닌 수용체 (67LR)는 세포막에서 KARS1과 결합하여 세포 이동을 증가시키는 역할을 한다. 67LR은 세포이동 및 세포사에서 중요한 인자로서 보고되었으며, KARS1은 67LR의 안정화를 통해 세포 이동을 가능하게 한다. 라미닌 시그널에 따라, p38MAPK에 의해 트레오닌 52에서 인산화된 KARS1은 MSC (multi-tRNA synthetase complex)로부터 분리되어 혈장 맴브레인으로 이동한다. 맴브레인에서, KARS1은 67LR에 결합하여 67LR의 E3 유비퀴틴 리가아제인 Nedd4와의 상호작용을 저해하고, 이에 따라 67LR의 유비퀴틴화-매개 분해가 억제된다. 이에 따라, ALS 조건에서 67LR 단백질 안정성이 신경 세포사와 관련이 있는지를 확인하였다.The 67kDa laminin receptor (67LR), a dimer form of p40/37LRP, plays a role in increasing cell migration by binding to KARS1 in the cell membrane. 67LR has been reported as an important factor in cell migration and cell death, and KARS1 enables cell migration through stabilization of 67LR. According to the laminin signal, KARS1 phosphorylated at threonine 52 by p38MAPK is separated from the multi-tRNA synthetase complex (MSC) and migrated to the plasma membrane. In the membrane, KARS1 binds to 67LR and inhibits the interaction of 67LR with Nedd4, an E3 ubiquitin ligase, thereby inhibiting ubiquitination-mediated degradation of 67LR. Accordingly, it was confirmed whether 67LR protein stability was related to neuronal cell death in ALS conditions.
67 LR 단백질 안정성의 효과를 연구하기 위해, 인간 신경모세포종 세포인 SK-N-SH 세포를 ALS-유사 조건인 SOD1-G93A로 트랜스펙션시키고, 67 LR 수준을 분석하였다. 그 결과, 혈장 맴브레인에서 67LR 단백질 발현이 ALS-유사 조건에서 상당히 감소됨을 확인하였다 (도 10A 및 10B). 또한, 67LR은 MAPK/ERK (Extracellular Signal-regulated Kinase) 시그널을 활성화시키고 세포 이동을 증가시키는 것으로 잘 알려져 있다. 이에, ALS-유사 조건에서 67LR 단백질 수준이 세포 이동 및 다운스트림 시그널링 경로에 영향을 미치는지를 확인하였다. 도 10C 및 10D에서 볼 수 있는 바와 같이, 라미닌 수용체에 대한 기질로서 라미닌-1에 의한 세포 이동 및 다운스트림 시그널링 경로의 감소가 ALS 조건에서 관찰되었다. To study the effect of 67 LR protein stability, SK-N-SH cells, a human neuroblastoma cell, were transfected with SOD1-G93A, an ALS-like condition, and 67 LR levels were analyzed. As a result, it was confirmed that the expression of 67LR protein in the plasma membrane was significantly reduced under ALS-like conditions ( FIGS. 10A and 10B ). In addition, it is well known that 67LR activates MAPK/ERK (Extracellular Signal-regulated Kinase) signaling and increases cell migration. Accordingly, it was confirmed whether the 67LR protein level affects cell migration and downstream signaling pathways under ALS-like conditions. As can be seen in FIGS. 10C and 10D , cell migration and reduction of downstream signaling pathways by laminin-1 as a substrate for laminin receptor were observed in ALS conditions.
아노이키스는 세포외 매트릭스 (ECM)과 세포 맴브레인 단백질 사이의 접촉이 줄어들어 유발되는 세포사멸의 한 종류이며 아노이키스의 내성은 세포 생존에서 중요한 역할을 한다. 세포를 poly HEMA가 코팅된 플레이트에 접종하여 KARS1 및 라미닌 처리가 아노이키스에 의해 유도되는 세포 생존율에 영향을 주는지를 관찰하였다. 그 결과, 93A 트랜스펙션 세포의 세포사는 WT 세포에 비해 라미닌-1 처리에 의한 세포 사멸 억제 효과가 현저하게 떨어지는 것을 확인하였다 (도 10E). 그리고, KARS1 발현 수준은 아노이키스-유도 세포사에 영향을 미치지 않았다(도 10F). 