WO2013036031A2 - Composition pharmaceutique comprenant un inhibiteur de l'expression du miarn486 en tant que principe actif dans la prévention et le traitement de troubles neurologiques - Google Patents

Composition pharmaceutique comprenant un inhibiteur de l'expression du miarn486 en tant que principe actif dans la prévention et le traitement de troubles neurologiques Download PDF

Info

Publication number
WO2013036031A2
WO2013036031A2 PCT/KR2012/007112 KR2012007112W WO2013036031A2 WO 2013036031 A2 WO2013036031 A2 WO 2013036031A2 KR 2012007112 W KR2012007112 W KR 2012007112W WO 2013036031 A2 WO2013036031 A2 WO 2013036031A2
Authority
WO
WIPO (PCT)
Prior art keywords
mir486
expression
neurod6
disease
spinal cord
Prior art date
Application number
PCT/KR2012/007112
Other languages
English (en)
Korean (ko)
Other versions
WO2013036031A3 (fr
Inventor
강수경
Original Assignee
서울대학교 산학협력단
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 서울대학교 산학협력단 filed Critical 서울대학교 산학협력단
Publication of WO2013036031A2 publication Critical patent/WO2013036031A2/fr
Publication of WO2013036031A3 publication Critical patent/WO2013036031A3/fr

Links

Classifications

    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • C12N2310/113Antisense targeting other non-coding nucleic acids, e.g. antagomirs

