WO2023132673A1 - Composition comprising nucleic acid complex for prevention or treatment of degenerative brain disease - Google Patents

Composition comprising nucleic acid complex for prevention or treatment of degenerative brain disease Download PDF

Info

Publication number
WO2023132673A1
WO2023132673A1 PCT/KR2023/000259 KR2023000259W WO2023132673A1 WO 2023132673 A1 WO2023132673 A1 WO 2023132673A1 KR 2023000259 W KR2023000259 W KR 2023000259W WO 2023132673 A1 WO2023132673 A1 WO 2023132673A1
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
disease
acid complex
bioactive
carrier peptide
Prior art date
Application number
PCT/KR2023/000259
Other languages
French (fr)
Korean (ko)
Inventor
옥예진
박민정
김혜주
유지연
박희경
Original Assignee
주식회사 시선테라퓨틱스
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 시선테라퓨틱스 filed Critical 주식회사 시선테라퓨틱스
Publication of WO2023132673A1 publication Critical patent/WO2023132673A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/549Sugars, nucleosides, nucleotides or nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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

Definitions

  • the present invention relates to a nucleic acid complex having blood-brain barrier penetrating ability and a pharmaceutical composition for preventing or treating degenerative brain disease containing the complex as an active ingredient, and more specifically, to a bioactive nucleic acid targeting NLRP3 gene; And a carrier peptide nucleic acid (Carrier Peptide Nucleic Acid) relates to a complementary nucleic acid complex and a pharmaceutical composition for preventing or treating degenerative brain disease containing the same as an active ingredient.
  • Carrier Peptide Nucleic Acid Carrier Peptide Nucleic Acid
  • Parkinson's disease is a degenerative brain disease caused by a decrease in dopamine, a neurotransmitter, in the striatum and substantia nigra. .
  • the prevalence is second only to Alzheimer's disease, and it is reported that about 1% of the population aged 60 or older suffer from this disease (Ali Samii, et al., Lancet. 2004 May 29; 363(9423):1783-93).
  • Parkinson's disease is characterized by the loss of dopaminergic neurons due to the accumulation and diffusion of Lewy bodies composed of misfolded ⁇ -synuclein. .
  • Lewy bodies which are the main cause of Parkinson's disease, are neurotoxic substances that propagate to peripheral areas of the brain and cause dopaminergic degeneration (Werner Poewe, et al., Nat Rev Dis Primers. 2017 Mar 23;3:17013). It is known that chronic microglia neuroinflammation occurs in the substantia nigra region of patients with Parkinson's disease in the early stages of onset, and this feature is also known to be prominent in the brains of patients with Parkinson's disease confirmed postmortem (Alexander Gerhard, et al., Neurobiol Dis.
  • NLRP3 NLR family pyrin domain containing 3; NACHT, leucine-rich repeat, and pyrin domain (PYD)-containing protein 3 (NALP3)
  • NACHT nucleotide-binding and oligomerization domain-like receptors
  • PYD pyrin domain
  • NALP3 pyrin domain-containing protein 3
  • the NLRP3 inflammasome is a NLRP3 sensor and signal adapter ASC (apoptosis-associated speck-like protein containing a CARD (caspase activation and recruitment) domain)) and pro-caspase-1.
  • ASC apoptosis-associated speck-like protein containing a CARD (caspase activation and recruitment) domain
  • caspase-1 is activated, triggering the release of interleukin-1 ⁇ (IL-1 ⁇ ) and IL-18, which are inflammatory cytokines, to initiate an inflammatory response.
  • alpha-synuclein aggregation activates the NLRP3 inflammasome and induces dopamine degeneration due to an increase in the inflammatory response of microglia (Richard Gordon, et al., Sci Transl Med. 2018 Oct. 31;10(465):eaah4066).
  • a significant number of drugs developed for the treatment of brain diseases have a problem in that they do not pass through the blood-brain barrier well.
  • the penetration mechanism of the blood-brain barrier has not yet been elucidated, and even if a disorder occurs in the central nervous system, it is a reality that drugs cannot reach the target area of the central nervous system, and effective treatment methods have not yet been developed.
  • nucleic acid drugs suppress the expression of target-specific messenger RNA (mRNA), so it is possible to address research areas that could not be treated with existing protein-targeting drugs (Ryszard Kole, et al. al., Nat Rev Drug Discov. 2012 Jan 20;11(2):125-40). Due to its performance and advantages as a drug, various clinical trials using nucleic acids are in progress, and despite the increasing use of nucleic acid-based therapeutics, the use of carriers for intracellular introduction or blood-brain barrier penetration is extremely limited.
  • mRNA messenger RNA
  • the present inventors have found that a nucleic acid complex in which a bioactive nucleic acid and a carrier peptide nucleic acid modified to have a positive charge as a whole are complementarily coupled have surprisingly improved cell permeability, and using this It was confirmed that the expression of the target gene can be controlled very efficiently, and a patent has been registered for a new structure with low cytotoxicity and improved cell permeability of bioactive nucleic acids and the ability to regulate gene expression (Korean Registered Patent No. 10). -1963885). In addition, the present inventors have continuously conducted research on the function of the construct to develop a new construct having the ability to penetrate the blood-brain barrier with excellent efficiency (Korean Patent Application No. 10-2019-0128465).
  • the present inventors have made diligent efforts to use nucleic acid complexes with excellent blood-brain barrier penetration ability to apply them to the treatment of degenerative brain diseases. It was found that it exhibits excellent effects in preventing or treating brain diseases, and the present invention was completed.
  • An object of the present invention is to provide a nucleic acid complex having blood-brain barrier penetrating ability and a pharmaceutical composition for preventing or treating degenerative brain disease containing the same as an active ingredient.
  • the present invention is a bioactive nucleic acid targeting the NLRP3 gene (Bioactive Nucleic Acid); And a carrier peptide nucleic acid (Carrier Peptide Nucleic Acid) provides a complementary nucleic acid complex.
  • the present invention also provides a pharmaceutical composition for preventing or treating degenerative brain disease containing the nucleic acid complex as an active ingredient.
  • the present invention also provides a method for preventing or treating a degenerative brain disease comprising administering the nucleic acid complex.
  • the present invention also provides the use of the nucleic acid complex for the prevention or treatment of degenerative brain diseases.
  • the present invention also provides the use of the nucleic acid complex for the preparation of a drug for preventing or treating degenerative brain disease.
  • 1 is a diagram confirming the ability of nucleic acid complexes to inhibit the expression of target genes and subgenes in an LPS-induced Parkinson's disease-like cell model.
  • FIG. 2 is a diagram confirming the ability of a nucleic acid complex to suppress intracellular expression of a target gene in a Parkinson's disease-like cell model induced by LPS.
  • Figure 3 is a diagram confirming the efficacy of the nucleic acid complex in a primary microglia model isolated from tissue.
  • Figure 4 is a view confirming the ability to improve animal behavior according to the administration of the nucleic acid complex in the MPTP/P Parkinson's disease animal model.
  • 5 is a diagram confirming the ability to inhibit target gene expression and subgene expression by brain region according to the administration of the nucleic acid complex in the MPTP/P Parkinson's disease animal model.
  • 6a is a view showing changes in expression of Tyrosine Hydroxylase (TH) in the striatum of the brain according to the administration of the nucleic acid complex in the MPTP/P Parkinson's disease animal model.
  • TH Tyrosine Hydroxylase
  • 6B is a diagram showing changes in expression of Tyrosine Hydroxylase (TH) in the substantia nigra of the brain according to administration of the nucleic acid complex in the MPTP/P Parkinson's disease animal model.
  • TH Tyrosine Hydroxylase
  • Figure 6c is a view confirming the expression suppression ability of ⁇ -Synuclein in the striatum (striatum) of the brain region according to the administration of the nucleic acid complex in the MPTP/P Parkinson's disease animal model.
  • Figure 6d is a view confirming the expression suppression ability of ⁇ -Synuclein in the substantia nigra of the brain region according to the administration of the nucleic acid complex in the MPTP/P Parkinson's disease animal model.
  • Figure 6e is a view confirming the expression inhibition ability of ionized calcium-binding adapter molecule 1 (IBA-1) in the striatum (striatum) of the brain region according to the administration of the nucleic acid complex in the MPTP/P Parkinson's disease animal model.
  • IBA-1 ionized calcium-binding adapter molecule 1
  • 6f is a diagram confirming the expression inhibition ability of ionized calcium-binding adapter molecule 1 (IBA-1) in the substantia nigra of the brain according to the administration of the nucleic acid complex in the MPTP/P Parkinson's disease animal model.
  • IBA-1 ionized calcium-binding adapter molecule 1
  • Parkinson's disease causes of Parkinson's disease are very diverse. About 5% of all patients are caused by genetic factors, and external environmental factors such as inflammation and oxidative stress play an important role. Parkinson's disease is caused by abnormal protein aggregation in dopaminergic neurons, which is the aggregation of alpha-synuclein protein. Aggregation of these proteins is further promoted by oxidative stress. Until now, the treatment of Parkinson's disease has been made by supplementing the dopamine neurotransmitter, but this is not a fundamental treatment for Parkinson's disease.
  • the number of patients with Parkinson's disease in Korea was 61,565 in 2010 and increased to 85,888 in 2014 with an average annual growth rate of 8.7%.
  • the proportion of patients aged 60 years or older was 95.7%, and the prevalence showed a correlation with the patient's age. Parkinson's disease patient and treatment cost increase, young doctors, 2015).
  • the onset of Parkinson's disease is very early, around 60 years of age, and because of this, the progression of the disease lasts for more than 20 years, so a treatment that slows the disease progression through early detection is actively required.
  • nucleic acid complex in which a bioactive nucleic acid targeting the NLRP3 gene and a carrier peptide nucleic acid are complementaryly bound can be used for the prevention and treatment of Parkinson's disease.
  • the present invention in one aspect, a bioactive nucleic acid targeting the NLRP3 gene (Bioactive Nucleic Acid); and a nucleic acid complex in which a carrier peptide nucleic acid is complementarily bound.
  • a nucleic acid complex in which a bioactive nucleic acid targeting the NLRP3 gene and a carrier peptide are complementaryly bound may have a structure of the following structural formula (1).
  • A is a bioactive nucleic acid having a sequence capable of binding to a gene of interest
  • C is a carrier peptide nucleic acid capable of binding to a bioactive nucleic acid
  • means a complementary bond between a bioactive nucleic acid and a carrier peptide nucleic acid
  • the bioactive nucleic acid represented by A has an overall negative charge or neutral charge
  • the carrier peptide nucleic acid includes one or more peptide nucleic acid monomers modified to have a positive charge throughout the carrier peptide nucleic acid.
  • the bioactive nucleic acid and the carrier peptide nucleic acid in the nucleic acid complex according to the present invention may have anti-parallel binding or parallel binding.
  • the complementary binding form of the nucleic acid can be separated in the presence of a target sequence of the bioactive nucleic acid (a sequence complementary to the bioactive nucleic acid).
  • Bioactive Nucleic Acid binds to a target gene and a nucleotide sequence containing the target gene in vitro or in vivo to determine the unique function of the gene (e.g., For example, activating or inhibiting transcript expression or protein expression), or regulating pre-mRNA splicing (eg, exon skipping), etc.
  • the nucleotide sequence may be characterized as a gene regulatory sequence, a gene coding sequence, or a splicing regulatory sequence.
  • the bioactive nucleic acid is expressed
  • a nucleic acid having a complementary sequence capable of binding to a target gene of interest to be reduced in particular, a complementary sequence capable of binding to the mRNA of such a target gene of interest, and suppressing the expression of the gene. It means a nucleic acid involved in regulation, and may be a nucleic acid having a sequence complementary to a target gene whose expression is to be reduced.
  • the bioactive nucleic acid in the present invention is preferably an antisense peptide nucleic acid of the NLRP3 (NLR family pyrin domain containing 3) gene, a target gene related to Parkinson's disease, and more preferably the nucleotide sequence represented by the sequence of SEQ ID NO: 2 It may include, but is not limited thereto.
  • the bioactive nucleic acids include DNA, RNA, or modified nucleic acids such as PNA (peptide nucleic acid), PMO (phosphorodiamidate morpholino oligonucleotide), LNA (locked nucleic acid), GNA (glycol nucleic acid) and TNA (threose nucleic acid), antisense It may be selected from the group consisting of oligonucleotide, aptamer, small interfering RNA (siRNA), short hairpin RNA (shRNA), ribozyme and DNAzyme, preferably the bioactive
  • the nucleic acid is selected from the group consisting of DNA, RNA, or modified nucleic acid PNA (peptide nucleic acid), PMO (phosphorodiamidate morpholino oligonucleotide), LNA (locked nucleic acid), GNA (glycol nucleic acid) and TNA (threose nucleic acid) it may be selected from the group consisting of DNA, RNA
  • Carrier Peptide Nucleic Acid refers to a nucleic acid to which a bioactive nucleic acid and some or all bases are complementaryly combined to impart functionality, and the carrier peptide nucleic acid used in the present invention is In addition to peptide nucleic acid (PNA), modified nucleic acids similar thereto may be used, and peptide nucleic acids are preferred, but are not limited thereto.
  • PNA peptide nucleic acid
  • the carrier peptide nucleic acid preferably includes one or more gamma- or alpha-backbone modified peptide nucleic acid monomers so that the entire carrier peptide nucleic acid is positively charged, and the gamma- or alpha-backbone modified peptide nucleic acid It is more preferable that monomers having amino acids having a positive charge are included more than monomers having amino acids having a negative charge so that the overall charge of the carrier peptide nucleic acid is positive.
  • the carrier peptide nucleic acid preferably includes the nucleotide sequence represented by SEQ ID NO: 4 or SEQ ID NO: 5, but is not limited thereto.
  • the "nucleic acid complex” can penetrate the bioactive substance into the body and ultimately into the cell through extracellular treatment, and specifically has the ability to deliver the bioactive nucleic acid targeting the NLRP3 gene into the cell. .
  • the nucleic acid complex is a bioactive nucleic acid represented by the sequence of SEQ ID NO: 2; and a carrier peptide nucleic acid represented by the sequence of SEQ ID NO: 4 or 5, but is not limited thereto.
  • the binding ability (melting temperature, Tm) of the bioactive nucleic acid targeting the NLRP3 gene and the carrier peptide nucleic acid is lower than that of the bioactive nucleic acid and the target NLRP3 gene.
  • the binding force is obtained by parallel binding or partial specific binding between the bioactive nucleic acid and the carrier peptide nucleic acid according to the 5'-direction and the 3'-direction of each nucleic acid, so that the bioactive nucleic acid and the carrier peptide nucleic acid
  • the binding force (Tm) of may be lower than the binding force between the bioactive nucleic acid and the target gene of the bioactive nucleic acid.
  • the bioactive nucleic acid or carrier peptide nucleic acid may be characterized by additionally binding a substance that helps endosome escape to the 5'-end or 3'-end of each nucleic acid. That is, it may be characterized in that it has a structure of the following Structural Formula (2) by further including a material that helps the endosome escape of the bioactive nucleic acid and the carrier peptide nucleic acid.
  • 'm' means a substance that helps endosome escape of bioactive nucleic acid and carrier peptide nucleic acid.
  • “substances that help endosomes escape” can be characterized in that they help the escape of bioactive nucleic acids from endosomes by increasing the osmotic pressure inside the endosomes or by destabilizing the membranes of endosomes. there is. It means that bioactive nucleic acids move more efficiently and rapidly to the nucleus or cytoplasm to meet and act on target genes (Daniel W Pack, et al., Nat Rev Drug Discov. 2005 Jul;4(7):581-93 ).
  • the material that helps the endosome escape is a peptide, lipid nanoparticles, conjugate nanoparticles (polyplex nanoparticles), polymer nanospheres (polymer nanospheres), inorganic nanomaterials (inorganic nanoparticles), cationic lipids It may be characterized in that at least one selected from the group consisting of nanomaterials (cationic lipid-based nanoparticles), cationic polymers (cationic polymers) and pH sensitive polymers (pH sensitive polymers).
  • a peptide (GLFDIIKKIAESF, SEQ ID NO: 6) may be linked to the bioactive nucleic acid via a linker, and Histidine (10) to the carrier peptide nucleic acid via a linker. It may be characterized by combining, but is not limited thereto.
  • the lipid nanoparticles may be selected from the group consisting of Lipid, phospholipids, acetyl palmitate, poloxamer 18, Tween 85, tristearin glyceride and Tween 80.
  • the polyplex nanoparticles may be poly(amidoamine) or polyethylenimine (PEI).
  • the polymer nanospheres are selected from the group consisting of polycaprolactone, poly(lactide-co-glycolide, polylactide, polyglycolide, poly(d,l-lactide), chitosan, and PLGA-polyethylene glycol. can be characterized.
  • the inorganic nanoparticles may be selected from the group consisting of Fe2O3, Fe3O4, WO3 and WO2.9.
  • the cationic lipid-based nanoparticles are 1- (aminoethyl) iminobis [N- (oleicylcysteinyl-1-amino-ethyl) propionamide], N-alkylated derivative of PTA and 3, 5- It may be characterized in that it is selected from the group consisting of didodecyloxybenzamidine.
  • the cationic polymer may be selected from the group consisting of vinylpyrrolidone-N, N-dimethylaminoethyl methacrylate acid copolymer diethyl sulphate, polyisobutylene and poly(N-vinylcarbazole).
  • the pH sensitive polymers may be selected from the group consisting of polyacids, poly(acrylic acid), poly(methacrylic acid), and hydrolyzed polyacrylamide.
  • the bioactive nucleic acid and the carrier peptide nucleic acid each comprise 2 to 50, preferably 5 to 30, more preferably 10 to 25, most preferably 15 to 17 nucleic acid monomers. that can be characterized.
  • the bioactive nucleic acid may be characterized in that it consists of natural nucleic acid bases and/or modified nucleic acid monomers.
  • the monomer used for the bioactive nucleic acid is PNA, it is referred to as a bioactive peptide nucleic acid, and when other monomers are used, it is referred to in the same way.
  • the bioactive nucleic acid and the carrier peptide nucleic acid are phosphodiester, 2' O-methyl, 2' methoxy-ethyl, phosphor It may be characterized by further comprising at least one functional group selected from the group consisting of amidate, methylphosphonate, and phosphorothioate.
  • the carrier peptide nucleic acid may be characterized in that a part or all of the base sequence of the bioactive nucleic acid is composed of a complementary sequence.
  • the carrier peptide nucleic acid may include one or more universal bases, and all of the carrier peptide nucleic acids may consist of universal bases.
  • each of the bioactive nucleic acid and the carrier peptide nucleic acid in the nucleic acid complex may be a complex characterized by having a positive charge (positive), negative charge (negative) or neutral charge as a whole.
  • the meaning of "overall” means the electrical properties of the entire bioactive nucleic acid or carrier peptide nucleic acid as a whole, not the electrical properties of individual bases, when viewed from the outside. Even if some of the monomers in the sexual nucleic acid have a positive charge, if there are more monomers with a negative charge, the bioactive nucleic acid will have a negative charge when looking at the electrical properties “as a whole”, and some bases and/or Even if the backbone has a negative charge, when a larger number of bases and/or backbones having a positive charge are present, the carrier peptide nucleic acid has a positive charge when looking at the electrical characteristics “as a whole”.
  • the nucleic acid complex of the present invention may be characterized as having a positive charge as a whole.
  • the bioactive nucleic acid has negative or neutral electrical characteristics as a whole
  • the carrier peptide nucleic acid has positive electrical characteristics as a whole. It is not limited thereto.
  • the electrical properties of the bioactive nucleic acid and the carrier peptide nucleic acid can be imparted using a modified peptide nucleic acid monomer
  • the modified peptide nucleic acid monomer is a carrier peptide nucleic acid having a positive charge, such as lysine (Lysine, Lys, K) , arginine (Arg, R), histidine (Histidine, His, H), diamino butyric acid (DAB), ornithine (Orn), and any one or more positive charges selected from the group consisting of amino acid analogs
  • a carrier peptide nucleic acid that includes amino acids of and has a negative charge, and may be characterized in that it includes a negatively charged amino acid such as glutamic acid (Glu, E) or an amino acid analog that is negatively charged.
  • the carrier peptide nucleic acid may be characterized by including one or more gamma- or alpha-backbone modified peptide nucleic acid monomers so as to have a positive charge as a whole.
  • the gamma- or alpha-backbone modified peptide nucleic acid monomer has lysine (Lysine, Lys, K), arginine (Arginine, Arg, R), histidine (His, H), diamino butyric acid (Diamino butyric acid) to have an electrical positivity. acid, DAB), ornithine (Orn), and amino acids having at least one positive charge selected from the group consisting of amino acid analogs.
  • the modification of the peptide nucleic acid monomer for charge imparting may use a peptide nucleic acid monomer having a modified nucleobase in addition to the backbone modification.
  • a peptide nucleic acid monomer having a modified nucleobase in addition to the backbone modification.
  • an amine, triazole, or imidazole moiety may be included in the nucleobase to have an electronegative property, or a carboxylic acid may be included in the base to have an electronegative property.
  • the modified peptide nucleic acid monomer of the carrier peptide nucleic acid may further contain a negative charge in the backbone or nucleobase, but the modified peptide nucleic acid monomer contains more positively charged monomers than monomers having negative charges, so that the carrier peptide as a whole is formed. It is preferred that the charge of the nucleic acid be positive.
  • the nucleic acid complex according to the present invention has a positive charge as a whole.
  • At least one material selected from the group consisting of a hydrophobic moiety, a hydrophilic moiety, a target antigen-specific antibody, an aptamer, or a fluorescent/luminescent marker is a bioactive nucleic acid And / or may be characterized in that it is bound to a carrier peptide nucleic acid, preferably the hydrophobic moiety, the hydrophilic moiety, a target antigen-specific antibody, an aptamer, and a fluorescent / luminescent marker for imaging
  • a carrier peptide nucleic acid preferably the hydrophobic moiety, the hydrophilic moiety, a target antigen-specific antibody, an aptamer, and a fluorescent / luminescent marker for imaging
  • One or more substances selected from the group consisting of and the like may be bound to the carrier peptide nucleic acid.
  • the binding of the carrier peptide nucleic acid may be characterized as a simple covalent bond or a covalent bond mediated by a linker, but is not limited thereto.
  • substances related to cell permeation, solubility, stability, delivery and imaging eg, hydrophobic residues, etc. bound to the nucleic acid carrier exist independently of the bioactive nucleic acid that regulates the expression of the target gene.
  • the complementary binding form of the bioactive nucleic acid and the carrier peptide nucleic acid may be largely characterized by having the form of antiparallel binding and parallel binding.
  • the complementary binding form has a structure that separates in the presence of a target sequence of a bioactive nucleic acid (a sequence complementary to the bioactive nucleic acid).
  • the antiparallel bond and the parallel bond are determined according to the 5'-direction and the 3'-direction in the binding method of DNA-DNA or DNA-PNA.
  • Antiparallel coupling is a general DNA-DNA or DNA-PNA coupling method.
  • the bioactive nucleic acid is in the 5' to 3' direction and the carrier peptide nucleic acid is in the 3' to 5' direction. It means that the shape is connected to each other in the direction.
  • Parallel binding is a form in which binding force is somewhat lower than that of anti-parallel binding, and refers to a form in which both the bioactive nucleic acid and the carrier peptide nucleic acid are bonded to each other in the 5' to 3' direction or the 3' to 5' direction.
  • the binding force of the bioactive nucleic acid and the carrier peptide nucleic acid is lower than the binding force of the bioactive nucleic acid and the target gene, particularly the mRNA of the target gene.
  • the binding force is determined by melting temperature, melting temperature or Tm.
  • the above Parallel binding or partial specific binding between the bioactive nucleic acid and the carrier peptide nucleic acid may be characterized, but is not limited thereto.
  • the carrier peptide nucleic acid has at least one peptide nucleic acid base selected from the group consisting of a linker, a universal base, and a peptide nucleic acid base having a base that is not complementary to a corresponding base of a bioactive nucleic acid. It can be, but is not limited thereto.
  • the universal base binds to natural bases such as adenine, guanine, cytosine, thymine, and uracil without selectivity, and is complementary to
  • One or more bases selected from the group consisting of inosine PNA, indole PNA, nitroindole PNA, and abasic can be used as a base having a lower binding force than the binding force, preferably. It can be characterized by using inosine PNA.
  • a combination of the binding form and electrical properties of nucleic acids for function control of the nucleic acid complex is provided, and the combination of the binding form and electrical properties of the nucleic acids controls the particle size and the time point of action, cell permeability, solubility and specificity It can be characterized as improving the degree.
  • the binding force of the bioactive nucleic acid and the carrier peptide nucleic acid is controlled, in the presence of the target gene, when the bioactive nucleic acid binds to the target sequence (when the bioactive nucleic acid is substituted with the target sequence, the target specific It is possible to adjust the timing of enemy separation and joining).
  • the control of the strand displacement time point and the target specific release and bind time point of the bioactive nucleic acid into the target gene is a carrier peptide for non-specific binding of the complex It can be characterized in that it can be controlled by the presence, number, and location of non-specific bases, universal bases, and linkers in nucleic acids. It may be characterized in that control is possible by a combination of the above conditions, such as a parallel or antiparallel bond, which is a form of complementary bond of the peptide complex.
  • the particle size of the nucleic acid complex may be characterized in that it is controlled by adjusting the charge balance between the bioactive nucleic acid and the carrier peptide nucleic acid. Specifically, when the positive charge of the carrier peptide nucleic acid increases, the particle size decreases, but when the positive charge of the carrier peptide nucleic acid exceeds a certain level, the particle size increases.
  • the particle size is determined by an appropriate charge balance with the overall carrier peptide nucleic acid according to the charge of the bioactive nucleic acid forming the complex as another important factor determining the particle size.
  • the positive charge of the carrier peptide nucleic acid according to the present invention is 1 to 7 (meaning that 1 to 7 monomers having a positive charge are included), preferably 2 to 5, most preferably 2 to 3
  • the charge of the bioactive nucleic acid may be characterized in that the net charge of the charge balance is 0 to 5 negative charges, preferably 0 to 3.
  • the nucleic acid complex can be prepared by hybridizing a bioactive nucleic acid and a carrier peptide nucleic acid under appropriate conditions.
  • Hybridization in the present invention means that complementary single-stranded nucleic acids form a double-stranded nucleic acid. Hybridization can occur when the complementarity between the two nucleic acid strands is perfect (perfect match) or even when some mismatch bases are present. The degree of complementarity required for hybridization may vary depending on hybridization conditions, and may be particularly controlled by binding temperature.
  • the nucleic acid complex may have blood-brain barrier penetrating ability.
  • blood-brain barrier or "BBB (Blood-Brain Barrier)” is used interchangeably herein, which closely regulates and severely restricts the exchange between blood and brain tissue and is circulated through brain tissue. Therefore, it is used to refer to the permeability barrier present in the blood.
  • Components of the blood brain barrier include the endothelial cells that form the deepest lining of all blood vessels, the dense junctions between adjacent endothelial cells that are structurally correlated with the BBB, the basement membrane of endothelial cells, and almost all of the exposed outer surface of blood vessels. An enlarged foot process of the overlying astrocytes is included.
  • the BBB prevents most substances in the blood, including most large molecules, such as Ig, antibodies, complement, albumin, and drugs and small molecules, from entering brain tissue.
  • the nucleic acid complex suggests that it has BBB penetrating ability.
  • target gene refers to a nucleic acid sequence (base sequence) to be activated, inhibited, or labeled, and is not different from the term “target nucleic acid”, and is used interchangeably herein.
  • bioactive nucleic acid when a target nucleic acid (base sequence) containing a target gene is contacted (bound) with the complex in vitro or in vivo , bioactive nucleic acid is separated from the carrier peptide nucleic acid and exhibit biological activity.
  • diseases that can be prevented or treated using the nucleic acid complex may be determined according to the target gene of the bioactive nucleic acid in the nucleic acid complex.
  • the target gene of the bioactive nucleic acid may be characterized in that NLRP3.
  • the present invention from another point of view, a bioactive nucleic acid targeting the NLRP3 gene (Bioactive Nucleic Acid); And a pharmaceutical composition for preventing or treating degenerative brain disease containing a nucleic acid complex complementary to a carrier peptide nucleic acid as an active ingredient.
  • a bioactive nucleic acid targeting the NLRP3 gene Bioactive Nucleic Acid
  • a pharmaceutical composition for preventing or treating degenerative brain disease containing a nucleic acid complex complementary to a carrier peptide nucleic acid as an active ingredient.
  • the disease that can be prevented and treated using the nucleic acid complex may preferably be a degenerative brain disease
  • the degenerative brain disease includes Parkinson's disease, Alzheimer's disease, Niemann-Pick disease, Creutzfeldt-Jakob disease, Huntington's disease, amyotrophic lateral sclerosis (amyotrophic lateral sclerosis), multiple sclerosis or dementia ( dementia), but is not limited thereto.
  • prevention refers to any action that prevents the onset of a disease or delays its progression by administering a composition containing the nucleic acid complex.
  • treatment used in the present invention refers to all activities in which symptoms of a disease are improved, symptoms are alleviated, or cured by administration of a composition containing the nucleic acid complex.
  • a pharmaceutical composition comprising a nucleic acid complex according to the present invention may include a pharmaceutically effective amount of the nucleic acid complex alone, or may include one or more pharmaceutically acceptable carriers, excipients or diluents.
  • the pharmaceutically effective amount refers to an amount sufficient to prevent, improve, and treat symptoms of degenerative brain disease.
  • pharmaceutically acceptable refers to a composition that is physiologically acceptable and does not usually cause allergic reactions such as gastrointestinal disorders and dizziness or similar reactions when administered to humans.
  • the carrier, excipient and diluent include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia gum, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, polyvinylpyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil.
  • the pharmaceutical composition may further include fillers, anti-coagulants, lubricants, wetting agents, flavoring agents, emulsifiers and preservatives.
  • carrier is defined as a compound that facilitates the addition of a nucleic acid complex into a cell or tissue.
  • DMSO dimethylsulfoxide
  • carrier facilitates the introduction of many organic compounds into the cells or tissues of living organisms.
  • diot is defined as a compound that is diluted in water which will dissolve the compound as well as stabilize the biologically active form of the subject compound. Salts dissolved in buffer solutions are used as diluents in the art. A commonly used buffer solution is phosphate buffered saline because it mimics the salt state of human solutions. Because buffer salts can control the pH of a solution at low concentrations, buffer diluents rarely modify the biological activity of a compound.
  • a substance containing a nucleic acid complex in the present invention can be administered to a patient by itself or as a mixed pharmaceutical composition together with other active ingredients, such as in combination therapy, or with suitable carriers or excipients.
  • compositions of the present invention may be formulated using methods known in the art to provide rapid, sustained or delayed release of the active ingredient after administration.
  • the dosage form may be in the form of a powder, granule, tablet, emulsion, syrup, aerosol, soft or hard gelatin capsule, sterile injectable solution, or sterile powder.
  • composition of the present invention can be administered through various routes including oral, transdermal, subcutaneous, intravenous or intramuscular, and the dosage of the active ingredient depends on various factors such as the route of administration, age, sex, weight and severity of the patient. can be selected appropriately.
  • compositions suitable for use in the present invention include compositions in which the active ingredients are contained in effective amounts to achieve their intended purpose. More specifically, a therapeutically effective amount refers to an amount of a compound effective to prolong the survival of the subject being treated or to prevent, alleviate or ameliorate symptoms of a disease. Determination of a therapeutically effective amount is well within the ability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • any nucleic acid delivery method known in the art may be used.
  • suitable delivery reagents include, for example, Mirus Transit TKO lipophilic reagent, lipofectin, lipofectamine, cellfectin, polycations (eg, polylysine), atelocollagen, nanoplexes, and liposomes.
  • atelocollagen as a delivery vehicle for nucleic acid molecules has been described by Minakuchi et al. Nucleic Acids Res., 32(13):e109 (2004); Hanai et al. Ann NY Acad Sci., 1082:9-17 (2006); and Kawata et al. Mol Cancer Ther., 7(9):2904-12 (2008).
  • Exemplary interfering nucleic acid delivery systems are provided in U.S. Patent Nos. 8,283,461, 8,313,772, and 8,501,930.
  • the nucleic acid complex may be administered using a delivery system such as a liposome.
  • a delivery system such as a liposome.
  • the liposome can help target the complex to a specific tissue, such as lymphoid tissue, or selectively target infected cells, and can also help increase the half-life of a composition containing the complex.
  • Liposomes include emulsions, foams, micelles, insoluble monolayers, liquid crystals, phospholipid dispersions, lamellar layers, and the like.
  • the complex to be delivered is a liposome.
  • a molecule that binds to a receptor prevalent in lymphocytes such as a monoclonal antibody that binds to the CD45 antigen, or in combination with other therapeutic compositions, is a liposome.
  • liposomes filled or decorated with a given complex of the present invention to deliver the nucleic acid complex composition can be directed to the site of lymphocytes.
  • Liposomes for use in accordance with the present invention are generally formed from standard vesicle-forming lipids, including neutral and negatively charged phospholipids and sterols such as cholesterol.
  • lipids are selected in consideration of, for example, stability of liposomes in the blood stream, acid lability, and size of liposomes.
  • a variety of methods can be used to prepare liposomes. See, eg, Szoka, et al., Ann. Rev. Biophys. Bioeng., 9:467, 1980), and US Pat. Nos. 4,235,871, 4,501,728, 4,837,028 and 5,019,369.
  • the present invention provides a bioactive nucleic acid targeting the NLRP3 gene; And a carrier peptide nucleic acid (Carrier Peptide Nucleic Acid) It relates to a method for preventing or treating degenerative brain disease comprising administering to a subject a nucleic acid complex complementary thereto.
  • the present invention provides a bioactive nucleic acid targeting the NLRP3 gene for the prevention or treatment of degenerative brain diseases; And it relates to the use of a nucleic acid complex in which a carrier peptide nucleic acid is complementarily bound.
  • the present invention provides a bioactive nucleic acid targeting the NLRP3 gene for the preparation of drugs for preventing or treating degenerative brain diseases; And it relates to the use of a nucleic acid complex in which a carrier peptide nucleic acid is complementaryly bound.
  • subject means a mammal suffering from or at risk of a condition or disease that can be alleviated, suppressed or treated by administering a nucleic acid complex according to the present invention, and preferably means a human.
  • the dose of the nucleic acid complex of the present invention to the human body may vary depending on the patient's age, weight, sex, dosage form, health condition and disease severity.
  • Toxicity and therapeutic efficacy of compositions comprising the nucleic acid complexes described herein can be measured by, for example, the LD50 (lethal dose to 50% of the population), the ED50 (the dose that is therapeutically effective in 50% of the population), the IC50 It can be estimated by standard pharmaceutical procedures in cell culture or laboratory animals to determine (the dose that has a therapeutically inhibitory effect on 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between the LD50 and the ED50 (or IC50).
  • Compounds exhibiting high therapeutic indices are preferred. Data obtained from these cell culture assays can be used to estimate a range of human doses.
  • the dosage or applied amount of such compounds lies preferably within a range of circulating concentrations that include the ED50 (or IC50) with little or no toxicity.
  • administration refers to the act of introducing the pharmaceutical composition of the present invention to a subject by any appropriate method, and the route of administration may be administered through various oral or parenteral routes as long as it can reach the target tissue. there is.
  • the administration route of the pharmaceutical composition of the present invention may be administered through any general route as long as it can reach the target tissue.
  • the pharmaceutical composition of the present invention is not particularly limited thereto, but may be administered intraperitoneally, intravenously, intramuscularly, subcutaneously, intradermally, orally, intranasally, intrapulmonaryly, or intrarectally, as desired.
  • the composition may be administered by any device capable of transporting an active substance to a target cell.
  • the pharmaceutical composition of the present invention may be administered as an individual therapeutic agent or in combination with other therapeutic agents, and may be administered sequentially or simultaneously with conventional therapeutic agents. And it can be single or multiple administrations. It is important to administer the amount that can obtain the maximum effect with the minimum amount without side effects in consideration of all the above factors.
  • Example 1 Preparation of bioactive peptide nucleic acids and carrier peptide nucleic acids, and complexes using them
  • NLRP3 was used as a target gene to test the effect of the nucleic acid complex on Parkinson's disease, and to confirm the therapeutic effect of Parkinson's disease, antisense peptide nucleic acid (Bioactive Peptide Nucleic Acid) for NLRP3 was used. antisense PNA) was used.
  • the bioactive nucleic acid (antisense PNA) used as a control of the present invention includes the sequence represented by SEQ ID NO: 1, and the bioactive peptide nucleic acid (antisense PNA) used to confirm the therapeutic effect of Parkinson's disease has the sequence represented by SEQ ID NO: 2 includes
  • the carrier peptide nucleic acids used in the examples of the present invention are composed of sequences represented by SEQ ID NOs: 3 to 5 (Table 1). All peptide nucleic acids used in the present invention were synthesized by HPLC purification method from PANAGENE (Korea).
  • Modification of the monomers converts the backbone of the peptide nucleic acid to lysine (indicated by Lysine, Lys, K, (+) ) for electrical properties and glutamic acid (Glutamic acid, Glu, E, (-)) for electrical negative to impart electrical properties. ) was constructed to have a modified peptide backbone.
  • Each combination of the bioactive nucleic acid and the carrier peptide nucleic acid was hybridized in the presence of DMSO, and as a result, a complex composed of the bioactive nucleic acid and the carrier peptide nucleic acid was prepared.
  • Example 1 the therapeutic effect of Parkinson's disease was analyzed using a nucleic acid complex comprising a carrier peptide nucleic acid and a bioactive peptide nucleic acid targeting NLRP3, which was prepared according to the structure shown in Table 2 below.
  • Example 2-1 Cell culture
  • HMC-3 Human-derived microglia obtained from ATCC (American Type Culture Collection, USA) were mixed with 10% (v/v) fetal bovine serum, penicillin 100 units/ml, and streptomycin in MEM culture medium (Wellgene, Korea). 100 ⁇ g/ml was added and cultured under conditions of 37°C and 5% (v/v) CO 2 .
  • Example 2-2 Analysis of gene expression using Western blot assay
  • Human-derived microglial cell lines were seeded in 1x10 5 cells in a 6-well plate and cultured for 24 hours, and treated with 1 ⁇ g/ml of Lipopolysaccharide (LPS, Sigma, USA) to induce an immune response. After 4 hours of LPS treatment, the experiment was conducted under the conditions of Example 2-1, and the complex containing the bioactive peptide nucleic acid and the carrier peptide nucleic acid was treated and cultured for 24, 48, 72, 96, and 120 hours, respectively, and RIPA buffer Protein lysate was obtained by adding 30 ⁇ L to each well.
  • LPS Lipopolysaccharide
  • Protein lysate was quantified using a BCA assay kit (Thermo Fisher, USA), and 30 ⁇ g of protein was separated by size through electrophoresis, and the protein was transferred to a PVDF membrane.
  • Pro Cas-1 (abcam, USA) and Pro IL-1 ⁇ (abcam, USA) were treated at a ratio of 1:1000 and left at 4°C for one day. It was washed with 1X TBS-T, treated with a secondary antibody, Goat Anti-Rabbit (Cell signaling Technology, USA) at a ratio of 1:2000, and left at room temperature for 1 hour.
  • Supersignal TM West Femto Maximum Sensitivity Substrate was treated, and the expression inhibition efficiency of the target gene was analyzed using Image600 (Amersham, Germany) equipment.
  • Human-derived microglial cell lines were seeded in 3x10 3 cells in an 8-well plate and cultured for 24 hours, and treated with 1 ⁇ g/ml of Lipopolysaccharide (LPS, Sigma, USA) to induce an immune response. After 4 hours of LPS treatment, the experiment was conducted under the conditions of Example 2-1, and the complex containing the bioactive peptide nucleic acid and the carrier peptide nucleic acid was treated, cultured for 24 and 72 hours, respectively, and cells were treated with 4% paraformaldehyde (Sigma, USA). was fixed.
  • LPS Lipopolysaccharide
  • the fixed cells were permeabilized with 0.1% Triton X-100 (Sigma, USA) dissolved in PBS for 10 minutes, blocked with 1% bovine serum albumin (BSA, Sigma, USA) for 1 hour, and then The antibody, NLRP3 (ABclonal, USA) was treated at a ratio of 1:100 and left at 4° C. for one day. After washing with 1X PBS, the secondary antibody, Alexa 488-goat Anti-Mouse (Abcam Technology, USA) was treated at a ratio of 1:200 and allowed to stand at room temperature for 1 hour. After diluting the DAPI solution (Sigma, USA) to 1x and leaving it for 10 minutes, the expression inhibition efficiency of the target gene was analyzed using a fluorescence microscope (Leica, Germany).
  • Example 3 Analysis of Parkinson's disease treatment effect using nucleic acid complex in primary cultured microglial cells isolated from brain tissue
  • Example 2 After selecting the nucleic acid combination whose effect was verified through Example 2, the treatment effect of Parkinson's disease was analyzed using primary microglia isolated from neonatal rats having the most similar characteristics to the body in culture conditions. .
  • Human-derived neuroblastoma obtained from KCLB (Korean Cell Line Bank, Korea) was mixed with 10% (v/v) fetal bovine serum, penicillin 100 units/ml, and streptomycin 100 in DMEM culture medium (Wellgene, Korea). [mu]g/ml was added and cultured under the conditions of 37°C and 5% (v/v) CO 2 .
  • Example 3-2 Isolation and culture of primary microglia from mouse brain tissue
  • mice In order to isolate and culture primary microglia from brain tissues of mice, 1-day-old neonatal SD rats (Nara Biotech, Korea) were purchased and brain tissues were isolated. The cortex from which the meninges and other tissue parts were removed was transferred to a Petri dish containing iced 1x HBSS (gibco, USA), and then the tissue was dissociated using a pipette and centrifuged at 1200 rpm for 5 minutes.
  • the supernatant containing only floating cells was collected and centrifuged at 1200 rpm for 8 minutes. After centrifugation, the cells were seeded in 1x10 5 in a 6-well plate, and cultured under conditions of 37°C and 5% (v/v) CO 2 .
  • 100 ng/mL of LPS was treated for 4 hours, followed by nucleic acid complex treatment and culture.
  • Example 3-3 Analysis of gene expression using Western blot assay
  • Example 3-2 Primary microglia cultured under the culture conditions of Example 3-2 were treated with 100 ng/ml of Lipopolysaccharide (LPS, sigma, USA) to induce an immune response. After 4 hours of LPS treatment, complexes containing bioactive peptide nucleic acids and carrier peptide nucleic acids were treated, and after incubation for 24, 48, and 72 hours, respectively, 30 ⁇ L of RIPA buffer was added to each well to obtain protein lysate. Protein lysate was quantified using a BCA assay kit (Thermo Fisher, USA), and 30 ⁇ g of protein was separated by size through electrophoresis, and the protein was transferred to a PVDF membrane.
  • LPS Lipopolysaccharide
  • Pro Cas-1 (abcam, USA) and Pro IL-1 ⁇ (abcam, USA) were treated at a ratio of 1:1000 and left at 4°C for one day. It was washed with 1X TBS-T, treated with a secondary antibody, Goat Anti-Rabbit (Cell signaling Technology, USA) at a ratio of 1:2000, and left at room temperature for 1 hour. Supersignal TM West Femto Maximum Sensitivity Substrate (Thermo Fisher, USA) was treated, and the expression inhibition efficiency of the target gene was analyzed using Image600 (Amersham, Germany) equipment.
  • Example 3-4 Analysis of cell viability of neuroblastoma cell lines using MTT assay
  • Example 3-2 Primary microglia cultured under the culture conditions of Example 3-2 were treated with a complex containing a bioactive peptide nucleic acid and a carrier peptide nucleic acid, and 4 hours later, LPS was treated with 100 ng/ml and cultured for 24, 48, and 72 hours, respectively. did After filtering the culture medium obtained at the end of each culture using a syringe filter (Millipore, USA), it was treated with human-derived neuroblastoma. Human-derived neuroblastoma was seeded in 2x10 4 in a 96-well plate, and after 24 hours, the medium was replaced with the medium obtained from primary microglia, and cultured for 24, 48, and 72 hours, respectively.
  • MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide, sigma, USA) solution was prepared at a concentration of 5 mg/mL in 1X PBS, treated at 20 ⁇ L per well and incubated for 4 hours, then the OD (optical density) was measured and analyzed with a spectrophotometer.
  • Example 4 Analysis of Parkinson's disease treatment effect using nucleic acid complex in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/probenecid-induced mouse model
  • Example 4-1 Production of MPTP/probenecid-induced Parkinson's disease animal model
  • levodopa levodopa, sigma, USA
  • carbidopa carbidopa, sigma, USA
  • Example 4-2 Analysis of motor function change using rotarod test analysis
  • a rotarod test was performed to evaluate the effect of improving motor deficits and balance maintenance, which are characteristics of lesions in animal models of Parkinson's disease, by nucleic acid complexes.
  • a rotarod apparatus (Jeongdo B&P, Korea) with a rotatable cylindrical rod composed of 5 compartments with a diameter of 7 cm and a height of 60 cm at 15 cm intervals was used.
  • the speed of the rotarod used in the test was set to increase constantly from 0 to 40 rpm, and the latency time taken until the animal was placed on the rotating rod and dropped was measured.
  • the maximum measurement time was limited to 300 seconds, and the therapeutic effect of the nucleic acid complex was analyzed using the average value after three repeated measurements.
  • a pole test was performed to evaluate the recovery of akinesia and spastic response using the Parkinson's disease animal disease model prepared under the conditions of Example 4-1.
  • the pole test was performed by placing the mouse facing the sky using a stick with a width of 0.8 cm and a height of 55 cm, and then rotating 180° to measure the total time required to reach the floor. The same number of training sessions were conducted for all experimental animals before the test, and this experiment was conducted after the administration of the nucleic acid complex was completed. The experiment was repeated three times, and the average value was used for analysis of the results.
  • Example 4-4 Analysis of motor function changes using cylinder test analysis
  • Example 4-1 In order to confirm the effect of sensorimotor function treatment in an animal model induced with Parkinson's disease by the method of Example 4-1, a cylinder test was performed.
  • the group of animals exposed to MPTP, a neurotoxin, and administered with only MPTP/probenecid decreased the number of times of using the paws compared to normal mice.
  • the group treated with the complex (PNA 2) increased the number of times of using the forelimbs, confirming the therapeutic efficacy of sensorimotor impairment in an animal model of Parkinson's disease (FIG. 4c).
  • Example 5 Gene expression analysis in Parkinson's disease induced animal model using MPTP/probenecid
  • Parkinson's disease animal model induced by the method of Example 4-1 changes in the expression of Parkinson's disease-related genes were confirmed in the striatum and substantia nigra regions of brain tissue where dopamine production and metabolism occur. .
  • Example 5-1 Analysis of gene expression in Parkinson's disease animal model tissue using Western blot assay
  • the striatum and substantia nigra were separated from the brain tissue extracted on the end day after the experiment was conducted by the method of Example 4-1, and protein lysate was obtained by adding RIPA buffer. Protein lysate was quantified using a BCA assay kit (Thermo Fisher, USA), and 30 ⁇ g of protein was separated by size through electrophoresis, and the protein was transferred to a PVDF membrane. Pro IL-1 ⁇ (abcam, USA), Tyrosine Hydroxylase (TH, abcam, USA), and alpha-synuclein ( ⁇ -synuclein, abcam, USA) were treated at a ratio of 1:1000 and left at 4°C for one day.
  • NLRP3 and lower-order gene expression changes and the expression levels of dopaminergic neurons and alpha-synuclein in Parkinson's disease-induced animal models were analyzed, and the nucleic acid complex combinations used are the same as those in Table 3 above.
  • the expression in the substantia nigra region is shown in FIG. 5B, and the expression of the target gene NLRP3 and IL-1 ⁇ , a sub-pathway gene, increased in the disease induction group (MPTP/p) in the second combination of nucleic acid complexes (PNA). 2), and the decreased dopaminergic neurons in the disease model were recovered with increased expression in the group treated with the nucleic acid complex of No. 2 (combination of SEQ ID NOs: 2 and 5, PNA 2) confirmed that In addition, the expression of alpha-synuclein decreased after treatment with the nucleic acid complex in combination No. 2, confirming the therapeutic efficacy of the nucleic acid complex of SEQ ID NOs: 2 and 5 (FIG. 5b).
  • Example 5-2 Analysis of expression changes in tissues in Parkinson's disease animal models using immunostaining
  • mouse brain tissue was separated on the end day, fixed in 4% formalin solution for one day, immersed in 30% sugar solution, dehydrated for 3 days, and then embedded in OCT (Leica, USA) solution (embedding) was done.
  • the embedded tissue was stored and frozen at -20°C, and then the tissue was sectioned at 20 ⁇ m using a cryostat (Cryostat, Leica, USA) and then mounted on a slide glass.
  • the mounted tissues were blocked for 2 hours using 1% BSA solution, and primary antibodies were used to detect Tyrosine Hydroxylase (abcam, USA), alpha-synuclein ( ⁇ -synuclein, abcam, USA), and IBA1 (wako, Japan).
  • the expression levels of dopaminergic neurons, alpha-synuclein, and immune cells, which are onset factors, were analyzed in brain tissues of Parkinson's disease-induced animal models, and comparative analysis was performed in the striatum and substantia nigra, which are brain regions important for onset.
  • the nucleic acid complex of the present invention in which a bioactive nucleic acid targeting NLRP3 and a carrier peptide nucleic acid are complementaryly bound, has the ability to penetrate the blood-brain barrier and can efficiently inhibit the expression of NLRP3, thereby preventing degenerative brain diseases, particularly Parkinson's disease. Useful for prevention or treatment.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Hospice & Palliative Care (AREA)
  • Genetics & Genomics (AREA)
  • Psychiatry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Biochemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to a nucleic acid complex having an ability to penetrate the blood-brain barrier and a pharmaceutical composition comprising same as an active ingredient for prevention or treatment of degenerative brain diseases and, more specifically, to a nucleic acid complex in which a NLRP3 gene-targeting bioactive nucleic acid complementarily binds to a carrier peptide nucleic acid, and a pharmaceutical composition comprising same as an active ingredient for prevention or treatment of degenerative brain diseases. The nucleic acid complex in which the NLRP3-targeting bioactive nucleic acid complementarily binds to the carrier peptide nucleic acid has an ability to penetrate the blood-brain barrier and can efficiently inhibit the expression of NLRP3 and as such, is useful for preventing or treating degenerative brain diseases, especially Parkinson's disease.

