WO2015023054A1 - Animal model with obsessive compulsive disorder using ninjurin 1 deficiency - Google Patents

Animal model with obsessive compulsive disorder using ninjurin 1 deficiency Download PDF

Info

Publication number
WO2015023054A1
WO2015023054A1 PCT/KR2014/005531 KR2014005531W WO2015023054A1 WO 2015023054 A1 WO2015023054 A1 WO 2015023054A1 KR 2014005531 W KR2014005531 W KR 2014005531W WO 2015023054 A1 WO2015023054 A1 WO 2015023054A1
Authority
WO
WIPO (PCT)
Prior art keywords
ninjurin1
mice
ninjurin
ocd
animal model
Prior art date
Application number
PCT/KR2014/005531
Other languages
French (fr)
Korean (ko)
Inventor
김규원
리호앙
안범주
오구택
전세진
Original Assignee
서울대학교산학협력단
이화여자대학교 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 서울대학교산학협력단, 이화여자대학교 산학협력단 filed Critical 서울대학교산학협력단
Publication of WO2015023054A1 publication Critical patent/WO2015023054A1/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
    • A61K31/13Amines
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0276Knockout animals
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70571Receptors; Cell surface antigens; Cell surface determinants for neuromediators, e.g. serotonin receptor, dopamine receptor
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/035Animal model for multifactorial diseases
    • A01K2267/0356Animal model for processes and diseases of the central nervous system, e.g. stress, learning, schizophrenia, pain, epilepsy

Definitions

  • the present invention relates to an obsessive-compulsive animal model prepared by deficiency of ninjurin 1, and a method for searching for a candidate drug for preventing or treating OCD.
  • Phylogenetic trees have been reported using proteins known in GenBank to find proteins that have homology with Ninjurin. In vertebrates, two proteins are known, Ninjurin 1 and Ninjurin 2. Ninjurin 1 and Ninjurin 2 have been found in vertebrates such as humans, mice, and rats.
  • Ninjurin 1 is expressed in various tissues.
  • Ninjurin 1 so far known are related to cell adhesion, nerve outgrowth, cellular senescence and cancer.
  • Obsessive-Compulsive Spectrum Disorder is an involuntary and persistent anxiety disease, and impulsive and repetitive behaviors are the main phenomena to solve this problem.
  • OCD is a neurosis characterized by the presence of repetitive thoughts, illusions (obsessive) and repetitive urges or actions (forces) that the patient perceives as pathological or is strongly resistant within.
  • the present inventors first disclosed the relationship between OCD and Ninjurin 1, and suggests the possibility of using Ninjurin 1 KO mice as a new model for OCD studies and a target protein for treating OCD.
  • an object of the present invention is to provide an obsessive-compulsive animal model prepared by deficiency of ninjurin 1 (Ninjurin 1).
  • an object of the present invention is to provide a method for searching for a candidate drug for preventing or treating OCD by using an OCD animal model prepared by deficient in ninjurin 1 (Ninjurin 1).
  • the present invention provides an obsessive-compulsive animal model prepared by deficiency of ninjurin 1.
  • the animal is characterized in that except for humans.
  • the ninjurin 1 deficiency is characterized by knock-out of the ninjurin 1 gene.
  • the present invention provides a method for searching for a candidate drug for preventing or treating OCD by using an OCD animal model prepared by deficient in ninjurin 1 (Ninjurin 1).
  • the detection method is characterized in that it comprises the step of determining whether the N-Methyl-D-aspartate receptor (NMDA receptor) is inhibited after administration of the candidate drug to the animal model.
  • NMDA receptor N-Methyl-D-aspartate receptor
  • the candidate drug is characterized in that the NMDA receptor antagonist.
  • the NMDA receptor antagonist is characterized in that the memantine (Memantine).
  • a Ninjurin 1 KO mouse can be provided as a new model for OCD.
  • the present invention can provide a novel target protein for the treatment of OCD.
  • the present invention can provide a method for searching for a drug candidate for OCD.
  • the present invention can provide a drug candidate for the treatment of OCD.
  • the present invention can provide a treatment for OCD as a novel use of NMDA receptor antagonists.
  • FIG. 2 shows the results of anxiety and repetitive behaviors, which are compulsive disorders of Ninjurin1 KO mice, through marble-burying test.
  • Figure 7 shows the results of confirming the expression of Ninjurin1 in synaptosomes of Puncta structure using immunocytochemical analysis.
  • Figure 10 shows the results of confirming the expression of NMDAR and Src expression increase in Ninjurin 1 KO mice using Western blotting.
  • the present inventors have predicted that OCD and Ninjurin 1 may be related to each other, and as a result of research, OCD-like behaviors such as self-made injury, repetitive behavior, and increased anxiety in Ninjurin 1-removed mice (Ninjurin 1 KO mice) And anatomical analysis confirmed abnormalities in the formation of cortical-stiatal circuits.
  • NMDA receptors are neuroreceptors known to regulate cell death and signaling between normal cells. Direct interaction with the dopamine D1 receptor regulates cell death or induces communication between normal cells. When the NMDA receptor interacts with the D1 receptor, cell death and signaling regulation between cells are independently driven by different mechanisms. Conventionally, various studies on NMDA receptors have been made as part of treating various diseases such as stroke, osteoporosis, epilepsy and dementia.
  • NMDA receptor antagonists are substances that inhibit the function of the NMDA receptor, and (1) antagonistically bind to binding sites with promoters such as glutamic acid or NMDA (e.g., D-2-amino-5-phosphonoyl acetate) (2) antagonically binding to glycine binding sites essential for activation by NMDA receptor promoters (e.g., 7-chloroquinurene acid), (3) antagonistically binding to binding sites of polyamines, which are active enhancers (Eg, alkane), (4) binding to ion channel sites to block the flow of ions (eg MK-801, Mg2 +) is known.
  • promoters such as glutamic acid or NMDA (e.g., D-2-amino-5-phosphonoyl acetate)
  • antagonically binding to glycine binding sites essential for activation by NMDA receptor promoters e.g., 7-chloroquinurene acid
  • polyamines which are
  • the present invention provides a composition for the prevention or treatment of obsessive-compulsive disorder comprising memantine (Memantine) or a pharmaceutically acceptable salt thereof as an NMDA receptor antagonist.
  • memantine memantine
  • a pharmaceutically acceptable salt thereof as an NMDA receptor antagonist.
  • Memantine (3,5-dimethyladamantan-1-amine) is one of the NMDA receptor antagonists that inhibits the excessive secretion of glutamate, a chemical in the brain. Glutamic acid plays a role in memory and learning when the amount is normal, but if the amount is excessive, it stimulates nerve cells to induce nerve cell destruction, which is related to stroke, epilepsy and dementia. It turned out. Memantine is one of the most commonly used treatments for Alzheimer's disease (dementia), but it is not known that it can be used to treat OCD.
  • composition of the present invention may further comprise components such as existing therapeutically active ingredients, other adjuvants, pharmaceutically acceptable carriers and the like.
  • pharmaceutically acceptable carriers include saline, sterile water, Ringer's solution, buffered saline, dextrose solution, maltodextrin solution, glycerol, ethanol and the like.
  • the present invention also provides a method of preventing or treating OCD by administering to a subject a pharmaceutical composition containing a pharmaceutically effective amount of an NMDA receptor antagonist.
  • “individual” means a subject in need of treatment for a disease, and more specifically, human or non-human primates, mice, rats, dogs, cats, horses and cattle, etc. Mean mammal.
  • “pharmaceutically effective amount” means the type and severity of the disease to be administered, the age and sex of the patient, the sensitivity to the drug, the time of administration, the route of administration and the rate of administration, the duration of treatment, the factors including the concurrent drug and other medicines It is determined according to factors well known in the art and can be easily determined by those skilled in the art in such an amount that the maximum effect can be obtained without side effects in consideration of all the above factors.
  • composition of the present invention is not limited in the method of administration as long as it can reach the target tissue.
  • examples include oral administration, arterial injection, intravenous injection, transdermal injection, intranasal administration, coronary administration or intramuscular administration.
  • the daily dosage is about 0.0001 to 100 mg / kg, preferably 0.001 to 10 mg / kg, preferably administered once to several times a day.
  • the Ninjurin1 KO mouse (C57BL / 6) used in the experiment was distributed at Ewha Womens University and kept aseptically in the animal laboratory at the College of Pharmacy, Seoul National University. All experiments were performed with the approval of the Seoul National University Laboratory Animal Management Committee (SNU-090316-). 9).
  • mice of 3-5 months of age were placed in cages (25 x 20 x 20 cm) arranged regularly (5 x 4) of 20 marbles (size: 15 mm), and mouse behavior patterns were recorded as moving images. After 30 minutes the number of hidden marbles was analyzed and considered to be meaningful at least 2/3 hidden (FIGS. 2A and 2C). ****: p ⁇ 0.0001; Comparison of Ninjurin1 KO mice to Ninjurin1 WT (FIG. 2B) or Hetero (FIG. 2D) mice, unpaired student's t-test.
  • Example 2 The marble burying test of Example 2 confirmed that anxiety was increased in Ninjurin1 KO mice. To further confirm this, an elevated-plus-maze test was performed as follows to measure the degree of anxiety.
  • FIGS. 3B and 3C The results are shown in FIGS. 3B and 3C, where the time of staying in each arm (FIG. 3B) and the number of times of entry (FIG. 3C) are indicative of the degree of anxiety.
  • Wild-type mice were placed in cages 3 days before the experiment and mice to be tested were placed 1 hour before.
  • Three chambers were constructed as chambers with three transparent and removable chambers. The overall dimensions consist of 60 cm x 45 cm x 25 cm, with each chamber consisting of 20 cm x 45 cm x 25 cm. Each chamber is connected by one hole (5cm x 5cm) for easy opening and closing.
  • the cage containing the Stranger mouse is made of steel cylindrical (height; 12cm, 9cm in diameter). In order to prevent the mice under test from rising above the cage, a transparent glass of water was put together (FIG. 4A).
  • mice were allowed to move freely in three chambers for 5 minutes and then in contact with stranger 1 mice for 5 minutes.
  • the new mouse (stranger 2) was placed in an empty cage and recorded as a video for 5 minutes.
  • the time spent in each chamber (FIGS. 4C and 4F), the time of direct contact with the mice (FIGS. 4D and 4G), and the heat maps (FIGS. 4B and 4E) were analyzed using the Noldus Ethovision XT8.5 program.
  • the cortex and striatum of Ninjurin1 KO mice via Golgi staining are as follows. Anatomical analysis of the structure was performed.
  • ninjurin1 influences the neural cell network formation
  • Western blotting was used to investigate the expression of ninjurin1 in the developing brain.
  • Example 6-1 sample preparation
  • Example 6-2 Western blotting
  • each primary antibody was incubated overnight at 4 °C. The next day, HRP-labeled anti-mouse IgG was treated as a secondary antibody at room temperature for 1 hour, washed with PBST (PBS, 1% Tween-20), and then detected using ECL solution (Intron, Westzol).
  • the primary antibodies used were PSD-95 (Millipore, mouse) and Synaptophysin (Millipore, mouse). Both antibodies were used as markers of synaptogenesis.
  • ⁇ -actin serum, rabbit
  • ⁇ -tubulin sigma, mouse
  • mice To investigate intracellular expression of ninjurin1, synaptosomes of mice were isolated as follows.
  • the brains of 8-week-old female mice were extracted and ground with scissors and then ground in cold HEPES-Sucrose buffer (Hom, homogenate).
  • the pellet was centrifuged (1000 g, 5 min) to obtain a pellet (P1, nuclear fraction), and the supernatant (S1, synaptosome) was centrifuged again (10000 g, 15 min) to pellet (P2, crude synaptosomal fraction) and supernatant ( S2, light membrane & cytosolic fraction).
  • P2 was then resuspended in cold HEPES buffer, and then centrifuged again (10000 g, 15 min) to obtain pellet (P2 ', washed synaptosomal fraction).
  • GFP-Ninj1 and Flag-mNinj1 were transfected (Lipofectamine 2000, Invitrogen) into primary cortical neurons DIV4 and DIV8 cultured for 4 days and 8 days, respectively. After 24 hours, fixed and stored using PFA (4%, 10 min) (PBS, 4 ° C). After washing twice with cold PBS for immunocytochemical analysis, the reaction of PFA was inhibited with 20 mM glycine / PBS. After permeabilization with 1% Triton X-100, nonspecific binding was inhibited via blocking solution (1% BSA (w / v), 5% FBS (v / v)).
  • Ninjurin1 is mainly present in the synaptosome fractions (P2, P2 ', P3), but not in the synaptic vesicle fractions (S3, S3') (Fig. 7a), and synapses in primary cultured cortical neurons. It was confirmed that it was present in the tosome (FIG. 7B). In addition, as a result of immunocytochemical analysis, it was confirmed that exogenously overexpressed Ninjurin1 is expressed in puncta-like structures (FIG. 7C).
  • ninjurin1 is expressed in the synaptosome region with puncta structure
  • the distribution of Ninjurin1 expression in synapses was examined in detail through several synaptic markers and dual immunostaining.
  • Primary antibodies used for dual immunostaining are as follows.
  • Pre-synapse markers Synaptophysin (Millipore, mouse), vGlut1 (Abcam, sheep),
  • Post-synapse marker Homer1 (Synaptic systems, mouse)
  • Brain tissue and lysate were prepared from wild-type (WT) and Ninjurin1 KO mice (3 months old male), and the expression levels of Fos B (Santa cruz) were compared by Western blotting (40ug, 12% gel). .
  • mice were perfused with PBS and fixed at 4 ° C. overnight with 4% PFA. After dehydration using 10%, 20% and 30% sucrose, OCT blocks were made.
  • the microtomes (HM 525, Thermo Scientific) were cut to 20 ⁇ m and placed in free-floating solution and stored at ⁇ 70 ° C. until use.
  • the cut brain tissue was washed with PBS (5 min, 3 times) and then blocked with blocking solution (0.03% Triton X-100, 10% BSA, 5% FBS) and Fos B antibody (Santa Cruz) overnight at 4 ° C. Was treated to tissue. The following day, after washing three times with PBS for 10 minutes, the secondary antibody was treated for 1 hour at room temperature. Nuclei were stained with Hechst33442 (Molecular probe, Invitrogen) and observed under confocal microscopy.
  • FIG. 9A Immunohistochemical staining of FIG. 9A and Western blot of FIG. 9B confirmed that the expression of Fos B was increased in the brain of Ninjurin1 KO mouse. Therefore, it can be seen that the neuronal activity is enhanced in Ninjurin1 KO mice.
  • Example 11-1 Barbering vs. skin damage
  • Memantine hydrochloride (sigma, 5 mg / kg) was intraperitoneally injected daily for 10 days in Barbering and skin injury Ninjurin1 KO mice (5 months old). Two weeks later, Barbering and the degree of skin damage were compared with before injection (FIG. 11A).

