JP2017057173A - Composition for treating brain damage disease targeting TIM-3 and screening method thereof - Google Patents

Composition for treating brain damage disease targeting TIM-3 and screening method thereof Download PDF

Info

Publication number
JP2017057173A
JP2017057173A JP2015184356A JP2015184356A JP2017057173A JP 2017057173 A JP2017057173 A JP 2017057173A JP 2015184356 A JP2015184356 A JP 2015184356A JP 2015184356 A JP2015184356 A JP 2015184356A JP 2017057173 A JP2017057173 A JP 2017057173A
Authority
JP
Japan
Prior art keywords
tim
hif
expression
cells
mice
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
JP2015184356A
Other languages
Japanese (ja)
Other versions
JP6653054B2 (en
Inventor
パク,ウン−ジョン
Eun-Jung Park
コ,ハン−ソク
Han-Seok Koh
ジョン,ソン−ホ
Sung-Ho Jeon
チャン,チ−ヨン
Chi-Young Chang
ユン,ヒ−ジョン
He-Jeong Yoon
ジョン,セ−ボム
Sae-Bom Jeon
キム,ヒョン−ソク
Hyung-Seok Kim
アン,イェ−ヒョン
Ye-Hyeon Ahn
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
NAT CANCER CT
NATIONAL CANCER CENTER
Original Assignee
NAT CANCER CT
NATIONAL CANCER CENTER
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 NAT CANCER CT, NATIONAL CANCER CENTER filed Critical NAT CANCER CT
Priority to JP2015184356A priority Critical patent/JP6653054B2/en
Publication of JP2017057173A publication Critical patent/JP2017057173A/en
Application granted granted Critical
Publication of JP6653054B2 publication Critical patent/JP6653054B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Abstract

PROBLEM TO BE SOLVED: To provide a pharmaceutical composition for preventing or treating brain damage diseases, the composition comprising TIM-3 (T-cell immunoglobulin and mucin domain protein 3) inhibitor as an active ingredient, and also provide a screening method for a therapeutic agent for brain damage disease, the method using TIM-3.SOLUTION: The present invention provides a pharmaceutical composition for preventing or treating brain damage diseases, based on the following findings: TIM-3 protein plays a role as a modulator in brain injury induced during the state of hypoxia caused by ischemia; and expression of TIM-3 is regulated by HIF-1, which regulates the expression of genes occurring in hypoxia. Therefore, the composition according to the present invention can be usefully used for the treatment and prevention of cerebral nervous system diseases associated with hypoxia.SELECTED DRAWING: Figure 1

Description

本発明は、TIM−3(T−cellimmunoglobulin and mucin domain protein 3)抑制剤を有効成分として含む脳損傷疾患の予防または治療用薬学的組成物及びTIM−3を利用した脳損傷疾患治療剤のスクリーニング方法に関する。   The present invention relates to a pharmaceutical composition for the prevention or treatment of brain injury diseases comprising a TIM-3 (T-cell immunoglobulin and mucin domain protein 3) inhibitor as an active ingredient, and a screening for therapeutic agents for brain injury diseases using TIM-3. Regarding the method.

脳虚血(cerebral ischaemia)は、複雑な病態生理学的変化を引き起こし、究極的に、特に虚血組織の中心部位(ischaemiccore)を取り囲む半陰影領域(penumbralarea)で脳損傷を引き起こす。このような変化には、常在細胞(resident cell)の活性化、炎症性メディエーター(inflammatory mediators)の生成及び炎症細胞の浸潤(infiltration)が含まれる。臨床実験結果によると、脳虚血による炎症反応は、脳損傷の発病と関連があるように見えるが、これと関連した炎症反応については未だ多く知られていない。   Cerebral ischemia causes complex pathophysiological changes, ultimately causing brain damage, particularly in the penumbrial area surrounding the ischemic core of ischemic tissue. Such changes include activation of resident cells, generation of inflammatory mediators and infiltration of inflammatory cells. According to the results of clinical experiments, the inflammatory response due to cerebral ischemia seems to be related to the onset of brain injury, but much is not yet known about the inflammatory response associated with this.

T−細胞免疫グロブリン及びムチンドメインタンパク質ファミリ(T−cell immunoglobulin and mucin domain protein family)のメンバーであるTIM−3は、TH1−依存的免疫反応を陰性的に調節する第1型ヘルパー(helper)T細胞(TH1)−特異的表面分子として初めて同定されたが、後続の研究において、TIM−3は、TH17細胞、Tregs、NK細胞、タンパク白血球(monocytes)、樹脂状細胞、肥満細胞(mast cells)及び小膠細胞(microglia)を含む多様な類型の兔疫細胞から発現されて、適応免疫(adaptive immunity)だけでなく、先天免疫(innate immunity)も調節するという事実が明かされた。最近の研究結果によると、TIM−3は、先天性兔疫細胞の活性化を調節することに重要な役割をし、環境によって活性化マーカーまたは活性化制限因子として作用する。動物モデル及び人体で、TIM−3は、感染、自己免疫疾患及び癌を含む多様な免疫関連疾病と密接な関連があることが表れた。興味深いことに、TIM−3は、細胞の種類と環境によって多様な機能を表すとされる(非特許文献1)。例えば、晩成ウイルス感染及び腫瘍でTIM−3の抑制は、枯渇されたT細胞のエフェクター(effector)機能を増加させる一方、TIM−3信号伝逹の増加は、Th−1−媒介されたEAE(experimental autoimmune encephalomyelitis)を改善することが表れた。また、自己兔疫性肝炎でCD4+CD25−T細胞上のTIM−3水準の減少は、免疫調節の損傷に寄与した一方、晩成C型肝炎では、CD4+及びCD8+T細胞のTIM−3が過発現された。   TIM-3, a member of the T-cell immunoglobulin and mucin domain protein family, is a first-type helper T that negatively regulates TH1-dependent immune responses. Although identified for the first time as a cell (TH1) -specific surface molecule, in subsequent studies, TIM-3 was found to be TH17 cells, Tregs, NK cells, protein leukocytes (monocytes), resinous cells, mast cells. And is expressed from various types of epithelial cells, including microglia, and regulates not only adaptive immunity but also innate immunity It has been revealed. According to recent research results, TIM-3 plays an important role in regulating the activation of congenital epithelial cells and acts as an activation marker or activation limiting factor depending on the environment. In animal models and the human body, TIM-3 has been shown to be closely related to a variety of immune related diseases including infections, autoimmune diseases and cancer. Interestingly, TIM-3 is said to exhibit various functions depending on the cell type and environment (Non-Patent Document 1). For example, suppression of TIM-3 in epigenetic virus infection and tumors increases the effector function of depleted T cells, while increased TIM-3 signaling is Th-1-mediated EAE ( It has been shown to improve experimental autoimmune encephalomyelitis). In addition, decreased TIM-3 levels on CD4 + CD25-T cells in autocidal hepatitis contributed to impaired immune regulation, whereas TIM-3 on CD4 + and CD8 + T cells was overexpressed in late hepatitis C .

低酸素症(hypoxia)に対する生理学的反応は、酸素−調節性アルファ−サブユニット(oxygen−regulated α−subunit)と構成的ベータ−サブユニット(constitutive β−subunit)とからなるヘテロ二量体の(heterodimeric)転写因子である、HIF(hypoxia−inducible factor)−1によって主に媒介すると知られている。HIF−1複合体は、低酸素症への適応と関連した様々な遺伝子の低酸素−反応部位(hypoxic−response elements、HREs)に結合する。興味深いことに、HIF−1は、低酸素環境下だけでなく炎症環境下でも細胞反応を調節し、多くの炎症関連の疾病の発病にも重要な役割をするとされる。生体内(in vivo)及び試験管内(in vitro)の実験において、HIF−1は、骨髄細胞の移動のような骨髄細胞媒介の炎症反応に必須であることが表れた。また、HIF−1活性は、虚血性肺及び腸の損傷後の病原性炎症反応と関連があった。従って、HIF−1は、炎症関連の信号伝逹を調節する核心的な調節因子とされる。   The physiological response to hypoxia is a heterodimer consisting of an oxygen-regulated alpha-subunit and a constitutive beta-subunit (constitutive β-subunit). It is known to be mainly mediated by HIF (hypoxia-inducible factor) -1, which is a heterodimeric transcription factor. HIF-1 complexes bind to hypoxic-response elements (HREs) of various genes associated with adaptation to hypoxia. Interestingly, HIF-1 regulates cellular responses not only in a hypoxic environment but also in an inflammatory environment and is thought to play an important role in the pathogenesis of many inflammation-related diseases. In in vivo and in vitro experiments, HIF-1 has been shown to be essential for bone marrow cell-mediated inflammatory responses such as bone marrow cell migration. HIF-1 activity was also associated with pathogenic inflammatory responses following ischemic lung and intestinal damage. Therefore, HIF-1 is regarded as a core regulator that regulates inflammation-related signal transmission.

一方、中枢神経系(CNS)は、免疫寛容地域(immune−privileged regions)であると知られていたが、最近の研究結果で先天性及び後天的適応性免疫反応(subsequent adaptive immune responses)を速めに誘発することができる精巧な監視システム(sentinel system)を備えていると報告された。CNSの免疫反応において、主な兔疫細胞として機能する膠細胞(glial cell)は、脳の微細な変化を認知し、病態生理学的刺激に速く反応する。   The central nervous system (CNS), on the other hand, was known to be an immune-privileged region, but recent studies have accelerated innate and acquired adaptive immune responses. It has been reported that it has an elaborate monitoring system that can be triggered. In the immune response of the CNS, glial cells that function as the main epithelial cells recognize minute changes in the brain and respond quickly to pathophysiological stimuli.

上記のような従来の報告に基づいて研究した結果、本発明者らは、低酸素症環境で小膠細胞(microglia)及び星状細胞(astrocyte)のTIM−3発現が増加(upregulated)し、このようなTIM−3の発現増加が好中球(neutrophils)の低酸素性半陰影(hypoxic penumbra)への浸潤(infiltration)に影響を及ぼすという事実を新たに発見した。このような浸潤は、虚血性脳損傷の主な原因として知られている。また、本発明者らは、神経膠細胞でHIF−1がTIM−3の酸素−依存的発現を調節するという事実を明らかにし、このような実験結果から本発明を完成した。   As a result of studies based on the conventional reports as described above, the present inventors have increased TIM-3 expression in microglia and astrocytes in a hypoxic environment, It was newly discovered that such increased expression of TIM-3 affects the infiltration of neutrophils into hypoxic penumbra. Such infiltration is known as a major cause of ischemic brain damage. The present inventors have also clarified the fact that HIF-1 regulates the oxygen-dependent expression of TIM-3 in glial cells, and completed the present invention from the results of such experiments.