Description

핵산 복합체를 포함하는 퇴행성 뇌질환의 예방 또는 치료용 조성물Composition for preventing or treating degenerative brain diseases containing nucleic acid complexes
본 발명은 혈뇌장벽 투과능을 가지는 핵산 복합체 및 이를 유효성분으로 함유하는 퇴행성 뇌질환 예방 또는 치료용 약학 조성물에 관한 것으로, 더욱 자세하게는 NLRP3 유전자를 표적으로 하는 생활성 핵산(Bioactive Nucleic Acid); 및 캐리어 펩티드 핵산(Carrier Peptide Nucleic Acid)이 상보적으로 결합된 핵산 복합체 및 이를 유효성분으로 함유하는 퇴행성 뇌질환 예방 또는 치료용 약학 조성물에 관한 것이다.The present invention relates to a nucleic acid complex having blood-brain barrier penetrating ability and a pharmaceutical composition for preventing or treating degenerative brain disease containing the complex as an active ingredient, and more specifically, to a bioactive nucleic acid targeting NLRP3 gene; And a carrier peptide nucleic acid (Carrier Peptide Nucleic Acid) relates to a complementary nucleic acid complex and a pharmaceutical composition for preventing or treating degenerative brain disease containing the same as an active ingredient.
파킨슨병은 선조체(striatum)와 흑질(substantia nigra) 부위에서 신경 전달 물질인 도파민(dopamine)의 감소로 발생하는 퇴행성 뇌질환으로 운동이상증, 안정시떨림, 경직 및 보행장애를 주된 증상으로 하는 질병이다. 퇴행성 뇌질환 중 유병률이 알츠하이머 질환에 이어 2위에 해당하는 질환으로서, 60세 이상의 인구 중에서 약 1% 정도가 이 질환으로 고통 받는 것으로 보고되어 있다(Ali Samii, et al., Lancet. 2004 May 29;363(9423):1783-93).Parkinson's disease is a degenerative brain disease caused by a decrease in dopamine, a neurotransmitter, in the striatum and substantia nigra. . Among degenerative brain diseases, the prevalence is second only to Alzheimer's disease, and it is reported that about 1% of the population aged 60 or older suffer from this disease (Ali Samii, et al., Lancet. 2004 May 29; 363(9423):1783-93).
파킨슨병은 잘못 접힌(mis-folded) 형태의 알파-시누클레인(α-synuclein)으로 구성된 루이소체(Lewy body)의 축적 및 확산에 의한 도파민성 뉴런(neuron)의 손실이 나타나는 병리학적 특징을 보인다. 파킨슨병을 유발하는 주요 원인인 루이소체는 신경독성(neurotoxic) 물질이며, 뇌의 주변 영역으로 전파(propagation)되고 도파민성 퇴행을 유발한다(Werner Poewe, et al., Nat Rev Dis Primers. 2017 Mar 23;3:17013). 발병 초기의 파킨슨병 환자의 흑질 부위에서는 만성적인 미세아교세포(microglia) 신경염증이 발생하며, 사후에 확인한 파킨슨병 환자의 뇌에서도 이러한 특징은 두드러지게 나타나는 것으로 알려져 있다(Alexander Gerhard, et al., Neurobiol Dis. 2006 Feb;21(2):404-12). 도파민의 퇴행이 진행되는 동안 이러한 만성적인 신경염증은 지속해서 발생하며 병증을 악화시키는데 기여하지만, 알파-시누클레인과 도파민의 감소를 연결하는 명확한 기전은 아직 밝혀지지 않았다.Parkinson's disease is characterized by the loss of dopaminergic neurons due to the accumulation and diffusion of Lewy bodies composed of misfolded α-synuclein. . Lewy bodies, which are the main cause of Parkinson's disease, are neurotoxic substances that propagate to peripheral areas of the brain and cause dopaminergic degeneration (Werner Poewe, et al., Nat Rev Dis Primers. 2017 Mar 23;3:17013). It is known that chronic microglia neuroinflammation occurs in the substantia nigra region of patients with Parkinson's disease in the early stages of onset, and this feature is also known to be prominent in the brains of patients with Parkinson's disease confirmed postmortem (Alexander Gerhard, et al., Neurobiol Dis. 2006 Feb;21(2):404-12). During dopamine degeneration, this chronic neuroinflammation continues to occur and contributes to exacerbation of the disease, but a clear mechanism linking the decrease in alpha-synuclein and dopamine has not yet been identified.
NLRP3(NLR family pyrin domain containing 3; NACHT, leucine-rich repeat, and pyrin domain(PYD)-containing protein 3(NALP3))은 단백질-코딩 유전자로서, 뉴클레오티드-결합 및 올리고머화 도메인-유사 수용체(nucleotide-binding oligomerization domain-like receptors; NLR)의 패밀리에 속한다. 인플라마좀(inflammasome)은 세포 내의 스트레스 및 환경에 반응하는 센서로 기능하며, 이 중 NLRP3 인플라마좀은 NLRP3 센서, 신호 어댑터인 ASC(apoptosis-associated speck-like protein containing a CARD(caspase activation and recruitment domain)) 및 프로-카스파아제-1(pro-caspase-1)로 구성되어 있다. NLRP3 인플라마좀이 세포의 스트레스에 의해 활성화될 경우 카스파아제-1이 활성화되며, 염증성 사이토카인(cytokine)인 interleukin-1β(IL-1β) 및 IL-18의 방출을 유발하여 염증반응을 시작하게 된다(Shuo Wang, et al., Int Immunopharmacol. 2019 Feb;67:458-464).NLRP3 (NLR family pyrin domain containing 3; NACHT, leucine-rich repeat, and pyrin domain (PYD)-containing protein 3 (NALP3)) is a protein-coding gene that binds to nucleotide-binding and oligomerization domain-like receptors (nucleotide- It belongs to the family of binding oligomerization domain-like receptors (NLRs). The inflammasome functions as a sensor that responds to intracellular stress and environment. Among them, the NLRP3 inflammasome is a NLRP3 sensor and signal adapter ASC (apoptosis-associated speck-like protein containing a CARD (caspase activation and recruitment) domain)) and pro-caspase-1. When the NLRP3 inflammasome is activated by cellular stress, caspase-1 is activated, triggering the release of interleukin-1β (IL-1β) and IL-18, which are inflammatory cytokines, to initiate an inflammatory response. (Shuo Wang, et al., Int Immunopharmacol. 2019 Feb; 67:458-464).
최근 파킨슨병 환자의 뇌에서 NLRP3 인플라마좀의 활성화에는 잘못 접힌 알파-시누클레인 단백질의 축적 및 활성화된 미세아교세포가 원인이 될 수 있음이 확인되었다(Gaia Codolo, et al., PLoS One. 2013;8(1):e55375). 이는 잘못 응집된(aggregation) 불용성(insoluble) 알파-시누클레인이 NLRP3 인플라마좀을 활성화한다는 결과와 더불어 미세아교세포의 활성화에 의한 도파민의 감소와 직접적인 상관관계가 있음을 확인한 결과라 할 수 있다. 이러한 최근 연구를 통해 알파-시누클레인 응집으로 NLRP3 인플라마좀이 활성화되고, 미세아교세포의 염증반응 증가로 인해 도파민 퇴화가 유도된다는 것이 확인되었다(Richard Gordon, et al., Sci Transl Med. 2018 Oct 31;10(465):eaah4066).Recently, it was confirmed that the accumulation of misfolded alpha-synuclein protein and activated microglia could be responsible for the activation of the NLRP3 inflammasome in the brains of patients with Parkinson's disease (Gaia Codolo, et al., PLoS One. 2013 ;8(1):e55375). This can be said to be the result of confirming that there is a direct correlation with the decrease in dopamine due to the activation of microglia, together with the result that insoluble alpha-synuclein, which is mis-aggregated, activates the NLRP3 inflammasome. Through these recent studies, it was confirmed that alpha-synuclein aggregation activates the NLRP3 inflammasome and induces dopamine degeneration due to an increase in the inflammatory response of microglia (Richard Gordon, et al., Sci Transl Med. 2018 Oct. 31;10(465):eaah4066).
뇌질환 치료제로 개발된 상당수의 약이 뇌혈관 장벽의 통과가 잘 이루어지지 않는 문제점을 갖고 있다. 이러한 혈뇌장벽의 투과 메커니즘은 아직 밝혀지지 않았으며, 중추 신경계에 장애가 발생해도 약물을 중추 신경계의 목적으로 하는 영역에 도달시킬 수 없는 현실이며, 효과적인 치료 방법들은 아직 개발되지 않았다.A significant number of drugs developed for the treatment of brain diseases have a problem in that they do not pass through the blood-brain barrier well. The penetration mechanism of the blood-brain barrier has not yet been elucidated, and even if a disorder occurs in the central nervous system, it is a reality that drugs cannot reach the target area of the central nervous system, and effective treatment methods have not yet been developed.
한편, 전통적인 약제와 다르게 핵산 약제는 표적 특이적 전령 RNA(messenger RNA, mRNA)의 발현을 억제하므로, 단백질을 표적으로 하는 기존 약제로 치료가 불가능 했던 연구 영역을 다룰 수 있게 되었다(Ryszard Kole, et al., Nat Rev Drug Discov. 2012 Jan 20;11(2):125-40). 약제로서의 성능 및 장점으로 인하여 핵산을 이용한 다양한 임상시험이 진행되고 있고, 증가하는 핵산 기반 치료제의 용도에도 불구하고 세포내 도입 또는 혈뇌장벽 투과를 위한 운반체의 사용은 극히 제한적이다.On the other hand, unlike traditional drugs, nucleic acid drugs suppress the expression of target-specific messenger RNA (mRNA), so it is possible to address research areas that could not be treated with existing protein-targeting drugs (Ryszard Kole, et al. al., Nat Rev Drug Discov. 2012 Jan 20;11(2):125-40). Due to its performance and advantages as a drug, various clinical trials using nucleic acids are in progress, and despite the increasing use of nucleic acid-based therapeutics, the use of carriers for intracellular introduction or blood-brain barrier penetration is extremely limited.
이와 관련하여, 본 발명자들은 생활성 핵산과 전체적으로 양전하를 가지도록 변형된 캐리어 펩티드 핵산(Carrier Peptide Nucleic Acid)이 상보적으로 결합된 핵산 복합체가 세포 투과성(cell permeability)이 놀랍게 향상되며, 이를 이용하여 표적 유전자의 발현을 매우 효율적으로 조절할 수 있음을 확인하고, 이러한 세포 독성이 낮고, 생활성 핵산의 세포투과성 및 유전자 발현 조절능력이 향상된 새로운 구조체에 대한 특허를 등록 받은 바 있다(대한민국 등록특허 제10-1963885호). 또한, 본 발명자들은 상기 구조체의 기능에 대한 연구를 지속적으로 수행하여, 우수한 효율로 혈뇌장벽을 투과할 수 있는 능력을 가지는 새로운 구조체를 개발하게 되었다(대한민국 특허출원 제10-2019-0128465호).In this regard, the present inventors have found that a nucleic acid complex in which a bioactive nucleic acid and a carrier peptide nucleic acid modified to have a positive charge as a whole are complementarily coupled have surprisingly improved cell permeability, and using this It was confirmed that the expression of the target gene can be controlled very efficiently, and a patent has been registered for a new structure with low cytotoxicity and improved cell permeability of bioactive nucleic acids and the ability to regulate gene expression (Korean Registered Patent No. 10). -1963885). In addition, the present inventors have continuously conducted research on the function of the construct to develop a new construct having the ability to penetrate the blood-brain barrier with excellent efficiency (Korean Patent Application No. 10-2019-0128465).
이에, 본 발명자들은 혈뇌장벽 투과능이 우수한 핵산 복합체를 이용하여, 퇴행성 뇌질환 치료에 적용하고자 예의 노력한 결과, NLRP3 유전자를 표적으로 하는 생활성 핵산과 캐리어 펩티드 핵산이 상보적으로 결합된 핵산 복합체가 퇴행성 뇌질환의 예방 또는 치료에 우수한 효과를 나타냄을 규명하고, 본 발명을 완성하게 되었다.Accordingly, the present inventors have made diligent efforts to use nucleic acid complexes with excellent blood-brain barrier penetration ability to apply them to the treatment of degenerative brain diseases. It was found that it exhibits excellent effects in preventing or treating brain diseases, and the present invention was completed.
본 배경기술 부분에 기재된 상기 정보는 오직 본 발명의 배경에 대한 이해를 향상시키기 위한 것이며, 이에 본 발명이 속하는 기술분야에서 통상의 지식을 가지는 자에게 있어 이미 알려진 선행기술을 형성하는 정보를 포함하지 않을 수 있다.The above information described in this background section is only for improving the understanding of the background of the present invention, and therefore does not include information that forms prior art known to those skilled in the art to which the present invention belongs. may not be
발명의 요약Summary of Invention
본 발명의 목적은 혈뇌장벽 투과능을 가지는 핵산 복합체 및 이를 유효성분으로 함유하는 퇴행성 뇌질환 예방 또는 치료용 약학 조성물을 제공하는 데 있다.An object of the present invention is to provide a nucleic acid complex having blood-brain barrier penetrating ability and a pharmaceutical composition for preventing or treating degenerative brain disease containing the same as an active ingredient.
상기 목적을 달성하기 위하여, 본 발명은 NLRP3 유전자를 표적으로 하는 생활성 핵산(Bioactive Nucleic Acid); 및 캐리어 펩티드 핵산(Carrier Peptide Nucleic Acid)이 상보적으로 결합된 핵산 복합체를 제공한다.In order to achieve the above object, the present invention is a bioactive nucleic acid targeting the NLRP3 gene (Bioactive Nucleic Acid); And a carrier peptide nucleic acid (Carrier Peptide Nucleic Acid) provides a complementary nucleic acid complex.
본 발명은 또한, 상기 핵산 복합체를 유효성분으로 함유하는 퇴행성 뇌질환 예방 또는 치료용 약학 조성물을 제공한다.The present invention also provides a pharmaceutical composition for preventing or treating degenerative brain disease containing the nucleic acid complex as an active ingredient.
본 발명은 또한, 상기 핵산 복합체를 투여하는 단계를 포함하는 퇴행성 뇌질환의 예방 또는 치료방법을 제공한다.The present invention also provides a method for preventing or treating a degenerative brain disease comprising administering the nucleic acid complex.
본 발명은 또한, 퇴행성 뇌질환의 예방 또는 치료를 위한 상기 핵산 복합체의 용도를 제공한다.The present invention also provides the use of the nucleic acid complex for the prevention or treatment of degenerative brain diseases.
본 발명은 또한, 퇴행성 뇌질환의 예방 또는 치료용 약제 제조를 위한 상기 핵산 복합체의 사용을 제공한다.The present invention also provides the use of the nucleic acid complex for the preparation of a drug for preventing or treating degenerative brain disease.
도 1은 LPS로 유도한 파킨슨병 유사 세포 모델에서 핵산 복합체의 표적 유전자 및 하위 유전자 발현 억제능을 확인한 도면이다.1 is a diagram confirming the ability of nucleic acid complexes to inhibit the expression of target genes and subgenes in an LPS-induced Parkinson's disease-like cell model.
도 2는 LPS로 유도한 파킨슨병 유사 세포 모델에서 핵산 복합체의 표적 유전자의 세포 내 발현 억제능을 확인한 도면이다.2 is a diagram confirming the ability of a nucleic acid complex to suppress intracellular expression of a target gene in a Parkinson's disease-like cell model induced by LPS.
도 3은 조직으로부터 분리한 일차 미세아교세포 모델에서 핵산 복합체의 효능을 확인한 도면이다.Figure 3 is a diagram confirming the efficacy of the nucleic acid complex in a primary microglia model isolated from tissue.
도 4는 MPTP/P 파킨슨병 동물모델에서 핵산 복합체 투여에 따른 동물 행동 개선능을 확인한 도면이다.Figure 4 is a view confirming the ability to improve animal behavior according to the administration of the nucleic acid complex in the MPTP/P Parkinson's disease animal model.
도 5는 MPTP/P 파킨슨병 동물모델에서 핵산 복합체 투여에 따른 뇌 부위별 표적 유전자 발현 및 하위 유전자 발현 억제능을 확인한 도면이다.5 is a diagram confirming the ability to inhibit target gene expression and subgene expression by brain region according to the administration of the nucleic acid complex in the MPTP/P Parkinson's disease animal model.
도 6a는 MPTP/P 파킨슨병 동물 모델에서 핵산 복합체 투여에 따른 뇌 부위 중 선조체(striatum)에서의 Tyrosine Hydroxylase(TH)의 발현 변화를 나타낸 도면이다.6a is a view showing changes in expression of Tyrosine Hydroxylase (TH) in the striatum of the brain according to the administration of the nucleic acid complex in the MPTP/P Parkinson's disease animal model.
도 6b는 MPTP/P 파킨슨병 동물 모델에서 핵산 복합체 투여에 따른 뇌 부위 중 흑질(Substantia Nigra)에서의 Tyrosine Hydroxylase(TH)의 발현 변화를 나타낸 도면이다.6B is a diagram showing changes in expression of Tyrosine Hydroxylase (TH) in the substantia nigra of the brain according to administration of the nucleic acid complex in the MPTP/P Parkinson's disease animal model.
도 6c는 MPTP/P 파킨슨병 동물 모델에서 핵산 복합체 투여에 따른 뇌 부위 중 선조체(striatum)에서의 α-Synuclein의 발현 억제능을 확인한 도면이다.Figure 6c is a view confirming the expression suppression ability of α-Synuclein in the striatum (striatum) of the brain region according to the administration of the nucleic acid complex in the MPTP/P Parkinson's disease animal model.
도 6d는 MPTP/P 파킨슨병 동물 모델에서 핵산 복합체 투여에 따른 뇌 부위 중 흑질(Substantia Nigra)에서의 α-Synuclein의 발현 억제능을 확인한 도면이다.Figure 6d is a view confirming the expression suppression ability of α-Synuclein in the substantia nigra of the brain region according to the administration of the nucleic acid complex in the MPTP/P Parkinson's disease animal model.
도 6e는 MPTP/P 파킨슨병 동물 모델에서 핵산 복합체 투여에 따른 뇌 부위 중 선조체(striatum)에서의 Ionized calcium-Binding Adapter molecule 1(IBA-1)의 발현 억제능을 확인한 도면이다.Figure 6e is a view confirming the expression inhibition ability of ionized calcium-binding adapter molecule 1 (IBA-1) in the striatum (striatum) of the brain region according to the administration of the nucleic acid complex in the MPTP/P Parkinson's disease animal model.
도 6f는 MPTP/P 파킨슨병 동물 모델에서 핵산 복합체 투여에 따른 뇌 부위 중 흑질(Substantia Nigra)에서의 Ionized calcium-Binding Adapter molecule 1(IBA-1)의 발현 억제능을 확인한 도면이다.6f is a diagram confirming the expression inhibition ability of ionized calcium-binding adapter molecule 1 (IBA-1) in the substantia nigra of the brain according to the administration of the nucleic acid complex in the MPTP/P Parkinson's disease animal model.
발명의 상세한 설명 및 바람직한 구현예DETAILED DESCRIPTION OF THE INVENTION AND PREFERRED EMBODIMENTS
다른 식으로 정의되지 않는 한, 본 명세서에서 사용된 모든 기술적 및 과학적 용어들은 본 발명이 속하는 기술분야에서 숙련된 전문가에 의해서 통상적으로 이해되는 것과 동일한 의미를 갖는다. 일반적으로 본 명세서에서 사용된 명명법은 본 기술분야에서 잘 알려져 있고 통상적으로 사용되는 것이다.Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In general, the nomenclature used herein is one well known and commonly used in the art.
파킨슨병의 발병 원인은 매우 다양하다. 전체 환자의 5% 정도가 유전적 요인에 의해서 나타나며, 염증과 산화적 스트레스와 같은 외부 환경적인 요인이 중요하게 작용한다. 파킨슨병은 도파민성 신경세포에 단백질의 비정상적인 응집 현상이 일어나 발병하는데, 알파-시뉴클레인 단백질의 응집이다. 이러한 단백질의 응집은 산화적 스트레스에 의해서 더 촉진된다. 지금까지 파킨슨병의 치료제로는 도파민 신경전달 물질을 보충하는 방법으로 이루어졌으나, 이는 파킨슨병의 근원적인 치료가 되지 못한다. 파킨슨병의 근본적 치료를 위해 질병을 수정하는 유전자를 직접적으로 표적하는 저분자 치료 약물, 유전자 치료, 단일클론항체, 관련 징후를 표적화한 면역요법, 줄기세포 및 유도만능줄기세포 등의 생물학적 제제 연구가 활발해지고 있다.The causes of Parkinson's disease are very diverse. About 5% of all patients are caused by genetic factors, and external environmental factors such as inflammation and oxidative stress play an important role. Parkinson's disease is caused by abnormal protein aggregation in dopaminergic neurons, which is the aggregation of alpha-synuclein protein. Aggregation of these proteins is further promoted by oxidative stress. Until now, the treatment of Parkinson's disease has been made by supplementing the dopamine neurotransmitter, but this is not a fundamental treatment for Parkinson's disease. For the fundamental treatment of Parkinson's disease, research on biological agents such as small-molecule therapeutic drugs that directly target disease-modifying genes, gene therapy, monoclonal antibodies, immunotherapy targeting related symptoms, and stem cells and induced pluripotent stem cells are actively being conducted. It's getting done.
한편, 대한민국 내 파킨슨병 환자 현황으로는 2010년 6만 1,565명이였으며 연평균 8.7%로 성장하여 2014년 8만 5,888명으로 증가하였다. 2014년 환자의 경우 60세 이상 환자 비율이 95.7%로, 유병율은 환자의 나이와 상관관계를 보였으며, 성비율의 경우 남성이 3만 3,831명이고 여성은 5만 2,057명을 나타내었다(최근 5년 파킨슨병 환자·진료비 증가세, 청년의사, 2015년). 파킨슨병은 알츠하이머병에 비해 발병 시기가 60세 정도로 매우 빠른 편이며 이로 인해 질환의 진행이 20년 이상 지속되기에 조기 발견을 통한 질환 진행을 늦추는 치료법이 적극적으로 요구된다.On the other hand, the number of patients with Parkinson's disease in Korea was 61,565 in 2010 and increased to 85,888 in 2014 with an average annual growth rate of 8.7%. In the case of patients in 2014, the proportion of patients aged 60 years or older was 95.7%, and the prevalence showed a correlation with the patient's age. Parkinson's disease patient and treatment cost increase, young doctors, 2015). Compared to Alzheimer's disease, the onset of Parkinson's disease is very early, around 60 years of age, and because of this, the progression of the disease lasts for more than 20 years, so a treatment that slows the disease progression through early detection is actively required.
본 발명에서는 NLRP3 유전자를 표적으로 하는 생활성 핵산과 캐리어 펩티드 핵산이 상보적으로 결합된 핵산 복합체가 파킨슨병의 예방 및 치료에 활용될 수 있음을 확인하였다.In the present invention, it was confirmed that a nucleic acid complex in which a bioactive nucleic acid targeting the NLRP3 gene and a carrier peptide nucleic acid are complementaryly bound can be used for the prevention and treatment of Parkinson's disease.
본 발명의 일 실시예에서, 본 발명자들의 대한민국 등록특허 제10-1963885호를 기반으로, NLRP3 유전자를 표적으로 하는 생활성 핵산과 캐리어 펩티드 핵산이 상보적으로 결합된 핵산 복합체를 미세아교세포주에 처리한 경우에, 효과적으로 NLRP3 및 하위단계 유전자의 발현이 억제되는 것을 확인하였으며, 상기 핵산 복합체를 투여한 파킨슨병 동물모델에서 행동 장애, 운동기능 장애 및 감각 운동 장애가 개선됨을 확인하였다.In one embodiment of the present invention, based on Korean Patent Registration No. 10-1963885 of the present inventors, treatment of a nucleic acid complex in which a bioactive nucleic acid targeting the NLRP3 gene and a carrier peptide nucleic acid are complementaryly coupled to a microglial cell line In one case, it was confirmed that the expression of NLRP3 and sublevel genes were effectively suppressed, and it was confirmed that behavioral disorders, motor function disorders, and sensorimotor disorders were improved in a Parkinson's disease animal model to which the nucleic acid complex was administered.
따라서, 본 발명은 일 관점에서, NLRP3 유전자를 표적으로 하는 생활성 핵산(Bioactive Nucleic Acid); 및 캐리어 펩티드 핵산(Carrier Peptide Nucleic Acid)이 상보적으로 결합된 핵산 복합체에 관한 것이다.Therefore, the present invention, in one aspect, a bioactive nucleic acid targeting the NLRP3 gene (Bioactive Nucleic Acid); and a nucleic acid complex in which a carrier peptide nucleic acid is complementarily bound.
본 발명에 있어서, NLRP3 유전자를 표적으로 하는 생활성 핵산과 캐리어 펩티드가 상보적으로 결합된 핵산 복합체는 하기 구조식 (1)의 구조를 가지는 것을 특징으로 할 수 있다.In the present invention, a nucleic acid complex in which a bioactive nucleic acid targeting the NLRP3 gene and a carrier peptide are complementaryly bound may have a structure of the following structural formula (1).
구조식 (1)structural formula (1)
[ A ≡ C(+) ][ A ≡ C (+) ]
상기 구조식 (1)에 있어서,In the structural formula (1),
A는 목적하는 유전자와 결합할 수 있는 서열을 가지는 생활성 핵산(Bioactive Nucleic Acid)이고,A is a bioactive nucleic acid having a sequence capable of binding to a gene of interest,
C는 생활성 핵산과 결합할 수 있는 캐리어 펩티드 핵산(Carrier Peptide Nucleic Acid)이고,C is a carrier peptide nucleic acid capable of binding to a bioactive nucleic acid;
‘≡’는 생활성 핵산과 캐리어 펩티드 핵산 간의 상보적인 결합을 의미하며,‘≡’ means a complementary bond between a bioactive nucleic acid and a carrier peptide nucleic acid,
A로 표시되는 생활성 핵산은 전체적으로 음전하 또는 중성을 가지며,The bioactive nucleic acid represented by A has an overall negative charge or neutral charge,
C(+)는 캐리어 펩티드 핵산이 전체적으로 양전하를 가진다는 것을 의미하며,C (+) means that the carrier peptide nucleic acid has an overall positive charge,
캐리어 펩티드 핵산은 캐리어 펩티드 핵산 전체적으로 양전하를 띠도록 변형된 펩티드 핵산 단량체를 하나 이상 포함한다.The carrier peptide nucleic acid includes one or more peptide nucleic acid monomers modified to have a positive charge throughout the carrier peptide nucleic acid.
본 발명에 따른 핵산 복합체에서의 생활성 핵산과 캐리어 펩티드 핵산은 역평행결합(anti-parallel binding) 또는 평행결합(parallel binding) 형태를 가질 수 있다. 본 발명에 있어서, 상기 핵산의 상보적인 결합 형태는 생활성 핵산의 목적 서열(생활성 핵산과 상보적인 서열) 존재 하에서 분리될 수 있다.The bioactive nucleic acid and the carrier peptide nucleic acid in the nucleic acid complex according to the present invention may have anti-parallel binding or parallel binding. In the present invention, the complementary binding form of the nucleic acid can be separated in the presence of a target sequence of the bioactive nucleic acid (a sequence complementary to the bioactive nucleic acid).
본 발명에 있어서, “생활성 핵산(Bioactive Nucleic Acid)”은 생체외(in vitro) 또는 생체내(in vivo)에서 표적 유전자, 및 이를 포함하는 염기서열과 결합하여 해당 유전자의 고유 기능(예를 들어, 전사체(transcript) 발현 또는 단백질 발현)을 활성화시키거나 또는 저해하거나, pre-mRNA의 스플라이싱(splicing)을 조절(예를 들어, 엑손스키핑(exon skipping) 하는 등의 기능을 수행하며, 상기 염기서열은 유전자 조절부위(gene regulatory sequence) 또는 유전자 부위(gene coding sequence) 또는 스플라이싱 조절 부위(splicing regulatory sequence)인 것을 특징으로 할 수 있다. 바람직하게는, 상기 생활성 핵산은 발현을 감소시키고자 하는 목적하는 표적 유전자와 결합할 수 있는 상보적인 서열, 특히 그러한 목적하는 표적 유전자의 mRNA에 결합할 수 있는 상보적인 서열을 가지는 핵산으로, 해당 유전자의 발현을 억제하는 등의 유전자 발현조절에 관여하는 핵산을 의미하며, 발현을 감소시키고자 하는 표적 유전자에 상보적인 서열을 가지는 핵산일 수 있다.In the present invention, "Bioactive Nucleic Acid" binds to a target gene and a nucleotide sequence containing the target gene in vitro or in vivo to determine the unique function of the gene (e.g., For example, activating or inhibiting transcript expression or protein expression), or regulating pre-mRNA splicing (eg, exon skipping), etc. , The nucleotide sequence may be characterized as a gene regulatory sequence, a gene coding sequence, or a splicing regulatory sequence. Preferably, the bioactive nucleic acid is expressed A nucleic acid having a complementary sequence capable of binding to a target gene of interest to be reduced, in particular, a complementary sequence capable of binding to the mRNA of such a target gene of interest, and suppressing the expression of the gene. It means a nucleic acid involved in regulation, and may be a nucleic acid having a sequence complementary to a target gene whose expression is to be reduced.