Abstract

The present invention relates to an animal model with obsessive compulsive disorder, manufactured by deficiency of Ninjurin 1, and a method for searching candidate drugs for preventing or treating obsessive compulsive disorder using the same. According to the present invention, a Ninjurin 1 KO mouse can be provided as a new model for researching obsessive compulsive disorder, and memantine or a salt thereof, which is an NMDA receptor antagonist, can be provided as a new use for treating obsessive compulsive disorder

Description

Ninjurin1 결핍을 이용한 강박증 동물모델OCD Animal Model Using Ninurin1 Deficiency
본 발명은 닌쥬린 1(Ninjurin 1)을 결핍시켜 제조된 강박증 동물모델, 및 이를 이용하여 강박증을 예방 또는 치료하기 위한 후보약물을 탐색하는 방법에 관한 것이다.The present invention relates to an obsessive-compulsive animal model prepared by deficiency of ninjurin 1, and a method for searching for a candidate drug for preventing or treating OCD.
Ninjurin은 1996년 Toshiyuki Araki 등에 의해 처음으로 보고된 것으로, 좌골 신경(sciatic nerve)의 원위부를 절단(transection)하거나 압착(crush)하여 손상을 일으킨 후 신경 섬유초 세포(Schwann cell)에서 발현이 증가하는 유전자(gene)를 찾는 과정에서 발견되었다.Ninjurin was first reported in 1996 by Toshiyuki Araki et al., Whose expression is increased in Schwann cells after injury by transection or crushing of the distal sciatic nerve. It was discovered in the process of finding a gene.
GenBank에 알려져 있는 단백질 정보를 이용하여 Ninjurin과 상동성(homology)을 가지는 단백질을 찾아 계통수(phylogenetic tree)가 보고되었다. 척추동물에서는 Ninjurin 1과 Ninjurin 2라는 두 가지의 단백질이 알려져 있다. 상기 Ninjurin 1과 Ninjurin 2는 인간, 마우스 및 랫트 등의 척추 동물에서 발견된 바 있다. Phylogenetic trees have been reported using proteins known in GenBank to find proteins that have homology with Ninjurin. In vertebrates, two proteins are known, Ninjurin 1 and Ninjurin 2. Ninjurin 1 and Ninjurin 2 have been found in vertebrates such as humans, mice, and rats.
인간 Ninjruin 1은 염색체 9q22에 위치하고 있으며 152개의 아미노산으로 구성되어 있다. 마우스 Ninjurin 1의 경우는 염색체 13에 위치하고 있으며 152개의 아미노산으로 구성되어 있다. Ninjurin 1의 아미노산 서열에서 2곳의 막 통과 영역(transmembrane domain)을 예측할 수 있으며, 실험을 통하여 Ninjrurin 1이 세포막에 위치한 단백질임이 알려져 있다. Human Ninjruin 1 is located on chromosome 9q22 and consists of 152 amino acids. Mouse Ninjurin 1 is located on chromosome 13 and consists of 152 amino acids. Two transmembrane domains can be predicted from the amino acid sequence of ninjurin 1. Through experiments, it is known that Ninjrurin 1 is a protein located in the cell membrane.
Ninjurin 1은 다양한 조직에서 발현되고 있다. 예를 들어, RNA 단계에서는 심장(heart), 뇌(brain), 태반(placenta), 폐(lung), 간(liver), 골격근(sk.Muscle), 콩팥(kidney), 이자(pancreas), 비장(spleen), 흉선(thymus), 전립선(prostate), 고환(testis), 난소(ovary), 소장(sm.int.), 대장(colon), 혈액(blood), 부신(adrenal gland) 및 후근절(Dorsal Root Ganglia, DRG)에서 발현되고, 단백질 단계에서는 간, 콩팥, 흉선, 자궁(uterus), 부신, 망막(retina) 및 후근절에서 발현되는 것이 보고된 바 있다.Ninjurin 1 is expressed in various tissues. For example, in the RNA stage, the heart, brain, placenta, lung, liver, skeletal muscle (sk.Muscle), kidneys, pancreas, spleen (spleen), thymus (prostate), prostate (testis), ovary (ovary), small intestine (sm.int.), large intestine (colon), blood (adrenal gland) and resection (Dorsal Root Ganglia, DRG), and protein levels have been reported in the liver, kidneys, thymus, uterus, adrenal gland, retina and dorsal root.
또한, 지금까지 알려진 Ninjurin 1의 기능으로는 세포 결합(cell adhesion), 신경 증식(neurite outgrowth), 세포 노화(cellular senescence) 및 암과 관련성 이 있다.In addition, the functions of Ninjurin 1 so far known are related to cell adhesion, nerve outgrowth, cellular senescence and cancer.
한편, 강박증(Obsessive-Compulsive Spectrum Disorder; OCD)은 비자발적이고 지속적으로 불안감(anxiety)을 느끼는 질병으로, 이를 해소하기 위해 충동적이고 반복적인 행동이 그 주요 현상으로 나타나게 된다. 강박증은 환자가 병적인 것으로 인식하거나 내면에서 강하게 저항하고 있는 반복적인 생각, 환상(강박) 및 반복적인 충동 또는 행동(강제)의 존재를 특징으로 하는 신경증이다. Obsessive-Compulsive Spectrum Disorder (OCD), on the other hand, is an involuntary and persistent anxiety disease, and impulsive and repetitive behaviors are the main phenomena to solve this problem. OCD is a neurosis characterized by the presence of repetitive thoughts, illusions (obsessive) and repetitive urges or actions (forces) that the patient perceives as pathological or is strongly resistant within.
과거 강박장애의 원인은 심리학적 요인에 근거하여 설명하려는 노력이 많았으나 최근의 약물 연구 및 뇌 영상 연구의 결과들은 생물학적 요인이 강박장애 발생과 연관성이 깊음을 보여주고 있다. 대표적인 예를 들면, 뇌의 대표적인 신경 전달물질인 세로토닌 시스템과의 연관성이다. 임상 약물 실험 상 세로토닌 시스템에 작용하는 약물들이 강박장애 치료에 뚜렷한 효과가 있고, 다른 임상 연구 결과도 세로토닌 및 세로토닌 연관 물질과 강박장애 간의 연관성이 있음을 보여주고 있다. 또한 많은 뇌 영상 연구가 강박장애에서 특정 신경회로 영역에 문제가 있음을 보여주고 있고, 약물치료 내지는 행동요법치료 후 이러한 영역의 문제가 정상화 됨을 보여주는 연구 결과도 있다.In the past, there have been many efforts to explain the causes of OCD based on psychological factors, but the results of recent drug studies and brain imaging studies show that biological factors are closely related to OCD. A representative example is the association with the serotonin system, a representative neurotransmitter of the brain. In clinical trials, drugs acting on the serotonin system have a pronounced effect on the treatment of OCD, and other clinical studies show that there is an association between serotonin and serotonin-related substances and OCD. In addition, many brain imaging studies have shown that certain neurocirculatory areas have problems in obsessive-compulsive disorder, and studies show that these areas are normalized after medication or behavioral therapy.
최근들어 강박증, 우울증, 자폐증 등의 각종 정신질환들에 대한 연구와 치료제 개발 요구가 증가하고 있지만, 아직까지 구체적인 연구모델과 방법이 정립되어 있지 않을 뿐 아니라 이들 정신질환을 일으키는 분자적 기전도 잘 알려져 있지 않다.Recently, there is an increasing demand for research and treatment of various mental disorders such as OCD, depression, and autism, but there are no specific research models and methods yet, and the molecular mechanisms causing these mental diseases are well known. Not.
이에, 본 발명자는 강박증과 Ninjurin 1과의 관련성을 최초로 밝혀, 강박증 연구를 위한 새로운 모델로서의 Ninjurin 1 KO 마우스의 이용 가능성과, 강박증 치료를 위한 타깃 단백질을 제시하고자 한다.Accordingly, the present inventors first disclosed the relationship between OCD and Ninjurin 1, and suggests the possibility of using Ninjurin 1 KO mice as a new model for OCD studies and a target protein for treating OCD.
구체적으로, 본 발명은 닌쥬린 1(Ninjurin 1)을 결핍시켜 제조된 강박증 동물모델을 제공하는 것을 목적으로 한다.Specifically, an object of the present invention is to provide an obsessive-compulsive animal model prepared by deficiency of ninjurin 1 (Ninjurin 1).
또한, 본 발명은 닌쥬린 1(Ninjurin 1)을 결핍시켜 제조된 강박증 동물모델을 이용하여 강박증을 예방 또는 치료하기 위한 후보약물을 탐색하는 방법을 제공하는 것을 목적으로 한다.In addition, an object of the present invention is to provide a method for searching for a candidate drug for preventing or treating OCD by using an OCD animal model prepared by deficient in ninjurin 1 (Ninjurin 1).
그러나, 본 발명이 이루고자 하는 기술적 과제는 이상에서 언급한 과제에 제한되지 않으며, 언급되지 않은 또 다른 과제들은 아래의 기재로부터 당업자에게 명확하게 이해될 수 있을 것이다.However, the technical problem to be achieved by the present invention is not limited to the above-mentioned problem, another task that is not mentioned will be clearly understood by those skilled in the art from the following description.
본 발명은, 닌쥬린 1(Ninjurin 1)을 결핍시켜 제조된 강박증 동물모델을 제공한다.The present invention provides an obsessive-compulsive animal model prepared by deficiency of ninjurin 1.
본 발명의 일 구현예로, 상기 동물은 인간을 제외한 것임을 특징으로 한다.In one embodiment of the invention, the animal is characterized in that except for humans.
본 발명의 다른 구현예로, 상기 닌쥬린 1 결핍은 닌쥬린 1 유전자의 넉아웃(Knock-out)에 의한 것임을 특징으로 한다.In another embodiment of the present invention, the ninjurin 1 deficiency is characterized by knock-out of the ninjurin 1 gene.
또한, 본 발명은, 닌쥬린 1(Ninjurin 1)을 결핍시켜 제조된 강박증 동물모델을 이용하여 강박증을 예방 또는 치료하기 위한 후보약물을 탐색하는 방법을 제공한다.In addition, the present invention provides a method for searching for a candidate drug for preventing or treating OCD by using an OCD animal model prepared by deficient in ninjurin 1 (Ninjurin 1).
본 발명의 일 구현예로, 상기 탐색방법은 동물모델에 후보약물을 투여한 후 NMDA 수용체(N-Methyl-D-aspartate receptor)가 저해되는지를 측정하는 단계를 포함하는 것을 특징으로 한다.In one embodiment of the present invention, the detection method is characterized in that it comprises the step of determining whether the N-Methyl-D-aspartate receptor (NMDA receptor) is inhibited after administration of the candidate drug to the animal model.
본 발명의 다른 구현예로, 상기 후보약물은 NMDA 수용체 길항제인 것을 특징으로 한다.In another embodiment of the present invention, the candidate drug is characterized in that the NMDA receptor antagonist.
본 발명의 또 다른 구현예로, 상기 NMDA 수용체 길항제는 메만틴(Memantine)인 것을 특징으로 한다.In another embodiment of the invention, the NMDA receptor antagonist is characterized in that the memantine (Memantine).
본 발명에 의하면, 강박증 연구를 위한 새로운 모델로서의 Ninjurin 1 KO 마우스를 제공할 수 있다.According to the present invention, a Ninjurin 1 KO mouse can be provided as a new model for OCD.
또한, 본 발명에 의하면 강박증 치료를 위한 신규 타깃 단백질을 제공할 수 있다.In addition, the present invention can provide a novel target protein for the treatment of OCD.
또한, 본 발명에 의하면 강박증 치료제 후보약물을 탐색하는 방법을 제공할 수 있다.In addition, the present invention can provide a method for searching for a drug candidate for OCD.
또한, 본 발명에 의하면 강박증 치료제 후보약물을 제공할 수 있다.In addition, the present invention can provide a drug candidate for the treatment of OCD.
또한, 본 발명에 의하면 NMDA 수용체 길항제의 신규용도로서 강박증 치료용도를 제공할 수 있다.In addition, the present invention can provide a treatment for OCD as a novel use of NMDA receptor antagonists.