US2014/0099254A1US2014 / 0099254A1

Han,G.,et al.(2013)Han, G .; , Et al. (2013) Cramer,T.et al.HIF−1alpha is essential for myeloid cell−mediated inflammation.Cell 112,645−57(2003)Cramer, T.A. et al. HIF-1 alpha is essential for myeloid cell-mediated inflation. Cell 112, 645-57 (2003) Zhang,L.et al.Estrogen stimulates microglia and brain recovery from hypoxia−ischemia in normoglycemic but not diabetic female mice.J.Clin.Invest.113,85−5(2004)Zhang, L.M. et al. Estrogens stimulates microglia and brain recovery from hypoxia-ischemia in normotic butt diabetic female. J. et al. Clin. Invest. 113, 85-5 (2004) Swanson,R.A.et al.A semiautomated method for measuring brain infarct volume.J.Cereb.Blood Flow Metab.10,290−93(1990)Swanson, R.M. A. et al. A semi-automated method for measuring brain inform volume. J. et al. Cereb. Blood Flow Metab. 10, 290-93 (1990) Frank,M.G.,Wieseler−Frank,J.L.,Watkins,L.R.&Maier,S.F. isolation of highly enriched and quiescent microglia from adult rat hippocampus:immunophenotypic and functional characteristics.J.Neurosci.Methods 151,121−30(2006)Frank, M.M. G. Wieseler-Frank, J .; L. , Watkins, L .; R. & Maier, S .; F. isolation of high enriched and quiet microglia from adult hippocampus: immunophenotypic and functional charactaristics. J. et al. Neurosci. Methods 151, 121-30 (2006) Weinstein,D.Isolation and purification of primary rodent astrocytes.Curr.Protoc.Neurosci.Chapter3,Unit35(2001)Weinstein, D.C. Isolation and purification of primary rodent astrocycles. Curr. Protoc. Neurosci. Chapter 3, Unit 35 (2001) Chang,C.Y.et al.Dualfunctionality of myeloperoxidase in rotenone−exposed brain−resident immune cells.Am.J.Pathol.179,964−79(2011)Chang, C.I. Y. et al. Dual function of myeloperoxidase in rotenone-exposed brain-resident immuno cells. Am. J. et al. Pathol. 179, 964-79 (2011) Huang,Z.et al.Effects of cerebral ischemia in mice deficient in neuronal nitric oxide synthase.Science 265,1883−885(1994)Huang, Z .; et al. Effects of cerebral ischemia in mificant in neuron nitrite synthase. Science 265, 1883-885 (1994) Blouw,B.et al.The hypoxic response of tumors is dependent on their microenvironment.Cancer Cell 4,133−46(2003)Bloom, B.W. et al. The hypoxic response of tumors is dependent on the microenvironment. Cancer Cell 4, 133-46 (2003) Kutner,R.H.,Zhang,X.Y.&Reiser,J.Production,concentration and titration of pseudotyped HIV−1−based lentiviral vectors.Nat.Protoc.4,495−05(2009)Kutner, R.A. H. , Zhang, X. Y. & Reiser, J .; Production, contention and titration of pseudotyped HIV-1-based lenticular vectors. Nat. Protoc. 4,495-05 (2009) Zhang,L.et al.Estrogen stimulates microglia and brain recovery from hypoxia−ischemia in normoglycemic but not diabetic female mice.J.Clin.Invest.113,85−95(2004)Zhang, L.M. et al. Estrogens stimulates microglia and brain recovery from hypoxia-ischemia in normotic butt diabetic female. J. et al. Clin. Invest. 113, 85-95 (2004) Wang,G.L.&Semenza,G.L.Characterization of hypoxia−inducible factor 1 and regulation of DNA binding activity by hypoxia.J.Biol.Chem.268,21513−21518(1993)Wang, G .; L. & Semenza, G .; L. Characteristic of hypoxia-inducible factor 1 and regulation of DNA binding activity by hypoxia. J. et al. Biol. Chem. 268, 21513-21518 (1993) Bergeron、M.,Yu,A.Y.,Solway,K.E.,Semenza,G.L.&Sharp,F.R.Induction of hypoxia−inducible factor−1(HIF−1) and its target genes following focal ischaemia in rat brain.Eur.J.Neurosci.11,4159−4170(1999)Bergeron, M.M. Yu, A .; Y. , Solway, K .; E. , Semenza, G .; L. & Sharp, F.A. R. Induction of hypoxia-inducible factor-1 (HIF-1) and it's target genes following focal ischaemia in rat brain. Eur. J. et al. Neurosci. 11, 4159-4170 (1999) Williams,R.The Mouse Brain Library http://www.mbl.org/atlas165/atlas165_start(1999)Williams, R.A. The Mouse Brain Library http: // www. mbl. org / atlas165 / atlas165_start (1999) Franklin,K.B.J.&Paxinos,G.The Mouse Brain in Stereotaxic Coordinates 3rd edn(Elsevier/Academic Press,2008)Franklin, K.M. B. J. et al. & Paxinos, G.M. The Mouse Brain in Stereocoordinates 3rd edn (Elsevier / Academic Press, 2008) Gerriets,T.et al.Noninvasive quantification of brain edema and the spaceoccupying effect in rat stroke models using magnetic resonance imaging.Stroke35,566−571(2004)Gerriets, T .; et al. Noninvasive quantification of brain edema and the spacecapping effect in rat stroke models using magnetic resonance imaging. Stroke 35, 566-571 (2004) Le,D.A.et al.Caspase activation and neuroprotection in caspase−3−deficient mice after in vivo cerebral ischemia and in vitro oxygen glucose deprivation.Proc.Natl Acad.Sci.USA99,15188−15193(2002)Le, D.C. A. et al. Caspase activation and neuroprotection in caspase-3-definitive microphone after in vivo cerebral ischemia and in vitro oxygen glucose deprivation. Proc. Natl Acad. Sci. USA99, 15188-15193 (2002) Broughton、B.R.,Reutens,D.C.&Sobey,C.G.Apoptotic mechanisms after cerebral ischemia.Stroke40,e331−e339(2009)Broughton, B.M. R. Reutens, D .; C. & Sovey, C.I. G. Apoptotic machinery after cerebral ischemia. Stroke40, e331-e339 (2009) Chaitanya,G.V.,Steven,A.J.&Babu,P.P.PARP−1 cleavage fragments:signatures of cell−death proteases in neurodegeneration.CellCommun.Signal.8,31(2010)Chaitanya, G .; V. Steven, A .; J. et al. & Babu, P.A. P. PARP-1 cleavage fragments: signatures of cell-death protocols in neurogeneration. Cell Commun. Signal. 8, 31 (2010) McColl,B.W.,Rothwell,N.J.&Allan,S.M.Systemic inflammatory stimulus potentiates the acute phase and CXC chemokine responses to experimental stroke and exacerbates brain damage via interleukin−1 and neutrophildependent mechanisms.J.Neurosci.27,4403−.4412(2007)McColl, B.M. W. Rothwell, N .; J. et al. & Allan, S .; M.M. Systematic stimulus potentials the exact phase and CXC chemokin responses to experiential brains and excurbate brain vein dimensions. J. et al. Neurosci. 27, 4403-. 4412 (2007) Chen、H.et al.Anti−CD11b monoclonal antibody reduces ischemic cell damage after transient focal cerebral ischemia in rat.Ann.Neurol.35,458−.463(1994)Chen, H.C. et al. Anti-CD11b monoclonal antibodies redismatic cell damage after transient focal cerebral ischemia in rat. Ann. Neurol. 35,458-. 463 (1994) Kobayashi,S.D.,Voyich,J.M.,Burlak,C.&DeLeo,F.R.Neutrophils in the innate immune response.Arch.Immunol.Ther.Exp.(Warsz).53,505−517(2005)Kobayashi, S .; D. , Voyich, J. et al. M.M. Burlake, C .; & DeLeo, F.A. R. Neutrophils in the innate immune response. Arch. Immunol. Ther. Exp. (Warsz). 53,505-517 (2005) Murikinati,S.et al.Activation of cannabinoid 2 receptors protects against cerebral ischemia by inhibiting neutrophil recruitment.FASEB J.24,788−798(2010)Murkinati, S.M. et al. Activation of cannabinoid 2 receptors protects against cerebral ischemia by inhibiting neutropil recruitment. FASEB J.H. 24, 788-798 (2010) Muir,K.W.,Tyrrell,P.,Sattar,N.&Warburton,E.Inflammation and ischaemic stroke.Curr.Opin.Neurol.20,334−.342(2007)Muir, K.M. W. Tyrrell, P .; Sattar, N .; & Warburton, E .; Inflammation and ischemic stroke. Curr. Opin. Neurol. 20, 334-. 342 (2007) Saijo,K.&Glass,C.K.Microglial cell origin phenotypes in health and disease.Nat.Rev.Immunol.11,775−.787(2011)Saijo, K .; & Glass, C.I. K. Microcellular cell origin phenotypes in health and disease. Nat. Rev. Immunol. 11,775-. 787 (2011) Ren,X.et al.Regulatory B cells limit CNS inflammation and neurologic deficits in murine experimental stroke.J.Neurosci.31,8556−.8563(2011)Ren, X. et al. Regulatory B cells limit CNS inflation and neurological defects in murine experomental stroke. J. et al. Neurosci. 31, 8556-. 8563 (2011) Kleinschnitz,C.et al.Post−stroke inhibition of induced NADPH oxidase type 4 prevents oxidative stress and neurodegeneration.PLoS Biol.8,pii:e1000479(2010)Kleinschnitz, C.I. et al. Post-stroke inhibition of induced NADPH oxidase type 4 presents oxidative stress and neurodegeneration. PLoS Biol. 8, pii: e1000479 (2010)

本発明の目的は、TIM−3(T−cell immunoglobulin and mucin domain protein 3)をターゲットとしてこの発現または活性を抑制させる脳損傷疾患の予防または治療用薬学的組成物を提供することにある。   An object of the present invention is to provide a pharmaceutical composition for preventing or treating a brain injury disease that suppresses this expression or activity targeting TIM-3 (T-cell immunoglobulin and mucin domain protein 3).

本発明の他の目的は、TIM−3を利用して脳損傷疾患の治療剤をスクリーニングする方法を提供することにある。   Another object of the present invention is to provide a method for screening for a therapeutic agent for brain injury using TIM-3.

上記目的を達成するために、本発明は、TIM−3(T−cell immunoglobulin and mucin domain protein 3)抑制剤を有効成分として含む脳損傷疾患の予防または治療用薬学的組成物を提供する。   In order to achieve the above object, the present invention provides a pharmaceutical composition for preventing or treating a brain injury disease comprising an inhibitor of TIM-3 (T-cell immunoglobulin and mucin domain protein 3) as an active ingredient.