따라서, 본 발명에서의 생활성 핵산은 파킨슨병의 관련 표적 유전자인 NLRP3(NLR family pyrin domain containing 3) 유전자의 안티센스 펩티드 핵산인 것이 바람직하며, 더욱 바람직하게는 서열번호 2의 서열로 표시되는 염기서열을 포함할 수 있으나, 이에 한정되는 것은 아니다.Therefore, the bioactive nucleic acid in the present invention is preferably an antisense peptide nucleic acid of the NLRP3 (NLR family pyrin domain containing 3) gene, a target gene related to Parkinson's disease, and more preferably the nucleotide sequence represented by the sequence of SEQ ID NO: 2 It may include, but is not limited thereto.
상기 생활성 핵산은 DNA, RNA, 또는 변형된 핵산인 PNA(peptide nucleic acid), PMO(phosphorodiamidate morpholino oligonucleotide), LNA(locked nucleic acid), GNA(glycol nucleic acid) 및 TNA(threose nucleic acid), 안티센스 올리고뉴클레오티드(antisense oligonucleotide), 앱타머(aptamer), siRNA(small interfering RNA), shRNA(short hairpin RNA), 리보자임(ribozyme) 및 DNAzyme로 구성된 군에서 선택되는 것일 수 있으며, 바람직하게는 상기 생활성 핵산은 DNA, RNA, 또는 변형된 핵산인 PNA(peptide nucleic acid), PMO(phosphorodiamidate morpholino oligonucleotide), LNA(locked nucleic acid), GNA(glycol nucleic acid) 및 TNA(threose nucleic acid)로 구성된 군에서 선택되는 것일 수 있다.The bioactive nucleic acids include DNA, RNA, or modified nucleic acids such as PNA (peptide nucleic acid), PMO (phosphorodiamidate morpholino oligonucleotide), LNA (locked nucleic acid), GNA (glycol nucleic acid) and TNA (threose nucleic acid), antisense It may be selected from the group consisting of oligonucleotide, aptamer, small interfering RNA (siRNA), short hairpin RNA (shRNA), ribozyme and DNAzyme, preferably the bioactive The nucleic acid is selected from the group consisting of DNA, RNA, or modified nucleic acid PNA (peptide nucleic acid), PMO (phosphorodiamidate morpholino oligonucleotide), LNA (locked nucleic acid), GNA (glycol nucleic acid) and TNA (threose nucleic acid) it may be
본 발명에 있어서, “캐리어 펩티드 핵산(Carrier Peptide Nucleic Acid)”은 생활성 핵산과 일부 혹은 전부의 염기가 상보적으로 결합하여 기능성을 부여하는 핵산을 의미하며, 본 발명에서 사용되는 캐리어 펩티드 핵산은 펩티드 핵산(PNA: Peptide Nucleic Acid) 뿐 아니라, 이와 유사한 변형된 핵산을 사용할 수 있으며, 펩티드 핵산이 바람직하지만, 이에 한정되는 의미는 아니다.In the present invention, "Carrier Peptide Nucleic Acid" refers to a nucleic acid to which a bioactive nucleic acid and some or all bases are complementaryly combined to impart functionality, and the carrier peptide nucleic acid used in the present invention is In addition to peptide nucleic acid (PNA), modified nucleic acids similar thereto may be used, and peptide nucleic acids are preferred, but are not limited thereto.
본 발명에 있어서, 상기 캐리어 펩티드 핵산은 캐리어 펩티드 핵산 전체적으로 양전하를 띠도록 1개 이상의 gamma- 또는 alpha-backbone 변형 펩티드 핵산 단량체를 하나 이상 포함하는 것이 바람직하며, 상기 gamma- 혹은 alpha-backbone 변형 펩티드 핵산 단량체는 양전하를 가지는 아미노산을 가지는 단량체가 음전하를 가지는 아미노산을 가지는 단량체에 비해 더 많이 포함되어 전체적인 캐리어 펩티드 핵산의 전하가 양성이 되도록 하는 것이 더욱 바람직하다.In the present invention, the carrier peptide nucleic acid preferably includes one or more gamma- or alpha-backbone modified peptide nucleic acid monomers so that the entire carrier peptide nucleic acid is positively charged, and the gamma- or alpha-backbone modified peptide nucleic acid It is more preferable that monomers having amino acids having a positive charge are included more than monomers having amino acids having a negative charge so that the overall charge of the carrier peptide nucleic acid is positive.
특히, 본 발명에 있어서, 상기 캐리어 펩티드 핵산은 서열번호 4 또는 서열번호 5의 서열로 표시되는 염기서열을 포함하는 것이 바람직하나, 이에 한정되는 것은 아니다.In particular, in the present invention, the carrier peptide nucleic acid preferably includes the nucleotide sequence represented by SEQ ID NO: 4 or SEQ ID NO: 5, but is not limited thereto.
본 발명에 있어서 “핵산 복합체”는 세포 외 처리를 통해 생활성 물질을 체내, 궁극적으로 세포 내로 침투시킬 수 있으며, 구체적으로는 세포 내로 NLRP3 유전자를 표적으로 하는 생활성 핵산을 전달할 수 있는 능력을 가진다.In the present invention, the "nucleic acid complex" can penetrate the bioactive substance into the body and ultimately into the cell through extracellular treatment, and specifically has the ability to deliver the bioactive nucleic acid targeting the NLRP3 gene into the cell. .
본 발명에 있어서, 상기 핵산 복합체는 서열번호 2의 서열로 표시되는 생활성 핵산; 및 서열번호 4 또는 5의 서열로 표시되는 캐리어 펩티드 핵산을 포함하는 것이 바람직하나, 이에 한정되는 것은 아니다.In the present invention, the nucleic acid complex is a bioactive nucleic acid represented by the sequence of SEQ ID NO: 2; and a carrier peptide nucleic acid represented by the sequence of SEQ ID NO: 4 or 5, but is not limited thereto.
또한, 상기 NLRP3 유전자를 표적으로 하는 생활성 핵산 및 캐리어 펩티드 핵산의 결합력(융해온도, melting temperature, Tm)은 생활성 핵산과 생활성 핵산의 표적인 NLRP3 유전자와의 결합력보다 낮은 것을 특징으로 할 수 있다.In addition, the binding ability (melting temperature, Tm) of the bioactive nucleic acid targeting the NLRP3 gene and the carrier peptide nucleic acid is lower than that of the bioactive nucleic acid and the target NLRP3 gene. there is.
상기 결합력은 생활성 핵산 및 캐리어 펩티드 핵산은 각각의 핵산의 5'-방향성 및 3'-방향성에 따라 평행 결합(Parallel binding) 또는 부분 특이결합(Partial specific binding)함으로써, 생활성 핵산 및 캐리어 펩티드 핵산의 결합력(Tm)이 생활성 핵산과 생활성 핵산의 목적하는 유전자와의 결합력보다 낮게 할 수 있다.The binding force is obtained by parallel binding or partial specific binding between the bioactive nucleic acid and the carrier peptide nucleic acid according to the 5'-direction and the 3'-direction of each nucleic acid, so that the bioactive nucleic acid and the carrier peptide nucleic acid The binding force (Tm) of may be lower than the binding force between the bioactive nucleic acid and the target gene of the bioactive nucleic acid.
본 발명에 있어서, 상기 생활성 핵산 또는 캐리어 펩티드 핵산은 각각의 핵산의 5'-말단 또는 3'-말단에 엔도좀 탈출을 도와주는 물질이 추가로 결합된 것을 특징으로 할 수 있다. 즉, 생활성 핵산과 캐리어 펩티드 핵산의 엔도좀 탈출(endosome escape)을 도와주는 물질을 더 포함하여 하기 구조식 (2)의 구조를 가지는 것을 특징으로 할 수 있다.In the present invention, the bioactive nucleic acid or carrier peptide nucleic acid may be characterized by additionally binding a substance that helps endosome escape to the 5'-end or 3'-end of each nucleic acid. That is, it may be characterized in that it has a structure of the following Structural Formula (2) by further including a material that helps the endosome escape of the bioactive nucleic acid and the carrier peptide nucleic acid.
구조식 (2)structural formula (2)
[ mA ≡ mC(+) ][ mA ≡ mC (+) ]
상기 구조식 (2)에 있어서,In the structural formula (2),
'm'은 생활성 핵산과 캐리어 펩티드 핵산의 엔도좀 탈출(endosome escape)을 도와주는 물질을 의미한다.'m' means a substance that helps endosome escape of bioactive nucleic acid and carrier peptide nucleic acid.
본 발명에 있어서, “엔도좀 탈출을 도와주는 물질”은 엔도좀 내부의 삼투압을 증가시키거나, 엔도좀의 막을 불안정화 시키는 방법에 의하여 생활성 핵산의 엔도좀에서 탈출을 도와주는 것을 특징으로 할 수 있다. 생활성 핵산이 보다 효율적이고 빠르게 핵이나 세포질로 이동하여 표적 유전자를 만나 작용하도록 도와주는 것을 의미한다(Daniel W Pack, et al., Nat Rev Drug Discov. 2005 Jul;4(7):581-93).In the present invention, "substances that help endosomes escape" can be characterized in that they help the escape of bioactive nucleic acids from endosomes by increasing the osmotic pressure inside the endosomes or by destabilizing the membranes of endosomes. there is. It means that bioactive nucleic acids move more efficiently and rapidly to the nucleus or cytoplasm to meet and act on target genes (Daniel W Pack, et al., Nat Rev Drug Discov. 2005 Jul;4(7):581-93 ).
본 발명에 있어서, 상기 엔도좀 탈출을 도와주는 물질은 펩티드, 지질 나노물질(lipid nanoparticles), 접합체 나노물질(polyplex nanoparticles), 고분자 나노구(polymer nanospheres), 무기물 나노물질(inorganic nanoparticles), 양이온 지질 나노물질(cationic lipid-based nanoparticles), 양이온 고분자(cationic polymer) 및 pH 감응 고분자(pH sensitive polymers)로 구성된 군에서 선택되는 어느 하나 이상인 것을 특징으로 할 수 있다.In the present invention, the material that helps the endosome escape is a peptide, lipid nanoparticles, conjugate nanoparticles (polyplex nanoparticles), polymer nanospheres (polymer nanospheres), inorganic nanomaterials (inorganic nanoparticles), cationic lipids It may be characterized in that at least one selected from the group consisting of nanomaterials (cationic lipid-based nanoparticles), cationic polymers (cationic polymers) and pH sensitive polymers (pH sensitive polymers).
본 발명에 있어서, 상기 엔도좀 탈출을 도와주는 물질로서 생활성 핵산에는 펩티드(GLFDIIKKIAESF, 서열번호 6)가 링커 매개로 연결될 수 있으며, 캐리어 펩티드 핵산에는 Histidine(10)을 링커 매개로 연결하는 방법으로 결합하는 것을 특징으로 할 수 있으나, 이에 제한되는 것은 아니다.In the present invention, as a material that helps the endosome escape, a peptide (GLFDIIKKIAESF, SEQ ID NO: 6) may be linked to the bioactive nucleic acid via a linker, and Histidine (10) to the carrier peptide nucleic acid via a linker. It may be characterized by combining, but is not limited thereto.
본 발명에 있어서, 상기 지질 나노물질(lipid nanoparticles)은 Lipid, phospholipids, acetyl palmitate, poloxamer 18, Tween 85, tristearin glyceride 및 Tween 80로 구성된 군에서 선택되는 것을 특징으로 할 수 있다.In the present invention, the lipid nanoparticles may be selected from the group consisting of Lipid, phospholipids, acetyl palmitate, poloxamer 18, Tween 85, tristearin glyceride and Tween 80.
본 발명에 있어서, 상기 접합체 나노물질(polyplex nanoparticles)은 poly(amidoamine) 또는 polyethylenimine(PEI)인 것을 특징으로 할 수 있다.In the present invention, the polyplex nanoparticles may be poly(amidoamine) or polyethylenimine (PEI).
본 발명에 있어서, 상기 고분자 나노구(polymer nanospheres)는 polycaprolactone, poly(lactide-co-glycolide, polylactide, polyglycolide, poly(d,l-lactide), chitosan 및 PLGA-polyethylene glycol로 구성된 군에서 선택되는 것을 특징으로 할 수 있다.In the present invention, the polymer nanospheres are selected from the group consisting of polycaprolactone, poly(lactide-co-glycolide, polylactide, polyglycolide, poly(d,l-lactide), chitosan, and PLGA-polyethylene glycol. can be characterized.
본 발명에 있어서, 상기 무기물 나노물질(inorganic nanoparticles)은 Fe2O3, Fe3O4, WO3 및 WO2.9로 구성된 군에서 선택되는 것을 특징으로 할 수 있다.In the present invention, the inorganic nanoparticles may be selected from the group consisting of Fe2O3, Fe3O4, WO3 and WO2.9.
본 발명에 있어서, 상기 양이온 지질 나노물질(cationic lipid-based nanoparticles)은 1-(aminoethyl)iminobis[N-(oleicylcysteinyl-1-amino-ethyl)propionamide], N-alkylated derivative of PTA 및 3, 5-didodecyloxybenzamidine로 구성된 군에서 선택되는 것을 특징으로 할 수 있다.In the present invention, the cationic lipid-based nanoparticles are 1- (aminoethyl) iminobis [N- (oleicylcysteinyl-1-amino-ethyl) propionamide], N-alkylated derivative of PTA and 3, 5- It may be characterized in that it is selected from the group consisting of didodecyloxybenzamidine.
본 발명에 있어서, 상기 양이온 고분자(cationic polymer)는 vinylpyrrolidone-N, N-dimethylaminoethyl methacrylate acid copolymer diethyl sulphate, polyisobutylene 및 poly(N-vinylcarbazole)로 구성된 군에서 선택되는 것을 특징으로 할 수 있다.In the present invention, the cationic polymer may be selected from the group consisting of vinylpyrrolidone-N, N-dimethylaminoethyl methacrylate acid copolymer diethyl sulphate, polyisobutylene and poly(N-vinylcarbazole).
본 발명에 있어서, 상기 pH 감응 고분자(pH sensitive polymers)는 polyacids, poly(acrylic acid), poly(methacrylic acid) 및 hydrolyzed polyacrylamide로 구성된 군에서 선택되는 것을 특징으로 할 수 있다.In the present invention, the pH sensitive polymers may be selected from the group consisting of polyacids, poly(acrylic acid), poly(methacrylic acid), and hydrolyzed polyacrylamide.
본 발명에 있어서, 상기 생활성 핵산과 캐리어 펩티드 핵산은 각각 2 내지 50개, 바람직하게는 5 내지 30개, 더욱 바람직하게는 10 내지 25개, 가장 바람직하게는 15 내지 17개의 핵산 단량체를 포함하는 것을 특징으로 할 수 있다.In the present invention, the bioactive nucleic acid and the carrier peptide nucleic acid each comprise 2 to 50, preferably 5 to 30, more preferably 10 to 25, most preferably 15 to 17 nucleic acid monomers. that can be characterized.
상기 생활성 핵산은 천연(natural) 핵산 염기 및/또는 변형된 핵산 단량체로 이루어져 있는 것을 특징으로 할 수 있다.The bioactive nucleic acid may be characterized in that it consists of natural nucleic acid bases and/or modified nucleic acid monomers.
본 발명에 있어, 상기 생활성 핵산에 사용된 단량체가 PNA일 경우, 생활성 펩티드 핵산이라고 하며, 다른 단량체가 사용된 경우에도 동일한 방식으로 부른다.In the present invention, when the monomer used for the bioactive nucleic acid is PNA, it is referred to as a bioactive peptide nucleic acid, and when other monomers are used, it is referred to in the same way.
본 발명에 있어서, 상기 생활성 핵산과 캐리어 펩티드 핵산은 포스포디에스테르(phosphodiester), 2' O-메틸(2' O-methyl), 2' 메톡시-에틸(2' methoxy-ethyl), 포스포르아미데이트(phosphoramidate), 메틸포스포네이트(methylphosphonate) 및 포스포로티오에이트(phosphorothioate)로 구성된 군에서 선택되는 하나 이상의 작용기를 추가로 포함하는 것을 특징으로 할 수 있다.In the present invention, the bioactive nucleic acid and the carrier peptide nucleic acid are phosphodiester, 2' O-methyl, 2' methoxy-ethyl, phosphor It may be characterized by further comprising at least one functional group selected from the group consisting of amidate, methylphosphonate, and phosphorothioate.
본 발명에 있어서, 상기 캐리어 펩티드 핵산은 상기 생활성 핵산과 염기서열 일부 혹은 전부가 상보적인 서열로 구성되는 것을 특징으로 할 수 있다. 특히, 캐리어 펩티드 핵산은 유니버설 염기(universal base)를 하나 이상 포함할 수 있으며, 캐리어 펩티드 핵산 모두가 유니버설 염기로 이루어질 수도 있다.In the present invention, the carrier peptide nucleic acid may be characterized in that a part or all of the base sequence of the bioactive nucleic acid is composed of a complementary sequence. In particular, the carrier peptide nucleic acid may include one or more universal bases, and all of the carrier peptide nucleic acids may consist of universal bases.
본 발명에 있어서, 상기 핵산 복합체 내의 상기 생활성 핵산 및 캐리어 펩티드 핵산 각각은 전기적 특성이 전체적으로, 양전하(양성), 음전하(음성) 또는 중성 전하를 가지는 것을 특징으로 하는 복합체일 수 있다.In the present invention, each of the bioactive nucleic acid and the carrier peptide nucleic acid in the nucleic acid complex may be a complex characterized by having a positive charge (positive), negative charge (negative) or neutral charge as a whole.
상기 전기적 특성의 표현에 있어, “전체적으로”의 의미는 개별 염기의 전기적 특성이 아닌 전체적인 생활성 핵산 또는 캐리어 펩티드 핵산 각각의 전하가 외부에서 볼 때 전체적인 전기적 특성을 의미하는 것으로, 예를 들어, 생활성 핵산 내의 일부 단량체가 양성을 가지더라도 음성을 가지는 단량체의 개수가 더 많이 존재하는 경우에는 생활성 핵산은 “전체적으로” 전기적 특성을 볼 때 음전하를 가지게 되는 것이며, 캐리어 펩티드 핵산 내의 일부 염기 및/또는 백본(backbone)이 음성을 가지더라도 양성을 가지는 염기 및/또는 백본의 개수가 더 많이 존재하는 경우에는 캐리어 펩티드 핵산은 “전체적으로” 전기적 특성을 볼 때 양전하를 가지게 되는 것이다.In the expression of the electrical properties, the meaning of "overall" means the electrical properties of the entire bioactive nucleic acid or carrier peptide nucleic acid as a whole, not the electrical properties of individual bases, when viewed from the outside. Even if some of the monomers in the sexual nucleic acid have a positive charge, if there are more monomers with a negative charge, the bioactive nucleic acid will have a negative charge when looking at the electrical properties “as a whole”, and some bases and/or Even if the backbone has a negative charge, when a larger number of bases and/or backbones having a positive charge are present, the carrier peptide nucleic acid has a positive charge when looking at the electrical characteristics “as a whole”.
이러한 관점에서, 본 발명의 핵산 복합체는 전체적으로 양전하를 가지는 것을 특징으로 할 수 있다. 상기 핵산 복합체에 있어서, 바람직하게는 상기 생활성 핵산은 전체적으로 전기적 특성을 볼 때 음전하 또는 중성의 특성을 가지며, 상기 캐리어 펩티드 핵산은 전체적으로 전기적 특성을 볼 때 양전하 특성을 가지는 것을 특징으로 할 수 있으나, 이에 제한되는 것은 아니다.From this point of view, the nucleic acid complex of the present invention may be characterized as having a positive charge as a whole. In the nucleic acid complex, preferably, the bioactive nucleic acid has negative or neutral electrical characteristics as a whole, and the carrier peptide nucleic acid has positive electrical characteristics as a whole. It is not limited thereto.
본 발명에 있어서, 상기 생활성 핵산과 캐리어 펩티드 핵산의 전기적 특성의 부여는 변형된 펩티드 핵산 단량체를 사용할 수 있고, 변형된 펩티드 핵산 단량체는 양전하를 가지는 캐리어 펩티드 핵산으로 리신(Lysine, Lys, K), 아르기닌(Arginine, Arg, R), 히스티딘(Histidine, His, H), 디아미노 부티르산(Diamino butyric acid, DAB), 오르니틴(Ornithine, Orn) 및 아미노산 유사체로 구성된 군에서 선택되는 어느 하나 이상의 양전하의 아미노산을 포함하고, 음전하를 가지는 캐리어 펩티드 핵산으로 음전하의 아미노산인 글루타민산(Glutamic acid, Glu, E) 또는 아미노산 유사체의 음전하의 아미노산을 포함하는 것을 특징으로 할 수 있다.In the present invention, the electrical properties of the bioactive nucleic acid and the carrier peptide nucleic acid can be imparted using a modified peptide nucleic acid monomer, and the modified peptide nucleic acid monomer is a carrier peptide nucleic acid having a positive charge, such as lysine (Lysine, Lys, K) , arginine (Arg, R), histidine (Histidine, His, H), diamino butyric acid (DAB), ornithine (Orn), and any one or more positive charges selected from the group consisting of amino acid analogs It is a carrier peptide nucleic acid that includes amino acids of and has a negative charge, and may be characterized in that it includes a negatively charged amino acid such as glutamic acid (Glu, E) or an amino acid analog that is negatively charged.
본 발명에 있어서, 상기 캐리어 펩티드 핵산은 전체적으로 양전하를 가지도록 1개 이상의 gamma- 또는 alpha-backbone 변형 펩티드 핵산 단량체를 포함하는 것을 특징으로 할 수 있다.In the present invention, the carrier peptide nucleic acid may be characterized by including one or more gamma- or alpha-backbone modified peptide nucleic acid monomers so as to have a positive charge as a whole.
상기 gamma- 혹은 alpha-backbone 변형 펩티드 핵산 단량체는 전기적 양성을 가지도록 리신(Lysine, Lys, K), 아르기닌(Arginine, Arg, R), 히스티딘(Histidine, His, H), 디아미노 부티르산(Diamino butyric acid, DAB), 오르니틴(Ornithine, Orn) 및 아미노산 유사체로 구성된 군에서 선택되는 어느 하나 이상의 양전하를 가지는 아미노산을 backbone에 포함하는 것을 특징으로 할 수 있다.The gamma- or alpha-backbone modified peptide nucleic acid monomer has lysine (Lysine, Lys, K), arginine (Arginine, Arg, R), histidine (His, H), diamino butyric acid (Diamino butyric acid) to have an electrical positivity. acid, DAB), ornithine (Orn), and amino acids having at least one positive charge selected from the group consisting of amino acid analogs.
본 발명에 있어서, 전하 부여를 위한 펩티드 핵산 단량체의 변형은 상기 backbone 변형 이외에도 nucleobase가 변형된 펩티드핵산 단량체를 사용할 수 있다. 바람직하게는 전기적 양성을 가지도록 amine, triazole, imidazole moiety를 nucleobase에 포함하거나, 전기적 음성을 가지도록 carboxylic acid를 염기에 포함하는 것을 특징으로 할 수 있다.In the present invention, the modification of the peptide nucleic acid monomer for charge imparting may use a peptide nucleic acid monomer having a modified nucleobase in addition to the backbone modification. Preferably, an amine, triazole, or imidazole moiety may be included in the nucleobase to have an electronegative property, or a carboxylic acid may be included in the base to have an electronegative property.
본 발명에 있어서, 상기 캐리어 펩티드 핵산의 변형 펩티드 핵산 단량체는 음전하를 backbone 혹은 nucleobase에 더 포함할 수 있지만, 변형 펩티드 핵산 단량체는 양전하를 가지는 단량체가 음전하를 가지는 단량체에 비해 더 많이 포함되어 전체적으로 캐리어 펩티드 핵산의 전하가 양성이 되는 것이 바람직하다.In the present invention, the modified peptide nucleic acid monomer of the carrier peptide nucleic acid may further contain a negative charge in the backbone or nucleobase, but the modified peptide nucleic acid monomer contains more positively charged monomers than monomers having negative charges, so that the carrier peptide as a whole is formed. It is preferred that the charge of the nucleic acid be positive.
바람직하게는 본 발명에 따른 상기 핵산 복합체는 전체적으로 양의 전하를 가지는 것을 특징으로 한다.Preferably, the nucleic acid complex according to the present invention has a positive charge as a whole.
본 발명에 따른 상기 핵산 복합체에 있어, 소수성 잔기(hydrophobic moiety), 친수성 잔기(hydrophilic moiety), 표적 항원 특이적 항체, 앱타머 또는 형광/발광 표지자 등으로 구성된 군에서 선택된 하나 이상의 물질이 생활성 핵산 및/또는 캐리어 펩티드 핵산에 결합된 것을 특징으로 할 수 있으며, 바람직하게는 상기 소수성 잔기(hydrophobic moiety), 친수성 잔기(hydrophilic moiety), 표적 항원 특이적 항체, 앱타머 및 이미징을 위한 형광/발광 표지자 등으로 구성된 군에서 선택된 하나 이상의 물질은 상기 캐리어 펩티드 핵산에 결합된 것일 수 있다.In the nucleic acid complex according to the present invention, at least one material selected from the group consisting of a hydrophobic moiety, a hydrophilic moiety, a target antigen-specific antibody, an aptamer, or a fluorescent/luminescent marker is a bioactive nucleic acid And / or may be characterized in that it is bound to a carrier peptide nucleic acid, preferably the hydrophobic moiety, the hydrophilic moiety, a target antigen-specific antibody, an aptamer, and a fluorescent / luminescent marker for imaging One or more substances selected from the group consisting of and the like may be bound to the carrier peptide nucleic acid.
본 발명에서 상기 소수성 잔기(hydrophobic moiety), 친수성 잔기(hydrophilic moiety), 표적 항원 특이적 항체, 앱타머, 소광자, 형광 표지자 및 발광 표지자로 구성된 군에서 선택된 하나 이상의 물질과 생활성 핵산 및/또는 캐리어 펩티드 핵산의 결합은 단순 공유결합 또는 링커로 매개된 공유결합인 것을 특징으로 할 수 있으나, 이에 한정되는 것은 아니다. 바람직하게는, 상기 핵산 운반체에 결합된 세포투과, 용해도, 안정성, 운반 및 이미징 관련 물질(예컨대, 소수성 잔기 등)은 표적 유전자의 발현을 조절하는 생활성 핵산과 독립적으로 존재하게 된다.In the present invention, at least one material selected from the group consisting of a hydrophobic moiety, a hydrophilic moiety, a target antigen-specific antibody, an aptamer, a quencher, a fluorescent marker, and a luminescent marker, and a bioactive nucleic acid and/or The binding of the carrier peptide nucleic acid may be characterized as a simple covalent bond or a covalent bond mediated by a linker, but is not limited thereto. Preferably, substances related to cell permeation, solubility, stability, delivery and imaging (eg, hydrophobic residues, etc.) bound to the nucleic acid carrier exist independently of the bioactive nucleic acid that regulates the expression of the target gene.
본 발명에 있어서, 앞서 기술한 바와 같이, 상기 생활성 핵산 및 캐리어 펩티드 핵산의 상보적인 결합 형태는 크게 역평행결합(antiparallel binding)과 평행결합(parallel binding)의 형태를 가지는 것을 특징으로 할 수 있다. 상기 상보적인 결합 형태는 생활성 핵산의 목적서열(생활성 핵산과 상보적인 서열) 존재 하에서 분리되는 구조를 가진다.In the present invention, as described above, the complementary binding form of the bioactive nucleic acid and the carrier peptide nucleic acid may be largely characterized by having the form of antiparallel binding and parallel binding. . The complementary binding form has a structure that separates in the presence of a target sequence of a bioactive nucleic acid (a sequence complementary to the bioactive nucleic acid).