이러한 결과는 라미닌 수용체에 의한 아노이키스-유도 세포사멸의 저해가 라미닌 수용체와 ECM의 상호작용을 통한 다운스트림 시그널링의 증가에 의한 것임을 시사하는 것이다.Anoikis is a type of apoptosis induced by reduced contact between the extracellular matrix (ECM) and cell membrane proteins, and the resistance of anoikis plays an important role in cell survival. Cells were inoculated on poly HEMA-coated plates to observe whether KARS1 and laminin treatment affect cell viability induced by anoikis. As a result, it was confirmed that the cell death of 93A-transfected cells was significantly lower than that of WT cells (FIG. 10E). And, KARS1 expression level did not affect the anoikis-induced cell death (FIG. 10F). These results suggest that the inhibition of anoikis-induced apoptosis by the laminin receptor is due to an increase in downstream signaling through the interaction of the laminin receptor with the ECM.
3-2. DX2의 ALS-유사 세포 조건에서 67 LR 발현 회복 확인 3-2. Confirmation of Restoration of 67 LR Expression in ALS-Like Cell Conditions of DX2
67LR의 발현이 혈장 맴브레인에서 감소하고 DX2에 의해 회복되는지를 확인하였다. 그 결과, ALS-유도 유전자를 발현하는 세포에서 DX2의 과발현은 혈장 맴브레인 (도 11A) 및 전체 세포 (도 11B)에서 67LR 단백질을 증가시켰으며, 67LR의 다운스트림 시그널 또한 DX2 유전자 도입에 의해 회복됨이 관찰되었다 (도 11C). 그런 다음 각 플라스미드로 트랜스펙션된 세포의 부착을 시험하였다. 도 11D에서 볼 수 있는 바와 같이, DX2 발현 세포는 세포 부착력이 증가되었으며, 이는 DX2가 세포의 분리와 아노이키스를 예방함을 시사하는 것이다.It was confirmed whether the expression of 67LR was decreased in the plasma membrane and restored by DX2. As a result, overexpression of DX2 in cells expressing ALS-induced genes increased 67LR protein in plasma membrane (FIG. 11A) and whole cells (FIG. 11B), and the downstream signal of 67LR was also recovered by DX2 gene introduction. was observed (FIG. 11C). The adhesion of cells transfected with each plasmid was then tested. As can be seen in FIG. 11D , DX2 expressing cells had increased cell adhesion, suggesting that DX2 prevents cell separation and anoikis.
<실시예 4> scAAV와 ssAAV의 비교<Example 4> Comparison of scAAV and ssAAV
본 발명의 TRS 제거 ITR을 포함하는 scAAV와 종래의 ssAAV의 비교를 위해 AAV-GFP의 형질전환 효율 테스트를 실시하였다. SK-SY5Y (neuroblastoma) 세포를 scAAV-GFP 또는 ssAAV-GFP (single-stranded AAV-GFP)로 감염시키고 48시간 후에, ssAAV-GFP 및 scAAV-GFP의 GFP 발현을 형광 현미경으로 관찰하였다. 그 결과, 도 12에서 볼 수 있는 바와 같이, 본 발명의 scAAV와 종래의 ssAAV에 비해 더 우수한 GFP 발현 수준을 보임을 확인하였다. 즉, 중추신경계에서 AIMP2-DX2의 발현을 증가시키면서, 혈액 또는 다른 기관에서의 발현을 제한하고 중추신경계에서만 특이적으로 발현시키기 위하여, 본 발명의 AAV 벡터가 최적화된 벡터임을 간접적으로 확인하였다.In order to compare the scAAV containing the TRS-removed ITR of the present invention and the conventional ssAAV, the AAV-GFP transformation efficiency test was performed. SK-SY5Y (neuroblastoma) cells were infected with scAAV-GFP or ssAAV-GFP (single-stranded AAV-GFP), and 48 hours later, GFP expression of ssAAV-GFP and scAAV-GFP was observed under a fluorescence microscope. As a result, as can be seen in FIG. 12 , it was confirmed that the scAAV of the present invention and the conventional ssAAV showed a better GFP expression level. That is, it was indirectly confirmed that the AAV vector of the present invention was an optimized vector in order to increase the expression of AIMP2-DX2 in the central nervous system, restrict expression in blood or other organs, and express it specifically only in the central nervous system.