Definitions

  • composition for preventing and treating neurological diseases including microRNA486 expression inhibitors as an active ingredient
  • the present invention relates to a pharmaceutical composition for preventing and treating neurological diseases, including miRNA486 expression inhibitors as an active ingredient.
  • RNAs small, unencoded RNAs (microRNAs, miRNAs), a novel class of regulatory molecules as factors involved in gene expression, are known to be involved in tissue regeneration. miRNAs are emerging as important factors in translational regulation, involved in regulating the function of cell fate, and alterations in functional gene expression have been shown to play an important role in the secondary damage process in some disease models.
  • miRNAs can regulate the expression of a specific set of functional genes, they are useful candidates as regulators upstream of the secondary spinal cord injury process among disease model miRNAs signaling mediators. Many miRNAs play an important role in neurodevelopment and are believed to be important mediators of cell differentiation. Based on the specific interaction of miRNAs with their target genes, RNA-based techniques have potential as therapeutic strategies. miRNAs expression will be a novel therapeutic target for the treatment of various diseases, including cancer, cardiovascular disease and wounded invasion. As one of the diseases, neurodestruction is due to various factors that depend on the inherent properties of the underlying neurodestructive disorder. This indicates the homeostasis of unstable cells along with the cytotoxicity of by-products mediated by overloaded immune cells. In particular, previous studies have explored the potential for miRNAs in neurodestruction. It has been shown that altered expression of miRNAs contributes to secondary damage following trauma to the central nervous system.
  • NeuroD6 is a factor associated with nerve regeneration.
  • NeuroD6 is known to have a neuroprotective role in the central nervous system against R0S-mediated secondary injury.
  • the NeuroD6 protein promotes neuronal differentiation and survival in the pathological microenvironment and stimulates the biomass of the mitochondria to initiate anti-apoptosis and chaperone responses.
  • the NeuroD6 expression interference is associated with the induction of motor neuron apoptosis initiated by actively proliferating macrophages and the secretion of pro-inflammatory factors mediating apoptosis and myelin destruction of motor neurons. These successive pathological events result in secondary damage for a long time after traumatic primary injury to the spinal cord.
  • NeuroD6 promotes neuronal survival through expression of anti-apoptotic regulators that maintain mitochondrial preservation.
  • miR miR-133b a miRNA during spinal cord regeneration of zebrafish, has been studied, and an increase in miR-133b expression in brain regeneration has been identified.
  • MiR-486 has been reported to be ubiquitous in rat brain, spinal cord, liver and heart. And identifying miR-486 or miR-422a, which regulates cell survival in the heart, and treating or preventing cardiac hypertrophy, heart failure, or myocardial infarction following increased expression of miR-486 and / or miR-422a in heart tissue.
  • miR486 motor neurons related to pathophysiology mediated by reactive oxygen species (ROS) in sustained nerve injury Since miR486 was specifically overexpressed in the eye, miR486 inhibitor expression interference was found to induce spinal cord injury (SCI) improvement by restoring the inhibition of NeuroD6.
  • SCI spinal cord injury
  • An object of the present invention is a pharmaceutical composition for preventing and treating neurological diseases, a method for treating neurological diseases, a method for screening candidate substances for preventing and treating neurological diseases, a kit for diagnosing neurological diseases, and a diagnostic for neurological diseases. It provides a gene detection method and a neurological disease diagnosis method for providing information.
  • the present invention provides a pharmaceutical composition for preventing and treating neurological diseases, comprising miR486 expression inhibitor as an active ingredient.
  • the present invention also provides a method for preventing and treating cancer, comprising administering a pharmaceutically effective amount of a miR486 expression inhibitor to an individual suffering from a neurological disease.
  • the present invention provides a pharmaceutical composition for preventing and treating neurological diseases, comprising miR486 expression inhibitor as an active ingredient.
  • the present invention also provides a method for preventing and treating cancer, comprising administering a pharmaceutically effective amount of a miR486 expression inhibitor to an individual suffering from a neurological disease.
  • It provides a method for screening a candidate substance for preventing and treating neurological diseases, comprising selecting a test substance whose NeuroD6 expression amount is increased compared to a control group not treated with the test substance.
  • the present invention also provides a kit for diagnosing a neurological disease, comprising a nucleotide of the miR486 gene, a nucleotide having a sequence complementary to the nucleotide, or a fragment thereof.
  • step 2) comparing the expression level of miR486 of step 1) with the expression level of miR486 of a sample derived from a normal individual as a control;
  • step 3 Provides a gene detection method for providing information for diagnosing neurological disease, comprising determining that the miR486 expression level of step 2) is higher than that of the control group.
  • step 2) comparing the expression level of miR486 of step 1) and the expression level of miR486 of a sample derived from a normal individual as a control;
  • step 3 When the miR486 expression level of step 2) is increased compared to the control group provides a method for diagnosing a neurological disease comprising the step of determining that the subject has a neurological disease.
  • the present invention also provides a pharmaceutical composition for the prevention and treatment of neurological diseases containing NeuroD6 as an active ingredient.
  • Advantageous Effects In the present invention, expression interference of miR486 using miR486 expression inhibitor
  • the niiR486 inhibitor can be used as an active ingredient of a pharmaceutical composition for preventing and treating neurological diseases, and miR486 expresses NeuroD6.
  • Mechanism to control the can be used to screen for candidates for the prevention and treatment of neurological diseases, can be used as a marker for diagnosing neurological diseases.
  • FIG. 1A shows the expression of NeuroD6, a miR486 target gene, before spinal cord injury and after 7 days of injection of miR486 or scrambled RNA;
  • FIG. 1C shows miR486 region ⁇ “in motor neurons in injured lesions in spinal cord tissue
  • FIG. 1D shows immunohistochemical images of NeuroD6 expression of NF160 + motor neurons in spinal cord injury lesions (T10), and arrows represent NeuroD6 expressing in NF160 + motor neurons;
  • FIG. 1E shows a schematic flow diagram of an experimental procedure for treating and evaluating spinal cord injury.
  • FIG. 2 (a) shows the functional outcome of antisense ⁇ m iR486 (antisense TM miR486) treatment in the injured spinal cord assessed with Beattie and Bresnahan (BBB);
  • FIG. 2 (b) shows antisense-injured spinal cord in the damaged spinal cord assessed on the Basso mouse scale (BMS). miR486 treatment results are shown;
  • Figure 2 (c) shows the induction of traumatic spinal cord injury symptoms when injecting miR486
  • Figure 2 (d) shows histological analysis and apoptotic cell death evaluation of neuronal destruction when miR486 or scrambled RNA treated normal and damaged spinal cord;