상기 역평행결합과 평행결합은 DNA-DNA 또는 DNA-PNA의 결합 방식에 있어, 5'-방향성과 3'-방향성에 따라 결정된다. 역평행 결합은 일반적인 DNA-DNA 또는 DNA-PNA의 결합 방식으로, 본 발명에 따른 핵산 복합체를 예로 들어 설명하면, 생활성 핵산은 5'에서 3' 방향으로, 캐리어 펩티드 핵산은 3'에서 5' 방향으로 서로 결합되는 형태를 의미한다. 평행결합은 역평행결합에 비해서는 결합력이 다소 떨어지는 형태로, 생활성 핵산과 캐리어 펩티드 핵산 모두가 5'에서 3' 방향 또는 3'에서 5' 방향으로 서로 결합되는 형태를 의미한다.The antiparallel bond and the parallel bond are determined according to the 5'-direction and the 3'-direction in the binding method of DNA-DNA or DNA-PNA. Antiparallel coupling is a general DNA-DNA or DNA-PNA coupling method. Taking the nucleic acid complex according to the present invention as an example, the bioactive nucleic acid is in the 5' to 3' direction and the carrier peptide nucleic acid is in the 3' to 5' direction. It means that the shape is connected to each other in the direction. Parallel binding is a form in which binding force is somewhat lower than that of anti-parallel binding, and refers to a form in which both the bioactive nucleic acid and the carrier peptide nucleic acid are bonded to each other in the 5' to 3' direction or the 3' to 5' direction.
본 발명에 따른 핵산 복합체에 있어서, 바람직하게는 상기 생활성 핵산 및 캐리어 펩티드 핵산의 결합력은 생활성 핵산과 생활성 핵산의 목적하는 유전자, 특히 목적 유전자의 mRNA와의 결합력보다 낮은 것을 특징으로 할 수 있다. 상기 결합력은 융해온도, melting temperature 또는 Tm에 의해 결정된다.In the nucleic acid complex according to the present invention, preferably, the binding force of the bioactive nucleic acid and the carrier peptide nucleic acid is lower than the binding force of the bioactive nucleic acid and the target gene, particularly the mRNA of the target gene. . The binding force is determined by melting temperature, melting temperature or Tm.
상기 생활성 핵산 및 캐리어 펩티드 핵산의 결합력(융해온도, melting temperature, Tm)이 생활성 핵산과 생활성 핵산의 목적하는 유전자, 특히 목적 유전자의 mRNA와의 결합력보다 낮게 하기 위한 구체적인 방법의 예로는, 상기 생활성 핵산과 캐리어 펩티드 핵산이 평행 결합(Parallel binding) 또는 부분 특이결합(Partial specific binding)하는 것을 특징으로 할 수 있으나, 이에 한정되는 것은 아니다.As an example of a specific method for making the binding strength (melting temperature, Tm) of the bioactive nucleic acid and the carrier peptide nucleic acid lower than the binding strength of the bioactive nucleic acid and the bioactive nucleic acid to the target gene, in particular, the mRNA of the target gene, the above Parallel binding or partial specific binding between the bioactive nucleic acid and the carrier peptide nucleic acid may be characterized, but is not limited thereto.
또 다른 예로서 상기 캐리어 펩티드 핵산이 링커, 유니버설 염기(universal base) 및 생활성 핵산의 대응되는 염기와 상보적이지 않은 염기를 가지는 펩티드 핵산 염기로 구성된 군에서 선택된 하나 이상의 펩티드 핵산 염기를 갖는 것을 특징으로 할 수 있으나, 이에 한정되는 것은 아니다.As another example, the carrier peptide nucleic acid has at least one peptide nucleic acid base selected from the group consisting of a linker, a universal base, and a peptide nucleic acid base having a base that is not complementary to a corresponding base of a bioactive nucleic acid. It can be, but is not limited thereto.
본 발명에 있어서, 상기 유니버설 염기(universal base)는 아데닌(adenine), 구아닌(guanine), 사이토신(cytosine), 티민(thymine), 우라실(Uracil) 등의 천연 염기와 선택성 없이 결합하고, 상보적인 결합력보다 낮은 결합력을 가지는 염기로 이노신 PNA(inosine PNA), 인돌 PNA(indole PNA), 나이트로인돌 PNA(nitroindole PNA) 및 무염기(abasic)로 구성된 군에서 선택된 하나 이상 사용할 수 있으며, 바람직하게는 이노신 PNA를 사용하는 것을 특징으로 할 수 있다.In the present invention, the universal base binds to natural bases such as adenine, guanine, cytosine, thymine, and uracil without selectivity, and is complementary to One or more bases selected from the group consisting of inosine PNA, indole PNA, nitroindole PNA, and abasic can be used as a base having a lower binding force than the binding force, preferably. It can be characterized by using inosine PNA.
본 발명에 있어서, 상기 핵산 복합체의 기능 조절을 위한 핵산들의 결합 형태와 전기적 성질 조합을 제공하고, 상기 핵산의 결합 형태와 전기적 성질 조합으로 입자 크기 및 작용 시점을 조절하고, 세포투과성, 용해도 및 특이도를 향상시키는 것을 특징으로 할 수 있다.In the present invention, a combination of the binding form and electrical properties of nucleic acids for function control of the nucleic acid complex is provided, and the combination of the binding form and electrical properties of the nucleic acids controls the particle size and the time point of action, cell permeability, solubility and specificity It can be characterized as improving the degree.
본 발명에 있어서, 상기 생활성 핵산과 캐리어 펩티드 핵산의 결합력 조절을 통해, 목적 유전자의 존재 하에서, 생활성 핵산이 목적 서열과 결합되는 시점(생활성 핵산의 목적 서열로의 치환되는 시점, 표적 특이적 분리 및 결합 시점) 등의 조절이 가능하다.In the present invention, the binding force of the bioactive nucleic acid and the carrier peptide nucleic acid is controlled, in the presence of the target gene, when the bioactive nucleic acid binds to the target sequence (when the bioactive nucleic acid is substituted with the target sequence, the target specific It is possible to adjust the timing of enemy separation and joining).
본 발명에 따른 핵산 복합체에 있어서, 생활성 핵산의 목적 유전자로의 치환(strand displacement) 시점, 표적 특이적 분리 및 결합(target specific release and bind) 시점의 조절은 복합체의 비특이 결합을 위한 캐리어 펩티드 핵산의 비특이 염기, 유니버설 염기 및 linker의 유무, 개수 및 위치에 의하여 조절이 가능한 것을 특징으로 할 수 있다. 상기 펩티드 복합체의 상보적인 결합의 형태인 평행(parallel) 또는 역평행(antiparallel) 형태의 결합 등과 상기 조건의 조합에 의하여 조절이 가능한 것을 특징으로 할 수 있다.In the nucleic acid complex according to the present invention, the control of the strand displacement time point and the target specific release and bind time point of the bioactive nucleic acid into the target gene is a carrier peptide for non-specific binding of the complex It can be characterized in that it can be controlled by the presence, number, and location of non-specific bases, universal bases, and linkers in nucleic acids. It may be characterized in that control is possible by a combination of the above conditions, such as a parallel or antiparallel bond, which is a form of complementary bond of the peptide complex.
본 발명에 있어서, 상기 핵산 복합체의 입자 크기는 생활성 핵산과 캐리어 펩티드 핵산의 전하 밸런스(charge balance)를 조절함으로써 조절되는 것을 특징으로 할 수 있다. 구체적으로는 캐리어 펩티드 핵산의 양전하가 증가하면 입자의 크기가 작아지나, 캐리어 펩티드 핵산의 양전하가 일정 수준을 넘게 되면 입자의 크기가 커지는 특징을 가지게 된다. 또한, 입자 크기를 결정하는 다른 중요한 요소로 복합체를 이루는 생활성 핵산 전하에 따른 전반적인 캐리어 펩티드 핵산과의 적절한 전하 밸런스에 의해서 입자 크기가 결정된다.In the present invention, the particle size of the nucleic acid complex may be characterized in that it is controlled by adjusting the charge balance between the bioactive nucleic acid and the carrier peptide nucleic acid. Specifically, when the positive charge of the carrier peptide nucleic acid increases, the particle size decreases, but when the positive charge of the carrier peptide nucleic acid exceeds a certain level, the particle size increases. In addition, the particle size is determined by an appropriate charge balance with the overall carrier peptide nucleic acid according to the charge of the bioactive nucleic acid forming the complex as another important factor determining the particle size.
본 발명에 따른 캐리어 펩티드 핵산의 양전하는 1개 내지 7개(양전하를 가지는 단량체가 1개 내지 7개 포함됨을 의미한다), 바람직하게는 2개 내지 5개, 가장 바람직하게는 2개 내지 3개이며, 생활성 핵산의 전하는 전하 밸런스의 넷 차지(net charge)가 음전하 0개 내지 5개, 바람직하게는 0개 내지 3개인 것을 특징으로 할 수 있다.The positive charge of the carrier peptide nucleic acid according to the present invention is 1 to 7 (meaning that 1 to 7 monomers having a positive charge are included), preferably 2 to 5, most preferably 2 to 3 And, the charge of the bioactive nucleic acid may be characterized in that the net charge of the charge balance is 0 to 5 negative charges, preferably 0 to 3.
본 발명에 있어서, 상기 핵산 복합체는 생활성 핵산 및 캐리어 펩티드 핵산이 적절한 조건에서 혼성화됨으로써 제조될 수 있다.In the present invention, the nucleic acid complex can be prepared by hybridizing a bioactive nucleic acid and a carrier peptide nucleic acid under appropriate conditions.
본 발명의 “혼성화”는 상보적인 단일가닥 핵산들이 이중-가닥 핵산을 형성하는 것을 의미한다. 혼성화는 2개의 핵산 가닥 간의 상보성이 완전할 경우(perfect match) 일어나거나 또는 일부 부정합(mismatch) 염기가 존재하여도 일어날 수 있다. 혼성화에 필요한 상보성의 정도는 혼성화 조건에 따라 달라질 수 있으며, 특히 결합 온도에 의하여 조절될 수 있다."Hybridization" in the present invention means that complementary single-stranded nucleic acids form a double-stranded nucleic acid. Hybridization can occur when the complementarity between the two nucleic acid strands is perfect (perfect match) or even when some mismatch bases are present. The degree of complementarity required for hybridization may vary depending on hybridization conditions, and may be particularly controlled by binding temperature.
본 발명에 있어서, 상기 핵산 복합체는 혈뇌장벽 투과능을 갖는 것을 특징으로 할 수 있다.In the present invention, the nucleic acid complex may have blood-brain barrier penetrating ability.
본 발명에 있어서, 용어 "혈뇌장벽" 또는 "BBB(Blood-Brain Barrier)"는 본원에서 상호 교환적으로 사용되고, 이는 혈액과 뇌 조직 간에 교환되는 것을 면밀히 조절하고 심하게 제한하는 뇌 조직을 통하여 순환되기 때문에 혈액 내에 존재하는 투과성 장벽을 지칭하기 위해 사용된다. 혈액 뇌 장벽 성분에는 모든 혈관의 가장 깊은 내막을 형성하는 내피 세포, BBB와 구조적으로 상관이 있는 인접한 내피 세포들 간의 조밀한 연접부, 내피 세포의 기저 막 및 혈관의 노출된 외부 표면의 거의 모두를 덮고 있는 가까운 성상세포의 확장된 족양 돌기(foot process)가 포함된다. BBB는 혈액 내의 대부분의 물질, 예를 들어 대부분의 대분자, 예를 들면 Ig, 항체, 보체, 알부민 및 약물 및 소분자가 뇌 조직 내로 유입되지 못하게 한다.In the present invention, the term "blood-brain barrier" or "BBB (Blood-Brain Barrier)" is used interchangeably herein, which closely regulates and severely restricts the exchange between blood and brain tissue and is circulated through brain tissue. Therefore, it is used to refer to the permeability barrier present in the blood. Components of the blood brain barrier include the endothelial cells that form the deepest lining of all blood vessels, the dense junctions between adjacent endothelial cells that are structurally correlated with the BBB, the basement membrane of endothelial cells, and almost all of the exposed outer surface of blood vessels. An enlarged foot process of the overlying astrocytes is included. The BBB prevents most substances in the blood, including most large molecules, such as Ig, antibodies, complement, albumin, and drugs and small molecules, from entering brain tissue.
본 발명의 일 실시예에서, 본 발명에 따른 핵산 복합체를 마우스 미정맥에 투여한 결과, 뇌 조직 내 표적 유전자 및 하위 유전자 발현 억제능을 확인하였으며, 파킨슨병에서 나타나는 운동기능 상실을 개선하는 효과를 나타냄을 확인한 바, 상기 핵산 복합체는 BBB 투과능을 갖는 것을 시사한다.In one embodiment of the present invention, as a result of administering the nucleic acid complex according to the present invention to the tail vein of a mouse, the ability to inhibit the expression of target genes and subgenes in brain tissue was confirmed, and the effect of improving motor function loss in Parkinson's disease was confirmed As confirmed, the nucleic acid complex suggests that it has BBB penetrating ability.
본 발명의 “표적 유전자”는 활성, 억제 또는 표지하고자 하는 핵산 서열(염기서열)을 의미하며, 용어 “표적 핵산”과 차이가 없으며, 본 명세서에서 혼용된다.The “target gene” of the present invention refers to a nucleic acid sequence (base sequence) to be activated, inhibited, or labeled, and is not different from the term “target nucleic acid”, and is used interchangeably herein.
본 발명에 있어서, 표적 유전자를 포함하는 표적 핵산(염기서열)이 생체 외(in vitro) 또는 생체 내(in vivo)에서 상기 복합체와 접촉(결합)하게 되면, 캐리어 펩티드 핵산으로부터 생활성 핵산이 분리되어 생물학적 활성을 나타내게 된다.In the present invention, when a target nucleic acid (base sequence) containing a target gene is contacted (bound) with the complex in vitro or in vivo , bioactive nucleic acid is separated from the carrier peptide nucleic acid and exhibit biological activity.
본 발명에 있어서, 상기 핵산 복합체를 이용하여 예방, 치료할 수 있는 질병은, 상기 핵산 복합체 내의 생활성 핵산의 표적 유전자에 따라 결정될 수 있다. 본 발명에서는 생활성 핵산의 표적 유전자는 NLRP3인 것을 특징으로 할 수 있다.In the present invention, diseases that can be prevented or treated using the nucleic acid complex may be determined according to the target gene of the bioactive nucleic acid in the nucleic acid complex. In the present invention, the target gene of the bioactive nucleic acid may be characterized in that NLRP3.
따라서, 본 발명은 다른 관점에서, NLRP3 유전자를 표적으로 하는 생활성 핵산(Bioactive Nucleic Acid); 및 캐리어 펩티드 핵산(Carrier Peptide Nucleic Acid)이 상보적으로 결합된 핵산 복합체를 유효성분으로 함유하는 퇴행성 뇌질환 예방 또는 치료용 약학 조성물에 관한 것이다.Therefore, the present invention from another point of view, a bioactive nucleic acid targeting the NLRP3 gene (Bioactive Nucleic Acid); And a pharmaceutical composition for preventing or treating degenerative brain disease containing a nucleic acid complex complementary to a carrier peptide nucleic acid as an active ingredient.
본 발명에서 상기 핵산 복합체를 이용하여 예방, 치료할 수 있는 질병은 바람직하게는 퇴행성 뇌질환(Degenerative Brain Disease)일 수 있으며, 상기 퇴행성 뇌질환은 파킨슨병(Parkinson's disease), 알츠하이머병(Alzheimer's disease), 니만-피크 병(Niemann-Pick disease), 크로이츠펠트-야콥병(Creutzfeldt-Jakob disease), 헌팅턴병(Huntington's disease), 루게릭병(amyotrophic lateral sclerosis; 근위축성 측삭경화증), 다발성 경화증(multiple sclerosis) 또는 치매(dementia)인 것을 특징으로 할 수 있으나, 이에 한정되는 것은 아니다.In the present invention, the disease that can be prevented and treated using the nucleic acid complex may preferably be a degenerative brain disease, and the degenerative brain disease includes Parkinson's disease, Alzheimer's disease, Niemann-Pick disease, Creutzfeldt-Jakob disease, Huntington's disease, amyotrophic lateral sclerosis (amyotrophic lateral sclerosis), multiple sclerosis or dementia ( dementia), but is not limited thereto.
본 발명에서 사용되는 용어 “예방”은, 상기 핵산 복합체를 포함하는 조성물의 투여로 질환의 발병을 막거나, 이의 진행을 지연시키는 모든 행위를 의미한다. 또한, 본 발명에서 사용되는 용어 “치료”는, 상기 핵산 복합체를 포함하는 조성물의 투여로 질환의 증세가 호전되거나 증상의 경감 또는 완치되는 모든 행위를 의미한다.As used herein, the term "prevention" refers to any action that prevents the onset of a disease or delays its progression by administering a composition containing the nucleic acid complex. In addition, the term “treatment” used in the present invention refers to all activities in which symptoms of a disease are improved, symptoms are alleviated, or cured by administration of a composition containing the nucleic acid complex.
본 발명에 따른 핵산 복합체를 포함하는 약학 조성물은 약학적으로 유효한 양의 상기 핵산 복합체를 단독으로 포함하거나, 하나 이상의 약학적으로 허용되는 담체, 부형제 또는 희석제를 포함할 수 있다. 상기에서 약학적으로 유효한 양이란 퇴행성 뇌질환의 증상을 예방, 개선 및 치료하기에 충분한 양을 말한다.A pharmaceutical composition comprising a nucleic acid complex according to the present invention may include a pharmaceutically effective amount of the nucleic acid complex alone, or may include one or more pharmaceutically acceptable carriers, excipients or diluents. In the above, the pharmaceutically effective amount refers to an amount sufficient to prevent, improve, and treat symptoms of degenerative brain disease.
상기 “약학적으로 허용되는”이란 생리학적으로 허용되고 인간에게 투여될 때, 통상적으로 위장 장애, 현기증과 같은 알레르기 반응 또는 이와 유사한 반응을 일으키지 않는 조성물을 말한다. 상기 담체, 부형제 및 희석제의 예로는, 락토즈, 덱스트로즈, 수크로즈, 솔비톨, 만니톨, 자일리톨, 에리스리톨, 말티톨, 전분, 아카시아 고무, 알지네이트, 젤라틴, 칼슘 포스페이트, 칼슘 실리케이트, 셀룰로즈, 메틸 셀룰로즈, 폴리비닐피롤리돈, 물, 메틸하이드록시벤조에이트, 프로필하이드록시벤조에이트, 탈크, 마그네슘 스테아레이트 및 광물유를 들 수 있다. 또한 상기 약학 조성물은 충진제, 항응집제, 윤활제, 습윤제, 향료, 유화제 및 방부제 등을 추가로 포함할 수 있다.The term "pharmaceutically acceptable" refers to a composition that is physiologically acceptable and does not usually cause allergic reactions such as gastrointestinal disorders and dizziness or similar reactions when administered to humans. Examples of the carrier, excipient and diluent include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia gum, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, polyvinylpyrrolidone, water, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil. In addition, the pharmaceutical composition may further include fillers, anti-coagulants, lubricants, wetting agents, flavoring agents, emulsifiers and preservatives.
용어 “담체(carrier)”는 세포 또는 조직 내로의 핵산 복합체의 부가를 용이하게 하는 화합물로 정의된다. 예를 들어, 디메틸술폭사이드(DMSO)는 생물체의 세포 또는 조직 내로의 많은 유기 화합물들의 투입을 용이하게 하는 통상 사용되는 담체이다.The term "carrier" is defined as a compound that facilitates the addition of a nucleic acid complex into a cell or tissue. For example, dimethylsulfoxide (DMSO) is a commonly used carrier that facilitates the introduction of many organic compounds into the cells or tissues of living organisms.
용어 “희석제(diluent)”는 대상 화합물의 생물학적 활성 형태를 안정화시킬 뿐만 아니라, 화합물을 용해시키게 되는 물에서 희석되는 화합물로 정의된다. 버퍼 용액에 용해되어 있는 염은 당해 분야에서 희석제로 사용된다. 통상 사용되는 버퍼 용액은 포스페이트 버퍼 식염수이며, 이는 인간 용액의 염 상태를 모방하고 있기 때문이다. 버퍼 염은 낮은 농도에서 용액의 pH를 제어할 수 있기 때문에, 버퍼 희석제가 화합물의 생물학적 활성을 변형하는 일은 드물다.The term “diluent” is defined as a compound that is diluted in water which will dissolve the compound as well as stabilize the biologically active form of the subject compound. Salts dissolved in buffer solutions are used as diluents in the art. A commonly used buffer solution is phosphate buffered saline because it mimics the salt state of human solutions. Because buffer salts can control the pH of a solution at low concentrations, buffer diluents rarely modify the biological activity of a compound.
본 발명에서의 핵산 복합체를 함유하는 물질은, 환자에게 그 자체로서 또는 병용 요법(combination therapy)에서와 같이 다른 활성 성분들과 함께 또는 적당한 담체나 부형제와 함께 혼합된 의약 조성물로서 투여될 수 있다.A substance containing a nucleic acid complex in the present invention can be administered to a patient by itself or as a mixed pharmaceutical composition together with other active ingredients, such as in combination therapy, or with suitable carriers or excipients.
본 발명의 조성물은 투여된 후 활성 성분의 신속, 지속 또는 지연된 방출을 제공할 수 있도록 당업계에 공지된 방법을 사용하여 제형화될 수 있다. 제형은 분말, 과립, 정제, 에멀젼, 시럽, 에어로졸, 연질 또는 경질 젤라틴 캅셀, 멸균 주사용액, 멸균 분말의 형태일 수 있다.Compositions of the present invention may be formulated using methods known in the art to provide rapid, sustained or delayed release of the active ingredient after administration. The dosage form may be in the form of a powder, granule, tablet, emulsion, syrup, aerosol, soft or hard gelatin capsule, sterile injectable solution, or sterile powder.
본 발명의 조성물은 경구, 경피, 피하, 정맥 또는 근육을 포함한 여러 경로를 통해 투여될 수 있으며, 활성 성분의 투여량은 투여 경로, 환자의 연령, 성별, 체중 및 환자의 중증도 등의 여러 인자에 따라 적절히 선택될 수 있다.The composition of the present invention can be administered through various routes including oral, transdermal, subcutaneous, intravenous or intramuscular, and the dosage of the active ingredient depends on various factors such as the route of administration, age, sex, weight and severity of the patient. can be selected appropriately.
본 발명에서 사용에 적합한 의약 조성물에는, 활성 성분들이 그것의 의도된 목적을 달성하기에 유효한 양으로 함유되어 있는 조성물이 포함된다. 더욱 구체적으로, 치료적 유효량은 치료될 객체의 생존을 연장하거나, 질환의 증상을 방지, 경감 또는 완화시키는데 유효한 화합물의 양을 의미한다. 치료적 유효량의 결정은, 특히, 여기에 제공된 상세한 개시 내용 측면에서, 통상의 기술자의 능력 범위 내에 있다.Pharmaceutical compositions suitable for use in the present invention include compositions in which the active ingredients are contained in effective amounts to achieve their intended purpose. More specifically, a therapeutically effective amount refers to an amount of a compound effective to prolong the survival of the subject being treated or to prevent, alleviate or ameliorate symptoms of a disease. Determination of a therapeutically effective amount is well within the ability of those skilled in the art, especially in light of the detailed disclosure provided herein.
본 발명의 핵산 복합체의 투여에 있어서, 당업계에 공지된 임의의 핵산 전달 방법이 사용될 수 있다. 이에 제한되는 것은 아니나, 적합한 전달 시약은 예를 들어, Mirus Transit TKO lipophilic reagent, lipofectin, lipofectamine, cellfectin, polycations(예를 들어, polylysine), atelocollagen, nanoplexes 및 리포좀이 있다. 핵산 분자의 전달 운반체로서 아테로콜라겐(atelocollagen)의 사용은 Minakuchi et al. Nucleic Acids Res., 32(13):e109 (2004); Hanai et al. Ann NY Acad Sci., 1082:9-17 (2006); and Kawata et al. Mol Cancer Ther., 7(9):2904-12 (2008)에 기술되어 있다. 예시적인 간섭 핵산 전달 시스템은 미국 특허 제8,283,461호, 제8,313,772호, 제8,501,930호에 제공된다.For administration of the nucleic acid complexes of the present invention, any nucleic acid delivery method known in the art may be used. Although not limited thereto, suitable delivery reagents include, for example, Mirus Transit TKO lipophilic reagent, lipofectin, lipofectamine, cellfectin, polycations (eg, polylysine), atelocollagen, nanoplexes, and liposomes. The use of atelocollagen as a delivery vehicle for nucleic acid molecules has been described by Minakuchi et al. Nucleic Acids Res., 32(13):e109 (2004); Hanai et al. Ann NY Acad Sci., 1082:9-17 (2006); and Kawata et al. Mol Cancer Ther., 7(9):2904-12 (2008). Exemplary interfering nucleic acid delivery systems are provided in U.S. Patent Nos. 8,283,461, 8,313,772, and 8,501,930.
본 발명에 있어서, 상기 핵산 복합체는 리포솜(liposome) 등의 전달체를 이용하여 투여할 수도 있다. 상기 리포솜은 림프 조직과 같은 특정 조직에 대해 상기 복합체를 표적화하거나, 감염 세포에 대해 선택적으로 표적화하는데 도움을 줄 수 있고, 또한 상기 복합체가 포함된 조성물의 반감기를 증가시키는데 도움을 줄 수 있다. 리포솜으로는 에멀션, 포움(foam), 마이셀(micelle), 불용성 단층, 액정, 인지질 분산물, 라멜라 층(lamellar layer) 등이 있다. 이러한 제제에 있어서, 송달되는 상기 복합체는, 단독으로 또는 특정 세포들을 대상으로, CD45 항원에 결합되는 모노클로날 항체와 같은 림프 세포 중 우세한 수용체에 결합하는 분자와 함께 또는 기타 치료 조성물과 함께, 리포솜의 일부로서 혼입시킨다. 따라서, 본 발명의 소정 복합체로 충진되거나 도포되어(decorated) 상기 핵산 복합체 조성물을 송달하는 리포솜은 림프 세포의 상기 부위로 지향될 수 있다.In the present invention, the nucleic acid complex may be administered using a delivery system such as a liposome. The liposome can help target the complex to a specific tissue, such as lymphoid tissue, or selectively target infected cells, and can also help increase the half-life of a composition containing the complex. Liposomes include emulsions, foams, micelles, insoluble monolayers, liquid crystals, phospholipid dispersions, lamellar layers, and the like. In such formulations, the complex to be delivered, alone or to specific cells, in combination with a molecule that binds to a receptor prevalent in lymphocytes, such as a monoclonal antibody that binds to the CD45 antigen, or in combination with other therapeutic compositions, is a liposome. incorporated as part of Thus, liposomes filled or decorated with a given complex of the present invention to deliver the nucleic acid complex composition can be directed to the site of lymphocytes.
본 발명에 따라 사용하기 위한 리포솜은 일반적으로 중성 및 음전하 인지질 및 콜레스테롤 등의 스테롤을 비롯한 표준 베시클(vesicle)-형성 지질로부터 형성된다. 일반적으로, 예를 들어 혈류 중의 리포솜의 안정성, 산 불안정성(acid lability) 및 리포솜의 크기 등을 고려하여 지질을 선택한다. 리포솜 제조에는 다양한 방법을 이용할 수 있다. 예를 들어, 문헌[Szoka, et al., Ann. Rev. Biophys. Bioeng., 9:467, 1980), 및 미국 특허 제4,235,871호, 제4,501,728호, 제4,837,028호 및 제5,019,369호]에 기재되어 있는 바와 같은 방법을 이용할 수 있다.Liposomes for use in accordance with the present invention are generally formed from standard vesicle-forming lipids, including neutral and negatively charged phospholipids and sterols such as cholesterol. In general, lipids are selected in consideration of, for example, stability of liposomes in the blood stream, acid lability, and size of liposomes. A variety of methods can be used to prepare liposomes. See, eg, Szoka, et al., Ann. Rev. Biophys. Bioeng., 9:467, 1980), and US Pat. Nos. 4,235,871, 4,501,728, 4,837,028 and 5,019,369.