Claims (20)

  1. 제 1 ITR(inverted terminal repeat)을 코딩하는 서열;a sequence encoding a first inverted terminal repeat (ITR);
    프로모터 서열;promoter sequence;
    상기 프로모터 서열과 작동가능하게 연결된 AIMP2-DX2를 코딩하는 서열;a sequence encoding AIMP2-DX2 operably linked to the promoter sequence;
    상기 AIMP2-DX2를 코딩하는 서열과 연결된 mir142-3p를 표적하는 서열; 및a sequence targeting mir142-3p linked to the sequence encoding AIMP2-DX2; and
    제 2 ITR을 코딩하는 서열sequence encoding the second ITR
    을 포함하되, including,
    상기 서열을 5'에서 3' 방향으로 순차적으로 포함하는, 아데노 부속 바이러스 벡터.An adeno-associated viral vector comprising the sequence sequentially in the 5' to 3' direction.
  2. 제 1항에 있어서, 상기 제 1 ITR을 코딩하는 서열 또는 제 2 ITR을 코딩하는 서열은 TRS(terminal resolution site) 부위가 제거된 서열인 것인, 벡터.The vector according to claim 1, wherein the sequence encoding the first ITR or the sequence encoding the second ITR is a sequence in which a terminal resolution site (TRS) region has been removed.
  3. 제 2항에 있어서, 상기 제 1 ITR을 코딩하는 서열(left ITR)은 서열번호 1로 표시되는 서열이고, 제 2 ITR을 코딩하는 서열(right ITR)은 서열번호 2로 표시되는 서열인 것인, 벡터.The method according to claim 2, wherein the first ITR-coding sequence (left ITR) is a sequence represented by SEQ ID NO: 1, and the second ITR-coding sequence (right ITR) is a sequence represented by SEQ ID NO: 2 , vector.
  4. 제 1항에 있어서, 상기 아데노 부속 바이러스는 scAAV(self complementary adeno-associated virus)인 것인, 벡터.The vector of claim 1, wherein the adeno-associated virus is a self complementary adeno-associated virus (scAAV).
  5. 제 1항에 있어서, 상기 프로모터 서열과 AIMP2-DX2를 코딩하는 서열 사이에 EagI-NotI, Eco53KI, SacI, EcoRI, acc65I, KpnI 및 SexAI로 이루어진 군 중 어느 하나 이상의 제한효소 사이트를 포함하는 것인, 벡터.According to claim 1, EagI-NotI, Eco53KI, SacI, EcoRI, acc65I, KpnI and SexAI between the promoter sequence and the sequence encoding AIMP2-DX2 to include any one or more restriction enzyme sites from the group consisting of, vector.
  6. 제 1항에 있어서, 상기 mir142-3P를 표적하는 서열의 3' 말단 쪽으로 제 1 종결코돈, 폴리 A를 코딩하는 서열 및 제 2 종결코돈이 순차적으로 위치하는 것인, 벡터.The vector according to claim 1, wherein the first stop codon, the poly A-encoding sequence and the second stop codon are sequentially positioned toward the 3' end of the mir142-3P-targeting sequence.
  7. 제 6항에 있어서, 상기 제 1 종결코돈 또는 제 2 종결코돈은 종결코돈이 3회 이상 반복된 것인, 벡터.The vector according to claim 6, wherein the first stop codon or the second stop codon has a stop codon repeated three or more times.