도 1은, Ninjurin1 KO 마우스의 강박증 유사행동인 자해(Self-made injury)를 관찰한 결과이다.1 is a result of observing self-made injury, which is an OCD-like behavior of Ninjurin1 KO mice.
도 2는, Ninjurin1 KO 마우스의 강박증 유사행동인 불안감(anxiety) 및 반복행동을 Marble-burying 테스트를 통해 확인한 결과이다.FIG. 2 shows the results of anxiety and repetitive behaviors, which are compulsive disorders of Ninjurin1 KO mice, through marble-burying test.
도 3은, Ninjurin1 KO 마우스의 불안감(anxiety) 정도를 Elevated-plus-maze를 이용하여 확인한 결과이다.3 is a result of confirming the degree of anxiety of the Ninjurin1 KO mouse using Elevated-plus-maze.
도 4는, Ninjurin1 KO 마우스와 자폐증(autism)과의 연관성을 Three-chamber 테스트를 이용하여 확인한 결과이다.4 shows the results of confirming the association between Ninjurin1 KO mice and autism using the Three-chamber test.
도 5는, Ninjurin1 KO 마우스의 Cortical-striatum 회로 형성 장애를 골지 염색법을 이용하여 확인한 결과이다.5 is a result of confirming the cortical-striatum circuit formation disorder of Ninjurin1 KO mouse using Golgi staining method.
도 6은, 뇌 발생 과정 중 Ninjurin1의 발현 패턴 변화를 웨스턴블랏팅을 이용하여 확인한 결과이다.6 is a result of confirming the change in the expression pattern of Ninjurin1 during brain development using Western blotting.
도 7은, Puncta 구조의 시냅토솜(synaptosome)에서 Ninjurin1의 발현을 면역세포화학 분석법을 이용하여 확인한 결과이다.Figure 7 shows the results of confirming the expression of Ninjurin1 in synaptosomes of Puncta structure using immunocytochemical analysis.
도 8은, Pre-synapse와 post-synapse에서 Ninjurin1의 발현을 이중 면역염색법으로 확인한 결과이다.8 shows the results of confirming the expression of Ninjurin1 in pre-synapse and post-synapse by double immunostaining.
도 9는, Ninjurin1 KO 마우스의 Neural 활동성 증가를 Fos B의 발현정도를 비교함으로써 확인한 결과이다.9 is a result confirmed by comparing the expression of Fos B to increase the Neural activity of Ninjurin1 KO mice.
도 10은, Ninjurin 1 KO 마우스에서 NMDAR의 발현과 Src의 발현 증가를 웨스턴블랏팅을 이용하여 확인한 결과이다.Figure 10 shows the results of confirming the expression of NMDAR and Src expression increase in Ninjurin 1 KO mice using Western blotting.
도 11은, 메만틴의 Ninjurin1 KO 마우스 강박증 저해효과를 Marble-burying 테스트를 통해 확인한 결과이다.11 is a result confirming the inhibitory effect of memantine Ninjurin1 KO mouse OCD through the Marble-burying test.
본 발명자들은, 강박증과 Ninjurin 1이 서로 관련되어 있을 것으로 예상하고 연구를 진행한 결과, Ninjurin 1을 제거한 마우스(Ninjurin 1 KO 마우스)에서 self-made injury, 반복적인 행동, 불안감 증가와 같은 강박증 유사행동을 관찰하였으며, 해부학적 분석을 통해 cortical-stiatal 회로의 형성에 이상이 있음을 확인하였다. The present inventors have predicted that OCD and Ninjurin 1 may be related to each other, and as a result of research, OCD-like behaviors such as self-made injury, repetitive behavior, and increased anxiety in Ninjurin 1-removed mice (Ninjurin 1 KO mice) And anatomical analysis confirmed abnormalities in the formation of cortical-stiatal circuits.
나아가, Ninjurin 1 결핍 마우스에서 NMDA 수용체의 발현과 Src의 활성이 증가됨을 확인하고, 메만틴(Memantine)을 처리하여 NMDA 수용체의 활성을 저해하였을 때 Ninjurin1 KO 마우스의 강박증 유사행동이 회복됨을 밝혔다. 즉, 강박증의 분자적 기전으로서, Ninjurin 1이 결핍되면 cortical-striatal 회로 형성에 이상이 생겨 NMDA 수용체 활성을 유도하고, 그 결과 강박증 유사행동이 일어남을 최초로 규명하였다. Furthermore, NMDA receptor expression and Src activity were increased in Ninjurin 1 deficient mice, and the treatment of obsessive compulsive behavior of Ninjurin1 KO mice was restored when Memantine treatment inhibited NMDA receptor activity. In other words, as a molecular mechanism of OCD, deficiency of Ninjurin 1 caused abnormalities in cortical-striatal circuit formation and induced NMDA receptor activity. As a result, it was first identified that OCD-like behavior occurred.
이와 동시에, 이러한 기작에 의해 유발되는 강박증을 치료할 수 있는 후보 약물을 탐색하는 방법과, 그 결과 얻어진 후보약물을 제시하고자 한다.At the same time, we propose a method of searching for candidate drugs that can treat OCD caused by this mechanism, and the resulting candidate drugs.
NMDA 수용체는, 세포의 사멸과 정상세포 간의 신호전달을 조절하는 것으로 알려진 신경수용체이다. 도파민 D1 수용체와 직접적인 상호작용을 하여 세포의 사멸을 조절하거나 정상적인 세포 간의 통신을 유도한다. NMDA 수용체가 D1 수용체와 상호작용할 때는 세포의 사멸과 세포 사이의 신호전달 조절기능이 서로 다른 기작을 통해 독립적으로 진행된다. 종래, 뇌졸중, 골다공증, 간질, 치매 등 각종 질환을 치료하기 위한 일환으로 NMDA 수용체에 대한 다양한 연구가 이루어져 왔다. NMDA receptors are neuroreceptors known to regulate cell death and signaling between normal cells. Direct interaction with the dopamine D1 receptor regulates cell death or induces communication between normal cells. When the NMDA receptor interacts with the D1 receptor, cell death and signaling regulation between cells are independently driven by different mechanisms. Conventionally, various studies on NMDA receptors have been made as part of treating various diseases such as stroke, osteoporosis, epilepsy and dementia.
NMDA 수용체 길항제는 NMDA 수용체의 기능을 저해하는 물질로서, (1) 글루탐산이나 NMDA 등의 촉진제와의 결합부위에 길항적으로 결합하는 것(예: D-2-아미노-5-포스포노길초산), (2) NMDA 수용체의 촉진제에 의한 활성화에 필수적인 글리신의 결합부위에 길항적으로 결합하는것(예: 7-클로로퀴누렌산), (3) 활성증강제인 폴리아민의 결합부위에 길항적으로 결합하는 것(예: 알카인), (4) 이온채널부위에 결합하여 이온의 흐름을 저지하는 것(예: MK-801, Mg2+) 등이 알려져 있다.NMDA receptor antagonists are substances that inhibit the function of the NMDA receptor, and (1) antagonistically bind to binding sites with promoters such as glutamic acid or NMDA (e.g., D-2-amino-5-phosphonoyl acetate) (2) antagonically binding to glycine binding sites essential for activation by NMDA receptor promoters (e.g., 7-chloroquinurene acid), (3) antagonistically binding to binding sites of polyamines, which are active enhancers (Eg, alkane), (4) binding to ion channel sites to block the flow of ions (eg MK-801, Mg2 +) is known.
이에, 본 발명은 NMDA 수용체 길항제로서 메만틴(Memantine) 또는 이의 약학적으로 허용가능한 염을 포함하는 강박증의 예방 또는 치료용 조성물을 제공한다.Accordingly, the present invention provides a composition for the prevention or treatment of obsessive-compulsive disorder comprising memantine (Memantine) or a pharmaceutically acceptable salt thereof as an NMDA receptor antagonist.
메만틴(3,5-dimethyladamantan-1-amine)은 NMDA 수용체 길항제 중 하나로서, 뇌 속의 화학물질인 글루타민산염의 과잉분비를 억제한다. 글루타민산은 그 양이 정상일때에는 기억이나 학습을 돕는 역할을 하지만, 만약 그 양이 과도할 경우에는 신경세포를 과자극하여 신경세포 파괴를 유도하기 때문에, 뇌졸중, 간질, 치매 등의 질환과의 관련성이 밝혀진 바 있다. 메만틴은 현재 가장 많이 사용되는 알츠하이머병(치매) 치료제 중 하나이나, 강박증 치료용도로 사용될 수 있음은 알려진 바 없다.Memantine (3,5-dimethyladamantan-1-amine) is one of the NMDA receptor antagonists that inhibits the excessive secretion of glutamate, a chemical in the brain. Glutamic acid plays a role in memory and learning when the amount is normal, but if the amount is excessive, it stimulates nerve cells to induce nerve cell destruction, which is related to stroke, epilepsy and dementia. It turned out. Memantine is one of the most commonly used treatments for Alzheimer's disease (dementia), but it is not known that it can be used to treat OCD.
상기 본 발명의 조성물은 기존 치료 활성 성분, 기타 보조제, 약제학적으로 허용가능한 담체 등의 성분을 추가로 포함할 수 있다. 상기 약제학적으로 허용가능한 담체는 식염수, 멸균수, 링거액, 완충 식염수, 덱스트로스 용액, 말토 덱스트린 용액, 글리세롤, 및 에탄올 등을 포함한다.The composition of the present invention may further comprise components such as existing therapeutically active ingredients, other adjuvants, pharmaceutically acceptable carriers and the like. Such pharmaceutically acceptable carriers include saline, sterile water, Ringer's solution, buffered saline, dextrose solution, maltodextrin solution, glycerol, ethanol and the like.
또한, 본 발명은 약학적으로 유효량의 NMDA 수용체 길항제를 함유하는 약학적 조성물을 개체에 투여하여 강박증을 예방 또는 치료하는 방법을 제공한다.The present invention also provides a method of preventing or treating OCD by administering to a subject a pharmaceutical composition containing a pharmaceutically effective amount of an NMDA receptor antagonist.
본 발명에서 "개체"란 질병의 치료를 필요로 하는 대상을 의미하고, 보다 구체적으로는 인간 또는 비-인간인 영장류, 생쥐 (mouse), 쥐 (rat), 개, 고양이, 말 및 소 등의 포유류를 의미한다. 또한 본 발명에서 "약제학적 유효량"은 투여되는 질환 종류 및 중증도, 환자의 연령 및 성별, 약물에 대한 민감도, 투여 시간, 투여 경로 및 배출 비율, 치료 기간, 동시 사용되는 약물을 포함한 요소 및 기타 의학 분야에 잘 알려진 요소에 따라 결정되며 상기 요소를 모두 고려하여 부작용 없이 최대 효과를 얻을 수 있는 양으로, 당업자에 의해 용이하게 결정될 수 있다.As used herein, "individual" means a subject in need of treatment for a disease, and more specifically, human or non-human primates, mice, rats, dogs, cats, horses and cattle, etc. Mean mammal. Also, in the present invention, "pharmaceutically effective amount" means the type and severity of the disease to be administered, the age and sex of the patient, the sensitivity to the drug, the time of administration, the route of administration and the rate of administration, the duration of treatment, the factors including the concurrent drug and other medicines It is determined according to factors well known in the art and can be easily determined by those skilled in the art in such an amount that the maximum effect can be obtained without side effects in consideration of all the above factors.
본 발명의 조성물은 목적 조직에 도달할 수 있는 한 투여방법에는 제한이 없다. 예를 들면, 경구 투여, 동맥 주사, 정맥 주사, 경피 주사, 비강 내 투여, 경기관지 투여 또는 근육 내 투여 등이 포함된다. 일일 투여량은 약 0.0001 내지 100mg/kg이고, 바람직하게는 0.001 내지 10mg/kg이며, 하루 일회 내지 수회 나누어 투여하는 것이 바람직하다.The composition of the present invention is not limited in the method of administration as long as it can reach the target tissue. Examples include oral administration, arterial injection, intravenous injection, transdermal injection, intranasal administration, coronary administration or intramuscular administration. The daily dosage is about 0.0001 to 100 mg / kg, preferably 0.001 to 10 mg / kg, preferably administered once to several times a day.