본 발명은 또 다른 관점에서, NLRP3 유전자를 표적으로 하는 생활성 핵산(Bioactive Nucleic Acid); 및 캐리어 펩티드 핵산(Carrier Peptide Nucleic Acid)이 상보적으로 결합된 핵산 복합체를 개체에 투여하는 단계를 포함하는 퇴행성 뇌질환 예방 또는 치료방법에 관한 것이다.In another aspect, the present invention provides a bioactive nucleic acid targeting the NLRP3 gene; And a carrier peptide nucleic acid (Carrier Peptide Nucleic Acid) It relates to a method for preventing or treating degenerative brain disease comprising administering to a subject a nucleic acid complex complementary thereto.
본 발명은 또 다른 관점에서, 퇴행성 뇌질환의 예방 또는 치료를 위한 NLRP3 유전자를 표적으로 하는 생활성 핵산(Bioactive Nucleic Acid); 및 캐리어 펩티드 핵산(Carrier Peptide Nucleic Acid)이 상보적으로 결합된 핵산 복합체의 용도에 관한 것이다.In another aspect, the present invention provides a bioactive nucleic acid targeting the NLRP3 gene for the prevention or treatment of degenerative brain diseases; And it relates to the use of a nucleic acid complex in which a carrier peptide nucleic acid is complementarily bound.
본 발명은 또 다른 관점에서, 퇴행성 뇌질환의 예방 또는 치료용 약제 제조를 위한 NLRP3 유전자를 표적으로 하는 생활성 핵산(Bioactive Nucleic Acid); 및 캐리어 펩티드 핵산(Carrier Peptide Nucleic Acid)이 상보적으로 결합된 핵산 복합체의 사용에 관한 것이다.In another aspect, the present invention provides a bioactive nucleic acid targeting the NLRP3 gene for the preparation of drugs for preventing or treating degenerative brain diseases; And it relates to the use of a nucleic acid complex in which a carrier peptide nucleic acid is complementaryly bound.
본 발명에서, “개체”는 본 발명에 따른 핵산 복합체를 투여하여 경감, 억제 또는 치료될 수 있는 상태 또는 질환을 앓고 있거나 그러한 위험이 있는 포유동물을 의미하며, 바람직하게 사람을 의미한다.In the present invention, “subject” means a mammal suffering from or at risk of a condition or disease that can be alleviated, suppressed or treated by administering a nucleic acid complex according to the present invention, and preferably means a human.
또한, 본 발명의 핵산 복합체의 인체에 대한 투여량은 환자의 나이, 몸무게, 성별, 투여 형태, 건강 상태 및 질환 정도에 따라 달라질 수 있다.In addition, the dose of the nucleic acid complex of the present invention to the human body may vary depending on the patient's age, weight, sex, dosage form, health condition and disease severity.
본 명세서에 기재되어 있는 핵산 복합체를 포함하는 조성물의 독성과 치료 효율성은, 예를 들어, LD50(군집의 50%에 대한 치사량), ED50(군집의 50%에 대해 치료 효과를 갖는 선량), IC50(군집의 50%에 대해 치료 억제 효과를 갖는 선량)을 결정하기 위하여, 세포 배양 또는 실험동물에서의 표준 제약 과정들에 의해 산정될 수 있다. 독성과 치료 효과 간의 선량 비가 치료 지수이고 이것은 LD50과 ED50(또는, IC50) 간의 비율로서 표현될 수 있다. 높은 치료 지수를 보이는 화합물들이 바람직하다. 이들 세포 배양 분석에서 얻어진 데이터는 인간에 사용하는 선량의 범위를 산정하는데 사용될 수 있다. 그러한 화합물들의 투여량(dosage) 또는 도포량은 바람직하게는 독성이 없거나 거의 없는 상태에서 ED50(또는, IC50)을 포함하는 순환 농도의 범위 내에 있다.Toxicity and therapeutic efficacy of compositions comprising the nucleic acid complexes described herein can be measured by, for example, the LD50 (lethal dose to 50% of the population), the ED50 (the dose that is therapeutically effective in 50% of the population), the IC50 It can be estimated by standard pharmaceutical procedures in cell culture or laboratory animals to determine (the dose that has a therapeutically inhibitory effect on 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between the LD50 and the ED50 (or IC50). Compounds exhibiting high therapeutic indices are preferred. Data obtained from these cell culture assays can be used to estimate a range of human doses. The dosage or applied amount of such compounds lies preferably within a range of circulating concentrations that include the ED50 (or IC50) with little or no toxicity.
본 발명의 용어 “투여”란, 어떠한 적절한 방법으로 개체에게 본 발명의 약학 조성물을 도입하는 행위를 의미하며, 투여 경로는 목적 조직에 도달할 수 있는 한 경구 또는 비경구의 다양한 경로를 통하여 투여될 수 있다.The term "administration" of the present invention refers to the act of introducing the pharmaceutical composition of the present invention to a subject by any appropriate method, and the route of administration may be administered through various oral or parenteral routes as long as it can reach the target tissue. there is.
본 발명의 약학 조성물의 투여 경로는 목적 조직에 도달할 수 있는 한 어떠한 일반적인 경로를 통하여도 투여될 수 있다. 본 발명의 약학 조성물은 특별히 이에 제한되지 않으나, 목적하는 바에 따라 복강내 투여, 정맥내 투여, 근육내 투여, 피하 투여, 피내 투여, 경구 투여, 비내 투여, 폐내 투여, 직장내 투여될 수 있다. 또한 상기 조성물은 활성 물질이 표적 세포로 이동할 수 있는 임의의 장치에 의해 투여될 수 있다.The administration route of the pharmaceutical composition of the present invention may be administered through any general route as long as it can reach the target tissue. The pharmaceutical composition of the present invention is not particularly limited thereto, but may be administered intraperitoneally, intravenously, intramuscularly, subcutaneously, intradermally, orally, intranasally, intrapulmonaryly, or intrarectally, as desired. In addition, the composition may be administered by any device capable of transporting an active substance to a target cell.
본 발명의 약학 조성물은 개별 치료제로 투여하거나 다른 치료제와 병용하여 투여될 수 있고 종래의 치료제와는 순차적 또는 동시에 투여될 수 있다. 그리고 단일 또는 다중 투여될 수 있다. 상기 요소를 모두 고려하여 부작용 없이 최소한의 양으로 최대 효과를 얻을 수 있는 양을 투여하는 것이 중요하다.The pharmaceutical composition of the present invention may be administered as an individual therapeutic agent or in combination with other therapeutic agents, and may be administered sequentially or simultaneously with conventional therapeutic agents. And it can be single or multiple administrations. It is important to administer the amount that can obtain the maximum effect with the minimum amount without side effects in consideration of all the above factors.
이하, 실시예를 통하여 본 발명을 더욱 상세히 설명하고자 한다. 이들 실시예는 오로지 본 발명을 예시하기 위한 것으로서, 본 발명의 범위가 이들 실시예에 의해 제한되는 것으로 해석되지 않는 것은 당업계에서 통상의 지식을 가진 자에 있어서 자명할 것이다.Hereinafter, the present invention will be described in more detail through examples. These examples are only for exemplifying the present invention, and it will be apparent to those skilled in the art that the scope of the present invention is not to be construed as being limited by these examples.
실시예 1: 생활성 펩티드 핵산 및 캐리어 펩티드 핵산, 및 이들을 이용한 복합체의 제조Example 1: Preparation of bioactive peptide nucleic acids and carrier peptide nucleic acids, and complexes using them
본 발명에서는 핵산 복합체의 파킨슨병에 대한 효과를 검정하기 위하여 표적 유전자로 NLRP3를 사용하였으며, 파킨슨병의 치료 효과를 확인하기 위하여 NLRP3에 대한 생활성 펩티드 핵산(Bioactive Peptide Nucleic Acid)으로 안티센스 펩티드 핵산(antisense PNA)을 사용하였다.In the present invention, NLRP3 was used as a target gene to test the effect of the nucleic acid complex on Parkinson's disease, and to confirm the therapeutic effect of Parkinson's disease, antisense peptide nucleic acid (Bioactive Peptide Nucleic Acid) for NLRP3 was used. antisense PNA) was used.
본 발명의 대조군으로 사용한 생활성 핵산(antisense PNA)은 서열번호 1로 표시되는 서열을 포함하며, 파킨슨병 치료 효과를 확인하기 위하여 사용한 생활성 펩티드 핵산(antisense PNA)은 서열번호 2로 표시되는 서열을 포함한다. 본 발명의 실시예에서 사용된 캐리어 펩티드 핵산은 서열번호 3 내지 5로 기재되는 서열로 구성되어 있다(표 1). 본 발명에서 사용된 모든 펩티드 핵산은 파나진(PANAGENE, 한국)에서 HPLC 정제 방법을 통해 합성하였다.The bioactive nucleic acid (antisense PNA) used as a control of the present invention includes the sequence represented by SEQ ID NO: 1, and the bioactive peptide nucleic acid (antisense PNA) used to confirm the therapeutic effect of Parkinson's disease has the sequence represented by SEQ ID NO: 2 includes The carrier peptide nucleic acids used in the examples of the present invention are composed of sequences represented by SEQ ID NOs: 3 to 5 (Table 1). All peptide nucleic acids used in the present invention were synthesized by HPLC purification method from PANAGENE (Korea).
Figure PCTKR2023000259-appb-img-000001
Figure PCTKR2023000259-appb-img-000001
단량체의 변형은 전기적인 성질을 부여하기 위하여 펩티드 핵산의 backbone을 전기적 양성은 리신(Lysine, Lys, K, (+)로 표기)으로, 전기적 음성은 글루타민산(Glutamic acid, Glu, E, (-)로 표기)으로 변형된 펩티드 backbone을 가지도록 제작하였다.Modification of the monomers converts the backbone of the peptide nucleic acid to lysine (indicated by Lysine, Lys, K, (+) ) for electrical properties and glutamic acid (Glutamic acid, Glu, E, (-)) for electrical negative to impart electrical properties. ) was constructed to have a modified peptide backbone.
각각의 생활성 핵산과 캐리어 펩티드 핵산 조합은 DMSO 하에서 혼성화 되었으며, 그 결과 생활성 핵산과 캐리어 펩티드 핵산으로 구성된 복합체가 제작되었다.Each combination of the bioactive nucleic acid and the carrier peptide nucleic acid was hybridized in the presence of DMSO, and as a result, a complex composed of the bioactive nucleic acid and the carrier peptide nucleic acid was prepared.
실시예 2: 핵산 복합체를 이용한 파킨슨병의 치료 효과 분석Example 2: Analysis of treatment effects of Parkinson's disease using nucleic acid complexes
실시예 1에 따라, 하기 표 2의 구조로 제조된 NLRP3를 표적 유전자로 하는 생활성 펩티드 핵산 및 캐리어 펩티드 핵산을 포함하는 핵산 복합체를 이용하여 파킨슨병의 치료 효과를 분석하였다.According to Example 1, the therapeutic effect of Parkinson's disease was analyzed using a nucleic acid complex comprising a carrier peptide nucleic acid and a bioactive peptide nucleic acid targeting NLRP3, which was prepared according to the structure shown in Table 2 below.
Figure PCTKR2023000259-appb-img-000002
Figure PCTKR2023000259-appb-img-000002
실시예 2-1: 세포 배양Example 2-1: Cell culture
ATCC(American Type Culture Collection, 미국)로부터 입수한 인간 유래 미세아교세포(HMC-3)를 MEM 배양 배지(웰진, 한국)에 10%(v/v) 소태아혈청, 페니실린 100units/㎖, 스트렙토마이신 100㎍/㎖을 첨가하고, 37℃, 5%(v/v) CO2의 조건하에서 배양하였다.Human-derived microglia (HMC-3) obtained from ATCC (American Type Culture Collection, USA) were mixed with 10% (v/v) fetal bovine serum, penicillin 100 units/ml, and streptomycin in MEM culture medium (Wellgene, Korea). 100 μg/ml was added and cultured under conditions of 37°C and 5% (v/v) CO 2 .
실시예 2-2: 웨스턴 블랏 분석(Western blot assay)을 이용한 유전자 발현의 분석Example 2-2: Analysis of gene expression using Western blot assay
인간 유래 미세아교세포주를 6웰 플레이트에 1x105으로 세포를 씨딩하고 24시간 배양 후, 면역 반응 유도를 위해 Lipopolysaccharide(LPS, Sigma, 미국)를 1μg/ml로 처리하였다. LPS 처리 4시간 뒤, 실시예 2-1의 조건으로 실험을 진행하여 생활성 펩티드 핵산 및 캐리어 펩티드 핵산을 포함하는 복합체를 처리하여 각각 24, 48, 72, 96, 120시간 배양하였고, RIPA buffer를 각 웰에 30μL씩 첨가하여 protein lysate를 얻었다. Protein lysate를 BCA assay kit(Thermo Fisher, 미국)를 사용하여 단백질 양을 정량하고 단백질 30μg을 전기영동을 통해 사이즈 별로 분리하여, PVDF membrane에 단백질을 옮긴 후 1차 항체인 NLRP3(ABclonal, 미국), Pro Cas-1(abcam, 미국), Pro IL-1β(abcam, 미국)를 1:1000으로 처리하고 4℃에서 하루 동안 방치하였다. 1X TBS-T를 이용하여 워싱하고 2차 항체인 Goat Anti-Rabbit(Cell signaling Technology, 미국)을 1:2000으로 처리하여 상온에서 1시간 방치하였다. SupersignalTM West Femto Maximum Sensitivity Substrate(Thermo Fisher, 미국)를 처리하고 Image600(Amersham, 독일) 장비를 이용하여 표적 유전자의 발현 억제 효율을 분석하였다.Human-derived microglial cell lines were seeded in 1x10 5 cells in a 6-well plate and cultured for 24 hours, and treated with 1 μg/ml of Lipopolysaccharide (LPS, Sigma, USA) to induce an immune response. After 4 hours of LPS treatment, the experiment was conducted under the conditions of Example 2-1, and the complex containing the bioactive peptide nucleic acid and the carrier peptide nucleic acid was treated and cultured for 24, 48, 72, 96, and 120 hours, respectively, and RIPA buffer Protein lysate was obtained by adding 30 μL to each well. Protein lysate was quantified using a BCA assay kit (Thermo Fisher, USA), and 30 μg of protein was separated by size through electrophoresis, and the protein was transferred to a PVDF membrane. Pro Cas-1 (abcam, USA) and Pro IL-1β (abcam, USA) were treated at a ratio of 1:1000 and left at 4°C for one day. It was washed with 1X TBS-T, treated with a secondary antibody, Goat Anti-Rabbit (Cell signaling Technology, USA) at a ratio of 1:2000, and left at room temperature for 1 hour. Supersignal TM West Femto Maximum Sensitivity Substrate (Thermo Fisher, USA) was treated, and the expression inhibition efficiency of the target gene was analyzed using Image600 (Amersham, Germany) equipment.
그 결과, 도 1에 나타낸 바와 같이 서열번호 2와 5의 조합의 핵산 복합체(조합 3, PNA 3)를 처리한 군에서 표적 유전자 및 하위 경로 유전자의 발현이 가장 많이 억제되는 것을 확인하였다.As a result, as shown in FIG. 1, it was confirmed that the expression of target genes and sub-pathway genes was most suppressed in the group treated with the nucleic acid complex of SEQ ID NOs: 2 and 5 (combination 3, PNA 3).
실시예 2-3: 면역 형광 염색(Immunofluorescence staining)을 이용한 유전자 발현의 분석Example 2-3: Analysis of gene expression using immunofluorescence staining
인간 유래 미세아교세포주를 8웰 플레이트에 3x103으로 세포를 씨딩하고 24시간 배양 후, 면역 반응 유도를 위해 Lipopolysaccharide(LPS, Sigma, 미국)를 1μg/ml로 처리하였다. LPS 처리 4시간 뒤 실시예 2-1의 조건으로 실험을 진행하여 생활성 펩티드 핵산 및 캐리어 펩티드 핵산을 포함하는 복합체를 처리하고 각각 24, 72시간 배양하였고, 4% paraformaldehyde(sigma, 미국)로 세포를 고정하였다. 고정한 세포를 PBS에 녹인 0.1% 트리톤 X-100(sigma, 미국)으로 10분 동안 투과하였고, 1% 소 혈청 알부민(bovine serum albumin; BSA, sigma, 미국)으로 1시간 동안 블로킹 한 후, 1차 항체인 NLRP3(ABclonal, 미국)를 1:100으로 처리하고 4℃에서 하루 동안 방치하였다. 1X PBS를 이용하여 워싱하고 2차 항체인 Alexa 488-goat Anti-Mouse(Abcam Technology, 미국)를 1:200으로 처리하여 상온에서 1시간 방치하였다. DAPI solution(Sigma, 미국)을 1x로 희석하여 10분 방치 후, 형광 현미경(Leica, 독일) 장비를 이용하여 표적 유전자의 발현 억제 효율을 분석하였다.Human-derived microglial cell lines were seeded in 3x10 3 cells in an 8-well plate and cultured for 24 hours, and treated with 1 μg/ml of Lipopolysaccharide (LPS, Sigma, USA) to induce an immune response. After 4 hours of LPS treatment, the experiment was conducted under the conditions of Example 2-1, and the complex containing the bioactive peptide nucleic acid and the carrier peptide nucleic acid was treated, cultured for 24 and 72 hours, respectively, and cells were treated with 4% paraformaldehyde (Sigma, USA). was fixed. The fixed cells were permeabilized with 0.1% Triton X-100 (Sigma, USA) dissolved in PBS for 10 minutes, blocked with 1% bovine serum albumin (BSA, Sigma, USA) for 1 hour, and then The antibody, NLRP3 (ABclonal, USA) was treated at a ratio of 1:100 and left at 4° C. for one day. After washing with 1X PBS, the secondary antibody, Alexa 488-goat Anti-Mouse (Abcam Technology, USA) was treated at a ratio of 1:200 and allowed to stand at room temperature for 1 hour. After diluting the DAPI solution (Sigma, USA) to 1x and leaving it for 10 minutes, the expression inhibition efficiency of the target gene was analyzed using a fluorescence microscope (Leica, Germany).
그 결과, 도 2a에 나타낸 바와 같이 LPS를 처리한 면역 유도군에서 표적 유전자인 NLRP3의 형광 발현이 증가하는 것을 확인하였으며, 이와 비교하였을 때 서열번호 2와 5의 핵산 복합체 조합(PNA 3)을 처리한 군의 형광 발현이 72시간까지 지속적으로 가장 많이 감소한 것을 확인하였다(도 2a). 해당 형광 세기를 ImageJ 프로그램을 사용하여 정량화한 결과, 면역 유도군과 대조군 핵산 복합체 처리군(조합 1, PNA 1) 대비 서열번호 2와 5의 조합의 핵산 복합체(PNA 3)의 형광 세기 정량 수치가 시간 의존적으로 감소하는 것을 확인하였다(도 2b).As a result, as shown in Figure 2a, it was confirmed that the fluorescence expression of the target gene, NLRP3, increased in the immune induction group treated with LPS. It was confirmed that the fluorescence expression of one group continuously decreased the most until 72 hours (Fig. 2a). As a result of quantifying the fluorescence intensity using the ImageJ program, the fluorescence intensity quantification of the nucleic acid complex (PNA 3) of the combination of SEQ ID NOs: 2 and 5 compared to the immune induction group and the control nucleic acid complex treatment group (combination 1, PNA 1) It was confirmed that it decreased in a time-dependent manner (Fig. 2b).
실시예 3: 뇌 조직으로부터 분리한 일차 미세아교세포(primary cultured microglial cell)에서 핵산 복합체를 이용한 파킨슨병의 치료 효과 분석Example 3: Analysis of Parkinson's disease treatment effect using nucleic acid complex in primary cultured microglial cells isolated from brain tissue
실시예 2를 통하여 효과가 검증된 핵산 조합체를 선별한 후, 배양 상태가 체내와 가장 유사한 특징을 가진 신생 랫드(neonatal rat)에서 분리한 일차 미세아교세포를 이용하여 파킨슨병의 치료 효과를 분석하였다.After selecting the nucleic acid combination whose effect was verified through Example 2, the treatment effect of Parkinson's disease was analyzed using primary microglia isolated from neonatal rats having the most similar characteristics to the body in culture conditions. .
사용한 핵산 복합체 조합은 하기 표 3과 같다.Nucleic acid complex combinations used are shown in Table 3 below.
Figure PCTKR2023000259-appb-img-000003
Figure PCTKR2023000259-appb-img-000003
실시예 3-1: 세포 배양Example 3-1: Cell culture
KCLB(Korean Cell Line Bank, 한국)로부터 입수한 인간 유래 신경아세포종(SH-SY5Y)을 DMEM 배양 배지(웰진, 한국)에 10%(v/v) 소태아혈청, 페니실린 100units/㎖, 스트렙토마이신 100㎍/㎖을 첨가하고, 37℃, 5%(v/v) CO2의 조건에서 배양하였다.Human-derived neuroblastoma (SH-SY5Y) obtained from KCLB (Korean Cell Line Bank, Korea) was mixed with 10% (v/v) fetal bovine serum, penicillin 100 units/ml, and streptomycin 100 in DMEM culture medium (Wellgene, Korea). [mu]g/ml was added and cultured under the conditions of 37°C and 5% (v/v) CO 2 .
실시예 3-2: 생쥐의 뇌 조직으로부터 일차 미세아교세포 분리 및 배양Example 3-2: Isolation and culture of primary microglia from mouse brain tissue
일차 미세아교세포를 생쥐의 뇌 조직에서 분리하여 배양하기 위해, 1일령 신생아 SD 랫드(나라바이오텍, 한국)를 구입하여, 뇌 조직을 분리하였다. 수막과 다른 조직 부위를 제거한 피질(Cortex)을 iced 1x HBSS(gibco, 미국)가 담긴 페트리디쉬에 옮긴 후 파이펫을 사용하여 조직을 해리시키고, 1200rpm에서 5분간 원심분리하였다. 원심분리 후, 상층액은 버리고 남은 pellet에 DMEM 배양배지(Dulbecco Modified Eagle Medium, 웰진, 한국)에 10%(v/v) 소태아혈청, 페니실린 100units/㎖, 스트렙토마이신 100㎍/㎖, GlutaMAX(Gibco, 미국) 0.1%를 첨가한 mixed glial cell 배양 배지를 넣어 세포를 풀어준 후, 전날 미리 PLL(Poly-L-lysine, Sigma, 미국)으로 코팅한 75T 플라스크에 세포를 씨딩하였다. 세포 배양 14일 후, mixed glial cell 배양 배지에 인슐린(Sigma, 미국) 5㎍/㎖를 첨가한 소교세포 배양 배지로 교체한 후 상기 플라스크를 2시간 동안 160rpm, 37℃의 조건에서 orbital shaker로 교반하고, 부유 세포만을 갖는 상층액을 수집하여 1200rpm에서 8분간 원심분리하였다. 원심분리 후, 6웰 플레이트에 1x105으로 세포를 씨딩하고, 37℃, 5%(v/v) CO2의 조건에서 배양하였다. 파킨슨병 유사 염증세포 모델을 만들기 위하여 LPS 100ng/mL 처리 4시간 후, 핵산 복합체 처리하여 배양하였다.In order to isolate and culture primary microglia from brain tissues of mice, 1-day-old neonatal SD rats (Nara Biotech, Korea) were purchased and brain tissues were isolated. The cortex from which the meninges and other tissue parts were removed was transferred to a Petri dish containing iced 1x HBSS (gibco, USA), and then the tissue was dissociated using a pipette and centrifuged at 1200 rpm for 5 minutes. After centrifugation, the supernatant was discarded, and 10% (v/v) fetal bovine serum, penicillin 100 units/ml, streptomycin 100 μg/ml, GlutaMAX ( After releasing the cells by adding mixed glial cell culture medium containing 0.1% of Gibco, USA), the cells were seeded in a 75T flask coated with PLL (Poly-L-lysine, Sigma, USA) the day before. After 14 days of cell culture, the mixed glial cell culture medium was replaced with the microglia culture medium in which 5 μg/ml of insulin (Sigma, USA) was added, and then the flask was stirred with an orbital shaker for 2 hours at 160 rpm and 37°C. Then, the supernatant containing only floating cells was collected and centrifuged at 1200 rpm for 8 minutes. After centrifugation, the cells were seeded in 1x10 5 in a 6-well plate, and cultured under conditions of 37°C and 5% (v/v) CO 2 . In order to create a Parkinson's disease-like inflammatory cell model, 100 ng/mL of LPS was treated for 4 hours, followed by nucleic acid complex treatment and culture.
실시예 3-3: 웨스턴 블랏 분석(Western blot assay)을 이용한 유전자 발현의 분석Example 3-3: Analysis of gene expression using Western blot assay
실시예 3-2의 배양 조건으로 배양된 일차 미세아교세포에 면역 반응 유도를 위해 Lipopolysaccharide(LPS, sigma, 미국) 100ng/ml을 처리하였다. LPS 처리 4시간 후 생활성 펩티드 핵산 및 캐리어 펩티드 핵산을 포함하는 복합체를 처리하였으며, 각각 24, 48, 72시간 배양한 후 RIPA buffer를 각 웰에 30μL씩 첨가하여 protein lysate를 얻었다. Protein lysate를 BCA assay kit(Thermo Fisher, 미국)를 사용하여 단백질 양을 정량하고 단백질 30μg을 전기영동을 통해 사이즈 별로 분리하여, PVDF membrane에 단백질을 옮긴 후 1차 항체인 NLRP3(ABclonal, 미국), Pro Cas-1(abcam, 미국), Pro IL-1β(abcam, 미국)를 1:1000으로 처리하고 4℃에서 하루 동안 방치하였다. 1X TBS-T를 이용하여 워싱하고 2차 항체인 Goat Anti-Rabbit(Cell signaling Technology, 미국)을 1:2000으로 처리하여 상온에서 1시간 방치하였다. SupersignalTM West Femto Maximum Sensitivity Substrate(Thermo Fisher, 미국)를 처리하고 Image600(Amersham, 독일) 장비를 이용하여 표적 유전자의 발현 억제 효율을 분석하였다.Primary microglia cultured under the culture conditions of Example 3-2 were treated with 100 ng/ml of Lipopolysaccharide (LPS, sigma, USA) to induce an immune response. After 4 hours of LPS treatment, complexes containing bioactive peptide nucleic acids and carrier peptide nucleic acids were treated, and after incubation for 24, 48, and 72 hours, respectively, 30 μL of RIPA buffer was added to each well to obtain protein lysate. Protein lysate was quantified using a BCA assay kit (Thermo Fisher, USA), and 30 μg of protein was separated by size through electrophoresis, and the protein was transferred to a PVDF membrane. Pro Cas-1 (abcam, USA) and Pro IL-1β (abcam, USA) were treated at a ratio of 1:1000 and left at 4°C for one day. It was washed with 1X TBS-T, treated with a secondary antibody, Goat Anti-Rabbit (Cell signaling Technology, USA) at a ratio of 1:2000, and left at room temperature for 1 hour. Supersignal TM West Femto Maximum Sensitivity Substrate (Thermo Fisher, USA) was treated, and the expression inhibition efficiency of the target gene was analyzed using Image600 (Amersham, Germany) equipment.
그 결과, 도 3a에 나타낸 바와 같이 파킨슨병 유사 세포 모델에서 서열번호 2와 5의 조합의 핵산 복합체를 처리한 군(조합 2, PNA 2)이 표적 유전자 및 하위 경로 유전자의 발현을 가장 많이 억제하는 것을 확인하였다(도 3a).As a result, as shown in FIG. 3a, the group treated with the nucleic acid complex of SEQ ID NOs: 2 and 5 in the Parkinson's disease-like cell model (combination 2, PNA 2) inhibited the expression of target genes and sub-pathway genes the most. It was confirmed (Fig. 3a).
실시예 3-4: MTT 분석(MTT assay)을 이용한 신경아세포종 세포주의 세포 생존율 분석Example 3-4: Analysis of cell viability of neuroblastoma cell lines using MTT assay
실시예 3-2의 배양 조건으로 배양된 일차 미세아교세포에 생활성 펩티드 핵산 및 캐리어 펩티드 핵산을 포함하는 복합체를 처리하고 4시간 후, LPS 100ng/ml을 처리하여 각각 24, 48, 72시간 배양하였다. 각각의 배양 종료 시점에 얻어진 배양 배지를 주사기 필터(Millipore, 미국)를 사용하여 여과한 후 인간 유래 신경아세포종에 처리하였다. 인간 유래 신경아세포종은 96웰 플레이트에 2x104으로 씨딩하였으며, 24시간 후 일차 미세아교세포에서 얻어진 배지로 교체한 뒤 각각 24, 48, 72시간 동안 배양하였다. MTT(3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide, sigma, 미국) 용액은 1X PBS에 5mg/mL 농도로 제조되었으며, 웰당 20μL씩 처리하여 4시간 배양 후 OD(optical density)를 spectrophotometer로 측정하여 분석하였다.Primary microglia cultured under the culture conditions of Example 3-2 were treated with a complex containing a bioactive peptide nucleic acid and a carrier peptide nucleic acid, and 4 hours later, LPS was treated with 100 ng/ml and cultured for 24, 48, and 72 hours, respectively. did After filtering the culture medium obtained at the end of each culture using a syringe filter (Millipore, USA), it was treated with human-derived neuroblastoma. Human-derived neuroblastoma was seeded in 2x10 4 in a 96-well plate, and after 24 hours, the medium was replaced with the medium obtained from primary microglia, and cultured for 24, 48, and 72 hours, respectively. MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide, sigma, USA) solution was prepared at a concentration of 5 mg/mL in 1X PBS, treated at 20 μL per well and incubated for 4 hours, then the OD (optical density) was measured and analyzed with a spectrophotometer.