  8. 제 6항에 있어서, 상기 제 1 종결코돈과 제 2 종결코돈 사이에 BspDI-ClaI, BamHI 및 StuI로 이루어진 군 중 어느 하나 이상의 제한효소 사이트를 포함하는 것인, 벡터.The vector according to claim 6, wherein one or more restriction enzyme sites from the group consisting of BspDI-ClaI, BamHI, and StuI are included between the first stop codon and the second stop codon.
  9. 제 1항에 있어서, 상기 miR142-3p를 표적하는 서열은 서열번호 5 또는 6으로 표시되는 서열을 포함하는 것인, 벡터.The vector according to claim 1, wherein the sequence targeting miR142-3p includes the sequence represented by SEQ ID NO: 5 or 6.
  10. 제 1항에 있어서, 상기 아데노 부속 바이러스 벡터는 서열번호 7로 표시되는 서열을 포함하는 것인, 벡터.The vector according to claim 1, wherein the adeno-associated virus vector comprises the sequence represented by SEQ ID NO: 7.
  11. 제 1항에 있어서, 상기 프로모터는 레트로바이러스(LTR) 프로모터, 사이토메갈로바이러스(CMV)프로모터, 라우스 육종 바이러스(RSV) 프로모터, MMT 프로모터, EF-1 알파 프로모터, UB6 프로모터, 치킨 베타-액틴 프로모터, CAG 프로모터, RPE65 프로모터, 옵신 프로모터 및 이들의 조합으로 이루어진 군으로부터 선택된 프로모터인 것인, 벡터.The method of claim 1, wherein the promoter is a retrovirus (LTR) promoter, a cytomegalovirus (CMV) promoter, a Rous sarcoma virus (RSV) promoter, an MMT promoter, an EF-1 alpha promoter, a UB6 promoter, a chicken beta-actin promoter, The vector is a promoter selected from the group consisting of CAG promoter, RPE65 promoter, opsin promoter, and combinations thereof.
  12. 제 1항에 있어서, 상기 miR-142-3p를 표적하는 핵산은 2 내지 10번 반복되는 것인, 벡터.The vector according to claim 1, wherein the nucleic acid targeting miR-142-3p is repeated 2 to 10 times.
  13. 제 1항 내지 제 12항 중 어느 한 항의 벡터를 유효성분으로 포함하는 퇴행성 신경질환 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating neurodegenerative diseases comprising the vector of any one of claims 1 to 12 as an active ingredient.
  14. 제 13 항에 있어서, 상기 퇴행성 신경질환은 루게릭 병(amyotrophic lateral sclerosis; ALS), 알츠하이머 질환(alzheimer's disease), 파킨슨 병(Parkinson’s disease), 망막 변성증(retinal degeneration), 경도인지 장애(mild cognitive impairment), 다발-경색성 치매((Multi-infarct dementia), 전두측두치매(fronto-temporal dementia), 루이소체 치매(dementia with Lewy bodies), 헌팅턴 질환(Huntington's disease), 신경퇴행질환, 대사성 뇌질환, 우울증, 간질, 다발성 경화증(Multiple sclerosis), 피질기저퇴행증(corticobasal degeneration), 다계통위축병(multiple system atrophy), 진행성핵상마비(progressive supranuclear palsy), 치상핵적핵담창구시상하핵위축증(dentatorubropallidoluysian atrophy), 척수소뇌실조병(spinocerebella ataxia), 원발성 측삭 경화증(primary lateral sclerosis), 척수근육위축병(spinal muscular atrophy), 중증 근무력증(myasthenia gravis) 및 뇌졸중(stroke)으로 구성된 군으로부터 선택된 1종 이상인, 조성물.According to claim 13, wherein the neurodegenerative disease is Lou Gehrig's disease (amyotrophic