  • FIG. 2 shows normal spinal cord (Normal SC), spinal cord treated with scrambled RNA (scramRNA SO, damaged spinal cord (SCI), damaged spinal cord treated with scrambled RNA (SCI / scramRNA), normal spinal cord treated with miR486 ( comparison of TUNEL-positive populations in injured spinal cords treated with itiiR486 / SC) and antisense_1 13 ⁇ 486 (3 (: 1 / ⁇ ⁇ 111? 486);
  • FIG. 2 (f) shows caspase-3 (caspase) in the normal spinal cord, spinal cord treated with scrambled RNA, damaged spinal cord, damaged spinal cord treated with scrambled RNA, normal spinal cord treated with miR486, and damaged spinal cord treated with antisense miR486. -3) express expression;
  • Figure 2 (g) shows ATP production in normal spinal cord, spinal cord treated with scrambled RNA, damaged spinal cord, damaged spinal cord treated with scrambled RNA, normal spinal cord treated with miR486 and damaged spinal cord treated with antisense-miR486;
  • Figure 2 (h) shows immunohistochemical analysis of neurodestructive phenotypes in spinal cord injury and miR486 interference in spinal cord injury mice 5 and 10 days after antisense miR486 or scrambled RNA treatment;
  • FIG. 2 (i) shows the evaluation of myelin destruction and neurodestruction by Luxal Fast Blue (LFB) staining of damaged spinal cord tissue
  • FIG. 2 (j) shows the expression of TuJ, MBP and GFAP of neuronal markers NeuroD6 and NF160 after post-treatment on days 0, 2 and 7 of miR486 expression interference in injured spinal cord tissue.
  • 3 shows that miR486 expression interference markedly inhibited pro-inflammatory factor secretion and also induced R0SVIII gene expression in spinal cord injury:
  • 3 (a) and 3 (b) show real-time RT-PCR analysis and histochemical analysis of other pro-inflammatory cytokine expression before interference with the damaged spinal cord and after 7 days of miR486 expression interference or scramble A treatment. Represents;
  • FIG. 3D shows real-time RT-PCR analysis of SEPN1, TXNL1, GPxl and GPx3, genes involved in the R0S-elimination system 7 days after interference or expression of miR486 on medullary spinal cord lesions;
  • FIG. 3 (f) shows an immunohistochemical analysis of the expression of the redox removal protein TXNL1 in motor neurons of injured spinal cord tissue (T10);
  • Figure 3 (g) shows an immunohistochemical analysis of the expression of the redox protein GPx3 in motor neurons of injured spinal cord tissue (T10);
  • Figure 3 (h) shows an immunohistochemical analysis of the expression of SEPN1, a redox removal protein in motor neurons of injured spinal cord tissue (T10).
  • FIG. 4 (a) shows a schematic flow chart of an experimental procedure for i R486 and miR486 expression interference and functional analysis
  • Figure 4 (b) shows the expression of Tuj, NF160, MBP and GFAP when injected miR486 and scrambled RNA into normal spinal cord, damaged spinal cord, normal spinal cord;
  • FIG. 4 (e) shows R0S expression levels in spinal cord lesions, the group injected with scrambled R A to the normal spinal cord, the group injected with miR486 to the normal spinal cord, and the group injected with miR486 and antisense -miR486 to the normal spinal cord;
  • Figure 4 (g) shows the expression of P-P13K, p-Akt, p-p38, p-JNK, Bax, caspase-3 and cytochrome C when treated with scrambled RNA and miR486 in the normal and damaged spinal cord ;
  • Figure 4 (h) shows the flow cytometry results and TUNEL analysis results when the scrambled RNA and miR486 injected into the cultured NPC;
  • FIG. 5 shows knockdown of NeuroD6 expression leading to traumatic spinal cord injury-like phenotype
  • Figure 5 shows the miR486-binding site of 3UTR of the ⁇ u-NeuroD6 gene of chromosome 6 and the base sequence complementary between NeuroD6 and the miR486 gene;
  • FIG. 5 shows the chip (CHIP) -PCR results for miR486 expression interference inducing NeuroD6 binding to regulatory sites of GPx3 and TXNL1 genes;
  • Figure 5 (c) shows the result of traumatic spinal cord injury-like symptoms such as hind limb paralysis in mice 1 day after injection of NeuroD6 into the normal spinal cord;
  • FIG. 5D shows in situ analysis (day 7) of NeuroD6 expression in spinal cord injury, miR486-implanted normal spinal cord and iniR486 / simiR486-implanted normal spinal cord;
  • FIG. 5 (e) shows Reactive Oxygen Species (ROS) expression in 7-day spinal cord tissue when NeuroD6 is knocked down in normal spinal cord tissue;
  • Figure 5 (g) shows the expression of inflammatory factors EDI, Cox2, iNOS and eNOS when NeuroD6 knocked down in normal spinal cord, 1 day after NeuroD6 injection;
  • Figure 5 (h) is the day of NeuroD6 injection when knocked down NeuroD6 in normal spinal cord Later IL-I ⁇ IL-6 and TNFa expression;
  • FIG. 5 (i) shows that NeuroD6 expression interference in normal spinal cord expresses SEPN1, TXNL1, TXNL2, Gpxl and Gpx3, factors related to the R0SVIII deletion system, compared to control (scrambled RNA in normal spinal cord);
  • FIG. 5 (q) shows a schematic flow chart of R0S / miR486-mediated neurodestruction of motor neurons after traumatic injury in the spinal cord.
  • Figure 6 shows the expression of R0S-removing enzymes and neurodegeneration of motor neurons when restoring NeuroD6 expression in spinal cord injury:
  • FIG. 6A is a flow chart illustrating an experimental procedure for processing miR486, antisense -miR486, siNeuroD6 and NeuroD6 and functional judgment;
  • Figure 6 (g) shows co-expression of NF160 and Cox2, co-expression of ED1 and GFAP, co-expression of NF160 and NeuroD6 when treated with scrambled RNA, NeuroD6, miR486, mir486 and NeuroD6, siNeuroD6 or siNeuroD6 and NeuroD6 in the injured spinal cord Expression;
  • the present invention prevents neurological diseases comprising miR486 expression inhibitor as an active ingredient And pharmaceutical compositions for treatment.
  • the present invention also provides a method of treating a neurological disorder comprising administering a pharmaceutically effective amount of a miR486 expression inhibitor to a subject suffering from the neurological disorder.
  • the present invention also provides a method for preventing a nervous system disease comprising administering to a subject a pharmaceutically effective amount of a miR486 expression inhibitor.
  • the present invention also provides a use of miR486 expression inhibitor in the manufacture of a pharmaceutical composition for preventing and treating neurological diseases.
  • the miR486 expression inhibitor may be selected from the group consisting of antisense oligonucleotides complementary to the miR486 gene, small interfering RNA (siRNA), short hairpin A and ribozyme, but are antisense nucleotides But may not be limited thereto.
  • antisense nucleotides as defined in the Watson-click base pair, bind (combine) to the complementary sequencing of DNA, immature -mR A or mature mR A to disrupt the flow of genetic information as a protein in DNA.
  • the nature of antisense nucleotides specific to the target sequence makes them exceptionally multifunctional. Since antisense nucleotides are long chains of monomeric units they can be easily synthesized for the target RNA sequence. Many recent studies have demonstrated the utility of antisense nucleotides as biochemical means for studying target proteins (Rothenberg et al., J. Natl. Cancer Inst., 81: 1539-1544, 1999).
  • antisense nucleotides can be considered as a novel form of inhibitor because of recent advances in nucleotide synthesis and in the field of nucleotide synthesis that exhibit improved cell adsorption, target binding affinity and nuclease resistance.
  • the siRNA is composed of a 15-30 mer sense sequence selected from the base sequence of the miR486 gene and an antisense sequence complementarily binding to the sense sequence, wherein the sense sequence is not particularly limited thereto. It may be composed of 25 bases, but is not limited thereto.
  • the miR486 expression inhibitor may be to increase the expression of NeuroD6, but is not limited thereto.
  • the neurological diseases include spinal cord injury, Parkinson's disease, stroke, muscular dystrophy, scoliosis, motor nerve injury, peripheral nerve injury due to trauma, ischemic brain injury, neonatal hypoxic ischemic brain injury, cerebral palsy, epilepsy, refractory epilepsy, Al Sheimer's disease, congenital metabolic neurological disease and traumatic brain injury may be any one selected from the group 5, but may be more specifically spinal cord injury, but not limited to this, may occur due to nerve damage Applicable to all neurological diseases present.