본 실시예에서는 파킨슨병 유사 염증세포 모델인 LPS를 처리한 일차 미세아교세포주로부터 얻어진 배지에 의한 신경아세포종 생존율의 변화를 분석하였으며 사용한 핵산 복합체 조합은 상기 표 3과 동일하다.In this example, changes in neuroblastoma survival rate by media obtained from primary microglial cell lines treated with LPS, a Parkinson's disease-like inflammatory cell model, were analyzed, and the nucleic acid complex combinations used are the same as those in Table 3 above.
그 결과, 도 3b에 나타낸 바와 같이 면역 유도군(LPS)과 대조군 서열의 핵산 복합체(조합 1, PNA 1)의 배양 배지를 처리한 군에서는 음성 대조군(NC) 대비 세포 생존율이 감소한 반면, 서열번호 2와 5의 핵산 복합체(조합 2, PNA 2)를 처리한 일차 미세아교세포주에의 배양 배지에 의해 신경아세포종의 세포 생존율이 72시간까지 증가하는 것을 확인하였다(도 3b).As a result, as shown in FIG. 3B, the cell viability decreased compared to the negative control group (NC) in the group treated with the culture medium of the nucleic acid complex of the immune induction group (LPS) and the control sequence (combination 1, PNA 1). It was confirmed that the cell viability of neuroblastoma was increased up to 72 hours by the culture medium of the primary microglial cell line treated with the nucleic acid complex of 2 and 5 (combination 2, PNA 2) ( FIG. 3B ).
실시예 4: MPTP(1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)/프로베네시드(probenecid)로 유도한 마우스 모델에서 핵산 복합체를 이용한 파킨슨병 치료 효과 분석Example 4: Analysis of Parkinson's disease treatment effect using nucleic acid complex in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/probenecid-induced mouse model
본 실시예에서는 실시예 3의 결과를 바탕으로 검증된 핵산 복합체 조합의 파킨슨병 치료 효과를 동물모델에서 평가하기 위하여 도파민 신경세포의 사멸을 유도하는 MPP+의 전구체인 MPTP(1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)와 흡수율을 높이는 프로베네시드(probenecid)를 마우스에 투여한 파킨슨병 동물모델에서 신경세포 사멸에 따른 운동기능의 저하가 핵산 복합체에 의해 회복되는지 검증하였다.In this example, in order to evaluate the Parkinson's disease therapeutic effect of the combination of nucleic acid complexes verified based on the results of Example 3 in an animal model, MPTP (1-methyl-4-phenyl -1,2,3,6-tetrahydropyridine) and probenecid, which increase uptake rate, were administered to mice, and in an animal model of Parkinson's disease, it was verified that the decline in motor function due to neuronal cell death was recovered by the nucleic acid complex.
실시예 4-1: MPTP/프로베네시드 유도성 파킨슨병 동물모델 제작Example 4-1: Production of MPTP/probenecid-induced Parkinson's disease animal model
MPTP/프로베네시드 파킨슨병 동물 모델에서 효능을 검증하기 위하여 7주령 수컷 C57BL/6 마우스(나라바이오텍, 한국)를 사용하였으며, 7일간 순화 후 실험에 사용하였다. 파킨슨병 유사 모델 구축을 위하여 MPTP 25mg/kg 그리고 프로베네시드 250mg/kg이 사용되었으며, 장기적 투여에 의한 손상 모델 제작을 위해 3.5일 간격으로 5주간 총 10회 복강내 주사를 진행하였다. MPTP와 프로베네시드의 4회 투여 이후 각 실험군당 10마리씩 무작위로 선별하여 사용하였으며, 핵산 복합체는 일주일에 두 번씩 4주간 마우스 미정맥에 투여하였다. 이때 양성 대조군에는 파킨슨병 환자의 표준 치료제로 사용되는 레보도파(Levodopa, sigma, 미국) 250mg/kg을 25mg/kg 칼비도파(carbidopa, sigma, 미국)와 혼합하여 핵산 복합체 투여 시작 시점부터 4주간 매일 경구 투여 진행하였다.To verify the efficacy of MPTP/probenecid in an animal model of Parkinson's disease, 7-week-old male C57BL/6 mice (Nara Biotech, Korea) were used, and after acclimatization for 7 days, they were used in experiments. 25 mg/kg of MPTP and 250 mg/kg of probenecid were used to construct a Parkinson's disease-like model, and intraperitoneal injections were performed a total of 10 times at 3.5 day intervals for 5 weeks to create a damage model due to long-term administration. After 4 administrations of MPTP and probenecid, 10 animals per experimental group were randomly selected and used, and the nucleic acid complex was administered to the tail vein of mice twice a week for 4 weeks. At this time, in the positive control group, 250 mg/kg of levodopa (Levodopa, sigma, USA), which is used as a standard treatment for patients with Parkinson's disease, was mixed with 25 mg/kg carbidopa (carbidopa, sigma, USA) and administered orally every day for 4 weeks from the start of nucleic acid complex administration. Dosing proceeded.
실시예 4-2: 로타로드 검사(rotarod test) 분석을 이용한 운동 기능 변화 분석Example 4-2: Analysis of motor function change using rotarod test analysis
본 실시예에서는 핵산 복합체에 의해 파킨슨병 동물모델의 병변 특징인 운동 결손과 균형 유지를 개선하는 효과를 평가하기 위해 로타로드 검사(rotarod test)를 진행하였다.In this example, a rotarod test was performed to evaluate the effect of improving motor deficits and balance maintenance, which are characteristics of lesions in animal models of Parkinson's disease, by nucleic acid complexes.
운동성 검증을 위해 지름 7cm, 15cm 간격으로 60cm 높이의 칸막이를 사용하여 5개의 칸으로 구성된 회전이 가능한 원통형의 봉이 있는 로타로드 장치(정도비앤피, 한국)를 사용하였다. 검사 진행 전 모든 실험동물은 3일 동안 충분한 훈련을 진행하였으며, 핵산 복합체 투여 종료 후 본 실험을 진행하였다. 검사에 사용된 로타로드는 속도가 0-40rpm까지 일정하게 증가하도록 설정하였으며, 회전하는 봉 위에 실험동물을 올려놓고 낙하하기까지 걸린 머무름 시간(Latency time)을 측정하였다. 최대 측정시간은 300초로 제한하였으며, 세 번 반복 측정 후 평균값을 사용하여 핵산 복합체의 치료 효과를 분석하였다.For the verification of motility, a rotarod apparatus (Jeongdo B&P, Korea) with a rotatable cylindrical rod composed of 5 compartments with a diameter of 7 cm and a height of 60 cm at 15 cm intervals was used. Before the test, all the experimental animals were sufficiently trained for 3 days, and this experiment was conducted after the administration of the nucleic acid complex was completed. The speed of the rotarod used in the test was set to increase constantly from 0 to 40 rpm, and the latency time taken until the animal was placed on the rotating rod and dropped was measured. The maximum measurement time was limited to 300 seconds, and the therapeutic effect of the nucleic acid complex was analyzed using the average value after three repeated measurements.
그 결과, 도 4a에 나타낸 바와 같이 회전하는 봉 위에서 오랜 시간 버티지 못하고 낙하한 파킨슨병 유도 모델 대비, 2번 조합의 핵산 복합체(서열번호 2와 5, PNA 2)를 처리한 그룹의 봉 위에 머무르는 시간이 증가하며 행동 장애가 개선된 것을 확인하였다(도 4a).As a result, as shown in FIG. 4a, the time spent on the rod in the group treated with the nucleic acid complex of No. 2 (SEQ ID NOs: 2 and 5, PNA 2) compared to the Parkinson's disease induction model that fell without being able to hold on to the rotating rod for a long time. As this increased, it was confirmed that the behavioral disorder was improved (FIG. 4a).
실시예 4-3: 폴 검사(pole test) 분석을 이용한 운동 기능 변화 분석Example 4-3: Analysis of motor function change using pole test analysis
실시예 4-1의 조건으로 제작된 파킨슨병 동물 질환 모델을 사용하여 무운동성과 경직반응 회복 여부를 평가하기 위해 폴 검사를 진행하였다.A pole test was performed to evaluate the recovery of akinesia and spastic response using the Parkinson's disease animal disease model prepared under the conditions of Example 4-1.
폴 검사는 폭 0.8cm, 높이 55cm 막대기를 사용하여 마우스가 하늘을 향하도록 올려놓은 뒤 180° 회전하여 바닥에 도착하는 데까지 소요되는 총 시간을 측정하는 방식으로 진행하였다. 검사 진행 전 모든 실험 동물에 대해 동일한 횟수의 훈련을 진행하였으며, 본 실험은 핵산 복합체 투여 종료 후 진행하였다. 실험은 세 번 반복 진행되었으며, 결과 분석은 평균값을 사용하였다.The pole test was performed by placing the mouse facing the sky using a stick with a width of 0.8 cm and a height of 55 cm, and then rotating 180° to measure the total time required to reach the floor. The same number of training sessions were conducted for all experimental animals before the test, and this experiment was conducted after the administration of the nucleic acid complex was completed. The experiment was repeated three times, and the average value was used for analysis of the results.
그 결과, 도 4b에 나타낸 바와 같이 파킨슨병을 유도한 MPTP/프로베네시드 동물 그룹은 지면 도착까지의 총 소요 시간이 정상 마우스에 비해(NC) 증가하였다. 반면 서열번호 2와 5의 핵산 복합체(PNA 2)를 처리한 그룹은 꼭대기에서 방향 회전 후 바닥에 도착하기까지의 시간이 감소하며 운동기능 장애가 개선된 것을 확인하였다(도 4b).As a result, as shown in FIG. 4b, the MPTP/probenecid animal group that induced Parkinson's disease had an increased total time to reach the ground compared to normal mice (NC). On the other hand, in the group treated with the nucleic acid complex of SEQ ID NOs: 2 and 5 (PNA 2), it was confirmed that the time from the directional rotation at the top to the bottom was reduced and motor dysfunction was improved (FIG. 4b).
실시예 4-4: 실린더 검사(cylinder test) 분석을 이용한 운동 기능 변화 분석Example 4-4: Analysis of motor function changes using cylinder test analysis
실시예 4-1의 방법으로 파킨슨병 유도한 동물모델에서의 감각 운동기능 치료 효과를 확인하기 위해 실린더 검사를 진행하였다.In order to confirm the effect of sensorimotor function treatment in an animal model induced with Parkinson's disease by the method of Example 4-1, a cylinder test was performed.
마우스는 새로운 환경에 놓이면 주변을 탐색하기 위해 앞발 두 개를 들어 벽을 짚는 행동을 취하는 것이 일반적이기 때문에 투명한 실린더에 마우스를 넣은 후 앞발 사용 횟수를 측정하였다. 실험은 핵산 복합체 투여 종료 다음 날 진행되었으며, 별도의 훈련 없이 실린더에서 3분 동안 앞발로 실린더 벽면을 짚는 횟수를 측정한 결과를 사용하였다.When a mouse is placed in a new environment, it is common to lift two forepaws and touch the wall to explore the surroundings, so the number of times the forepaws were used was measured after placing the mouse in a transparent cylinder. The experiment was conducted the day after the end of the nucleic acid complex administration, and the result of measuring the number of times of pressing the cylinder wall with the forepaw for 3 minutes in the cylinder without separate training was used.
그 결과, 도 4c에 나타낸 바와 같이 신경 독성물질인 MPTP에 노출된 MPTP/프로베네시드만 투여한 동물 그룹은 앞발 사용 횟수가 정상 마우스 대비 감소하였으며, 이와 대조적으로 서열번호 2와 5의 조합의 핵산 복합체(PNA 2)를 처리한 그룹은 앞발 사용 횟수가 증가하며 파킨슨병 동물 모델에서의 감각 운동 장애 치료 효능을 확인하였다(도 4c).As a result, as shown in FIG. 4c, the group of animals exposed to MPTP, a neurotoxin, and administered with only MPTP/probenecid decreased the number of times of using the paws compared to normal mice. The group treated with the complex (PNA 2) increased the number of times of using the forelimbs, confirming the therapeutic efficacy of sensorimotor impairment in an animal model of Parkinson's disease (FIG. 4c).
실시예 5: MPTP/프로베네시드(probenecid)를 이용한 파킨슨병 유도 동물 모델에서의 유전자 발현 분석Example 5: Gene expression analysis in Parkinson's disease induced animal model using MPTP/probenecid
본 실시예에서는 실시예 4-1의 방법으로 유도한 파킨슨병 동물 모델에서 도파민 생성과 대사가 일어나는 뇌 조직의 선조체(striatum)와 흑질(substantia nigra) 부위에서 파킨슨병 관련 유전자의 발현 변화를 확인하였다.In this example, in the Parkinson's disease animal model induced by the method of Example 4-1, changes in the expression of Parkinson's disease-related genes were confirmed in the striatum and substantia nigra regions of brain tissue where dopamine production and metabolism occur. .
실시예 5-1: 웨스턴 블랏 분석(Western blot assay)을 이용한 파킨슨병 동물 모델 조직에서의 유전자 발현 분석Example 5-1: Analysis of gene expression in Parkinson's disease animal model tissue using Western blot assay
실시예 4-1의 방법으로 실험 진행 후 종료일에 적출한 뇌 조직에서 선조체와 흑질을 분리하였으며, RIPA buffer를 첨가하여 protein lysate를 얻었다. Protein lysate를 BCA assay kit(Thermo Fisher, 미국)를 사용하여 단백질 양을 정량하고 단백질 30μg을 전기영동을 통해 사이즈 별로 분리하여, PVDF membrane에 단백질을 옮긴 후 1차 항체인 NLRP3(ABclonal, 미국), Pro IL-1β(abcam, 미국), Tyrosine Hydroxylase(TH, abcam, 미국), 알파-시누클레인(α-synuclein, abcam, 미국)을 1:1000으로 처리하고 4℃에서 하루 동안 방치하였다. 1X TBS-T를 이용하여 워싱하고 2차 항체인 Goat Anti-Rabbit(Cell signaling Technology, 미국)을 1:2000으로 처리하여 상온에서 1시간 방치하였다. SupersignalTM West Femto Maximum Sensitivity Substrate(Thermo Fisher, 미국)를 처리하고 Image600(Amersham, 독일) 장비를 이용하여 표적 유전자의 발현 억제 효율을 분석하였다.The striatum and substantia nigra were separated from the brain tissue extracted on the end day after the experiment was conducted by the method of Example 4-1, and protein lysate was obtained by adding RIPA buffer. Protein lysate was quantified using a BCA assay kit (Thermo Fisher, USA), and 30 μg of protein was separated by size through electrophoresis, and the protein was transferred to a PVDF membrane. Pro IL-1β (abcam, USA), Tyrosine Hydroxylase (TH, abcam, USA), and alpha-synuclein (α-synuclein, abcam, USA) were treated at a ratio of 1:1000 and left at 4°C for one day. It was washed with 1X TBS-T, treated with a secondary antibody, Goat Anti-Rabbit (Cell signaling Technology, USA) at a ratio of 1:2000, and left at room temperature for 1 hour. Supersignal TM West Femto Maximum Sensitivity Substrate (Thermo Fisher, USA) was treated, and the expression inhibition efficiency of the target gene was analyzed using Image600 (Amersham, Germany) equipment.
본 실시예에서는 파킨슨병 유도 동물 모델에서의 NLRP3 및 하위 단계 유전자 발현 변화 및 도파민성 신경세포와 알파-시누클레인의 발현 정도를 분석하였으며, 사용한 핵산 복합체 조합은 상기 표 3과 동일하다.In this example, NLRP3 and lower-order gene expression changes and the expression levels of dopaminergic neurons and alpha-synuclein in Parkinson's disease-induced animal models were analyzed, and the nucleic acid complex combinations used are the same as those in Table 3 above.
그 결과, 도 5a에 나타낸 바와 같이 실시예 4-1의 방법으로 진행된 실험에서 종료일에 적출한 뇌 조직의 선조체(striatum) 부위를 사용하여 발현 분석을 진행하였을 때, 2번 조합의 핵산 복합체(서열번호 2와 5 조합, PNA 2)를 처리한 군이 표적 유전자인 NLRP3 및 하위 경로 유전자인 IL-1β의 발현이 질환 동물군(MPTP/p) 대비 감소한 것이 확인되었다. 또한 도파민성 신경세포(TH, Tyrosine hydroxylase)는 MPTP/프로베네시드 유도 모델에서 가장 많이 감소하였으며, 2번 조합의 핵산 복합체를 처리한 군(PNA 2)에서는 증가되는 것을 확인하여 도파민성 신경 세포의 회복 현상을 확인하였다. 또한, MPTP/프로베네시드 유도군에서 증가하였던 파킨슨병의 병인 요인인 알파-시누클레인의 발현은 2번 조합의 핵산 복합체를 처리한 군에서는 발현이 억제되는 것을 확인하였다(도 5a).As a result, as shown in FIG. 5a, when expression analysis was performed using the striatum region of the brain tissue extracted on the end day of the experiment conducted by the method of Example 4-1, the nucleic acid complex of No. 2 combination (sequence It was confirmed that the expression of NLRP3, a target gene, and IL-1β, a sub-pathway gene, in the group treated with the combination of numbers 2 and 5, PNA 2) decreased compared to the diseased animal group (MPTP/p). In addition, dopaminergic neurons (TH, Tyrosine hydroxylase) decreased the most in the MPTP/probenecid-induced model, and increased in the group treated with the nucleic acid complex of No. 2 (PNA 2). The recovery phenomenon was confirmed. In addition, it was confirmed that the expression of alpha-synuclein, the etiological factor of Parkinson's disease, which was increased in the MPTP/probenecid induction group, was suppressed in the group treated with the nucleic acid complex of No. 2 combination (FIG. 5a).
흑질(substantia nigra) 부위에서의 발현은 도 5b에서 나타내었으며, 질환 유도군(MPTP/p)에서 증가된 표적 유전자인 NLRP3 및 하위 경로 유전자인 IL-1β의 발현이 2번 조합의 핵산 복합체(PNA 2)를 처리한 그룹에서 가장 많이 감소하였으며, 질환 모델에서 감소된 도파민성 신경세포는 2번 조합의 핵산 복합체를 처리한 군(서열번호 2와 5 조합, PNA 2)에서 발현이 증가하며 회복되는 것을 확인하였다. 또한, 알파-시누클레인의 발현은 2번 조합의 핵산 복합체 처리 후 감소하는 것을 통해 서열번호 2와 5 조합의 핵산 복합체에 의한 파킨슨병 치료 효능을 확인하였다(도 5b).The expression in the substantia nigra region is shown in FIG. 5B, and the expression of the target gene NLRP3 and IL-1β, a sub-pathway gene, increased in the disease induction group (MPTP/p) in the second combination of nucleic acid complexes (PNA). 2), and the decreased dopaminergic neurons in the disease model were recovered with increased expression in the group treated with the nucleic acid complex of No. 2 (combination of SEQ ID NOs: 2 and 5, PNA 2) confirmed that In addition, the expression of alpha-synuclein decreased after treatment with the nucleic acid complex in combination No. 2, confirming the therapeutic efficacy of the nucleic acid complex of SEQ ID NOs: 2 and 5 (FIG. 5b).
실시예 5-2: 면역염색을 이용한 파킨슨병 동물 모델에서의 조직 내 발현 변화 분석Example 5-2: Analysis of expression changes in tissues in Parkinson's disease animal models using immunostaining
실시예 4-1의 조건으로 실험 진행 후 종료일에 마우스 뇌 조직을 분리하여 4% 포르말린 용액에 하루 동안 고정하고 30% 설탕 용액에 담가 3일 동안 탈수한 후, OCT(Leica, 미국) 용액에 포매(embedding) 하였다. 포매한 조직은 -20℃에서 보관하여 얼린 후, 동결절편기(Cryostat, Leica, 미국)를 사용하여 조직을 20μm로 섹션한 후 슬라이드 글라스에 마운팅(mounting)하였다. 마운팅한 조직은 1% BSA 용액을 사용하여 2시간 동안 블락킹하였으며, Tyrosine Hydroxylase(abcam, 미국), 알파-시누클레인(α-synuclein, abcam, 미국), IBA1(wako, 일본) 1차 항체를 처리한 후 4℃에서 하루 동안 방치하였다. 이어서 1차 항체 용액을 제거하고 1X TBS로 씻어낸 후 2차 항체 용액 Goat Anti-Rabbit(Cell signaling Technology, 미국)을 1:2000으로 상온에서 2시간 처리하였다. 1X TBS를 사용하여 2차 항체 씻어낸 후 DAB 염색을 통해 발현을 분석하였다.After the experiment was conducted under the conditions of Example 4-1, mouse brain tissue was separated on the end day, fixed in 4% formalin solution for one day, immersed in 30% sugar solution, dehydrated for 3 days, and then embedded in OCT (Leica, USA) solution (embedding) was done. The embedded tissue was stored and frozen at -20°C, and then the tissue was sectioned at 20 μm using a cryostat (Cryostat, Leica, USA) and then mounted on a slide glass. The mounted tissues were blocked for 2 hours using 1% BSA solution, and primary antibodies were used to detect Tyrosine Hydroxylase (abcam, USA), alpha-synuclein (α-synuclein, abcam, USA), and IBA1 (wako, Japan). After treatment, it was left at 4° C. for one day. Subsequently, the primary antibody solution was removed, washed with 1X TBS, and then treated with a secondary antibody solution, Goat Anti-Rabbit (Cell signaling Technology, USA) at a ratio of 1:2000 at room temperature for 2 hours. After washing the secondary antibody with 1X TBS, expression was analyzed by DAB staining.
본 실시예에서는 파킨슨병 유도 동물 모델의 뇌 조직에서 도파민성 신경세포와 발병 요인인 알파-시누클레인 및 면역세포의 발현 정도를 분석하였으며, 발병에 중요한 뇌 영역인 선조체와 흑질 내에서 비교 분석하였다.In this example, the expression levels of dopaminergic neurons, alpha-synuclein, and immune cells, which are onset factors, were analyzed in brain tissues of Parkinson's disease-induced animal models, and comparative analysis was performed in the striatum and substantia nigra, which are brain regions important for onset.
도파민성 신경세포의 변화를 분석한 결과, 도 6a에 나타낸 바와 같이 선조체에서의 도파민성 신경세포를 나타내는 tyrosine hydroxylase의 발현은 MPTP/프로베네시드 파킨슨병 유도 모델에서 감소한 것을 확인하였으며, 2번 조합의 핵산 복합체(서열번호 2 및 5 조합, PNA 2)를 투여한 그룹에서는 발현이 증가한 것을 확인하였다(도 6a). 흑질 부위에서도 도 6b에서 나타낸 바와 같이 MPTP/프로베네시드만 투여한 그룹에서는 발현 정도가 감소한 것을 확인하였으며, 2번 핵산복합체를 투여한 그룹(PNA 2)에서는 발현이 증가하는 것을 확인하였다(도 6b).As a result of analyzing changes in dopaminergic neurons, it was confirmed that the expression of tyrosine hydroxylase representing dopaminergic neurons in the striatum was reduced in the MPTP/probenecid Parkinson's disease induction model, as shown in FIG. In the group administered with the nucleic acid complex (SEQ ID NO: 2 and 5 combination, PNA 2), it was confirmed that expression increased (FIG. 6a). In the substantia nigra region, as shown in FIG. 6b, it was confirmed that the level of expression decreased in the group administered with only MPTP/probenecid, and the expression increased in the group administered with nucleic acid complex No. 2 (PNA 2) (FIG. 6b). ).
병인 요인인 알파-시누클레인의 발현을 확인하였을 때, 선조체의 경우 파킨슨병 유도 모델(MPTP/p)에서 갈색 반응이 증가하여 발현이 증가한 것을 확인하였으며, 2번 조합의 핵산 복합체(PNA 2) 투여군에서는 알파-시누클레인의 갈색 반응이 현저하게 감소한 것이 관찰되었다. 이를 수치화해 비교하였을 때 양성 대조군을 처리한 그룹에 비해 2번 조합의 핵산 복합체를 투여한 그룹에서 알파-시누클레인의 발현이 감소하는 것이 나타났다(도 6c). 흑질에서의 알파-시누클레인 발현 정도를 비교한 결과에서도 선조체와 마찬가지로 MPTP/프로베네시드 질환 유도 그룹에서 증가하였던 갈색 반응이 2번 핵산 복합체를 투여 한 그룹에서는 현저히 감소하였다. 이를 수치화해 나타낸 그래프에서도 2번 조합 핵산 복합체를 처리한 그룹에서 발현 정도가 감소하는 것을 확인할 수 있었다(도 6d).When the expression of alpha-synuclein, a pathogenic factor, was confirmed, in the case of the striatum, it was confirmed that the brown reaction increased in the Parkinson's disease induction model (MPTP/p) and the expression increased. observed a significant reduction in the brown response of alpha-synuclein. When compared by quantification, it was found that the expression of alpha-synuclein was decreased in the group administered with the nucleic acid complex of No. 2 combination compared to the group treated with the positive control group (FIG. 6c). As a result of comparing the expression level of alpha-synuclein in the substantia nigra, as in the striatum, the brown reaction, which was increased in the MPTP/probenecid disease induction group, was remarkably reduced in the group administered with nucleic acid complex No. 2. In the graph showing this numerically, it was confirmed that the expression level decreased in the group treated with the combined nucleic acid complex No. 2 (FIG. 6d).
마지막으로 뇌 면역 세포인 미세아교세포(microglia)의 마커(IBA1)의 발현을 확인한 결과, 도 6e에 나타낸 바와 같이 선조체 내에서 MPTP/프로베네시드 유도 그룹에서 갈색 반응이 현저히 증가하였으며, 2번 핵산 복합체를 투여한 그룹에서는 갈색 반응이 감소하여 면역 세포의 감소를 확인하였다. 이를 수치화한 그래프에서도 2번 조합 핵산 복합체를 투여한 그룹에서 IBA의 반응이 감소한 것이 나타났다(도 6e). 도 6f에 나타낸 흑질에서의 IBA1 발현을 확인한 결과에서도 선조체와 동일하게 2번 조합 핵산 복합체를 처리한 그룹의 갈색 반응이 다른 질환 유도군(MPTP/p) 대비 가장 많이 감소한 것을 확인할 수 있었으며, 수치화한 그래프에서도 가장 많이 감소한 것으로 나타났다(도 6f).Finally, as a result of confirming the expression of a marker (IBA1) of microglia, which is a brain immune cell, as shown in FIG. In the group administered with the complex, the brown reaction was reduced, confirming the reduction of immune cells. It was also shown in the numerical graph that the IBA response was reduced in the group administered with the nucleic acid complex No. 2 (FIG. 6e). As a result of confirming the expression of IBA1 in the substantia nigra shown in FIG. 6f, it was confirmed that the brown reaction in the group treated with the combination nucleic acid complex No. 2 was the most reduced compared to the other disease-induced group (MPTP/p), as in the striatum. It was found to be the most decreased in the graph (FIG. 6f).
본 발명에 따른 NLRP3를 표적으로 하는 생활성 핵산과 캐리어 펩티드 핵산이 상보적으로 결합된 핵산 복합체는 혈뇌장벽 투과능을 가지며, NLRP3의 발현을 효율적으로 억제할 수 있어 퇴행성 뇌질환, 특히 파킨슨병의 예방 또는 치료에 유용하다.The nucleic acid complex of the present invention, in which a bioactive nucleic acid targeting NLRP3 and a carrier peptide nucleic acid are complementaryly bound, has the ability to penetrate the blood-brain barrier and can efficiently inhibit the expression of NLRP3, thereby preventing degenerative brain diseases, particularly Parkinson's disease. Useful for prevention or treatment.
이상으로 본 발명 내용의 특정한 부분을 상세히 기술하였는바, 당업계의 통상의 지식을 가진 자에게 있어서 이러한 구체적 기술은 단지 바람직한 실시 양태일 뿐이며, 이에 의해 본 발명의 범위가 제한되는 것이 아닌 점은 명백할 것이다. 따라서, 본 발명의 실질적인 범위는 첨부된 청구항들과 그것들의 등가물에 의하여 정의된다고 할 것이다.As above, specific parts of the present invention have been described in detail, and it will be clear to those skilled in the art that these specific descriptions are merely preferred embodiments, and the scope of the present invention is not limited thereby. will be. Accordingly, the substantial scope of the present invention will be defined by the appended claims and their equivalents.
전자파일 첨부하였음.Electronic file attached.