lateral sclerosis; ALS), Alzheimer's disease (alzheimer's disease), Parkinson's disease (Parkinson's disease), retinal degeneration (retinal degeneration), mild cognitive impairment (mild cognitive impairment) , Multi-infarct dementia, fronto-temporal dementia, dementia with Lewy bodies, Huntington's disease, neurodegenerative disease, metabolic brain disease, depression , epilepsy, multiple sclerosis, corticobasal degeneration, multiple system atrophy, progressive supranuclear palsy, dentatorubropallidoluysian atrophy , Spinocerebella ataxia, primary lateral sclerosis, spinal muscular atrophy, myasthenia gravis and at least one selected from the group consisting of stroke, composition .
  15. 제 14항에 있어서, 상기 퇴행성 신경질환은 루게릭 병(amyotrophic lateral sclerosis; ALS)인 것인, 조성물.15. The method of claim 14, wherein the neurodegenerative disease is Lou Gehrig's disease (amyotrophic lateral sclerosis; ALS), the composition.
  16. 제 15항에 있어서, 치료는 개체의 운동 능력을 개선시키거나 수명을 연장시키는 것인, 조성물.16. The composition of claim 15, wherein the treatment improves athletic performance or prolongs the lifespan of the subject.
  17. 제 13항에 있어서, 상기 조성물은 뇌 또는 척수로부터 선택되는 중추신경계에 투여되는 것인, 조성물.The composition of claim 13, wherein the composition is administered to the central nervous system selected from the brain or spinal cord.
  18. 제 17항에 있어서, 상기 조성물이 중추신경계에 투여되면, 벡터가 중추신경계 세포에서만 특이적으로 발현되는 것인, 조성물.The composition of claim 17, wherein when the composition is administered to the central nervous system, the vector is specifically expressed only in cells of the central nervous system.
  19. 제 1항 내지 제 12항 중 어느 한 항의 벡터를 약학적으로 유효한 양으로 퇴행성 신경질환에 걸린 개체에 투여하는 단계를 포함하는 퇴행성 신경질환의 치료 방법.13. A method of treating a neurodegenerative disease comprising administering the vector of any one of claims 1 to 12 to a subject suffering from a neurodegenerative disease in a pharmaceutically effective amount.
  20. 퇴행성 신경질환 예방 또는 치료용 약학 조성물의 제조에 사용하기 위한, 제 1항 내지 제 12항 중 어느 한 항의 벡터의 용도. Use of the vector of any one of claims 1 to 12 for use in the preparation of a pharmaceutical composition for the prevention or treatment of neurodegenerative diseases.
PCT/KR2021/012420 2020-09-14 2021-09-13 Adeno-associated virus vector for targeted gene delivery WO2022055320A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2020-0117739 2020-09-14
KR1020200117739A KR20220035693A (en) 2020-09-14 2020-09-14 Adeno-associated virus vector for targeted gene delivery