本発明の一実施例において、前記TIM−3抑制剤は、直接または間接的にTIM−3に結合するか、これと反応するか、またはこの発現を調節するなどの方法で、TIM−3の発現または活性を特異的に抑制または減少させることができる物質であり、有機または無機化合物、タンパク質、抗体、ペプチドまたは核酸分子などを含む。本発明の一実施例において、前記TIM−3抑制剤は、TIM−3に結合するか、これと反応して、TIM−3の活性を特異的に抑制または減少させる拮抗抗体またはその断片であるが、これに限定されない。本発明の一実施例において、前記TIM−3抑制剤は、TIM−3遺伝子の発現を直接または間接的に抑制する核酸分子であり、このような核酸分子の例としては、TIM−3遺伝子またはその断片に対するアンチセンスヌクレオチド、siRNA、shRNAまたはmiRNAなどがあるが、これに限定されない。   In one embodiment of the invention, the TIM-3 inhibitor binds to, reacts with, or modulates the expression of TIM-3, such as directly or indirectly binding to TIM-3. A substance that can specifically suppress or reduce expression or activity, and includes organic or inorganic compounds, proteins, antibodies, peptides, nucleic acid molecules, and the like. In one embodiment of the present invention, the TIM-3 inhibitor is an antagonistic antibody or a fragment thereof that specifically binds to or reacts with TIM-3 and specifically suppresses or decreases the activity of TIM-3. However, it is not limited to this. In one embodiment of the present invention, the TIM-3 inhibitor is a nucleic acid molecule that directly or indirectly suppresses the expression of the TIM-3 gene. Examples of such a nucleic acid molecule include a TIM-3 gene or Examples include, but are not limited to, antisense nucleotides, siRNA, shRNA, or miRNA for the fragment.

本発明の一実施例において、前記TIM−3タンパク質は、序列番号1のアミノ酸序列からなり、前記TIM−3遺伝子は、序列番号2の塩基序列からなる。   In one embodiment of the present invention, the TIM-3 protein consists of an amino acid sequence of sequence number 1, and the TIM-3 gene consists of a sequence of sequence number 2.

本発明の一実施例において、前記TIM−3抑制剤は、TIM−3遺伝子の上位(upstream)遺伝子またはTIM−3遺伝子の発現調節部位の発現または活性を抑制することで、TIM−3の発現を抑制する作用をする。   In one embodiment of the present invention, the TIM-3 inhibitor suppresses the expression or activity of an upstream gene of the TIM-3 gene or an expression regulatory site of the TIM-3 gene, thereby expressing TIM-3. The action which suppresses.

本発明の一実施例において、前記TIM−3抑制剤は、HIF−1(hypoxia−inducible factor−1)の発現または活性を抑制させる。   In one embodiment of the present invention, the TIM-3 inhibitor suppresses the expression or activity of HIF-1 (hypoxia-inducible factor-1).

本発明の一実施例において、前記TIM−3抑制剤は、好中球走化因子(neutrophil chemotactic factor)の発現または活性を減少させて、好中球の移動及び浸潤を阻害することで、脳損傷疾患の予防または治療効果を表す。   In one embodiment of the present invention, the TIM-3 inhibitor decreases the expression or activity of a neutrophil chemotactic factor to inhibit neutrophil migration and invasion, thereby It represents the preventive or therapeutic effect of the injured disease.

また、本発明は、(a)TIM−3が発現される細胞または動物モデルに候補物質を処理する段階と、(b)前記候補物質処理後、TIM−3の発現または活性程度を測定する段階と、(c)前記TIM−3の発現または活性程度が候補物質を処理しない対照群に比べて減少した候補物質を選別する段階とを含む脳損傷疾患治療剤のスクリーニング方法を提供する。   The present invention also includes (a) a step of treating a candidate substance in a cell or animal model in which TIM-3 is expressed, and (b) a step of measuring the expression or activity level of TIM-3 after the candidate substance treatment And (c) selecting a candidate substance having a decreased expression or activity level of TIM-3 as compared to a control group not treating the candidate substance, and a screening method for a therapeutic agent for a brain injury disease.

本発明の一実施例において、前記スクリーニング方法は、前記(c)段階で選別した候補物質が対照群に比べてHIF−1の発現または活性を抑制させるか否かを追加で分析する段階をさらに含む。   In one embodiment of the present invention, the screening method further includes the step of additionally analyzing whether the candidate substance selected in the step (c) suppresses the expression or activity of HIF-1 compared to the control group. Including.

本発明の一実施例において、前記(b)段階の測定及び/または前記HIF−1の発現または活性を分析する方法は、免疫組織化学染色、PCR、RT−PCR、ウエスタンブロット、ELISAまたはタンパク質チップで構成された群から選ばれる方法で行うが、これに限定されない。   In one embodiment of the present invention, the method of measuring the step (b) and / or analyzing the expression or activity of the HIF-1 is performed by immunohistochemical staining, PCR, RT-PCR, Western blot, ELISA or protein chip. However, the method is not limited to this.

本発明の一実施例において、前記TIM−3が発現される細胞は、神経膠細胞(glial cell)であるが、これに限定されない。本発明の一実施例において、前記動物モデルは、低酸素虚血性(hypoxia−ischemia)脳損傷疾患モデルであるが、これに限定されない。   In one embodiment of the present invention, the cell expressing TIM-3 is a glial cell, but is not limited thereto. In one embodiment of the present invention, the animal model is a hypoxia-ischemia brain injury disease model, but is not limited thereto.

本発明が適用可能な脳損傷疾患の例としては、脳梗塞、脳卒中、低酸素性脳損傷、虚血性脳疾患、中風などがあるが、これに限定されない。本発明の一実施例によると、前記脳損傷疾患は、低酸素(hypoxia)環境で発生した炎症(inflammation)関連の脳損傷である。   Examples of brain injury diseases to which the present invention can be applied include, but are not limited to, cerebral infarction, stroke, hypoxic brain injury, ischemic brain disease, and medium wind. According to one embodiment of the present invention, the brain injury disease is an inflammation-related brain injury that occurs in a hypoxia environment.

本発明者らは、虚血によって発生する低酸素(hypoxia)状態時に誘発する脳損傷でTIM−3タンパク質がモジュレータとして役割をし、TIM−3の発現が低酸素状態で発生する遺伝子発現を調節するHIF−1によって調節を受けることを確認した。そこで、本発明は、低酸素症が伴われる脳神経系疾患、例えば、脳梗塞、脳卒中、低酸素性脳損傷、虚血性脳疾患及び中風疾患の治療及び予防のために有用に使用されることができる。   The present inventors have the role of TIM-3 protein as a modulator in brain injury induced by hypoxia caused by ischemia, and the expression of TIM-3 regulates gene expression generated in hypoxia To be regulated by HIF-1. Therefore, the present invention may be usefully used for the treatment and prevention of cranial nervous system diseases associated with hypoxia, for example, cerebral infarction, stroke, hypoxic brain injury, ischemic brain disease and medium wind disease. it can.