Claims (12)

  1. NLRP3 유전자를 표적으로 하는 생활성 핵산(Bioactive Nucleic Acid); 및 캐리어 펩티드 핵산(Carrier Peptide Nucleic Acid)이 상보적으로 결합된 핵산 복합체.Bioactive Nucleic Acid targeting the NLRP3 gene; and a carrier peptide nucleic acid (Carrier Peptide Nucleic Acid) complementary nucleic acid complex.
  2. 제1항에 있어서, 상기 NLRP3 유전자를 표적으로 하는 생활성 핵산은 서열번호 2의 서열로 표시되는 염기서열을 포함하는 것을 특징으로 하는 핵산 복합체.The nucleic acid complex according to claim 1, wherein the bioactive nucleic acid targeting the NLRP3 gene comprises the nucleotide sequence represented by SEQ ID NO: 2.
  3. 제1항에 있어서, 상기 캐리어 펩티드 핵산은 서열번호 4 또는 서열번호 5의 서열로 표시되는 염기서열을 포함하는 것을 특징으로 하는 핵산 복합체.The nucleic acid complex according to claim 1, wherein the carrier peptide nucleic acid comprises a nucleotide sequence represented by SEQ ID NO: 4 or SEQ ID NO: 5.
  4. 제1항에 있어서, 상기 NLRP3 유전자를 표적으로 하는 생활성 핵산 또는 캐리어 펩티드 핵산은 각각의 핵산의 5'-말단 또는 3'-말단에 엔도좀 탈출을 도와주는 물질이 추가로 결합된 것을 특징으로 하는 핵산 복합체.The method of claim 1, wherein the bioactive nucleic acid or carrier peptide nucleic acid targeting the NLRP3 gene is additionally bound to a material that helps endosome escape to the 5'-end or 3'-end of each nucleic acid. nucleic acid complexes.
  5. 제4항에 있어서, 상기 엔도좀 탈출을 도와주는 물질은 펩티드, 지질 나노물질(lipid nanoparticles), 접합체 나노물질(polyplex nanoparticles), 고분자 나노구(polymer nanospheres), 무기물 나노물질(inorganic nanoparticles), 양이온 지질 나노물질(cationic lipid-based nanoparticles), 양이온 고분자(cationic polymer) 및 pH 감응 고분자(pH sensitive polymers)로 구성된 군에서 선택되는 어느 하나 이상인 것을 특징으로 하는 핵산 복합체.The method of claim 4, wherein the material that helps the endosomes escape is a peptide, lipid nanoparticles, conjugate nanomaterials (polyplex nanoparticles), polymer nanospheres (polymer nanospheres), inorganic nanomaterials (organic nanoparticles, cations) A nucleic acid complex, characterized in that at least one selected from the group consisting of cationic lipid-based nanoparticles, cationic polymers and pH sensitive polymers.
  6. 제5항에 있어서, 상기 펩티드는 GLFDIIKKIAESF(서열번호 6) 또는 Histidine(10)인 것을 특징으로 하는 핵산 복합체.The nucleic acid complex according to claim 5, wherein the peptide is GLFDIIKKIAESF (SEQ ID NO: 6) or Histidine (10).
  7. 제1항에 있어서, 상기 NLRP3 유전자를 표적으로 하는 생활성 핵산은 전체적으로 음전하 또는 중성을 가지는 것을 특징으로 하는 핵산 복합체.The nucleic acid complex according to claim 1, wherein the bioactive nucleic acid targeting the NLRP3 gene has a negative or neutral charge as a whole.
  8. 제1항에 있어서, 상기 캐리어 펩티드 핵산은 전체적으로 양전하를 가지는 것을 특징으로 하는 핵산 복합체.The nucleic acid complex according to claim 1, wherein the carrier peptide nucleic acid has an overall positive charge.
  9. 제1항에 있어서, 상기 핵산 복합체는 전체적으로 양전하를 가지는 것을 특징으로 하는 핵산 복합체.The nucleic acid complex according to claim 1, wherein the nucleic acid complex has a positive charge as a whole.
  10. 제1항에 있어서, 상기 핵산 복합체는 혈뇌장벽 투과능을 가지는 것을 특징으로 하는 핵산 복합체.The nucleic acid complex according to claim 1, wherein the nucleic acid complex has blood-brain barrier penetrating ability.
  11. 제1항 내지 제10항 중 어느 한 항에 따른 핵산 복합체를 유효성분으로 함유하는 퇴행성 뇌질환 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating degenerative brain disease, containing the nucleic acid complex according to any one of claims 1 to 10 as an active ingredient.
  12. 제11항에 있어서, 상기 퇴행성 뇌질환은 알츠하이머병, 파킨슨병, 니만-피크 병, 크로이츠펠트-야콥병, 헌팅턴병, 루게릭병, 다발성 경화증 또는 치매인 것을 특징으로 하는 약학 조성물.The pharmaceutical composition according to claim 11, wherein the degenerative brain disease is Alzheimer's disease, Parkinson's disease, Niemann-Pick disease, Creutzfeldt-Jakob disease, Huntington's disease, Lou Gehrig's disease, multiple sclerosis or dementia.
PCT/KR2023/000259 2022-01-06 2023-01-06 Composition comprising nucleic acid complex for prevention or treatment of degenerative brain disease WO2023132673A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR1020220002084A KR20230106325A (en) 2022-01-06 2022-01-06 Composition for Preventing or Treating Degenerative Brain Disease Comprising Nucleic Acid Complex
KR10-2022-0002084 2022-01-06