이하, 본 발명의 이해를 돕기 위하여 바람직한 실시예를 제시한다. 그러나 하기의 실시예는 본 발명을 보다 쉽게 이해하기 위하여 제공되는 것일 뿐, 하기 실시예에 의해 본 발명의 내용이 한정되는 것은 아니다.Hereinafter, preferred examples are provided to aid in understanding the present invention. However, the following examples are merely provided to more easily understand the present invention, and the contents of the present invention are not limited by the following examples.
실시예 1. Ninjurin1 KO 마우스의 강박증 유사 행동 : Self-made injuryExample 1 OCD-like Behavior of Ninjurin1 KO Mice: Self-made Injury
야생형(wt)과 Ninjurin1 KO 마우스(5-7개월령)의 외형적 차이를 관찰하고 행동패턴의 변화를 비교하였다. 실험에 사용한 Ninjurin1 KO 마우스(C57BL/6)는 이화여대에서 분양받아 서울대학교 약학대학 동물실의 무균상태로 유지하였으며, 모든 실험은 서울대학교 실험동물 관리 위원회의 승인을 얻어 수행되었다(SNU-090316-9). The differences between the wild-type (wt) and Ninjurin1 KO mice (5-7 months old) were observed and the behavioral patterns were compared. The Ninjurin1 KO mouse (C57BL / 6) used in the experiment was distributed at Ewha Womans University and kept aseptically in the animal laboratory at the College of Pharmacy, Seoul National University. All experiments were performed with the approval of the Seoul National University Laboratory Animal Management Committee (SNU-090316-). 9).
그 결과, 야생형과는 달리 Ninjurin1 KO 마우스에서 barbering(도 1a), self-made injury(도 1b), eye-scratching(도 1c)와 같은 손상이 관찰되었으며, 그 빈도는 barbering(47, 83%), self-made injury(7.25%), eye-scratching (12,32%)로 야생형에 비해 현저히 증가되어 있었다(도 1d). 이러한 표현형(phenotype)은 과도한 우울(glooming) 행동에 의한 유발되는 자해(Self-made injury)로 예상되며, 인간의 강박증(OCD)과 유사한 행동 패턴임을 확인하였다.As a result, unlike wild type, damages such as barbering (FIG. 1A), self-made injury (FIG. 1B), and eye-scratching (FIG. 1C) were observed in Ninjurin1 KO mice, and the frequency was barbering (47, 83%). , self-made injury (7.25%) and eye-scratching (12,32%) were significantly increased compared to wild type (FIG. 1D). This phenotype is expected to be a self-made injury caused by excessive glooming behavior, and it is confirmed that the phenotype is a behavioral pattern similar to that of human OCD.
실시예 2. Ninjurin1 KO 마우스의 강박증 유사행동 : Marble-burying 테스트Example 2 OCD-like Behavior of Ninjurin1 KO Mice: Marble-burying Test
Ninjurin1 KO 마우스(C57BL/6)와 OCD의 연관성을 확인하기 위해, 반복적인 행동(repetitive behavior), 불안감(anxiety-related behaviors)의 정도를 조사할 수 있는 marble-burying 테스트를 다음과 같이 수행하였다.To confirm the association between Ninjurin1 KO mice (C57BL / 6) and OCD, a marble-burying test was performed as follows to investigate the degree of repetitive behavior and anxiety-related behaviors.
3-5개월령의 마우스를 20개의 marble(크기 : 15mm)이 규칙적으로 배열(5 x 4) 되어 있는 케이지(25 x 20 x 20cm)에 넣은 후, 마우스 행동 패턴을 동영상으로 기록하였다. 30분 후 숨겨진 marble의 개수를 분석하고, 적어도 2/3 이상 숨겨진 것을 의미있는 것으로 간주하였다(도 2a 및 도 2c). **** : p<0.0001; Ninjurin1 WT (도 2b) 또는 Hetero(도 2d) 마우스에 대한 Ninjurin1 KO 마우스의 비교, unpaired student's t-test.Mice of 3-5 months of age were placed in cages (25 x 20 x 20 cm) arranged regularly (5 x 4) of 20 marbles (size: 15 mm), and mouse behavior patterns were recorded as moving images. After 30 minutes the number of hidden marbles was analyzed and considered to be meaningful at least 2/3 hidden (FIGS. 2A and 2C). ****: p <0.0001; Comparison of Ninjurin1 KO mice to Ninjurin1 WT (FIG. 2B) or Hetero (FIG. 2D) mice, unpaired student's t-test.
실험 결과, Ninjurin1 KO 마우스는 야생형(WT)과 Hetero 마우스에 비해 증가된 marble-burying 행동 패턴을 보였다. 따라서, Ninjurin1 KO 마우스는 정상 마우스보다 더 반복적인 행동을 가지고, 불안감을 많이 느낀다는 것을 확인할 수 있었다. 이처럼 Marble burying 테스트와 상기 실시예 1의 self-made injury 테스트를 함께 고려해 볼 때, Ninjurin1 KO 마우스는 OCD와 유사한 행동을 나타냄을 알 수 있다.As a result, Ninjurin1 KO mice showed an increased marble-burying behavior pattern compared to wild-type (WT) and Hetero mice. Therefore, it was confirmed that Ninjurin1 KO mice had more repetitive behaviors and felt more anxiety than normal mice. Considering the marble burying test and the self-made injury test of Example 1, it can be seen that the Ninjurin1 KO mouse exhibits a similar behavior to that of the OCD.
실시예 3. Ninjurin1 KO 마우스의 불안감(anxiety) 정도 비교 : Elevated-plus-maze 테스트Example 3. Comparison of Anxiety Degree in Ninjurin1 KO Mice: Elevated-plus-maze Test
상기 실시예 2의 marble burying 테스트를 통해 Ninjurin1 KO 마우스에서 불안감이 증가되어 있다는 것을 확인하였다. 이를 좀더 확인하기 위해, 불안감의 정도를 측정할 수 있는 elevated-plus-maze 테스트를 다음과 같이 수행하였다.The marble burying test of Example 2 confirmed that anxiety was increased in Ninjurin1 KO mice. To further confirm this, an elevated-plus-maze test was performed as follows to measure the degree of anxiety.
Ninjurin1 WT과 KO 마우스(2개월령)를 실험 1시간 전 이동시켜 환경에 적응시키고, 두 개의 open arm(25 x 5 x 0.5 cm)과 수직방향으로 두개의 close arm(25 x 5 x 16 cm)을 가진 50cm 높이의 미로(maze)를 제작하였다. Open arm은 마우스가 떨어지기 않도록 낮은 벽(0.5 cm)을 가지고, close arm는 높은 투명한 플라스틱 벽 (16cm)을 가지고 있다(도 3a). 테스트는 주위 소음이 없고 100ux 정도의 조명이 있는 방에서 이루어 졌으며, 8분 동안 미로내에서의 이동성을 동영상으로 기록하고, open arm과 close arm에 머무르고 있는 시간을 비교 분석하였다. Ninjurin1 WT and KO mice (2 months old) were moved one hour before the experiment to adapt to the environment, and two open arms (25 x 5 x 0.5 cm) and two close arms (25 x 5 x 16 cm) in the vertical direction. A maze with a height of 50 cm was produced. The open arm has a low wall (0.5 cm) so that the mouse does not fall off, and the close arm has a high transparent plastic wall (16 cm) (Figure 3a). The test was conducted in a room with 100 UX lighting without ambient noise, and recorded video of mobility within the maze for 8 minutes and compared the time spent in the open and close arms.
그 결과를 도 3b 및 도 3c에 나타내었으며, 각각의 arm에 머무는 시간(도 3b)과 들어가는 횟수(도 3c)는 불안감의 정도를 비교할 수 있는 지표가 된다. *: p<0.05; **:p<0.01 (WT과 KO의 비교) unpaired student's T-test, s.e.mThe results are shown in FIGS. 3B and 3C, where the time of staying in each arm (FIG. 3B) and the number of times of entry (FIG. 3C) are indicative of the degree of anxiety. *: p <0.05; **: p <0.01 (Comparison of WT and KO) unpaired student's T-test, s.e.m
실시예 4. Ninjurin1 KO 마우스의 자폐증(autism)과의 연관성: Three-chamber 테스트Example 4 Association with Autism in Ninjurin1 KO Mice: Three-chamber Test
Ninjurin1 KO 마우스의 자폐증과의 관련성을 조사하기 위해 자폐증의 주요 특징인 social communication의 정도를 다음과 같이 three-chamber 테스트를 통해 야행형과 비교하였다.To investigate the association of Ninjurin1 KO mice with autism, the degree of social communication, which is a major feature of autism, was compared with the nocturnal type through the three-chamber test as follows.
타겟 대상(stranger 1, stranger 2, 6-8 weeks) 야생형(WT) 마우스는 실험을 시작하기 3일전 케이지에 넣고, 테스트할 마우스는 1시간 전에 넣어 적응시켰다. Three chamber는 투명하고 제거 가능한 세 개의 방을 가진 챔버로 제작되었다. 전체 크기는 60 cm x 45 cm x 25 cm로 구성되어 각 챔버는 20 cm x 45 cm x 25 cm로 이루어져 있다. 각 챔버는 하나의 구멍(5cm x 5cm)으로 연결되어 쉽게 열고 닫을 수 있다. Stranger 마우스를 넣는 케이지는 steel cylindrical (높이; 12cm, 밑지름 9cm)로 제작되었다. 테스트 중인 마우스가 케이지 위로 올라가는 것을 막기 위해 투명한 물컵을 함께 넣어 주었다(도 4a). Target subjects (stranger 1, stranger 2, 6-8 weeks) Wild-type (WT) mice were placed in cages 3 days before the experiment and mice to be tested were placed 1 hour before. Three chambers were constructed as chambers with three transparent and removable chambers. The overall dimensions consist of 60 cm x 45 cm x 25 cm, with each chamber consisting of 20 cm x 45 cm x 25 cm. Each chamber is connected by one hole (5cm x 5cm) for easy opening and closing. The cage containing the Stranger mouse is made of steel cylindrical (height; 12cm, 9cm in diameter). In order to prevent the mice under test from rising above the cage, a transparent glass of water was put together (FIG. 4A).
마우스를 5분 동안 three chamber내에서 자유롭게 움직이게 한 후, 5분 동안 stranger 1 마우스와 접촉하게 하였다. 그리고 새로운 마우스(stranger 2)를 비어있는 케이지에 넣은 후, 5분 동안 동영상으로 기록하였다. 각 챔버에 머무는 시간(도 4c 및 도 4f), 마우스와 직접 접촉하고 있는 시간(도 4d 및 도 4g), heat maps(도 4b 및 4e)을 Noldus Ethovision XT8.5 프로그램을 이용하여 분석하였다. *: p<0.05; **: p<0.01; **** : p<0.0001 (WT과 Ninjurin1 KO 마우스의 비교), s.e.mMice were allowed to move freely in three chambers for 5 minutes and then in contact with stranger 1 mice for 5 minutes. The new mouse (stranger 2) was placed in an empty cage and recorded as a video for 5 minutes. The time spent in each chamber (FIGS. 4C and 4F), the time of direct contact with the mice (FIGS. 4D and 4G), and the heat maps (FIGS. 4B and 4E) were analyzed using the Noldus Ethovision XT8.5 program. *: p <0.05; **: p <0.01; ****: p <0.0001 (comparison of WT and Ninjurin1 KO mice), s.e.m
실험 결과, three chamber 테스트를 통해 Ninjurin1 KO 마우스의 social communication을 조사하였을 때, Ninjurin1 KO 마우스는 각 챔버에 머무는 시간과 새로운 마우스와 직접 접촉하는 시간에 있어서 WT 마우스와 유사한 패턴을 나타내는 것으로 보아, 정상적 social interaction을 나타내었다. 따라서 Ninjurin1 KO 마우스는 자폐증과는 다른 행동을 보인다는 것을 확인하였다.As a result of the three chamber test, the social communication of Ninjurin1 KO mice was found to be similar to that of WT mice in the time of staying in each chamber and in direct contact with new mice. The interaction is shown. Therefore, it was confirmed that Ninjurin1 KO mice exhibited a different behavior from autism.
실시예 5. Ninjurin1 KO 마우스의 Cotical-striatum 회로 형성 장애 : 골지염색법Example 5 Cotical-striatum Circuit Formation Disorders in Ninjurin1 KO Mice: Golgi Staining
뇌에서 피질선조(Cortico-striatal) 회로의 이상이 강박증의 발병과 연관되어 있는 것으로 알려져 있기 때문에, 다음과 같이 골지(Golgi) 염색 방법을 통해 Ninjurin1 KO 마우스의 대뇌피질(cortex), 선조체(striatum) 구조의 해부학적인 분석을 수행하였다.Since the abnormalities of the Cortico-striatal circuit in the brain are known to be associated with the development of OCD, the cortex and striatum of Ninjurin1 KO mice via Golgi staining are as follows. Anatomical analysis of the structure was performed.