TIM−3が低酸素虚血性脳卒中のマウスモデルの低酸素が誘導された脳領域で発現することを示す。(a)TIM−3転写レベルを低酸素虚血性脳卒中の誘導24時間後に、マウスモデルから対側性皮質(C,boxed region)と脳卒中が誘導された同側性皮質(I,boxed region)から脳組織を取り外して測定した。逆転写酵素遺伝子の増幅方法によって出た結果は、image Jプログラムを使用して量を測定し、actinの発現を反映して定量化した。HIF−1α転写レベルは、低酸素のpositive controlで表した。右側パネルは、低酸素虚血性脳卒中誘導のマウスモデルの脳断面組織のTTC染色を示す。(b)ウェスタンブロット分析法は、TIM−3とHIF−1タンパク質の発現を示す(n=3)。相対的なTIM−3発現程度は、独立した3回の実験から測定された有意値で示した。(c)低酸素虚血性脳卒中のマウス脳断面の同側性皮質と対側性皮質でTIM−3発現をTim−3抗体を使用して免疫組織化学法を通じて確認し、TIM−3を発現する細胞数をmm当たり個数で測定した。(d)免疫組織化学法は、低酸素虚血性脳卒中が誘導されたマウスで脳切片を取り外してTIM−3とhypoxyprobe−1(red、to detect hypoxic regions)抗体を使用して測定したものである。Scale bars、50μm(×20);50μm(×40).(e,f)グループごとに3匹から同側性部位と対側性部位から脳細胞を分離し、星状細胞を表すことができるGFAP(f)と小膠細胞を表すIba−1(e)、そしてTIM−3抗体を一斉に付けてFACS(蛍光利用細胞分類機)を使用して分析した。この結果は、ゲーティングされた小膠細胞で相対的なTIM−3レベルを表したもので、3回の独立した実験を通じて確認された。FIG. 5 shows that TIM-3 is expressed in hypoxia-induced brain regions of a mouse model of hypoxic ischemic stroke. (A) TIM-3 transcription level from 24 hours after induction of hypoxic-ischemic stroke, from contralateral cortex (C, boxed region) from mouse model and from ipsilateral cortex (I, boxed region) from which stroke was induced Brain tissue was removed and measured. The results obtained by the reverse transcriptase gene amplification method were measured using the image J program and quantified to reflect the expression of actin. HIF-1α transcription levels were expressed as a hypoxic positive control. The right panel shows TTC staining of brain cross-sectional tissue of a mouse model of hypoxic ischemic stroke induction. (B) Western blot analysis shows expression of TIM-3 and HIF-1 proteins (n = 3). The relative degree of TIM-3 expression was expressed as a significant value measured from three independent experiments. (C) TIM-3 expression is confirmed through immunohistochemistry using Tim-3 antibody in the ipsilateral cortex and contralateral cortex of the hypoxic-ischemic stroke mouse brain cross section and expresses TIM-3 The number of cells was measured by number per mm 2 . (D) The immunohistochemical method was measured using a TIM-3 and a hypoprobe-1 (red, to detect hypoxic regions) antibody after removing a brain section from a mouse in which hypoxic ischemic stroke was induced. . Scale bars, 50 μm (× 20); 50 μm (× 40). (E, f) Brain cells were separated from ipsilateral and contralateral sites from 3 animals per group, and GFAP (f) capable of representing astrocytes and Iba-1 (e ), And TIM-3 antibody was added all at once and analyzed using a FACS (fluorescence-based cell sorter). This result represented relative TIM-3 levels in gated microglia and was confirmed through three independent experiments. 一次培養した神経膠細胞において、HIF−1αがTIM−3のプロモータに結合し、その発現を調節することを示す。(a)BV2細胞に20%のOまたは1%のO環境を24時間維持した後、TIM−3の細胞表面発現様相をPEが結合されたTIM−3抗体で染色し、蛍光利用細胞分類機を使用して分析した。3回の独立した実験を通じて得た結果を棒グラフとして表し、平均値の変化(±s.d.)を正常環境サンプルと比較して表した。(b)マウスから培養した神経膠細胞を24時間の間正常環境と低酸素環境で培養し、その細胞をTim−3抗体を使用して兔疫細胞化学法を通じてTIM−3の発現を確認した。(c、d)マウスから一次培養した神経膠細胞と神経細胞を24時間の間正常環境と低酸素環境で培養し、逆転写酵素遺伝子増幅方法でTIM−3とactinの発現程度を測定した。発現変化を独立して3回繰り返した実験を通じて平均値変化(±s.d.)をグラフで示す(NS、統計が有効でない、Student−Newman−Keuls test)。(e)マウスから由来した神経膠細胞を24時間の間正常環境と低酸素環境で培養し、免疫沈降精製法をHIF−1抗体と対照群IgGを持って行った。結果は、3回の独立した実験を通じてグラフで示した。(f)HIF−1α+f/+fマウスから由来した神経膠細胞にAd−GFP or Ad−Cre/GFPウイルスを感染させ、この感染された細胞にTIM−3−luciferase reporter(Tim−3プロモータ遺伝子が入っているvector)constructsを形質注入し、24時間の間正常環境と低酸素環境で培養した。プロモータ遺伝子発現活性をratio of luciferase activity/β−galactosidase activityとして表現した。(g、h)逆転写酵素遺伝子増幅方法(g)とwestern blot分析法(h)は、primerと抗体を使用して24時間の間正常環境と低酸素環境下で行った。このデータは、独立して3回の繰り返した実験を通じて表した。グラフは、低酸素環境化でAd−GFPが感染された細胞に比べてTIM−3遺伝子転写とタンパク質の発現量を比べて示す。IP、免疫沈降FIG. 5 shows that HIF-1α binds to the TIM-3 promoter and regulates its expression in primary cultured glial cells. (A) BV2 cells are maintained in a 20% O 2 or 1% O 2 environment for 24 hours, and then the cell surface expression of TIM-3 is stained with PE-conjugated TIM-3 antibody to obtain fluorescence-using cells. Analysis was performed using a sorter. The results obtained through three independent experiments were represented as bar graphs, and the change in mean value (± sd) compared to normal environment samples. (B) Glial cells cultured from mice were cultured for 24 hours in a normal environment and a hypoxic environment, and the cells were confirmed for TIM-3 expression through a plague cytochemistry method using Tim-3 antibody. . (C, d) Glial cells and neurons primarily cultured from mice were cultured in normal and hypoxic environments for 24 hours, and the expression levels of TIM-3 and actin were measured by the reverse transcriptase gene amplification method. The mean value change (± sd) is shown graphically through experiments in which expression changes were repeated three times independently (NS, statistics not valid, Student-Newman-Keuls test). (E) Glial cells derived from mice were cultured in normal and hypoxic environments for 24 hours, and immunoprecipitation purification was carried out with HIF-1 antibody and control group IgG. The results are shown graphically through three independent experiments. (F) Glial cells derived from HIF-1α + f / + f mice were infected with Ad-GFP or Ad-Cre / GFP virus, and TIM-3-luciferase reporter (Tim-3 promoter gene was transferred to the infected cells). The containing vector) constructs were transfected and cultured in normal and hypoxic environments for 24 hours. Promoter gene expression activity was expressed as ratio of luciferase activity / β-galactosidase activity. (G, h) The reverse transcriptase gene amplification method (g) and the Western blot analysis method (h) were performed in a normal environment and a hypoxic environment for 24 hours using a primer and an antibody. This data was represented through three independent experiments independently. The graph shows TIM-3 gene transcription and protein expression levels compared to cells infected with Ad-GFP in a hypoxic environment. IP, immunoprecipitation TIM−3の遮断は、低酸素虚血性脳卒中後に誘導される脳損傷を有意味に減少させることを示す。(a)IgG(n=12)とTIM−3 blocking抗体(n=12)100μgを処理した低酸素虚血性脳卒中モデルマウスからTTC染色された脳切片のイメージを示す代表図。梗塞volumeは、image Jプログラムを通じて分析し、損傷された同側性部位を百分率で表現した。(b)低酸素虚血性脳卒中の誘導24時間後、TIM−3抗体を処理したマウス(n=4)とIgG(n=4)を処理したマウスから得たMRI(磁気共鳴映像法)の代表写真。(c)低酸素虚血性脳卒中の誘導24時間後、TIM−3抗体を処理したマウス(n=4)とIgG(n=4)を処理したマウスから得たT2−MRI(磁気共鳴映像法)の代表写真。(d)浮腫の形成程度をT2−weighted MRI images and ADC mapを通じて得た。(e)低酸素虚血性脳卒中の誘導24時間後、TIM−3抗体を処理したマウスと、IgGを処理したマウスから得た脳切片中でNeuN(神経細胞)cleaved caspase−3(細胞死滅をdetectionする抗体)で免疫組織化学法をし、confocal顕微鏡で測定した代表写真。Scale bar、50μm。グラフは、NeuNとcleaved caspase−3抗体で染色された細胞数をmm当たり平均個数を示す。(f)低酸素虚血性脳卒中の誘導24時間後、TIM−3抗体を処理したマウスと、IgGを処理したマウスから得た対側性皮質と同側性皮質の中でfull−length PARPタンパク質(細胞死滅を表すタンパク質)の発現を表したwestern blot写真。グラフは、full−length PARPのレベルを比べて示す。全てのデータは、3回の独立した実験から有意な値で表した。TIM-3 blockade indicates a significant reduction in brain damage induced after hypoxic-ischemic stroke. (A) Representative view showing an image of a TTC-stained brain section from a hypoxic-ischemic stroke model mouse treated with IgG (n = 12) and 100 μg of TIM-3 blocking antibody (n = 12). Infarct volume was analyzed through the image J program and the damaged ipsilateral site expressed as a percentage. (B) Representative of MRI (magnetic resonance imaging) obtained from mice treated with TIM-3 antibody (n = 4) and IgG (n = 4) 24 hours after induction of hypoxic-ischemic stroke Photo. (C) T2-MRI (magnetic resonance imaging) obtained from mice treated with TIM-3 antibody (n = 4) and IgG (n = 4) 24 hours after induction of hypoxic ischemic stroke Representative photo. (D) The degree of edema formation was obtained through T2-weighted MRI images and ADC map. (E) 24 hours after induction of hypoxic-ischemic stroke, NeuN (neuronal cell) cleaved caspase-3 (detection of cell death was detected in brain sections obtained from mice treated with TIM-3 antibody and mice treated with IgG. Representative photograph measured by a confocal microscope after immunohistochemical method. Scale bar, 50 μm. The graph shows the average number of cells per mm 2 stained with NeuN and the cleaved caspase-3 antibody. (F) 24 hours after induction of hypoxic-ischemic stroke, the full-length PARP protein (in the contralateral and ipsilateral cortex obtained from mice treated with TIM-3 antibody and mice treated with IgG ( Western blot photograph showing the expression of a protein representing cell death). The graph shows the level of full-length PARP in comparison. All data were expressed as significant values from three independent experiments. TIM−3遮断抗体が好中球の移動を減少させることを示す。逆転写酵素遺伝子増幅方法(a)及びウエスタンブロット分析法(b)をIgGとTIM−3 blocking抗体が処理された低酸素虚血性脳卒中モデルマウスでMPO発現を測定するために使用した。グラフは、MPOレベルを比べて示した。(c)低酸素虚血性脳卒中の誘導24時間後、TIM−3抗体を処理したマウスと、IgGを処理したマウスから得た脳切片中からMPOとGr−1抗体で免疫組織化学法をし、confocal顕微鏡で測定した代表写真。Scale bar,50μm。グラフは、MPOとGr−1抗体で染色された細胞数のmm当たり平均個数を示す(±s.d.)。低酸素虚血性脳卒中モデルマウスから由来した(d)大脳皮質と(e)基底部位で得た脳切片をMPO抗体とGr−1抗体を使用して免疫組織化学法を行い、MPOとGr−1が染色された細胞をmm当たり細胞数を数えた。FIG. 5 shows that TIM-3 blocking antibodies reduce neutrophil migration. Reverse transcriptase gene amplification method (a) and Western blot analysis method (b) were used to measure MPO expression in hypoxic ischemic stroke model mice treated with IgG and TIM-3 blocking antibodies. The graph shows the MPO level compared. (C) 24 hours after induction of hypoxic ischemic stroke, immunohistochemistry was performed with MPO and Gr-1 antibodies from brain sections obtained from mice treated with TIM-3 antibody and mice treated with IgG, Representative photograph measured with a confocal microscope. Scale bar, 50 μm. The graph shows the average number of cells stained with MPO and Gr-1 antibody per mm 2 (± sd). (D) Cerebral cortex derived from hypoxic-ischemic stroke model mice and (e) Brain sections obtained at the basal site were subjected to immunohistochemistry using MPO and Gr-1 antibodies, and MPO and Gr-1 The number of cells stained per mm 2 was counted. 生体内と生体外の両方でTIM−3の遮断は、代表的な二つの好中球化学走性因子の発現を減少させることを示す。(a)マウスから一次培養した神経膠細胞(2×10)をトランスウェルのlow chamberに敷いて、TIM−3と対照群IgG抗体を図示のように先ず処理した後、脾臓細胞5×10をupper chamberに載せる。低酸素状況で24時間培養後、脾臓細胞low chamberに移動された程度を蛍光利用細胞分類機を使用して分析した。独立した3回の実験を通じてlow chamberに移動したGr−1highCD11bhigh細胞の%を平均値±s.dで表現した。(b)Gr−1highCD11bhighである好中球をC57BL/6マウスの骨髄から分離し、IgGとTIM−3抗体を処理した神経膠細胞と共に低酸素環境で培養する。3回の独立した実験から得た結果は、IgGを処理した細胞を1と見た時、減少された程度を表す。(c)逆転写酵素遺伝子増幅法をIgGとTIM−3を処理した低酸素虚血性脳卒中モデルから得た組織に対して行った。(d)グラフは、actinで補正して表した結果を示す(n=3)。(e)マウスから由来した神経膠細胞にIgGとTIM−3抗体を処理し、正常環境と低酸素環境で24時間の間培養する。CXCL1とIL−1beta転写レベルは、逆転写酵素遺伝子増幅法によって決めた。グラフは、独立して3回実験した結果から出た。NS、有効性がない。Blocking TIM-3 both in vivo and in vitro shows that the expression of two representative neutrophil chemotaxis factors is reduced. (A) Glial cells (2 × 10 5 ) primarily cultured from mice were laid on a transchamber low chamber, and TIM-3 and control group IgG antibody were first treated as shown, followed by spleen cells 5 × 10 5 is placed on the upper chamber. After culturing for 24 hours in a hypoxic condition, the degree of migration to spleen cell low chamber was analyzed using a fluorescence-based cell sorter. The percentage of Gr-1 high CD11b high cells that migrated to the low chamber through three independent experiments was the mean ± s. Expressed with d. (B) Gr-1 high CD11b high neutrophils are isolated from bone marrow of C57BL / 6 mice and cultured in a hypoxic environment together with gliocytes treated with IgG and TIM-3 antibody. The results obtained from three independent experiments represent the degree of reduction when IgG treated cells are viewed as 1. (C) Reverse transcriptase gene amplification was performed on tissues obtained from a hypoxic-ischemic stroke model treated with IgG and TIM-3. (D) The graph shows the results corrected with actin (n = 3). (E) Glial cells derived from mice are treated with IgG and TIM-3 antibody and cultured for 24 hours in normal and hypoxic environments. CXCL1 and IL-1beta transcription levels were determined by reverse transcriptase gene amplification. The graph is derived from the results of three independent experiments. NS, not effective. 低酸素によって誘導される好中球の移動は、HIF−1が欠けた環境で減少することを示す。(a)HIF−1α+f/+fマウスから一次培養した神経膠細胞(2×10)にAd−GFP or Ad−Cre/GFPウイルスを感染させ、トランスウェルのlow chamberに載せ、5×10をupper chamberに載せる。低酸素状況で24時間培養後、脾臓細胞がlow chamberに移動した程度を蛍光利用細胞分類機を使用して分析した。(b)Gr−1highCD11bhighである好中球をC57BL/6マウスの骨髄から分離し、HIF−1α+f/+fマウスから一次培養した神経膠細胞をAd−GFPまたはAd−Cre/GFPウイルスで感染させて好中球細胞のように低酸素環境で培養する。3回の独立した実験を通じて得た結果は、Ad−GFP感染されたHIF−1α+f/+fマウスの神経膠細胞と比べて好中球細胞の移動変化を示した。(c)CXCL1とIL−1beta転写レベルは、Ad−GFPまたはAd−Cre/GFPウイルスを感染させた神経膠細胞を正常環境と低酸素環境で24時間の間培養してその発現を確認した。(d)グラフは、real−time quantitative PCRから得た結果である。Hypoxia-induced neutrophil migration is shown to decrease in an environment devoid of HIF-1. (A) Glial cells (2 × 10 5 ) primarily cultured from HIF-1α + f / + f mice were infected with Ad-GFP or Ad-Cre / GFP virus, and placed on the low well of the transwell, 5 × 10 5 Is placed on the upper chamber. After culturing for 24 hours under hypoxia, the extent to which spleen cells migrated to low chamber was analyzed using a fluorescence-based cell sorter. (B) Gr-1 high CD11b high neutrophils were isolated from bone marrow of C57BL / 6 mice, and glial cells primarily cultured from HIF-1α + f / + f mice were subjected to Ad-GFP or Ad-Cre / GFP virus. And incubate in a hypoxic environment like neutrophil cells. The results obtained through three independent experiments showed altered migration of neutrophil cells compared to glial cells from HIF-1α + f / + f mice infected with Ad-GFP. (C) CXCL1 and IL-1beta transcript levels were confirmed by culturing glial cells infected with Ad-GFP or Ad-Cre / GFP virus in normal and hypoxic environments for 24 hours. (D) The graph is the result obtained from real-time quantitative PCR. LysM−Hif−1α−/−形質転換マウスで低酸素虚血性脳卒中によって誘導された脳損傷が減少することを示す実験結果である。(a)HIF−1α+f/+fまたはLysM−Hif−1α−/−マウスから培養した神経膠細胞で表示されたプライマーを利用して逆転写酵素遺伝子増幅法を行った。(b)TIM−3遺伝子転写レベルをHIF−1α+f/+f or LysM−Hif−1α−/−マウス(n=3)の対側性皮質と虚血性脳卒中が誘導された同側性皮質から由来した脳組織から確認した。(c)24時間の間低酸素虚血性脳卒中が誘導されたHIF−1α+f/+f(n=12)or LysM−Hif−1α−/−マウス(n=12)からTTC染色された脳切片のイメージを示した代表図。梗塞volumeは、image Jプログラムを通じて分析し、損傷された同側性部位を百分率で表現した。(d)低酸素脳卒中24時間後、HIF−1α+f/+f or LysM−Hif−1α−/−マウスから得た脳切片中でNeuN(神経細胞をdetectionする抗体)cleaved caspase−3(細胞死滅をdetectionする抗体)で免疫組織化学法をし、confocal顕微鏡で測定した代表写真。Scale bar,50μm。グラフは、NeuNとcleaved caspase−3抗体で染色された細胞数のmm当たり平均個数を示す。(±s.d.3回独立して実験)It is an experimental result which shows that the brain damage induced | guided | derived by the hypoxic-ischemic-stroke is reduced in a LysM-Hif-1 (alpha) <-/-> transformed mouse. (A) Reverse transcriptase gene amplification was performed using primers displayed on glial cells cultured from HIF-1α + f / + f or LysM-Hif-1α − / − mice. (B) TIM-3 gene transcription levels from HIF-1α + f / + f or LysM-Hif-1 α − / − mice (n = 3) contralateral cortex and ipsilateral cortex in which ischemic stroke was induced Confirmed from the derived brain tissue. (C) TTC-stained brain section from HIF-1α + f / + f (n = 12) or LysM-Hif-1α − / − mice (n = 12) in which hypoxic ischemic stroke was induced for 24 hours A representative diagram showing an image. Infarct volume was analyzed through the image J program and the damaged ipsilateral site expressed as a percentage. (D) 24 hours after hypoxic stroke, NeuN (an antibody that detects nerve cells) cleaved caspase-3 (cell death) in brain slices obtained from HIF-1α + f / + for LysM-Hif-1α − / − mice A representative photograph measured by a confocal microscope after immunohistochemistry with a detection antibody). Scale bar, 50 μm. The graph shows the average number of cells per mm 2 stained with NeuN and the cleaved caspase-3 antibody. (± sd 3 independent experiments) LysM−Hif−1a−/−マウスにLV−TIM3−GFPの頭蓋腔接種は、脳梗塞範囲と神経学的後遺症を増加させることを示す実験結果である。(a)IVI spectrum system(Xenogen IVIS−200)を使用してPBS、GFPが過発現されるレンチウイルスを接種したマウス、そして、TIM−3とGFPが過発現されたレンチウイルスを接種したマウスの蛍光イメージを測定した代表図(excitation filter,from 445 to 490nm、and emission filter,from 515 to 575nm)。(b)LV−TIM3−GFPまたはLV−GFPを接種したマウスからTTC染色された脳切片のイメージを示した代表図。(c、d)梗塞大きさ(c、n=6 for LV−GFP or n=5 for LV−TIM3−GFP)and 神経学的後遺症(d、n=6 for each group)は、低酸素虚血性脳卒中の誘導24時間後に検査した。Experimental results show that LV-TIM3-GFP inoculation of LysM-Hif-1a-/-mice increases cerebral infarction area and neurological sequelae. (A) Using an IVI spectrum system (Xenogen IVIS-200), mice inoculated with lentivirus overexpressing PBS, GFP, and mice inoculated with lentivirus overexpressing TIM-3 and GFP The representative figure which measured the fluorescence image (excitation filter, from 445 to 490 nm, and emission filter, from 515 to 575 nm). (B) Representative view showing an image of a brain section stained with TTC from a mouse inoculated with LV-TIM3-GFP or LV-GFP. (C, d) Infarct size (c, n = 6 for LV-GFP or n = 5 for LV-TIM3-GFP) and neurological sequelae (d, n = 6 for reach group) are hypoxic-ischemic Examined 24 hours after induction of stroke. 低酸素脳環境で発生可能なTIM−3関連事件の模式図である。低酸素環境依存的HIF−1aの活性は、小膠細胞と星状細胞でTIM−3発現を増加させる。HIF−1/TIM−3軸の活性化は、好中球誘引物質の生成と低酸素地域で好中球の浸潤を誘導する。好中球の非正常的な浸潤現象は、過度な炎症反応を誘導し、引き継いで脳の病態生理学的環境に原因となる。It is a schematic diagram of a TIM-3 related event that can occur in a hypoxic brain environment. Hypoxia-dependent HIF-1a activity increases TIM-3 expression in microglia and astrocytes. Activation of the HIF-1 / TIM-3 axis induces neutrophil attractant production and neutrophil infiltration in hypoxic areas. Unusual neutrophil infiltration phenomena induce an excessive inflammatory response, which in turn is responsible for the pathophysiological environment of the brain. 1%の酸素条件及び20%の酸素条件下で一次培養膠細胞またはBV2小膠細胞にTIM−3のshRNAをそれぞれ細胞感染させた後、PCR及び免疫細胞化学法を通じて細胞内でTIM−3の発現程度を比較分析した結果を示し、10Aは、一次培養膠細胞の結果を、10Bは、BV2小膠細胞の結果を示し、10AにおけるaはPCR分析結果であり、bは免疫細胞化学法を表す。TIM-3 shRNA was infected with primary culture cells or BV2 microglia cells under 1% oxygen condition and 20% oxygen condition, respectively. 10A shows the result of primary cultured glial cells, 10B shows the result of BV2 microglia cells, 10A shows the results of PCR analysis, and b shows the results of immunocytochemistry. Represent.