  • the present inventors have identified the effect of miR486 overexpression on neuronal destruction of R0S mediated spinal cord injury in motor neurons. It was found that most target genes of miR486 were downregulated, including overexpression of miR486 10 and NeuroD6 in spinal cord injury tissues (see FIGS. La and lb). In spinal cord injury lesions, only miR486 was expressed in NF160 + motor neurons, and 2 ', 7'-dichlorodihydrofluorescein diacetate (2 ⁇ , 7'-dichlorodihydrof kiorescein diacetate, DCFDA) was produced in the cytoplasm and reactive oxygen species (Reactive) oxygen species (R0S) was confirmed to accumulate (see FIG. 15). NeuroD6 expression was detected in the nucleus and cytoplasm of lesions of spinal cord injury and motor neurons of normal spinal cord tissue. In addition, only ED1 + macrophages produced R0S, but did not express miR486 (see FIG. ID).
  • miR486 is a result of confirming the recovery of motor function according to the interference of miR486 expression.
  • Expression interfered spinal cord injury animals showed improved motor function. Three days after spinal cord injury, • 20 miR486 expression interference spinal cord injury animals despite the 'most there was a part that rotates during their feet walking, and parallel to the walking continued their were weighed maintained during the arrest phenotype was gradually restored to . In contrast, injured animals injected only with scrambled siRNA had limited joint movement (see FIGS. 2A and 2B).
  • miR486 interference effectively expressed the expression of i R486 and Cox2, EDI, ⁇ IL6 and TNFCL.
  • FIG. 3a the expression of redox-causing factors MP0 and eNOS was reduced, and the R0S-removing enzymes SEPNl, TXNLl, GPxl and GPx3 were significantly increased (FIGS. 3c, 3d, 3f and 3h).
  • the GPx3, TXNLl and SEPNl expression was detected in NF160 + motor neurons in normal spinal cord tissue (see FIG. 3E).
  • the number of ED1-positive inflammatory cells, macrophages, microglial cells was significantly reduced (see FIG. 3B).
  • NPC neural progenitor cells
  • knockdown of miR486 expression effectively induced p-PI3K / p-Akt expression, improved NPC survival, and allowed cells to escape neuronal cell death. It was. Interference with miR486 expression after antisense -miR486 injection in spinal cord injury mice resulted in increased target gene expression.
  • the present inventors confirmed that miR486 expression in the injured spinal cord and the initiation of anti-oxidative reaction through GPx3 and TXNL1 ol by insufficient NeuroD6 resulted in interference of miR486 expression induced NeuroD6 binding to the regulatory sites of GPx3 and TXNL1 (see FIG. 5B). ). It was confirmed that R0S accumulated significantly in spinal cord injury and miR486-injected spinal cord tissue and siNeuroD6-injected spinal cord tissue (see FIG. 5E). It was confirmed that in vivo expression of the motor neuron markers NF160, Tuj and MBP were downregulated in spinal cord tissue after siNeuroD6 injection (see FIG. 5F).
  • NeuroD6 expression knockdown in the intact spinal cord has cytotoxic effects on motor neurons, increased expression of TNF ⁇ , ⁇ , IL6, C0X2, iNOS and eNOS, and expression of the R0S clearance factors SEPNl, TXNL2, GPxl and GPx3 was down-regulated (see FIGS. 5G-5I).
  • NeuroD6 or R0S regulated miR486 expression As a result of confirming ATP expression, NeuroD6-mediated neuronal protection in spinal cord or cultured NPCs was also associated with caspase-3 downregulation and normalized ATP synthesis ability (see FIG. 5J). In particular, miR486 expression increased more than 3-fold (320%) in hydrogen peroxide treatment and NeuroD6 or ascorbic acid treatment significantly reduced hydrogen peroxide-mediated miR486 increase (194% and 132%, respectively) (see FIG. 5K). Hydrogen peroxide-induced R0S production was significantly inhibited by the expression of NeuroD6 (53%), SEPNl (37%), TXNL1 (23%) and GPx3 (40%).
  • NeuroD6 or ascorbic acid (5 / zg / ml) treatment significantly upregulated SEPN1, TXNL1 and GPx3 compared to the exposure effect with hydrogen peroxide alone (see FIGS. 51-5O).
  • exogenous NeuroD6 protected neurons from ROS-mediated neurotoxicity (see Figure 5p).
  • the composition containing the miR486 expression inhibitor of the present invention as an active ingredient may include, but is not limited to, the active ingredient in an amount of 0.0001 to 50 weight 3 ⁇ 4> based on the total weight of the composition.
  • composition of the present invention may contain one or more active ingredients exhibiting the same or similar function in addition to the miR486 expression inhibitor.
  • composition of the present invention may be prepared by including one or more pharmaceutically acceptable carriers in addition to the above-described active ingredients for administration.
  • Pharmaceutically acceptable carriers may be used in combination with saline, sterile water, Ringer's solution, saturated saline, textose solution, maltodextrin solution, glycerol, ethanol, liposomes, and one or more of these components.
  • other conventional additives such as buffers and bacteriostatic agents can be added.
  • diluents, dispersants, surfactants, binders and lubricants can be additionally added to formulate injectable formulations such as aqueous solutions, suspensions, emulsions, pills, capsules, granules or tablets, and can act specifically on target organs.
  • injectable formulations such as aqueous solutions, suspensions, emulsions, pills, capsules, granules or tablets, and can act specifically on target organs.
  • Target organ specific antibodies or other ligands may be used in combination with the carriers so as to be used.
  • it can be formulated according to each disease or component using an appropriate method in the art or using the method disclosed in Remington's Pharmaceutical Science (Recent Edition, Mack Publishing Company, Easton PA). ⁇
  • Nucleotides or nucleic acids used in the present invention can be prepared for oral, topical, parenteral, nasal, intravenous, intramuscular, subcutaneous, intraocular, transdermal and the like. More specifically, nucleic acids or vectors are used in injectable forms. Thus, in particular the area to be treated may be combined with any pharmaceutically acceptable vehicle for injectable compositions for direct infusion.
  • the compositions of the present invention may in particular comprise isotonic sterile solutions or lyophilized compositions which allow the composition of injectable solutions upon the addition of dry, in particular sterile water or appropriate physiological saline. Direct injection of nucleic acid into a patient's tumor is advantageous because it allows the treatment efficiency to be focused on the infected tissue.
  • the dosage of nucleic acid used can be adjusted by various parameters, in particular by gene, vector, mode of administration used, disease in question or alternatively desired duration of treatment. In addition, the range varies depending on the patient's weight, age, sex, health status, diet, administration time, administration method, excretion rate and the severity of the disease.
  • the daily dosage is specifically about 0.0001 to 100 mg / kg, more specifically 0.001 to 10 mg / kg, and may be administered once to several times a day.
  • the present invention also provides a method for preventing or treating a neurological disease, comprising administering to a subject a pharmaceutically effective amount of the composition.
  • the neurological disorders specifically include spinal cord injury, Parkinson's disease, stroke, muscular dystrophy, scoliosis, motor neuron injury, peripheral nerve injury due to trauma, ischemic brain injury, neonatal hypoxic ischemic brain injury, cerebral palsy, epilepsy, refractory epilepsy, Al-Hymer's disease, congenital metabolic nervous system disease and traumatic brain injury may be any one selected from the group, and more specifically, spinal cord injury, but not limited thereto, but may be caused by nerve injury. Applicable to all possible neurological diseases.