FD Rapid GolgiStain Kit(Neurodigitech)를 이용한 standard Golgi-Cox impregnation 방법을 사용하여 Golgi 염색을 수행하였다. WT과 Ninjurin 1 KO 마우스(8주령 수컷) 뇌조직을 분리 후 고정하고, Gogi-Cox 용액을 25℃에서 18일동안 관류하였다. 마이크로톰(Microtome)(HM 525, Thermo Scientific)을 이용하여 피질선조 부분을 150um 두께로 절단하고 광학 현미경(Leica, CTR 5000)을 이용하여 관찰하였다. Scale bar=100um (도 5a 및 도 5c), 50um (도 5b).Golgi staining was performed using standard Golgi-Cox impregnation method using FD Rapid GolgiStain Kit (Neurodigitech). WT and Ninjurin 1 KO mice (8-week-old male) brain tissue were separated and fixed, and Gogi-Cox solution was perfused at 25 ° C. for 18 days. Using a microtome (HM 525, Thermo Scientific), the cortical filigree was cut to 150 um in thickness and observed using an optical microscope (Leica, CTR 5000). Scale bar = 100 um (FIGS. 5A and 5C), 50 um (FIG. 5B).
그 결과, Ninjurin1 KO 마우스에서 선조체(striatum)(도 5a), 중형 돌기 뉴런(Medium Spiny Neurons; MSN)(도 5b), 피질선조(cortical-striatum) 신경세포 (도 5c)에서, 수지상 분지 복합성(dendritic arbor complexity)과 수지상 분지(dendritic branching)의 수가 현저히 감소되어 있는 것을 관찰할 수 있었다. 따라서, Ninjurin1에 의해 신경세포의 수지 복합성이 유도되며, Ninjurin1이 없을 경우 피질선조 회로의 형성에 이상이 발생함을 확인하였다.As a result, in the striatum (FIG. 5A), Medium Spiny Neurons (MSN) (FIG. 5B), and cortical-striatum neurons (FIG. 5C) in Ninjurin1 KO mice, dendritic branching complex ( Significant decreases in dendritic arbor complexity and dendritic branching were observed. Therefore, it was confirmed that Ninjurin1 induced the resin complex of neurons, and in the absence of Ninjurin1, abnormality occurred in the formation of cortical line.
실시예 6. 뇌 발생 과정 중 Ninjurin1의 발현 패턴 변화Example 6 Expression Pattern Change of Ninjurin1 during Brain Development
Ninjurin1이 신경세포의 네트워크 형성에 영향을 미치기 때문에, 웨스턴블롯팅(Western blotting)을 통해 발생 중 뇌에서 Ninjurin1의 발현을 조사하였다.Since ninjurin1 influences the neural cell network formation, Western blotting was used to investigate the expression of ninjurin1 in the developing brain.
실시예 6-1. 샘플 준비Example 6-1. sample preparation
마우스 뇌의 각 부분에서 Ninjurin1의 발현을 조사하기 위해, 2주령(도 6a), 8주령(도 6b)의 숫컷을 PBS를 사용하여 관류(perfusion)한 후 뇌를 적출하였다. PBS를 통해 2번 세정한 후, olfactory bulb (OB), cerebral cortex (Ctx), cerebellum (CB), medulla oblongata (MO), spinal cord (SC)를 분리하였다. 분리된 각 부분을 용해 완충액을 사용하여 단백질을 추출하였다. 발생 단계별로 Ninurin1의 발현을 조사하기 위해, 2, 4, 6, 8, 10, 14일령 마우스의 뇌를 적출하였다(도 6c). 또한 마우스 피질신경세포(cortical neuron)를 1차 배양하여 분화 시기별 (3, 5, 7, 12, 16일)로 세포를 준비한 후 단백질을 추출하였다(도 6d).In order to investigate the expression of Ninjurin1 in each part of the mouse brain, males at 2 weeks (FIG. 6A) and 8 weeks (FIG. 6B) were perfused using PBS and brains were extracted. After washing twice with PBS, olfactory bulb (OB), cerebral cortex (Ctx), cerebellum (CB), medulla oblongata (MO), and spinal cord (SC) were isolated. Each separated portion was extracted protein using lysis buffer. In order to investigate the expression of Ninurin1 in each developmental stage, brains of 2, 4, 6, 8, 10 and 14 day old mice were extracted (FIG. 6C). In addition, the mouse cortical neurons (cortical neuron) was first cultured to prepare the cells by differentiation time (3, 5, 7, 12, 16 days) and then extracted the protein (Fig. 6d).
실시예 6-2. 웨스턴 블롯팅(Western blotting)Example 6-2. Western blotting
추출된 단백질을 SDS-PAGE를 통해 분리한 후, 각 1차 항체를 4℃에서 밤새 배양하였다. 다음날 2차 항체로서 HRP-표지 항-마우스 IgG를 실온에서 1시간 처리한 후, PBST(PBS, 1% Tween-20)로 세정한 후 ECL 용액(Intron, Westzol)을 이용하여 검출하였다. 이때 사용한 1차 항체는 PSD-95(Millipore, mouse)와 Synaptophysin (Millipore, mouse)이며, 두 항체 모두 신경연접형성(synaptogenesis) 표지인자로 사용되었다. 대조군으로서 β-actin (sigma, rabbit)과 α-tubulin (sigma, mouse)을 사용하였다.After the extracted protein was isolated via SDS-PAGE, each primary antibody was incubated overnight at 4 ℃. The next day, HRP-labeled anti-mouse IgG was treated as a secondary antibody at room temperature for 1 hour, washed with PBST (PBS, 1% Tween-20), and then detected using ECL solution (Intron, Westzol). The primary antibodies used were PSD-95 (Millipore, mouse) and Synaptophysin (Millipore, mouse). Both antibodies were used as markers of synaptogenesis. Β-actin (sigma, rabbit) and α-tubulin (sigma, mouse) were used as controls.
실험 결과, 도 6에 나타낸 바와 같이, 마우스 뇌에서 발생이 진행되면서 Ninjurin1의 발현이 증가하는 것을 확인하였다. 즉, Ninjurin1의 발현은 뇌의 각 부분에서 골고루 발현되며, 발생 단계 중 신경연접형성이 일어나는 시기와 유사하게 그 발현이 증가하는 것을 관찰하였다.As a result, as shown in Figure 6, as the development progresses in the mouse brain it was confirmed that the expression of Ninjurin1 increases. In other words, the expression of Ninjurin1 is evenly expressed in each part of the brain, and the expression of Ninjurin1 is increased similarly to the time of neural junction formation during developmental stage.
실시예 7. Puncta 구조의 synaptosome에서의 Ninjurin1 분포 확인Example 7 Identification of Ninjurin1 Distribution in Synaptosomes of Puncta Structure
Ninjurin1의 세포 내 발현을 조사하기 위해, 마우스의 시냅토솜(synaptosome)을 다음과 같이 분리하였다. To investigate intracellular expression of ninjurin1, synaptosomes of mice were isolated as follows.
실시예 7-1. 샘플 준비Example 7-1. sample preparation
8주령 암컷 마우스의 뇌를 적출하여 가위로 분쇄한 후, 차가운 HEPES-수크로스 완충액에서 분쇄하였다(Hom, homogenate). 분쇄물을 원심분리(1000g, 5min)하여 펠렛(P1, nuclear fraction)을 얻었고, 상층액(S1, synaptosome)을 다시 원심분리 (10000g, 15min)하여 펠렛(P2, crude synaptosomal fraction)과 상층액(S2, light membrane & cytosolic fraction)을 얻었다. 다음 P2를 차가운 HEPES 완충액으로 재현탁한 후, 다시 원심분리(10000g, 15min)하여 펠렛(P2', washed synaptosomal fraction)을 얻었다. P2 펠렛을 9배 부피의 차가운 증류수를 이용한 저-삼투성 쇼크(hypo-osmotic shock)로 용해한 후, 4mM HEPES을 넣고 다시 원심분리(21000g, 20min)하여 펠렛 (P3, lysed synaptosomal membrane fraction)을 얻었다. 이후, 상층액(S3, crude synaptic vesicle fraction)을 다시 원심분리(55000 rpm)하여 펠렛(S3', synaptic vesicle fraction)을 얻었다(도 7a).The brains of 8-week-old female mice were extracted and ground with scissors and then ground in cold HEPES-Sucrose buffer (Hom, homogenate). The pellet was centrifuged (1000 g, 5 min) to obtain a pellet (P1, nuclear fraction), and the supernatant (S1, synaptosome) was centrifuged again (10000 g, 15 min) to pellet (P2, crude synaptosomal fraction) and supernatant ( S2, light membrane & cytosolic fraction). P2 was then resuspended in cold HEPES buffer, and then centrifuged again (10000 g, 15 min) to obtain pellet (P2 ', washed synaptosomal fraction). After dissolving P2 pellet by hypo-osmotic shock using 9 times volume of cold distilled water, 4mM HEPES was added and centrifuged again (21000g, 20min) to obtain pellet (P3, lysed synaptosomal membrane fraction). . Then, the supernatant (S3, crude synaptic vesicle fraction) was centrifuged again (55000 rpm) to obtain pellets (S3 ', synaptic vesicle fraction) (Fig. 7a).
16일 동안 배양한 1차 cortical 신경세포(DIV 16)에서, Syn-PER 시냅스 단백질 추출 시약(Pierce, Thermo scientific)을 사용하여 핵 분획(nuclear fraction), 분쇄물(homogenate), 시냅토솜(synaptosome), 세포질 분획(cytosolic fraction)을 얻었다. 1차 cortical 신경세포(DIV 16)를 Syn-PER 완충액으로 용해한 후 원심분리(1000g, 10min)하여 핵 분획과 상층액을 얻고, 상층액을 다시 원심분리(10000g, 20min)하여 시냅토솜과 세포질 분획을 얻었다. 이들을 기존의 웨스턴 블롯팅 방법을 사용하여 단백질 분석하였다(도 7b). In primary cortical neurons (DIV 16) incubated for 16 days, using the Syn-PER synaptic protein extraction reagent (Pierce, Thermo scientific), the nuclear fraction, homogenate, synaptosome The cytosolic fraction was obtained. After dissolving primary cortical neurons (DIV 16) with Syn-PER buffer, centrifugation (1000g, 10min) to obtain the nuclear fraction and supernatant, and the supernatant again centrifuged (10000g, 20min) to synaptosomes and cytoplasmic fraction Got. These were analyzed for proteins using conventional western blotting methods (FIG. 7B).
실시예 7-2. 면역세포화학분석(Immunocytochemistry)Example 7-2. Immunocytochemistry
Ninjurin1의 세포 내 발현 분포를 좀 더 확인하기 위해 면역세포화학분석을 수행하였다. 각각 4일 및 8일 동안 배양한 1차 cortical 신경세포인 DIV4와 DIV8에 GFP-Ninj1과 Flag-mNinj1을 트랜스펙션(Lipofectamine 2000, Invitrogen)하였다. 24시간 후, PFA(4%, 10min)를 사용하여 고정하고 보관하였다(PBS, 4℃). 면역세포화학분석을 위해 차가운 PBS로 2회 세정한 후, 20mM 글리신/PBS로 PFA의 반응을 저해하였다. Triton X-100 1%를 사용하여 permeabilization한 후, 블락킹 용액(1% BSA(w/v), 5% FBS(v/v))를 통해 비특이적 결합을 저해하였다. 4℃에서 밤새 각 1차 항체를 처리하였다. PBS로 2회 세정한 후, 실온에서 1시간동안 2차 항체(Alexa 488/546-conjugated donkey anti-rabbit/mouse IgG)를 처리한 후 공초점 현미경(confocal microscopy)(LSM 700, Carl Zeiss)을 이용하여 관찰하였다(도 7c). 사용한 1차 항체는 Anti-Flag(sigma), anti-GFP(Abcam), Ninjurin1(custom-made, Ab1-15)이다. Scale bar : 50um.In order to further confirm the expression distribution of ninjurin1 in cells, immunocytochemical analysis was performed. GFP-Ninj1 and Flag-mNinj1 were transfected (Lipofectamine 2000, Invitrogen) into primary cortical neurons DIV4 and DIV8 cultured for 4 days and 8 days, respectively. After 24 hours, fixed and stored using PFA (4%, 10 min) (PBS, 4 ° C). After washing twice with cold PBS for immunocytochemical analysis, the reaction of PFA was inhibited with 20 mM glycine / PBS. After permeabilization with 1% Triton X-100, nonspecific binding was inhibited via blocking solution (1% BSA (w / v), 5% FBS (v / v)). Each primary antibody was treated overnight at 4 ° C. After washing twice with PBS, a secondary antibody (Alexa 488 / 546-conjugated donkey anti-rabbit / mouse IgG) was treated for 1 hour at room temperature followed by confocal microscopy (LSM 700, Carl Zeiss). It was observed using (Fig. 7c). Primary antibodies used were Anti-Flag (sigma), anti-GFP (Abcam), Ninjurin1 (custom-made, Ab1-15). Scale bar: 50um.