本発明は、虚血性脳卒中など低酸素症による脳損傷疾患の治療用組成物及び脳損傷疾患治療剤のスクリーニング方法に関し、具体的には、TIM(T−cell immunoglobulin and mucin domain protein)−3抑制剤を有効成分として含む脳損傷疾患の治療用組成物、及び(a)TIM−3が発現される細胞または動物に候補物質を処理する段階と、(b)TIM−3の発現または活性程度を測定する段階と、(c)TIM−3の発現または活性程度が候補物質を処理しない対照群に比べて減少した候補物質を選別する段階とを含む脳損傷疾患治療剤のスクリーニング方法に関する。   TECHNICAL FIELD The present invention relates to a composition for treating brain injury diseases caused by hypoxia such as ischemic stroke and a screening method for a therapeutic agent for brain injury diseases, and specifically, TIM (T-cell immunoglobulin and mucin domain protein) -3 suppression. A composition for treating a brain injury disease comprising an agent as an active ingredient, and (a) a step of treating a candidate substance in a cell or animal in which TIM-3 is expressed, and (b) a level of expression or activity of TIM-3. The present invention relates to a screening method for a therapeutic agent for a brain injury disease, comprising a step of measuring, and (c) selecting a candidate substance having a decreased expression or activity level of TIM-3 compared to a control group not treated with the candidate substance.

大脳虚血(cerebral ischaemia)は、一連の病態生理学的変化を引き起こして脳損傷を誘発する。炎症媒介体(inflammatory mediator)の生産及び浸透は、脳損傷を引き起こす重要な段階で、大脳虚血による脳損傷程度は、炎症状態と非常に密接な関連があることを示唆する臨床及び研究結果が増加している。そこで、炎症調節をターゲットとする脳神経系疾患の治療剤の開発戦略に対する関心が高まっている。但し、現在までは、虚血性脳疾患時に伴われる炎症反応について知られた情報が非常に少ないという限界があった。   Cerebral ischemia causes a series of pathophysiological changes and induces brain damage. Inflammatory mediator production and penetration is an important step in causing brain damage, and clinical and research results suggest that the degree of brain damage due to cerebral ischemia is very closely related to the inflammatory condition. It has increased. Therefore, there is a growing interest in strategies for developing therapeutic agents for cranial nervous system diseases that target inflammation control. To date, however, there has been a limit that very little information is known about the inflammatory response associated with ischemic brain disease.