  • the pharmaceutically effective amount is 0.0001 to 100 mg / kg, 0.001 to 10 mg / kg, but is not limited thereto. Dosage may vary depending on the weight, age, sex, health status, diet, duration of administration, rate of administration, elimination rate, and severity of the particular patient.
  • composition can be administered orally or parenterally during clinical administration and intraperitoneal injection, rectal injection, subcutaneous injection, intravenous injection, intramuscular injection, intrauterine dural injection, cerebrovascular injection or intrathoracic injection during parenteral administration.
  • the subject is a spinal cord animal, specifically a mammal, more specifically an experimental object such as a rat, a rabbit, a guinea pig, a hamster, a dog or a cat, and more specifically, an ape-like animal such as a chimpanzee or a gorilla. Can be.
  • the pharmaceutical composition comprising the m iR486 inhibitor as an active ingredient can be usefully used for preventing or treating neurological diseases.
  • the present invention
  • 3) provides a method for screening a candidate substance for preventing and treating neurological diseases, including selecting a test substance whose expression level is reduced compared to a control group that does not process the test substance.
  • It provides a method for screening a candidate substance for preventing and treating neurological diseases, comprising selecting a test substance whose NeuroD6 expression amount is increased compared to a control group not treated with the test substance.
  • Expression measurement of step 2) is RT-PCR, Quantitative or semi-Quentitative RT-PCR, Quantitative or semi-Quentitative real-time RT-PCR -PCR), Northern blot, and DNA or RNA chip may be measured using any one method selected from the group consisting of, but is not limited thereto.
  • miR486 gene is overexpressed in the spinal cord injury and miR486 is overexpressed to inhibit the expression of NeuroD6, the miR486 and NeuroD6 prevent neurological diseases and 1 can be used for screening therapeutic candidates.
  • the present invention also provides a kit for diagnosing a neurological disease comprising a nucleotide of the miR486 gene, a nucleotide having a sequence complementary to the nucleotide, or a fragment thereof.
  • step 2) comparing the expression level of miR486 of step 1) with the expression level of n] iR486 in a sample isolated from normal individuals as a control;
  • step 3 Provides a gene detection method for providing information for diagnosing neurological disease, comprising determining that the miR486 expression level of step 2) is higher than that of the control group.
  • step 2) comparing the expression level of miR486 of step 1) with the expression level of miR486 of a sample isolated from a normal individual as a control;
  • step 2) When the miR486 expression level of step 2) is increased compared to the control group provides a method for diagnosing a neurological disease comprising the step of determining that the subject has a neurological disease.
  • the nervous system diseases include spinal cord injury, Parkinson's disease, stroke, muscular dystrophy, scoliosis, motor nerve injury, peripheral nerve injury due to trauma, ischemic brain injury, neonatal hypoxic ischemic brain injury, cerebral palsy, epilepsy, refractory epilepsy, Alzheimer's disease.
  • the disease may be one selected from the group consisting of congenital metabolic neurological disease and traumatic brain injury, but is not limited thereto.
  • the miR486 and NeuroD6 can be used in kits for providing information for diagnosing neurological disease, gene detection and diagnosing neurological disease.
  • the present invention also provides a pharmaceutical composition for preventing and treating neurological diseases, including NeuroD6 as an active ingredient.
  • the present invention also provides a method of treating a neurological disease, comprising administering a pharmaceutically effective amount of NeuroD6 to a subject having a neurological disease.
  • the present invention also provides a method for preventing neurological disease, comprising administering to a subject a pharmaceutically effective amount of NeuroD6.
  • the present invention also provides the use of NeuroD6 in the manufacture of a pharmaceutical composition for preventing and treating neurological diseases.
  • the nervous system diseases include spinal cord injury, Parkinson's disease, stroke, muscular dystrophy, scoliosis, motor nerve injury, peripheral nerve injury due to trauma, ischemic brain injury, neonatal hypoxic ischemic brain injury, cerebral palsy, epilepsy, intractable epilepsy, Alzheimer's disease.
  • the disease may be one selected from the group consisting of congenital metabolic neurological disease and traumatic brain injury, but is not limited thereto.
  • the present invention provides a vector comprising a polynucleotide encoding NeuroD6, or a pharmaceutical composition for preventing and treating neurological diseases containing the cells containing the vector as an active ingredient.
  • the nervous system diseases include spinal cord injury, Parkinson's disease, stroke, muscular dystrophy, scoliosis, motor neuron injury, peripheral nerve injury due to trauma, ischemic brain injury, neonatal hypoxic ischemic brain injury, cerebral palsy, epilepsy, intractable epilepsy, Alpheimer's disease.
  • the disease may be one selected from the group consisting of congenital metabolic. Nervous system disease and traumatic brain injury, but is not limited thereto.
  • the nervous system diseases include spinal cord injury, Parkinson's disease, stroke, muscular dystrophy, scoliosis, motor neuron injury, peripheral nerve injury due to trauma, ischemic brain injury, neonatal hypoxic ischemic brain injury, cerebral palsy, epileptic refractory epilepsy, Alpheimer's disease. It may be any one selected from the group consisting of congenital metabolic neurological disease and traumatic brain injury, but is not limited thereto.
  • a method for screening a candidate substance for preventing and treating neurological diseases comprising selecting a test compound or composition in which the activity of the NeuroD6 protein of step 2) is increased compared to the activity of the NeuroD6 protein untreated with the test compound or composition.
  • the neurological disorders include spinal cord injury, Parkinson's disease, stroke, muscular dystrophy, scoliosis, motor neuron injury, peripheral nerve injury due to trauma, ischemic brain injury, neonatal hypoxic ischemic brain injury, cerebral palsy, epilepsy, refractory epilepsy, egg ⁇ Hemer's disease, congenital metabolic nervous system disease and traumatic brain injury may be any one selected from the group consisting of, but is not limited thereto.
  • 3) Provides a protein detection method for providing information of neurological disease diagnosis, comprising the step of determining that the risk of neurological disease is high when the expression level of NeuroD6 protein is reduced compared to the control group.
  • the nervous system diseases include spinal cord injury, Parkinson's disease, stroke, muscular dystrophy, scoliosis, motor neuron injury, peripheral nerve injury due to trauma, ischemic brain injury, neonatal hypoxic ischemic brain injury, cerebral palsy, epilepsy, refractory epilepsy, Alzheimer's disease. It may be one selected from the group consisting of a disease, congenital metabolic nervous system disease and traumatic brain injury, but is not limited thereto.
  • NeuroD6 can be usefully used as an active ingredient in pharmaceutical compositions for preventing and treating neurological diseases.
  • the present invention will be described in detail by way of examples.