실험결과, Ninjurin1은 시냅토솜 분획(P2, P2', P3)에 주로 존재하지만, 시냅스 베시클 분획(S3, S3')에는 존재하지 않았으며(도 7a), 1차 배양한 cortical 신경세포에서도 시냅토솜에 존재하는 것을 확인하였다(도 7b). 또한, 면역세포화학 분석 결과, 외생적으로 과발현한 Ninjurin1이 puncta-유사 구조체에서 발현되고 있는 것을 확인할 수 있었다(도 7c).As a result, Ninjurin1 is mainly present in the synaptosome fractions (P2, P2 ', P3), but not in the synaptic vesicle fractions (S3, S3') (Fig. 7a), and synapses in primary cultured cortical neurons. It was confirmed that it was present in the tosome (FIG. 7B). In addition, as a result of immunocytochemical analysis, it was confirmed that exogenously overexpressed Ninjurin1 is expressed in puncta-like structures (FIG. 7C).
실시예 8. pre-synapse 와 post-synapse에서 Ninjurin1의 발현 확인Example 8. Expression of Ninjurin1 in pre-synapse and post-synapse
Ninjurin1이 puncta 구조를 가진 시냅토솜 부분에서 발현되기 때문에, 여러 시냅스 표지인자와 이중 면역염색을 통해 시냅스 내에서의 Ninjurin1 발현 분포를 좀더 상세히 조사하였다.Since ninjurin1 is expressed in the synaptosome region with puncta structure, the distribution of Ninjurin1 expression in synapses was examined in detail through several synaptic markers and dual immunostaining.
이중 면역염색에 사용한 1차 항체는 다음과 같다.Primary antibodies used for dual immunostaining are as follows.
Pre-synapse 표지인자 : Synaptophysin(Millipore, mouse), vGlut1(Abcam, sheep), Pre-synapse markers: Synaptophysin (Millipore, mouse), vGlut1 (Abcam, sheep),
Post-synapse 표지인자 : Homer1 (Synaptic systems, mouse)Post-synapse marker: Homer1 (Synaptic systems, mouse)
실험 결과, 도 8에 나타낸 바와 같이, Ninjurin1은 각 표지인자들과 부분적으로 함께 발현되는 것을 확인하였다. 하지만 Presynapse 표지인자인 Synaptophysin과 vGlut1와는 여러 부분에서 overlay되지만 Postsynapse 표지인자인 Homer1과는 상대적으로 적은 부분이 overlay되는 것을 관찰하였다. 따라서, Ninjurin1은 Pre-synapse와 Post-synapse 모두에서 발현되지만, Post-synapse 보다는 Pre-synapse에 많이 존재함을 알 수 있다.As a result of the experiment, as shown in Figure 8, it was confirmed that Ninjurin1 is partially co-expressed with each marker. However, the presynapse markers Synaptophysin and vGlut1 were overlaid in various parts, but the postsynapse markers Homer1 were overlaid. Therefore, Ninjurin1 is expressed in both pre-synapse and post-synapse, but it can be seen that it is present in pre-synapse more than post-synapse.
실시예 9. Ninjurin1 KO 마우스의 Neural 활동성 증가 확인Example 9 Confirmation of Increased Neural Activity of Ninjurin1 KO Mice
실시예 1 내지 8에서 Ninjurin1 KO 마우스가 강박증 유사행동을 보이며 Ninjurin1이 시냅스에서 발현됨이 확인되었으므로, 다음과 같이 Fos B를 면역염색하여 발현 정도를 비교함으로써 신경 활성을 비교하였다. Since Ninjurin1 KO mice showed OCD-like behavior and Ninjurin1 was expressed at synapses in Examples 1 to 8, nerve activity was compared by immunostaining Fos B and comparing expression levels as follows.
야생형(WT)과 Ninjurin1 KO 마우스(3개월령 숫컷)에서 뇌 조직과 용해물(lysate)을 준비하고, 웨스턴 블랏팅(40ug, 12% gel)을 통해 Fos B (Santa cruz)의 발현 정도를 비교하였다. 면역조직화학 분석(Immunohistochemistry)의 경우, 마우스를 PBS로 관류하고 4℃에서 밤새 4% PFA로 고정하였다. 10%, 20%, 30% 수크로스를 사용하여 탈수한 후, OCT 블록을 만들었다. 마이크로톰(HM 525, Thermo Scientific)을 이용하여 20um로 자른 후 free-floating 용액에 넣어 사용 전까지 -70℃에 보관하였다. 자른 뇌조직을 PBS로 세정한 후(5min, 3회) 블락킹 용액(0.03% Triton X-100, 10% BSA, 5% FBS)으로 블락킹하고, 4℃에서 밤새 Fos B 항체(Santa Cruz)를 조직에 처리하였다. 다음 날, PBS로 10분간 3회 세정한 후, 2차 항체를 실온에서 1시간 처리하였다. Hechst33442(Molecular probe, Invitrogen)로 핵을 염색한 후, 공초점 현미경을 통해 관찰하였다.Brain tissue and lysate were prepared from wild-type (WT) and Ninjurin1 KO mice (3 months old male), and the expression levels of Fos B (Santa cruz) were compared by Western blotting (40ug, 12% gel). . For immunohistochemistry, mice were perfused with PBS and fixed at 4 ° C. overnight with 4% PFA. After dehydration using 10%, 20% and 30% sucrose, OCT blocks were made. The microtomes (HM 525, Thermo Scientific) were cut to 20 μm and placed in free-floating solution and stored at −70 ° C. until use. The cut brain tissue was washed with PBS (5 min, 3 times) and then blocked with blocking solution (0.03% Triton X-100, 10% BSA, 5% FBS) and Fos B antibody (Santa Cruz) overnight at 4 ° C. Was treated to tissue. The following day, after washing three times with PBS for 10 minutes, the secondary antibody was treated for 1 hour at room temperature. Nuclei were stained with Hechst33442 (Molecular probe, Invitrogen) and observed under confocal microscopy.
도 9a의 면역조직화학염색 결과 및 도 9b의 웨스턴 블롯 결과, Fos B의 발현이 Ninjurin1 KO 마우스 뇌에서 증가되어 있음을 확인하였다. 따라서, Ninjurin1 KO 마우스에서 신경 활성이 항진되어 있음을 알 수 있다.Immunohistochemical staining of FIG. 9A and Western blot of FIG. 9B confirmed that the expression of Fos B was increased in the brain of Ninjurin1 KO mouse. Therefore, it can be seen that the neuronal activity is enhanced in Ninjurin1 KO mice.
실시예 10. Ninjurin1 KO 마우스에서 NMDAR의 발현과 Src의 발현 증가 확인Example 10 Confirmation of Increased Expression of NMDAR and Src in Ninjurin1 KO Mice
웨스턴 블롯팅을 이용하여, Ninjurin1 KO 마우스의 뇌와 대뇌피질(cortex)에서 NMDA 수용체의 발현을 조사하였다. 조직으로 brain, cortex, synaptosme을 각각 준비한 후 웨스턴 블롯팅을 실시하였으며, 이때 사용한 1차 항체는 다음과 같다.Western blotting was used to examine the expression of NMDA receptors in the brain and cortex of Ninjurin1 KO mice. Brain, cortex, and synaptosme were prepared as tissues, followed by Western blotting. The primary antibodies used were as follows.
1) Millipore : NMDAR2A (Rabbit), NMDAR2B (Rabbit), NMDAR1 (Rabbit), PSD-95 (Mouse), Synaptophysin (Mouse), Synapsin (Mouse), GABARA (Mouse)1) Millipore: NMDAR2A (Rabbit), NMDAR2B (Rabbit), NMDAR1 (Rabbit), PSD-95 (Mouse), Synaptophysin (Mouse), Synapsin (Mouse), GABAR A (Mouse)
2) Cell signaling : Src, p-Src (Y416)2) Cell signaling: Src, p-Src (Y416)
3) Sigma : β-actin, α-tubulin3) Sigma: β-actin, α-tubulin
실험 결과, Ninjurin1 KO 마우스에서 글루타메이트 서브유닛들(NMDAR2A, NMDAR2B, NMDAR1)의 발현은 증가되어 있었지만, 시냅스 형성을 조절하는 인자인 synapsin과 synaptophysin에는 변화가 관찰되지 않았다(도 10a, 도 10b). 또한 인산화된 Src의 양이 Ninjurin1 KO 마우스 시냅토솜에서 증가되어 있었다(도 10c). 따라서, Src의 활성화를 통한 글루타메이트 경로에 의해, Ninjurin1 KO 마우스의 강박증 유사 행동이 조절될 것으로 예상된다. As a result, the expression of glutamate subunits (NMDAR2A, NMDAR2B, NMDAR1) was increased in Ninjurin1 KO mice, but no change was observed in synapsin and synaptophysin, factors regulating synapse formation (FIGs. 10A and 10B). The amount of phosphorylated Src was also increased in Ninjurin1 KO mouse synaptosomes (FIG. 10C). Therefore, it is expected that the obsessive-like behavior of Ninjurin1 KO mice is regulated by the glutamate pathway through activation of Src.
실시예 11. Memantine의 Ninjurin1 KO 마우스 강박증 저해 효과 확인Example 11 Confirmation of Inhibitory Effect of Memantine on Ninjurin1 KO Mouse OCD
Ninjurin1 KO 마우스의 강박증 유사행동이 NMDAR의 증가와 관련있음을 확인하였기 때문에, NMDAR의 길항제(antagonist)로서 알츠하이머 질환 치료제인 메만틴염산염(Memantine hydrochloride)에 대하여 강박증 저해효과를 조사하였다.Obsessive-compulsive behavior of Ninjurin1 KO mice was associated with an increase in NMDAR. Therefore, the effect of inhibiting OCD against memantine hydrochloride, an antagonist of NMDAR, was treated for Alzheimer's disease.
실시예 11-1. Barbering과 피부 손상 비교Example 11-1. Barbering vs. skin damage
Barbering과 피부 손상을 보이는 Ninjurin1 KO 마우스(5개월령)에 메만틴염산염(sigma, 5mg/kg)을 10일 동안 매일 복강 주입하였다. 2주 후, Barbering과 피부손상의 정도를 약물 주입하기 전과 비교하였다(도 11a).Memantine hydrochloride (sigma, 5 mg / kg) was intraperitoneally injected daily for 10 days in Barbering and skin injury Ninjurin1 KO mice (5 months old). Two weeks later, Barbering and the degree of skin damage were compared with before injection (FIG. 11A).
실시예 11-2. Marble-burying 테스트Example 11-2. Marble-burying test
Ninjurin1 KO 마우스에 5mg/kg의 메만틴염산염을 복강 주입하였다. 1시간 후, 실시예 2의 방법과 동일하게 Marble-burying 테스트를 실시하였다(도 11d). Ninjurin1 KO mice were intraperitoneally injected with 5 mg / kg of memantine hydrochloride. After 1 hour, a marble-burying test was conducted in the same manner as in Example 2 (FIG. 11D).
실험 결과, 메만틴염산염을 주입하였을 경우 Ninjurin1 KO 마우스의 barbering(도 11b), self-made injury(도 11c), Marble-burying(도 11d)과 같은 강박증 유사행동이 현저히 감소하였다. 결론적으로, NMDAR는 Ninjurin1 KO 마우스의 OCD 유사행동을 매개하며, 이의 활성을 저해하는 메만틴염산염은 OCD 행동을 치료하기 위한 가능한 후보로 기대된다.As a result, the injection of memantine hydrochloride significantly reduced OCD-like behaviors such as barbering (FIG. 11B), self-made injury (FIG. 11C), and marble-burying (FIG. 11D) of Ninjurin1 KO mice. In conclusion, NMDAR mediates OCD-like behavior in Ninjurin1 KO mice, and memantine hydrochloride, which inhibits its activity, is expected to be a possible candidate for treating OCD behavior.
전술한 본 발명의 설명은 예시를 위한 것이며, 본 발명이 속하는 기술분야의 통상의 지식을 가진 자는 본 발명의 기술적 사상이나 필수적인 특징을 변경하지 않고서 다른 구체적인 형태로 쉽게 변형이 가능하다는 것을 이해할 수 있을 것이다. 그러므로 이상에서 기술한 실시예들은 모든 면에서 예시적인 것이며 한정적이 아닌 것으로 이해해야만 한다.The foregoing description of the present invention is intended for illustration, and it will be understood by those skilled in the art that the present invention may be easily modified in other specific forms without changing the technical spirit or essential features of the present invention. will be. Therefore, it should be understood that the embodiments described above are exemplary in all respects and not restrictive.