本発明は、虚血以後に発生するhypoxia(低酸素)状態による脳損傷に、TIM−3が非正常的な炎症細胞の浸透及び炎症反応と連関があり、TIM−3の制御は、炎症反応、脳細胞の死滅、脳梗塞部位の減少に影響を与えるということを糾明することにその特徴がある。本発明は、虚血によって発生する低酸素(hypoxia)状態時に誘発する脳損傷で、TIM−3タンパク質がモジュレータとしての役割をしており、TIM−3の発現が低酸素状態で発生する遺伝子発現を調節するHIF−1によって調節を受けるという研究結果を基盤としている。本発明の一実施例によると、低酸素虚血性脳卒中のマウスモデル(Hypoxia−ischemia mouse model)の低酸素が誘導された脳領域の神経膠細胞(glial cell)でTIM−3の発現は増加し(図1)、TIM−3の発現は、HIF−1によって調節された(図2)。また、TIM−3の遮断は、低酸素虚血症後に伴われる脳梗塞部位及び脳細胞の死滅を減少させ(図3)、好中球の脳への移動及び移動関連サイトカインを減少させることを確認した(図4)。また、低酸素によって誘導される好中球の移動及び脳損傷は、HIF−1欠乏マウスの低酸素虚血脳卒中モデルでも減少され(図6)、該マウスにTIM−3の発現を増加させれば脳損傷が再び増加した。このような結果は、低酸素環境でHIF−1/TIM−3軸と脳損傷の関連性を表す。   The present invention relates to brain injury caused by a hypoxia (hypoxia) state occurring after ischemia, which is associated with inflammatory cell penetration and inflammatory response, where TIM-3 is abnormal. It is characterized by demonstrating that it affects the death of brain cells and the reduction of cerebral infarction sites. The present invention relates to brain damage induced by hypoxia caused by ischemia, in which TIM-3 protein plays a role as a modulator, and TIM-3 expression is expressed in hypoxia. Based on the research results of being regulated by HIF-1 that regulates According to one embodiment of the present invention, expression of TIM-3 is increased in hypoxia-induced brain cells of a hypoxic-ischemic mouse model mouse model (Hypoxia-ischemia mouse model). (FIG. 1), TIM-3 expression was regulated by HIF-1 (FIG. 2). Also, TIM-3 blockade reduces cerebral infarction and brain cell death following hypoxic ischemia (FIG. 3), reduces neutrophil migration to the brain and migration-related cytokines. Confirmed (FIG. 4). Hypoxia-induced neutrophil migration and brain damage are also reduced in the hypoxic-ischemic stroke model of HIF-1-deficient mice (FIG. 6), which can increase TIM-3 expression in the mice. Brain damage increased again. Such a result represents an association between HIF-1 / TIM-3 axis and brain injury in a hypoxic environment.

従って、本発明は、TIM−3抑制剤を有効成分として含有する脳損傷疾患、例えば、脳梗塞、脳卒中、低酸素性脳損傷、虚血性脳疾患及び中風疾患の予防または治療用薬学的組成物を提供することができる。本明細書で使用された用語「予防」は、療法剤(例えば、予防剤または治療剤)または療法剤の組合物を投与して対象体で脳損傷疾患の兆候が表れるか再発または発展することを防ぐことを意味する。本明細書で使用された用語「治療」は、脳損傷疾患患者の症状やいずれか一つ以上の身体的パラメータを改善させるか調節するかその発生や進展を遅延させることを意味し、患者の認識有無は問わない。本発明の薬学的組成物は、一つ以上の薬剤学的に許容される担体、賦形剤または希釈剤を含む。前記担体、賦形剤及び希釈剤の例としては、ラクトース、デキストロース、スクロース、ソルビトール、マンニトール、キシリトール、エリスリトール、マルチトール、デンプン、アラビアガム、アルジネート、ゼラチン、カルシウムホスフェート、カルシウムシリケート、セルロース、メチルセルロース、ポリビニルピロリドン、水、メチルヒドロキシベンゾエート、プロピルヒドロキシベンゾエート、タルク、マグネシウムステアレート及び鉱物油が挙げられる。また、充填剤、抗凝集剤、滑剤、湿潤剤、香料、乳化剤及び防腐剤などをさらに含んでもよい。使用に適合した担体としては、食塩水、燐酸塩緩衝食塩水、最小必須培地(MEM)またはHEPES緩衝液のMEMを含む水性媒質を挙げられるが、これに限定されない。   Therefore, the present invention relates to a pharmaceutical composition for preventing or treating brain injury diseases, for example, cerebral infarction, stroke, hypoxic brain injury, ischemic brain disease and medium wind disease, comprising a TIM-3 inhibitor as an active ingredient. Can be provided. As used herein, the term “prophylaxis” refers to the administration of a therapeutic agent (eg, a prophylactic or therapeutic agent) or a combination of therapeutic agents that manifests, recurs, or develops in a subject with signs of brain injury. Means to prevent. As used herein, the term “treatment” means to improve or regulate symptoms or any one or more physical parameters of a patient with a brain injury disorder or delay its development or progression, It doesn't matter whether it is recognized or not. The pharmaceutical composition of the invention comprises one or more pharmaceutically acceptable carriers, excipients or diluents. Examples of the carriers, excipients and diluents include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, gum arabic, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methylcellulose, Polyvinyl pyrrolidone, water, methyl hydroxybenzoate, propyl hydroxybenzoate, talc, magnesium stearate and mineral oil. Further, it may further contain a filler, an anti-aggregating agent, a lubricant, a wetting agent, a fragrance, an emulsifier, an antiseptic and the like. Carriers suitable for use include, but are not limited to, aqueous media including saline, phosphate buffered saline, minimal essential medium (MEM) or MEM in HEPES buffer.

また、本発明の薬学的組成物は、哺乳動物に投与された後、活性成分の迅速、持続または遅延された放出を提供するように当業界に公知された方法を使用して剤形化することができる。剤形は、粉末、料粒、精製、エマルジョン、シロップ、エアゾル、軟質または硬質のゼラチンカプセル、滅菌注射溶液、滅菌粉末などの形態であってもよい。本発明の薬学的組成物は、筋肉、皮下、経皮、静脈、鼻腔内、腹腔内または経口の経路で投与されてもよく、好ましくは、筋肉内または皮下経路で投与される。組成物の投与量は、投与経路、動物の年齢、性別、体重及び重症度などの様々な因子によって適切に選択される。   In addition, the pharmaceutical compositions of the present invention are formulated using methods known in the art to provide rapid, sustained or delayed release of the active ingredient after administration to a mammal. be able to. Dosage forms may be in the form of powders, granules, refined, emulsions, syrups, aerosols, soft or hard gelatin capsules, sterile injectable solutions, sterile powders and the like. The pharmaceutical compositions of the present invention may be administered by intramuscular, subcutaneous, transdermal, intravenous, intranasal, intraperitoneal or oral route, preferably by intramuscular or subcutaneous route. The dosage of the composition is appropriately selected according to various factors such as the administration route, the age, sex, weight and severity of the animal.

本発明の薬学的組成物は、下記の多様な経口または非経口投与形態で剤形化するが、これに限定されない。先ず、経口投与のための固形製剤としては、錠剤、丸剤、散剤、顆粒剤、硬質または軟質カプセル剤などが含まれ、このような固形製剤は、本発明の有効成分に少なくとも一つ以上の賦形剤を交ぜて調剤される。また、単純な賦形剤の他にマグネシウムステアレート、タルクのような滑剤を使用してもよい。経口投与のための液状製剤としては、懸濁剤、内用液剤、乳剤またはシロップ剤などがあるが、よく使用される単純希釈剤である水、リキッドパラフィンの他に様々な賦形剤が含まれてもよい。また、本発明の薬学的組成物は、非経口投与も可能であり、非経口投与は、皮下注射剤、静脈注射剤、筋肉内注射剤または胸部内注射剤を注入する方法などによる。この場合、非経口投与用剤型に製剤化するために、本発明の有効成分を安定剤または緩衝制と共に水で混合して溶液または懸濁液に製造し、これをアンプルまたはバイアルの単位投与型で製造することができる。非経口投与のための製剤としては、滅菌した水溶液、非水性溶剤、懸濁液剤、乳剤、凍結乾燥製剤または坐剤などが含まれる。非水性溶剤、懸濁液剤としては、プロピレングリコール(propylene glycol)、ポリエチレングリコール、オリーブオイルのような植物性油またはエチルオレートのような注射可能なエステルなどが使用されてもよい。また、本発明の薬学的組成物は、マウス、ラット、家畜、人間などの哺乳動物に多様な経路で投与されてもよく、その例としては、経口、直腸、静脈、筋肉、皮下、子宮内硬膜または脳血管内注射などがある。本発明の薬学的組成物は、患者の年、性別、体重によって適切な方法を選択して投与する。   The pharmaceutical composition of the present invention is formulated in the following various oral or parenteral dosage forms, but is not limited thereto. First, solid preparations for oral administration include tablets, pills, powders, granules, hard or soft capsules, and such solid preparations contain at least one or more active ingredients of the present invention. Formulated with excipients. In addition to simple excipients, lubricants such as magnesium stearate and talc may be used. Liquid preparations for oral administration include suspensions, solutions for internal use, emulsions or syrups, but include various excipients in addition to commonly used simple diluents such as water and liquid paraffin. May be. The pharmaceutical composition of the present invention can also be administered parenterally, and parenteral administration is based on a method of injecting a subcutaneous injection, an intravenous injection, an intramuscular injection, or an intrathoracic injection. In this case, in order to formulate into a dosage form for parenteral administration, the active ingredient of the present invention is mixed with water together with a stabilizer or a buffer to produce a solution or suspension, which is then administered in unit of ampoules or vials. Can be manufactured in molds. Preparations for parenteral administration include sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, lyophilized formulations or suppositories. As the non-aqueous solvent or suspension, propylene glycol (polypropylene glycol), polyethylene glycol, vegetable oils such as olive oil, or injectable esters such as ethyl oleate may be used. In addition, the pharmaceutical composition of the present invention may be administered to mammals such as mice, rats, domestic animals and humans by various routes, and examples thereof include oral, rectal, intravenous, muscle, subcutaneous, intrauterine. For example, dura mater or intracerebral intravascular injection. The pharmaceutical composition of the present invention is administered by selecting an appropriate method according to the patient's year, sex and weight.

以下、本発明を実施例によってさらに詳しく説明する。下記の実施例は、単に本発明をより具体的に説明するためのもので、本発明の範囲がこれらの実施例に限らないということは、当業界で通常の知識を持った者において自明である。   Hereinafter, the present invention will be described in more detail with reference to examples. The following examples are merely for explaining the present invention more specifically, and it is obvious to those skilled in the art that the scope of the present invention is not limited to these examples. is there.

実験材料及び方法
<1−1>実験動物
Randall Johnson博士が製作したHIF−1α+f/+f(HIF−1α−floxed alleles)を持ったマウスを使用した。骨髄系統細胞でHIF−1αに欠けたマウスは、HIF−1α+f/+fマウスとLysM−Cre形質転換マウスの異種交配から製作した(非特許文献2)。8週令の雄C57BL/6マウス(Orient Bio)を生体内(in vivo)及び試験管内(in vitro)実験に使用した。
Experimental Materials and Methods <1-1> Experimental Animals Mice with HIF-1α + f / + f (HIF-1α-floxed alleles) produced by Dr. Randall Johnson were used. Bone marrow lineage cells lacking HIF-1α were produced from crossbreeding of HIF-1α + f / + f mice and LysM-Cre transformed mice (Non-patent Document 2). Eight week old male C57BL / 6 mice (Orient Bio) were used for in vivo and in vitro experiments.