  • mice were intraperitoneally administered in an amount of 25 ⁇ / g using 1.25% avertin and then anesthetized, and the back skin of the mouse was incised. ), The spinal cord at the T10 region was damaged to a size of about 1 mm deep in the dorsal direction and about 2 mm wide, and then sutured to receive antibiotics (amicin). Antibiotics were administered intramuscularly every two days.
  • mice were fixed with stereotaxic instruments under anesthesia.
  • DharmaFECT Dermata Inc., Chicago Antisense miR486 (anti _miR486 or anti miR; 20 ⁇ stock solution, Dhamacon) diluted with IL) was injected once 2 days after SCI with a 25 ⁇ Hamilton syringe with 26 gauge needles.
  • the antisense -miR486 injection dose finally used 10 ⁇ .
  • the rate of microinjector in the stereosuspension instrument was 1 / min.
  • the syringe was secured at an angle of about 90 degrees above the spinal cord of the stereosuspension instrument.
  • NeuroD6 (0pen Biosystems) was injected once 2 days after spinal cord injury. NeuroD6 infusion doses were 100, 200 and 400 ng. NeuroD6 was diluted with lipofectamin (Klinvitrogen, Carlsbad, Calif.) And NeuroD6 Vector DNA constructs were obtained from Open Biosystems (Thermo Scientific Co.). NeuroD6 injection method was carried out according to the same method as the antisense -miR486 injection method of Example ⁇ 1-2>. Under anesthesia by intravenous anesthesia (Avert in), the rat spinal cord was opened using a surgical knife, and the spinal cord bone was removed using surgical scissors and bone tongs. ⁇ 1-4> Infusion of miR486
  • miR486 (Dharmcon) was injected only once into the Hamilton syringe with a 26-gauge needle with a microinjector into the T11-L1 site.
  • concentration of miR486 stock solution was 20 ⁇ , which was diluted with DharmaFECTCDharmacon Inc., Chicago, IL).
  • the miR486 injection dose was finally 10 ⁇ and the total injection volume of miR486 was 5 per rat.
  • the miR486 injection rate of the microinjector was 1 ⁇ / tnin.
  • the syringe was fixed to a stereosuspension instrument 90 degrees above the spinal cord. The needle was injected into the T11-L1 site of the spinal cord and the syringe was left in the site for 3 minutes to prevent leakage after miR486 injection.
  • NeuroD6 siRNAs siNeuroD6; Dharmcon
  • mice were fixed in stereosuspension instruments under anesthesia via intravenous anesthesia injection.
  • NeuroD6 siRNAs were administered once by injection dose of NeuroD6 siRNAs (10 ⁇ ), and the 20 ⁇ stock solution of NeuroD6 siRNAs was also diluted with the same volume of DharmaFECT (Dharaacon Inc., Chicago, IL).
  • NeuroD6 siRNAs (10 ⁇ ) injection into the normal spinal cord NeuroD6 (100 ng) was injected once a day at the same site of NeuroD6 siRNAs injection.
  • NeuroD6 100 ng was injected once a day at the same site of NeuroD6 siRNAs injection.
  • PC Neural progenitor cells
  • NPCs Spinal cord-derived neural progenitor cells
  • the tissue was crushed into small pieces ( ⁇ 1 1 ⁇ 3 ) and digested with 0.025% trypsin at 371: for 30 minutes.
  • the enzyme activity of the trypsin was neutralized with DMEM containing 10% FBS, and the sample was centrifuged at 1500 rpm for 5 minutes to obtain a high concentration of cell pellet.
  • the NPC pellets were incubated overnight at 37 ° C.
  • DMEM containing 10% FBS fetal bovine serum
  • the medium was changed 48 hours and then every 4 days. After 2 or 3 passages, cells were used to characterize their differentiation capacity. The cells were placed in differentiation conditions to confirm the neural differentiation capacity of the cultured cells.
  • the cultured NPCs formed globular clumps of cells and floating neurospheres in the culture medium. Transfer the cells to a petri dish and add Neurobasal neuron-specific medium (B27 (Invitrogen, Gaithersburg, MD), 20 ng / ml bFGF and 20 ng / ml BDNF (Sigma, St Louis, M0)) NB, Invitrogen, Gaithersburg, MD) was incubated for 3 days.
  • Neurobasal neuron-specific medium B27 (Invitrogen, Gaithersburg, MD), 20 ng / ml bFGF and 20 ng / ml BDNF (Sigma, St Louis, M0)
  • the culture concentration of the spheres was maintained at 1 20 cells / cm 2 to prevent self-counseling.
  • neuron-derived neurospheres were placed in a cover-slip double-coated with PDL-laminin. During differentiation, 70% of the medium every 4 days Replaced. All data presented were run at least three times.
  • ⁇ 3-1> Confirmation of expression of miR486 and its target genes following spinal cord injury Extract RNA of whole cells with TrizoKLife Technologies, Frederick, Mass., USA), and PCR by using 20 pni specific oligo—dT primers. Amplified by cycle (95 ° C. 1 minute; 55 ° C. 1 minute; 72 ° C. 1 minute) and reverse transcribed into first strand cDNA.
  • the PCR reaction was performed using ABI 7700 Prism Sequence Detection System and SYBER green detection kit (Applied Biosystems, Foster, CA, USA).
  • the primer sequence was designed with Primer Express software (PE-Ai Bio Bios, Warrington, UK) using the gene sequence obtained from the GeneBank database (Table 1). For PCR product labeling, the SYBER green detection kit (Applied Biosystems) was used.
  • the labeled oligonucleotides were diluted in a shake buffer containing 50% ion free formamide, 4X SSC and sodium pyrophosphate and covered with parafilm.
  • the reaction was carried out in a humidified bath at 45 ° C for 12 hours. After reaction, the sections were washed with IX SSC for 10 minutes at room temperature, washed with IX SSC for 20 minutes at 60 t, and distilled water for 5 minutes at room temperature. It was washed, dehydrated with 70%, 95% and 100% isopropanol and finally dried in air. Controls missing specific probes did not have detectable staining.
  • the samples were evaluated by Leica fluorescence microscopy (Leica Microsystems, Exon, PA, USA). This test was repeated at least three times.
  • the sections were fixed for 30 minutes in 43 ⁇ 4 paraformaldehyde for immunohistochemical analysis of spinal cord tissue. The sections were then washed three times in PBS and the nonspecific binding was blocked with normal horse serum. The cut sections were reacted with the following antibodies at 4 ° C. for 12 hours; Anti-GFAP (1: 2000; Dako), anti-EDI (1: 1000; Cell Signal), anti-Tuj (1: 250; Sigma) and anti-NF160 (1: 250; Sigma). After washing off the primary antibody, the cut sections were incubated for 1 hour. After strong washing in PBS, the cells were reacted with FITC or Texas-red conjugated secondary antibody (1: 250; Molecular Probe; and 1: 250; Jackson Laboratory, respectively) for 30 minutes.
  • Controls using IgG with or without irrelevant primary antibody were not stained.
  • the samples were evaluated using a Leica fluorescence microscope (Leica Microsystems, Exon, PA, USA). Immunocytochemical analysis was repeated at least three times. Double-labeled cells were identified through 1 iffli cutting plane collection through the sample.
  • miR486 was expressed only in NF160 + motor nerve, and cytoplasm was accumulated in R0S by producing 2 ', 7'-dichlorodihydrofluorescein diacetate (2', 7'-dichlorodihydrof luorescein diacetate, DCFDA). It was confirmed (Fig. Lc). NeuroD6 expression was also detected at high levels in the nucleus and cytoplasm of motor neurons in lesions of spinal cord injury and in normal spinal cord tissue, and R0S in ED1 + macrophages. Produced, but did not express miR486 (FIG. Id).
  • BBB Beat tie and Bresnahan locomotor rating scale and Basso mouse scale
  • TdT in situ apoptosis detector kit (Roche, USA) according to the manufacturer's instructions. After the damaged spinal cord tissue is fixed, the damaged tissue. Sections were placed in 4% paraformaldehyde, 90% at 37 ° C with TUNEL reaction mixture containing deoxynucleot idyl transferase (TdT) buffer and biotinylated dUTP. Reaction in humid environment for minutes. Then, the secondary antibody bound to the fluorescent label After reaction, the results were analyzed using a fluorescence microscope (Leica Microsystem, PA).
  • TUNEL-positive apoptotic cells and cells at spinal cord lesion sites were quantified by counting positively stained cells. Three 100-fold microscopic images were randomly captured in the range where the positive cells were abundant around the lesion site for each cut. The number of positively stained cells of the three images was averaged. The results are expressed as relative cell ratios per field of view under the microscope.