Claims (7)

  1. 닌쥬린 1(Ninjurin 1)을 결핍시켜 제조된 강박증 동물모델.Obsessive-compulsive animal model produced by deficient in ninjurin 1 (Ninjurin 1).
  2. 제 1 항에 있어서, 상기 동물은 인간을 제외한 것을 특징으로 하는 동물모델.The animal model of claim 1, wherein the animal is not human.
  3. 제 1 항에 있어서, 상기 닌쥬린 1 결핍은 닌쥬린 1 유전자의 넉아웃(Knock-out)에 의한 것임을 특징으로 하는 동물모델.The animal model of claim 1, wherein the ninjurin 1 deficiency is caused by knock-out of the ninjurin 1 gene.
  4. 제 1 항의 동물모델을 이용하여 강박증을 예방 또는 치료하기 위한 후보약물 탐색방법.A method of searching for a candidate drug for preventing or treating OCD using the animal model of claim 1.
  5. 제 4 항에 있어서, 상기 탐색방법은 동물모델에 후보약물을 투여한 후 NMDA 수용체(N-Methyl-D-aspartate receptor)가 저해되는지를 측정하는 단계를 포함하는 것을 특징으로 하는 탐색방법.The method of claim 4, wherein the detection method comprises measuring whether the N-Methyl-D-aspartate receptor (NMDA) receptor is inhibited after administering the candidate drug to the animal model.
  6. 제 5 항에 있어서, 상기 후보약물은 NMDA 수용체 길항제인 것을 특징으로 하는 탐색방법.The method of claim 5, wherein the candidate drug is an NMDA receptor antagonist.
  7. 제 6 항에 있어서, 상기 NMDA 수용체 길항제는 메만틴(Memantine)인 것을 특징으로 하는 탐색방법.7. The method of claim 6, wherein said NMDA receptor antagonist is memantine.
PCT/KR2014/005531 2013-08-14 2014-06-23 Animal model with obsessive compulsive disorder using ninjurin 1 deficiency WO2015023054A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR20130096951A KR101484405B1 (en) 2013-08-14 2013-08-14 Pharmaceutical composition for the prevention or treatment of obssesive-compulsive spectrum disorder caused by ninjurin 1 deficiency
KR10-2013-0096951 2013-08-14