Publications (1)

Publication Number Publication Date
WO2022055320A1 true WO2022055320A1 (en) 2022-03-17

Family

ID=80632026

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2021/012420 WO2022055320A1 (en) 2020-09-14 2021-09-13 Adeno-associated virus vector for targeted gene delivery

Country Status (2)

Country Link
KR (1) KR20220035693A (en)
WO (1) WO2022055320A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023218430A1 (en) * 2022-05-13 2023-11-16 Generoath Co., Ltd. Methods of treating retinal degenerative diseases using aimp2-dx2 and optionally a target sequence for mir‑142 and compositions thereof

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20080041601A (en) * 2005-05-27 2008-05-13 폰다지오네 센트로 산 라파엘 델 몬테 테이보 Gene vector comprising mi-rna
JP2015523410A (en) * 2012-07-31 2015-08-13 イェダ リサーチ アンド デベロップメント カンパニー リミテッド Methods for diagnosing and treating motor neuron disease
WO2016115177A1 (en) * 2015-01-12 2016-07-21 Juno Therapeutics, Inc. Modified hepatitis post-transcriptional regulatory elements
KR20170041363A (en) * 2015-10-07 2017-04-17 원광대학교산학협력단 Pharmaceutical composition comprising AIMP2-DX2 for preventing or treating neuronal diseases and use thereof
US20180051300A1 (en) * 2006-01-31 2018-02-22 The Board Of Trustees Of The Leland Stanford Junior University Self-complementary parvoviral vectors, and methods for making and using the same
WO2020188472A1 (en) * 2019-03-15 2020-09-24 Generoath Co., Ltd. Vectors containing aimp2-dx2 and target nucleic acids for mir-142 and uses thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20080041601A (en) * 2005-05-27 2008-05-13 폰다지오네 센트로 산 라파엘 델 몬테 테이보 Gene vector comprising mi-rna
US20180051300A1 (en) * 2006-01-31 2018-02-22 The Board Of Trustees Of The Leland Stanford Junior University Self-complementary parvoviral vectors, and methods for making and using the same
JP2015523410A (en) * 2012-07-31 2015-08-13 イェダ リサーチ アンド デベロップメント カンパニー リミテッド Methods for diagnosing and treating motor neuron disease
WO2016115177A1 (en) * 2015-01-12 2016-07-21 Juno Therapeutics, Inc. Modified hepatitis post-transcriptional regulatory elements
KR20170041363A (en) * 2015-10-07 2017-04-17 원광대학교산학협력단 Pharmaceutical composition comprising AIMP2-DX2 for preventing or treating neuronal diseases and use thereof
WO2020188472A1 (en) * 2019-03-15 2020-09-24 Generoath Co., Ltd. Vectors containing aimp2-dx2 and target nucleic acids for mir-142 and uses thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023218430A1 (en) * 2022-05-13 2023-11-16 Generoath Co., Ltd. Methods of treating retinal degenerative diseases using aimp2-dx2 and optionally a target sequence for mir‑142 and compositions thereof

Also Published As

Publication number Publication date
KR20220035693A (en) 2022-03-22

Similar Documents

Publication Publication Date Title
CN108350467B (en) Gene construct
KR102616629B1 (en) Genetic constructs for use in the treatment of neurodegenerative disorders or stroke
JP7291423B2 (en) Vectors containing target nucleic acids for AIMP2-DX2 and miR-142, and uses thereof
AU2009216930A1 (en) Method of treating ocular diseases by gene therapy
WO2022055320A1 (en) Adeno-associated virus vector for targeted gene delivery
CN111936172A (en) Compositions and methods for treating retinal disorders
WO2019169371A1 (en) Drug stabilized therapeutic transgenes delivered by adeno-associated virus expression
KR20230110503A (en) Method for treating age-related macular disease using target sequences for AIMP2-DX2 and optionally miR-142 and compositions thereof
US20040166091A1 (en) Materials and methods for treating disorders of the ear
US20230374092A1 (en) METHODS OF TREATING NEURONAL DISEASES USING AIMP2-DX2 AND OPTIONALLY A TARGET SEQUENCE FOR miR-142 AND COMPOSITIONS THEREOF
WO2022080893A2 (en) Composition for delivering gene to brain tissue and use thereof
KR102626543B1 (en) Recombinant vector containing AIMP2-DX2 and target sequence for miR-142-3p
WO2021112597A1 (en) Composition for treating diabetic retinopathy, comprising raav containing soluble vegfr-1 variant cdna
US20240050520A1 (en) Gene therapy for treating usher syndrome
US20230181763A1 (en) Application of aav44.9 vector in gene therapy for the inner ear
WO2023163562A1 (en) Composition for preventing or treating alzheimer&#39;s disease comprising nucleic acid complex
US20230061456A1 (en) Methods and compositions for generating type 1 vestibular hair cells
AU2022259511A1 (en) Gene therapy for neuroprotection
WO2023064388A2 (en) Stereocilin promoters and uses thereof
CA3220122A1 (en) Granulin/epithelin modules and combinations thereof to treat neurodegenerative disease
CN117062629A (en) Methods for reducing degeneration of retinal ganglion cells
WO2014171721A1 (en) Ik factor and pharmaceutical use of nucleic acid encoding ik factor
WO2003070900A2 (en) Cftr with a partially deleted r domain and uses thereof
WO2012086474A1 (en) Therapeutic agent for central nervous system diseases
JPWO2022071974A5 (en)

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: 21867185

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21867185

Country of ref document: EP

Kind code of ref document: A1