  • caspase 3 activity was measured in traumatic spinal cord tissue 1 week after antisense-miR486 treatment.
  • caspase-3 activity analysis 10 proteins in a total 50 ⁇ volume were mixed with the equilibrated caspase-3 reagent (Promega). After reacting at room temperature for 1 hour, luminescence was measured using a TD 20/20 luminometer (LuminoiTieter) (Turner Designs, Sunnyvale, CA). Blank values were excluded and fold increase in activity was calculated based on activity measured from untreated cells. Each sample was measured three times.
  • the amount of protein was confirmed using the Protein Assay Kit (Bio-Rad, Hercules, CA, USA) according to the manufacturer's instructions.
  • Cells were 150 mmol / 1 KC1, 25 mmol / 1 Tris-HCl; pH 7.6, 2 mmol / 1 EDTA pH 7.4, 10 mmol / 1 KP0 4 pH 7.4, 0.1 ⁇ ol / l MgCl 2 and 1 mg protein concentration per ml of 0.1% (w / v) buffer Bovine Serum Albumin (BSA) Suspended in the containing buffer.
  • BSA Bovine Serum Albumin
  • ATP synthesis was carried out in substrate buffer 750 ⁇ ⁇ 10 mmol / 1 malate, 10 mmol / 1 pyruvate, 1 mmol / 1 ADP, 40 zg / ml digitonin and 0.15 mmol / 1 adenosine pentaphosphate.
  • the cell suspension was added to add 250 ⁇ . Cells were reacted at 37 ° C for 10 minutes. 50 ⁇ of the reaction mixture was removed at 0 and 10 minutes, and 100 mmol / 1 Tris-HCl, 4 mmol / 1 EDTA (pH 7.75) boiling for 2 minutes was removed. Digestion and the digested buffer were diluted to 1/10.
  • the amount of ATP was measured with an ATP Bioluminescence Assay Kit (Roche Diagnostics, Basel, Switzerland) according to the manufacturer's instructions on a luminometer (Berthold, Detection Systems, Pforzheim, Germany).
  • miR486 expression interference from spinal cord injury animals sikyeotgo significantly improved motor function, through the cascaded L-rise eu 3 activation, and ATP production dysfunction showed that protect the TU EL- positive cell death (Fig. 2f and 2g) .
  • LFB staining was performed as follows to confirm morphological changes.
  • the frozen section was first placed overnight in 1: 1 alcohol / chloroform and dehydrated using 93 ⁇ 4 ethyl alcohol.
  • the dehydrated sections were reacted overnight at 56 ° C. with 0.1% luxal fast blue solution (up to 16 hours).
  • excess dye was washed off with 95% ethyl alcohol and then washed with distilled water.
  • the slide was reacted with 0.05% Lithium carbonate solution for 30 seconds and then reacted with 70% ethyl alcohol for 30 seconds to distinguish between tissues. After washing with distilled water, it was dyed with cresyl violet solution for counterstaining.
  • miR486 interference effectively inhibited the expression of miR486 (88% of miR486 expression was downregulated) and the expression of the proinflammatory factors Cox2, EDI, ⁇ IL6 and TNF ⁇ (FIG. 3A).
  • miR486 expression interference After 7 days of miR486 expression interference, myeloperoxidase (MP0) and nitric oxide synthase (eNOS) expressions were markedly reduced, while 3 and 7 days after miR486 expression interference in spinal cord injury.
  • MP0 myeloperoxidase
  • eNOS nitric oxide synthase
  • TXNL1 and selenoprotein (Selenoprotein Nl, SEPN1) were induced.
  • P13k and p-Akt expression was induced when interference with ⁇ !? 486 expression in the injured spinal cord, but the expression of caspase-3 and cytochrome-C was reduced ( 3e).
  • CHIP chip analysis was performed to confirm NeuroD6 expression after miR486 interference.
  • the monoclonal antibody, anti-NeuroD6, was purchased from Santa Cruze and rabbit IgG (PP64B) antibody was purchased from Upstate.
  • the cells were centrifuged to make cell pellets and cryopreserved in liquid nitrogen.
  • the pellet was pre-IP lime buffer (10 mM Tris-HCl, 10 mM NaCl, 3 mM MgCl 2 , 1 mM CaCl 2> 4% GEPAL, 1 mM PMSF), 60 mL PMSF and additional components (100 mM PMSF, 256 Protease inhibitor, 20% SDS, 5 M NaCl, 3 ⁇ 40 2 ).
  • Cells were sonicated with a 1261 minute wave at 50% amplitude using a Branson Sonifier 450D and placed in ice water after 1 minute. Sonicated fractions ranged in size from 200-1000 bp.
  • Chip analysis after interference with miR486 expression confirmed the induction of NeuroD6 binding to regulatory sites of GPx3 and TXNL1 (FIG. 5B).
  • React ive oxygen species (ROS) accumulated significantly in spinal cord injury and miR486—injected spinal cord tissue and siNeuroD6-injected spinal cord tissue (FIG. 5E).
  • ROS React ive oxygen species
  • FIG. 5E After 48 hours of siNeuroD6 injection, protein expression levels were confirmed and in vivo expression of the motor neuron markers NF160, Tuj and MBP in spinal cord tissues was downregulated (FIG. 50.
  • NeuroD6 expression knockdown in intact spinal cord cells in motor neurons It has a toxic effect, increased the expression of TNFa, ⁇ , IL6, C0X2, iNOS and eNOS (FIG.) And confirmed gene expression.Knockdown of NeuroD6 expression in the intact spinal cord has a cytotoxic effect on motor neurons.
  • TNFa, ⁇ , IL6, C0X2, iNOS and eNOS and down-regulated expression of SEPNl, TXNL2, GPxl and GPx3, which are factors that eliminate R0S, and the composition of pathological microenvironment according to tissue R0S accumulation.
  • Fig. 5g-i NeuroD6-mediated neuronal protection in spinal cord or cultured NPCs was also associated with caspase-3 downregulation and normalized ATP synthesis ability (FIG. 5J).
  • m iR486 expression increased more than 3-fold (320%) in hydrogen peroxide treatment and NeuroD6 or ascorbic acid markedly decreased hydrogen peroxide-mediated miR486 increase (194% and 133 ⁇ 4 respectively) (FIG. 5K).
  • Hydrogen peroxide-induced R0S production was markedly inhibited by the expression of NeuroD6 (53%), SEPNl (37%), TXNL1 (23%) and GPx3 (40%).
  • NeuroD6 or ascorbic acid (5 ug / ml) treatment significantly upregulated SEPN1, TXNL1 and GPx3 compared to the exposure effect with hydrogen peroxide alone (FIGS. 51-5O).
  • exogenous NeuroD6 protected neurons from R0S-mediated neurotoxicity (FIG. 5P).
  • Example 7 Confirmation of neuroprotective effect of exogenous NeuroD6 in the damaged spinal cord
  • the expression of SEPNl, TX L1 and GPx3 and BBS and BMS were confirmed.
  • NeuroD6 overexpression significantly increased SEPN1, TXNL1 and GPx3 expression (FIGS. 6B-E).
  • NeuroD6-injected animals recovered from paralysis, maintained their weight consistently during parallel gait, and exhibited a mainly rotated foot position while walking.
  • injured animals injected with scrambled siRNA only had limited joint mobility (FIGS. 6H and 6I).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biomedical Technology (AREA)
  • Epidemiology (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biophysics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne une composition pharmaceutique comprenant un inhibiteur de l'expression de miARN486 en tant que principe actif dans la prévention et le traitement de troubles neurologiques. Plus particulièrement, comme il a été confirmé que l'interférence avec l'expression de miR486 au moyen d'un inhibiteur de miR486 surmontait l'inhibition de NeuroD6, induisant ainsi des améliorations dans les lésions médullaires ou SCI, l'inhibiteur du miR486 peut être utilisé en tant que principe actif dans une composition pharmaceutique destinée à la prévention et au traitement de troubles neurologiques; en outre, le mécanisme selon lequel le miR486 régule l'expression de NeuroD6 peut être appliqué dans le criblage de substances candidates destinées à la prévention et au traitement de troubles neurologiques.
PCT/KR2012/007112 2011-09-06 2012-09-05 Composition pharmaceutique comprenant un inhibiteur de l'expression du miarn486 en tant que principe actif dans la prévention et le traitement de troubles neurologiques WO2013036031A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2011-0090424 2011-09-06
KR1020110090424A KR20130026950A (ko) 2011-09-06 2011-09-06 microRNA486 발현 억제제를 유효성분으로 포함하는 신경계 질환 예방 및 치료용 약학적 조성물