<1−2>低酸素性脳虚血症モデル及び梗塞(infarct)の体積の測定
C57BL/6雄マウス(8週、Orient Bio)に対して非特許文献3のような方法でH/Iを誘導した。簡単に説明すると、マウスをZoletil(Virbac)及びRompun(Bayer)(4:1)で麻酔させ、それぞれのマウスの右側総頸動脈(common carotid artery)を露出させ、4−0手術用シルク(surgical silk)で二重接合した。切開部位を縫合し、過量の食べ物と水で2時間の間マウスを回復させた。全身性低酸素症(systemic hypoxia)は、温度調節低酸素チャンバ(BioSpherix、C−474)で8%の酸素/バランスN2に露出させて誘導した。このような一時的一側脳虚血症(transient unilateral cerebral ischaemia)モデルは、同側半球(ipsilateral hemisphere)で再生可能な脳損傷を発生させるが、対側半球(contralateral hemisphere)では発生させない。TIM−3−抑制(blocking)実験のために、H/I30分後にマウスに100μgのラット(rat)IgG2a、k isotype(eBioscience、16−4321)または抗TIM−3モノクロニル抗体(eBioscience、RMT−3−23)を静脈注射した。H/Iの24時間が経った後マウスを殺した後、脳を除去し、直ちに2mm厚さのセクションで切った後、TTCと共に37℃で30分間培養した。前記セクションのイメージは、カメラが装着された立体顕微鏡(Zeiss、Stereo Discovery.V20)で観察した。梗塞(infarct)体積は、梗塞組織の浮腫に対して償う間接的な方式で測定し、半球面積に対する損傷面積の割合の百分率で計算し、浮腫による半球の膨潤(swelling)は補正された。梗塞体積の計算式は、以下の通りである(非特許文献4):
梗塞体積(Infarct volume)(%)=[(対側性半球−同側性半球の健康な領域)/対側性半球]×100
<1-2> Measurement of volume of hypoxic cerebral ischemia model and infarction (infarct) H / I was measured for C57BL / 6 male mice (8 weeks, Orient Bio) by the method as described in Non-Patent Document 3. Induced. Briefly, mice are anesthetized with Zoletil (Virbac) and Rompun (Bayer) (4: 1) to expose the right common carotid artery of each mouse and 4-0 surgical silk. (silk). The incision site was sutured and the mice were allowed to recover for 2 hours with excess food and water. Systemic hypoxia was induced by exposure to 8% oxygen / balance N2 in a temperature controlled hypoxia chamber (BioSpherix, C-474). Such a transient unilateral cerebral ischemia model produces reproducible brain damage in the ipsilateral hemisphere, but not in the contralateral hemisphere. For TIM-3-blocking experiments, mice were treated with 100 μg rat IgG2a, kisotype (eBioscience, 16-4321) or anti-TIM-3 monoclonal antibody (eBioscience, RMT-) 30 min after H / I. 3-23) was injected intravenously. After 24 hours of H / I, the mice were killed, the brains were removed, immediately cut into 2 mm thick sections, and incubated with TTC at 37 ° C. for 30 minutes. The image of the section was observed with a stereo microscope (Zeiss, Stereo Discovery. V20) equipped with a camera. Infarct volume was measured in an indirect manner to compensate for edema of the infarcted tissue, calculated as a percentage of the ratio of damaged area to hemispheric area, and corrected for hemispheric swelling due to edema. The formula for calculating the infarct volume is as follows (Non-patent Document 4):
Infarct volume (%) = [(contralateral hemisphere-healthy area of ipsilateral hemisphere) / contralateral hemisphere] × 100

<1−3>磁気共鳴映像の測定(Magnetic resonance imaging assessments)
マウスを動物ベッドに固定させ、MRI測定装備(Bruker7T BioSpec)下に位置させた後、映像測定の間麻酔させる。Relaxation Enhancement sequenceを持ったRapid Acquisitionを利用してT2−加重された(weighted)イメージを得た。0.7mm厚さの18個の隣接軸スライスを得た[matrix256×256;field of view=20×20mm;TR(Repetition Time)=2,500ms;TE(Echo Time)=35ms;acquisition time=4分;no gap]。ADC(apparent diffusion coefficient)マップは、スピン−エコーシーケンスを利用して拡散加重された(diffusion−weighted)イメージによって得た。このため、8個の隣接軸イメージを得た[thickness 0.7mm、matrix256×128、field of view=20×20mm、TR=2,000ms、TE=26.936ms、acquisition time=16分、1average、b values=45,350、mm当たり1,000及び2,000s、no gap]。ADCマップは、スキャナで得た。浮腫体積は、T2−加重されたイメージから得、ADCマップは、Image J analyserから得た。浮腫体積(Oedema volume)(%)=[(同側性体積−対側性体積)/対側性体積]×100。
<1-3> Measurement of magnetic resonance imaging (Magnetic resonance imaging)
Mice are fixed to the animal bed and placed under the MRI measurement equipment (Bruker7T BioSpec) and then anesthetized during the video measurement. A T2-weighted image was obtained using Rapid Acquisition with a Relaxation Enhancement sequence. 18 adjacent axial slices of 0.7 mm thickness were obtained [matrix 256 × 256; field of view = 20 × 20 mm; TR (Repetition Time) = 2,500 ms; TE (Echo Time) = 35 ms; acquisition time = 4 Min; no gap]. The ADC (approximate diffusion coefficient) map was obtained by a diffusion-weighted image using a spin-echo sequence. For this reason, eight adjacent axis images were obtained [thickness 0.7 mm, matrix 256 × 128, field of view = 20 × 20 mm, TR = 2,000 ms, TE = 26.936 ms, acquisition time = 16 minutes, 1 average, b values = 45,350, 1,000 and 2,000 s per mm 2 , no gap]. The ADC map was obtained with a scanner. Edema volume was obtained from T2-weighted images and ADC maps were obtained from Image J analyzer. Oedema volume (%) = [(Ipsilateral volume-contralateral volume) / contralateral volume] x 100.

<1−4>マウス脳組織から小膠細胞(microglia)及び星状細胞(astrocytes)の分離
公知の方法によって脳組織から小膠細胞を分離した(非特許文献5)。簡単に説明すると、かん流された(perfused)マウスから脳を除去し、同側性(ipsilateral)及び対側性(contralateral)半球に分けた後、研いで250μgml−1のcollagenase IV/DNase Iを処理した後、37℃で45分間培養して分解した。その細胞分解産物を50/70%Percoll濃度勾配(gradients)で1,000gで25分間分画した。50及び70%バンド間の境界面で小膠細胞を集め、HBSS(hanks’ balanced salt solutions)で洗浄した(Welgene)。分離した小膠細胞の純度は、FACS分析で測定した。公知の方法によって星状細胞を分離した(非特許文献6)。簡単に説明すると、脳組織からの細胞サスペンション(suspensions)を30/60%のPercoll濃度勾配(gradients)で1,000gで25分間分画した。PBS/30%の境界面で星状細胞を収集した。分離した星状細胞の純度は、抗−GFAP抗体を利用したFACS分析で測定した(Cell Signaling Technology、#3670、1:500)。
<1-4> Separation of microglia and astrocytes from mouse brain tissue Microglia were separated from brain tissue by a known method (Non-patent Document 5). Briefly, brains were removed from perfused mice and divided into ipsilateral and contralateral hemispheres, then sharpened to 250 μgml −1 collagenase IV / DNase I. After the treatment, it was decomposed by incubation at 37 ° C. for 45 minutes. The cell lysate was fractionated at 1,000 g for 25 minutes with a 50/70% Percoll gradient. Microglia were collected at the interface between the 50 and 70% bands and washed with HBSS (hanks' balanced salt solutions) (Welgene). The purity of the separated microglia was measured by FACS analysis. Astrocytes were separated by a known method (Non-patent Document 6). Briefly, cell suspensions from brain tissue were fractionated at 1,000 g for 25 minutes with a 30/60% Percoll gradient. Astrocytes were collected at the PBS / 30% interface. The purity of the separated astrocytes was measured by FACS analysis using an anti-GFAP antibody (Cell Signaling Technology, # 3670, 1: 500).

<1−5>神経膠細胞及びニューロン強化(enriched)中脳培養
1ないし3日が過ぎたマウスの大脳皮質からマウス1次混合神経膠細胞(primary mixed glial cells)を培養した(非特許文献7)。抗CD11b抗体を使用したFACS分析によってマウスの混合神経膠細胞の培養で小膠細胞の割合は30.50%と測定された(eBioscience、11−0112、5μgml−1)。14胎児日(embryonic day)のマウスからニューロン強化(enriched)中脳細胞を培養した(非特許文献7)。簡単に説明すると、腹側の中脳組織(ventral mesencephalic tissues)を切開し、CMF−HBSS(Ca2+、Mg2+−free HBSS)で10分間培養し、CMF−HBSS内の0.01%のトリプシン(trypsin)で9分間37℃で培養した。培養物を10%ウシ胎児血清、6mgml−1グルコース、204mgml−1L−グルタミン及びトリプシンの阻害のための100Uml−1ペニシリン/ストレプトマイシン(P/S)を添加したDMEM(Dulbecco’s modified eagle’s medium)で二回洗浄した後、粉砕して単一細胞に分離させた。細胞をポリ−D−lysine(5mgml−1)及びラミニン(laminin)(0.2mgml−1)コーティングプレートに分注した(ウェル当たり2×10細胞)。
<1-5> Glial cells and neuron-enhanced midbrain culture Primary mixed glial cells were cultured from mouse cerebral cortex after 1 to 3 days (Non-patent Document 7). ). The percentage of microglia in mouse mixed glial cultures was determined to be 30.50% by FACS analysis using anti-CD11b antibody (eBioscience, 11-0112, 5 μgml −1 ). Neuron-enriched midbrain cells were cultured from mice on the 14th embryonic day (Non-patent Document 7). Briefly, ventral mesencephalic tissues are dissected, incubated with CMF-HBSS (Ca2 +, Mg2 + -free HBSS) for 10 minutes, and 0.01% trypsin in CMF-HBSS. ) For 9 minutes at 37 ° C. Cultures were supplemented with 10% fetal bovine serum, 6 mg ml −1 glucose, 204 mg ml −1 L-glutamine and 100 Uml −1 penicillin / streptomycin (P / S) for inhibition of trypsin (Dulbecco's modified Eagle's medium) and then crushed to separate into single cells. Cells were dispensed into poly-D-lysine (5 mg ml −1 ) and laminin (0.2 mg ml −1 ) coated plates (2 × 10 6 cells per well).