Publications (1)

Publication Number Publication Date
WO2023132673A1 true WO2023132673A1 (en) 2023-07-13

Family

ID=87073967

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/000259 WO2023132673A1 (en) 2022-01-06 2023-01-06 Composition comprising nucleic acid complex for prevention or treatment of degenerative brain disease

Country Status (2)

Country Link
KR (1) KR20230106325A (en)
WO (1) WO2023132673A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20180018405A (en) * 2016-08-09 2018-02-21 주식회사 시선바이오머티리얼스 Peptide Nucleic Acid Complex with Increased Cell Permeability and pharmaceutical composition comprising the same
US20180312542A1 (en) * 2015-10-20 2018-11-01 President And Fellows Of Harvard College Endosomal escape peptides
JP2018537528A (en) * 2015-11-04 2018-12-20 アイデラ・ファーマシューティカルズ,インコーポレーテッド Composition for inhibiting NLRP3 gene expression and use thereof
KR20190096148A (en) * 2018-02-08 2019-08-19 주식회사 시선테라퓨틱스 Peptide Nucleic Acid Complex with Endosome Escape Ability and Uses thereof
KR20210088446A (en) * 2020-01-06 2021-07-14 주식회사 시선테라퓨틱스 Composition for Preventing or Treating Macular Degeneration Comprising Cell Permeable Nucleic Acid Complex
KR20220041397A (en) * 2020-09-25 2022-04-01 주식회사 시선테라퓨틱스 Composition for Preventing or Treating Dmentia Comprising Peptide Nucleic Acid Complex

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180312542A1 (en) * 2015-10-20 2018-11-01 President And Fellows Of Harvard College Endosomal escape peptides
JP2018537528A (en) * 2015-11-04 2018-12-20 アイデラ・ファーマシューティカルズ,インコーポレーテッド Composition for inhibiting NLRP3 gene expression and use thereof
KR20180018405A (en) * 2016-08-09 2018-02-21 주식회사 시선바이오머티리얼스 Peptide Nucleic Acid Complex with Increased Cell Permeability and pharmaceutical composition comprising the same
KR20190096148A (en) * 2018-02-08 2019-08-19 주식회사 시선테라퓨틱스 Peptide Nucleic Acid Complex with Endosome Escape Ability and Uses thereof
KR20210088446A (en) * 2020-01-06 2021-07-14 주식회사 시선테라퓨틱스 Composition for Preventing or Treating Macular Degeneration Comprising Cell Permeable Nucleic Acid Complex
KR20220041397A (en) * 2020-09-25 2022-04-01 주식회사 시선테라퓨틱스 Composition for Preventing or Treating Dmentia Comprising Peptide Nucleic Acid Complex

Also Published As

Publication number Publication date
KR20230106325A (en) 2023-07-13

Similar Documents

Publication Publication Date Title
WO2019156365A1 (en) Peptide nucleic acid complex having endosomal escape capacity, and use thereof
US8007790B2 (en) Methods for treating polycystic kidney disease (PKD) or other cyst forming diseases
WO2018030608A1 (en) Nanoliposome carrier composition containing hybrid of cas9 protein and guide rna
WO2018030789A1 (en) Peptide nucleic acid complex having improved cell permeability and pharmaceutical composition comprising same
KR20120046099A (en) Methods of inhibiting fibrogenesis and treating fibrotic disease
WO2022065919A1 (en) Composition for preventing or treating dementia, containing peptide nucleic acid complex as active ingredient
WO2023132673A1 (en) Composition comprising nucleic acid complex for prevention or treatment of degenerative brain disease
WO2020096234A1 (en) Composition for preventing or treating atopic dermatitis comprising skin-penetrating nucleic acid complex as effective component
WO2021125762A1 (en) Composition for preventing or treating dementia, containing peptide nucleic acid complex with blood-brain barrier penetration ability as active ingredient
WO2023132674A1 (en) Composition comprising nucleic acid complex for prevention or treatment of degenerative brain disease
WO2021141368A2 (en) Composition for preventing or treating macular degeneration, containing cell permeable nucleic acid complex as active ingredient
WO2019066519A1 (en) Pharmaceutical composition for prevention or treatment of age-related macular degeneration containing rna complex targeting connective tissue growth factor
WO2023163562A1 (en) Composition for preventing or treating alzheimer's disease comprising nucleic acid complex
KR20140069038A (en) Method of treating mucoepidermoid carcinoma
WO2022260480A1 (en) Nanoparticle comprising peptide-lipid conjugate for delivering oligonucleotide into target cell and pharmaceutical composition comprising same
WO2021075880A1 (en) Peptide-nucleic acid complex having blood-brain barrier penetration ability and composition comprising same
WO2023163563A1 (en) Composition for preventing or treating lung cancer comprising nucleic acid complex
KR102572074B1 (en) Composition for Preventing or Treating Macular Degeneration Comprising Cell Permeable Nucleic Acid Complex
WO2022045763A1 (en) Composition comprising cell permeable nucleic acid complex as active ingredient for preventing or treating rheumatoid arthritis
WO2022131745A1 (en) Composition for preventing or treating glioblastoma comprising peptide nucleic acid complex as active ingredient
KR102156323B1 (en) Composition for Preventing or Treating Psoriasis Comprising Skin Permeable Nucleic Acid Complex
WO2024101915A1 (en) Improved peptide nucleic acid complex
WO2016178519A1 (en) Composition for inhibiting muscle aging or treating muscle aging related diseases
WO2022255819A1 (en) Composition for preventing or treating non-alcoholic steatohepatitis containing peptide nucleic acid complex as active ingredient
WO2020032680A1 (en) Recombinant herpes simplex virus and method for preparing same

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: 23737421

Country of ref document: EP

Kind code of ref document: A1