Publications (2)

Publication Number Publication Date
WO2013036031A2 true WO2013036031A2 (fr) 2013-03-14
WO2013036031A3 WO2013036031A3 (fr) 2013-07-11

Family

ID=47832704

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2012/007112 WO2013036031A2 (fr) 2011-09-06 2012-09-05 Composition pharmaceutique comprenant un inhibiteur de l'expression du miarn486 en tant que principe actif dans la prévention et le traitement de troubles neurologiques

Country Status (2)

Country Link
KR (1) KR20130026950A (fr)
WO (1) WO2013036031A2 (fr)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102304041B1 (ko) 2019-12-24 2021-09-24 경북대학교 산학협력단 microRNA 억제제를 유효성분으로 포함하는 염증성 질환 예방 또는 치료용 조성물

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20100049079A (ko) * 2007-07-18 2010-05-11 더 리젠트스 오브 더 유니버시티 오브 콜로라도 인간의 정상 심장과 기능부진 심장에서 마이크로 rna의 차등 발현
WO2010054386A2 (fr) * 2008-11-10 2010-05-14 Battelle Memorial Institute Procédés, compositions et dispositifs utilisant du microarn pour déterminer des conditions physiologiques

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20100049079A (ko) * 2007-07-18 2010-05-11 더 리젠트스 오브 더 유니버시티 오브 콜로라도 인간의 정상 심장과 기능부진 심장에서 마이크로 rna의 차등 발현
WO2010054386A2 (fr) * 2008-11-10 2010-05-14 Battelle Memorial Institute Procédés, compositions et dispositifs utilisant du microarn pour déterminer des conditions physiologiques

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MOHAMMAD M. GHAHRAMANI SENO ET AL.: 'Gene and miRNA expression profiles in autism spectrum disorders' BRAIN RESEARCH vol. 1380, 22 March 2011, pages 85 - 97, XP028161669 *

Also Published As

Publication number Publication date
WO2013036031A3 (fr) 2013-07-11
KR20130026950A (ko) 2013-03-14

Similar Documents

Publication Publication Date Title
Xie et al. miR-145-5p/Nurr1/TNF-α signaling-induced microglia activation regulates neuron injury of acute cerebral ischemic/reperfusion in rats
Miyazaki et al. Targeting 5-HT1A receptors in astrocytes to protect dopaminergic neurons in parkinsonian models
Zhao et al. Hypoxia‐driven proliferation of embryonic neural stem/progenitor cells–role of hypoxia‐inducible transcription factor‐1α
TWI761444B (zh) 己糖激酶2專一性抑制劑在急性中樞神經系統損傷疾病中的應用
Fu et al. Suppressing long noncoding RNA OGRU ameliorates diabetic retinopathy by inhibition of oxidative stress and inflammation via miR-320/USP14 axis
Wu et al. Chloroquine promotes the recovery of acute spinal cord injury by inhibiting autophagy-associated inflammation and endoplasmic reticulum stress
Chen et al. Hydrogen sulfide protects against TNF-α induced neuronal cell apoptosis through miR-485-5p/TRADD signaling
Wang et al. Astrocytic CCAAT/Enhancer-binding protein delta contributes to reactive oxygen species formation in neuroinflammation
Xiao et al. Reduction of lactoferrin aggravates neuronal ferroptosis after intracerebral hemorrhagic stroke in hyperglycemic mice
Zhu et al. Higd1a facilitates exercise-mediated alleviation of fatty liver in diet-induced obese mice
Zhou et al. Targeting RPTPσ with lentiviral shRNA promotes neurites outgrowth of cortical neurons and improves functional recovery in a rat spinal cord contusion model
Huang et al. Expression of peroxiredoxin 1 after traumatic spinal cord injury in rats
Zhao et al. NIPA2 regulates osteoblast function via its effect on apoptosis pathways in type 2 diabetes osteoporosis
Zhang et al. Long non-coding RNA KCNQ1OT1 promotes hydrogen peroxide-induced lens epithelial cell apoptosis and oxidative stress by regulating miR-223-3p/BCL2L2 axis
JP2010527375A (ja) Gpr12の阻害による認知障害の治療方法
CN110592222A (zh) Triml1作为肝癌的分子标记物的应用
Gong et al. Downregulation of Nogo-B ameliorates cerebral ischemia/reperfusion injury in mice through regulating microglia polarization via TLR4/NF-kappaB pathway
Ma et al. Electroacupuncture‐regulated miR‐34a‐3p/PDCD6 axis promotes post‐spinal cord injury recovery in both in vitro and in vivo settings
Sun et al. High hepcidin level accounts for the nigral iron accumulation in acute peripheral iron intoxication rats
Hao et al. Tetramethylpyrazine promotes angiogenesis and nerve regeneration and nerve defect repair in rats with spinal cord injury
WO2017182580A1 (fr) Procédés de diagnostic et de traitement de troubles liés à la reproduction et procédés de contraception
WO2013036031A2 (fr) Composition pharmaceutique comprenant un inhibiteur de l'expression du miarn486 en tant que principe actif dans la prévention et le traitement de troubles neurologiques
Yang et al. Vasoactive intestinal peptide in rats with focal cerebral ischemia enhances angiogenesis
KR101191958B1 (ko) Tle1 억제제를 유효성분으로 함유하는 활액막 육종 예방 및 치료용 약학적 조성물
JP2007517498A (ja) 骨形態形成タンパク質(bmp)2a及びその使用

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: 12829806

Country of ref document: EP

Kind code of ref document: A2

122 Ep: pct application non-entry in european phase

Ref document number: 12829806

Country of ref document: EP

Kind code of ref document: A2