Publications (1)

Publication Number Publication Date
WO2015023054A1 true WO2015023054A1 (en) 2015-02-19

Family

ID=52468421

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2014/005531 WO2015023054A1 (en) 2013-08-14 2014-06-23 Animal model with obsessive compulsive disorder using ninjurin 1 deficiency

Country Status (2)

Country Link
KR (1) KR101484405B1 (en)
WO (1) WO2015023054A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018038583A1 (en) * 2016-08-26 2018-03-01 에이비온 주식회사 Anti-ninj-1 antibody and use of same

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102379890B1 (en) 2019-12-13 2022-03-28 고려대학교 산학협력단 Animal model for obsessive compulsive disorder and a method for producing thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20090029200A (en) * 2006-05-22 2009-03-20 반다 파마슈티칼즈, 인코퍼레이티드. Treatment for depressive disorders

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2528622C (en) * 2003-05-27 2010-08-03 Forest Laboratories, Inc. Combination of an nmda receptor antagonist and a selective serotonin reuptake inhibitor for the treatment of depression and other mood disorders
US20090221554A1 (en) * 2008-02-28 2009-09-03 Zenyaku Kogyo Kabushiki Kaisha Method of treating cognitive impairment
FR2983732B1 (en) * 2011-12-09 2013-11-22 Servier Lab NOVEL ASSOCIATION BETWEEN 4- {3- [CIS-HEXAHYDROCYCLOPENTA [C] PYRROL-2 (1H) -YL] PROPOXY} PHARMACEUTICALS CONTAINING THE SAME

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20090029200A (en) * 2006-05-22 2009-03-20 반다 파마슈티칼즈, 인코퍼레이티드. Treatment for depressive disorders

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SEONG-JUN CHO ET AL.: "Ninjurinl, a target of p53, regulates p53 expression and p53- dependent cell survival, senescence, and radiation-induced mortality.", PROC NATL ACAD SCI U S A., vol. 110, no. 23, 4 June 2013 (2013-06-04), pages 9362 - 9367 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018038583A1 (en) * 2016-08-26 2018-03-01 에이비온 주식회사 Anti-ninj-1 antibody and use of same
US11286296B2 (en) 2016-08-26 2022-03-29 Abion Inc. Anti-human ninjurin-1 (NINJ-1) antibodies and methods for detecting human NINJ-1

Also Published As

Publication number Publication date
KR101484405B1 (en) 2015-01-19

Similar Documents

Publication Publication Date Title
Lee et al. Emerging roles of mitochondria in synaptic transmission and neurodegeneration
Pearson-Leary et al. Novel roles for the insulin-regulated glucose transporter-4 in hippocampally dependent memory
Hua et al. Ca2+‐dependent glutamate release involves two classes of endoplasmic reticulum Ca2+ stores in astrocytes
Gallos et al. Selective targeting of the α5-subunit of GABAA receptors relaxes airway smooth muscle and inhibits cellular calcium handling
Workman et al. Rapid antidepressants stimulate the decoupling of GABAB receptors from GIRK/Kir3 channels through increased protein stability of 14-3-3η
Kishimoto et al. Synergistic μ-opioid and 5-HT1A presynaptic inhibition of GABA release in rat periaqueductal gray neurons
Razy-Krajka et al. Monoaminergic modulation of photoreception in ascidian: evidence for a proto-hypothalamo-retinal territory
Bradley et al. Biased M1-muscarinic-receptor-mutant mice inform the design of next-generation drugs
Zaninetti et al. Presence of functional neuronal nicotinic acetylcholine receptors in brainstem motoneurons of the rat
Kinney et al. Synaptically evoked GABA transporter currents in neocortical glia
Farah Naquiah et al. Transgenerational effects of paternal heroin addiction on anxiety and aggression behavior in male offspring
Lewis et al. Bidirectional regulation of aggression in mice by hippocampal alpha-7 nicotinic acetylcholine receptors
Boehm et al. Glycine receptors in cultured chick sympathetic neurons are excitatory and trigger neurotransmitter release
Zou et al. The effects of quinine on neurophysiological properties of dopaminergic neurons
Han et al. Cordycepin improves behavioral‐LTP and dendritic structure in hippocampal CA1 area of rats
Al‐Romaiyan et al. Investigation of intracellular signalling cascades mediating stimulatory effect of a Gymnema sylvestre extract on insulin secretion from isolated mouse and human islets of Langerhans
WO2015023054A1 (en) Animal model with obsessive compulsive disorder using ninjurin 1 deficiency
Berghe et al. Synaptic transmission induces transient Ca2+ concentration changes in cultured myenteric neurones
Mah et al. Ethanol alters calcium signaling in axonal growth cones
Qi et al. mGluR 5 in the nucleus accumbens shell regulates morphine‐associated contextual memory through reactive oxygen species signaling
Naziroğlu et al. Role of TRPM2 cation channels in dorsal root ganglion of rats after experimental spinal cord injury
Rabe et al. The transcription factor Uncx4. 1 acts in a short window of midbrain dopaminergic neuron differentiation
Kapell et al. Neuron-oligodendrocyte potassium shuttling at nodes of Ranvier protects against inflammatory demyelination
Hesketh et al. Effects of chronic treatment with citalopram on cannabinoid and opioid receptor-mediated G-protein coupling in discrete rat brain regions
Zaika et al. Functional role of M‐type (KCNQ) K+ channels in adrenergic control of cardiomyocyte contraction rate by sympathetic neurons

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: 14836183

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14836183

Country of ref document: EP

Kind code of ref document: A1