<1−6>アデノウイルス形質導入(Adenoviral transduction)
Cre再組合酵素遺伝子がサイトメガロウイルスプロモータの調節下で発現される非増殖性アデノウイルス(AD−GFP/Cre)をVector Biolabsから購入した。レポーターAd−GFPを対照群として使用した(Vector Biolabs)。アデノウイルスの形質導入のために、1次混合神経膠細胞をHIF−1α+f/+fマウスから培養し、Ad−GFPまたはAd−GFP/Creで24時間の間感染させた[MOI(multiplicity of infection)=100]。フローサイトメトリーで測定された感染効率(infection efficiency)は、約50%であった。
<1-6> Adenovirus transduction (Adenoviral transduction)
Non-proliferating adenovirus (AD-GFP / Cre) in which the Cre recombinase gene is expressed under the control of the cytomegalovirus promoter was purchased from Vector Biolabs. Reporter Ad-GFP was used as a control group (Vector Biolabs). For adenovirus transduction, primary mixed glial cells were cultured from HIF-1α + f / + f mice and infected with Ad-GFP or Ad-GFP / Cre for 24 hours [MOI (multiplicity of infection). ) = 100]. The infection efficiency measured by flow cytometry was about 50%.

<1−7>ChIPアッセイ
ChIPアッセイキット(Upstate Biotechnology)を使用してChIPアッセイを行った。マウス1次混合神経膠細胞を低酸素環境で24時間の間培養し、直ちに1%ホルムアルデヒド/ホスフェート−バッファー食塩水(phosphate−buffered saline)で固定し、超音波処理して500ないし1,000−bp DNA断片を得た。クロマチン(chromatin)を5μgの抗HIF−1α(Novus、NB100−134)またはウサギIgGで免疫沈殿させた。免疫沈殿されたDNAをTIM−3−プロモータに特異的なプロモータ対で増幅させた[F,5’−CCTGCTGCTTTGGAATTTGC−3’(序列番号3);及びR,5’−GAGTACTTGGCAGGGGAAATC−3’(序列番号4)]。
<1-7> ChIP Assay A ChIP assay was performed using a ChIP assay kit (Upstate Biotechnology). Mouse primary mixed glial cells were cultured for 24 hours in a hypoxic environment, immediately fixed with 1% formaldehyde / phosphate-buffered saline, and sonicated for 500-1,000- A bp DNA fragment was obtained. Chromatin was immunoprecipitated with 5 μg anti-HIF-1α (Novus, NB100-134) or rabbit IgG. The immunoprecipitated DNA was amplified with a promoter pair specific for the TIM-3-promoter [F, 5′-CCTGCTGCCTTGGAATTTTGC-3 ′ (order number 3); and R, 5′-GAGTACTTGGCAGGGGAAATC-3 ′ (order number 4)].

<1−8>好中球移動測定(Neutrophil migration assay)
FITC−結合された抗CD11b(eBioscience、11−0112、5μgml−1)及びPE−結合された抗Gr−1(Ly6G)(eBioscience、12−5931、2ugml−1)の結合に基づいて、FACS Aria system(BD Bioscience)を使用して好中球を分離した。分類された好中球をトランスウェル(Transwell)の上側チャンバにマウス1次混合神経膠細胞が分注された24−ウェルプレート上に添加した。前記細胞を1%または20%の酸素条件で24時間の間培養した。移動(transmigration)は、ヘマサイトメータ(haematocytometer)及びフローサイトメトリー(flow cytometry)を利用して測定した。
<1-8> Neutrophil migration assay (Netrophil migration assay)
Based on the binding of FITC-conjugated anti-CD11b (eBioscience, 11-0112, 5 μgml −1 ) and PE-conjugated anti-Gr-1 (Ly6G) (eBioscience, 12-5931, 2 ugml −1 ), FACS Aria Neutrophils were isolated using a system (BD Bioscience). Sorted neutrophils were added to 24-well plates containing primary mouse mixed glia in the upper chamber of the Transwell. The cells were cultured for 24 hours at 1% or 20% oxygen conditions. The migration was measured using a haematocytometer and flow cytometry.

<1−9>神経学的後遺症(neurological deficits)の測定
神経学的後遺症は、神経学的スコアリングシステム(neurological scoring system)を使用して評価した(非特許文献8)。マウスの神経学的点数は、以下の通りである:0、正常運動機能(normal motor function);1、しっぽ持ち上げによる対側性胴体及び前肢の屈折(flexion ofcontralateral torso and forelimb upon lifting by tail);2、対側への回転(circling to the contralateral side when mouse was held by the tail、but normal posture at rest);3、休息期対側への偏向(leaning to contralateral side at rest);及び4、自発的運動能力の喪失(no spontaneous motor activity)。
<1-9> Measurement of neurological sequelae The neurological sequelae was evaluated using a neurological scoring system (Non-patent Document 8). The neurological scores of mice are as follows: 0, normal motor function; 1, contralateral torso and forelimb reflexes by lifting the tail; and forelimb lifting by; 2, turning to the contralateral side when the mouse was held by the tail; 3, and the biasing to the contralateral side; Loss of spontaneous motor activity.

<1−10>免疫組織化学(immunohistochemistry)
免疫組織化学のために脳を除去し、パラフィンに固定及び包埋した。ミクロトーム(microtome)を使用して梗塞部位を通じて冠状部(coronal sections)(10−mm厚さ)を切ってスライドにマウントした。パラフィンを除去し、セクションをPBS−Tで洗浄し、10%ウシ血清アルブミンで2時間の間ブロッキングした。その後、次の1次抗体を適用した:goat anti−TIM−3(Santa Cruz Biotechnology、sc−30326、2μgml−1)、rat anti−Gr−1(Ly6G)(eBioscience、MPO(Dako、A0398、10μgml−1)、rabbit anti−Iba−1(Wako、#019−19741、2μgml−1)、rabbit anti−cleaved caspase−3(Cell Signaling Technology、#9662S、1:300)、mouse anti−NeuN(Millipore、#MAB377、10μgml−1)。ピモニダゾール(pimonidazole)(Hypoxyprobe−1、Natural Pharmacia International)を使用して低酸素領域を検出した(非特許文献9)。共焦点顕微鏡(Carl Zeiss LSM510)を使用してイメージを得た。1次神経膠細胞でTIM−3発現の測定のために、マウス1次混合神経膠細胞をメタノールで固定し、PBS−Tで洗浄し、抗TIM−3抗体(R&D Systems、AF1529、1μgml−1)で4℃で培養した。
<1-10> Immunohistochemistry
Brains were removed for immunohistochemistry, fixed and embedded in paraffin. A microtome was used to cut coronal sections (10-mm thickness) through the infarct site and mounted on the slide. Paraffin was removed and sections were washed with PBS-T and blocked with 10% bovine serum albumin for 2 hours. The following primary antibodies were then applied: goat anti-TIM-3 (Santa Cruz Biotechnology, sc-30326, 2 μgml −1 ), rat anti-Gr-1 (Ly6G) (eBioscience, MPO (Dako, A0398, 10 μgml). -1 ), rabbit anti-Iba-1 (Wako, # 019-19741, 2 μgml −1 ), rabbit anti-cleaved case-3 (Cell Signaling Technology, # 9662S, 1: 300), mouse antiMileNeup # MAB377,10μgml -1). pimonidazole (pimonidazole) (Hypoxyprobe-1, Natural Pharma The hypoxic region was detected using ia International (Non-Patent Document 9), and images were obtained using a confocal microscope (Carl Zeiss LSM510), measuring TIM-3 expression in primary glial cells. For this purpose, mouse primary mixed glial cells were fixed with methanol, washed with PBS-T, and cultured at 4 ° C. with anti-TIM-3 antibody (R & D Systems, AF1529, 1 μgml −1 ).

<1−11>TIM−3プロモータアッセイ
ゲノムDNAからマウスTIM−3プロモータの1,517−bp断片(始めコドンに対して−1,517から+1)をPCR−増幅し、PGL3 basic vector(Promega)にクローニングした。突然変異プライマー及びPhusion High−Fidelity DNA重合酵素(NEB)を使用して、それぞれのHREの部位特異的突然変異(site−directed mutagenesis)を行った。全ての製作物(constructs)は、DNAシークエンシングで確認した。Lipofectamine2000(Invitrogen)を使用してマウス1次混合神経膠細胞(primary mixed glial cells)をトランスフェクションした。トランスフェクション後に、細胞を1%または20%の酸素条件で24時間の間培養し、luciferase assay system(Promega)でレポーター遺伝子活性を測定した。トランスフェクション効率の標準化(normalization)のためにベータ−ガラクトシダーゼ(β−Galactosidase)活性を測定した。
<1-11> TIM-3 Promoter Assay A 1,517-bp fragment of the mouse TIM-3 promoter (−1,517 to +1 with respect to the start codon) was PCR-amplified from genomic DNA, and PGL3 basic vector (Promega) Cloned into. Site-directed mutagenesis of each HRE was performed using mutant primers and Phusion High-Fidelity DNA polymerase (NEB). All constructs were confirmed by DNA sequencing. Lipofectamine 2000 (Invitrogen) was used to transfect mouse primary mixed glial cells (primary mixed glial cells). After transfection, the cells were cultured for 24 hours under 1% or 20% oxygen conditions, and the reporter gene activity was measured with a luciferase assay system (Promega). Beta-galactosidase activity was measured for normalization of transfection efficiency.

<1−12>ウエスタンブロットの分析
H/Iマウスの右側及び左側半球を切開し、プロテアーゼ阻害剤(protease inhibitors)[2mM phenylmethylsulphonyl fluoride、100μgml−1 leupeptin、10μgml−1 pepstatin、1μgml−1 aprotinin及び2mM EDTA]を含有した氷冷却したRIPAバッファーでpellet pestle(Fisher)で均質化した。均質化物を4℃で12,000rpmで30分間遠心分離し、上層液を収去した。サンプルをSDS−ポリアクリルアミドゲル電気泳動法で分離し、ニトロセルロース膜(nitrocellulose membranes)に移し、次の1次抗体と共に培養した:goat anti−TIM−3(R&D Systems、AF1529、0.1μgml−1)、mouse anti−PARP(Zymed、33−3100、2μgml−1)、rabbit anti−MPO(Dako、A0398、2μgml−1)、goat anti−Iba−1(Abcam、ab5076、0.5μgml−1)、mouse anti−GFAP(Cell Signaling Technology、#3670、1:1,000)、mouse anti−NeuN(Millipore、#MAB377、1μgml−1)、mouse anti−α−tubulin(Sigma、T5168、1:5,000)、microtubule−associated protein 2(Millipore、#MAB3418、1μgml−1)、glutamate decarboxylase(Abcam、ab11070、1μgml−1)、peroxidase−conjugated goat anti−rabbit(Bio−Rad,#170−6515,1:5,000)、peroxidase−conjugated rabbit anti−goat(Zymed,R−21459,1:5,000)、peroxidase−conjugated goat anti−mouse(Bio−Rad,#170−6516,1:5,000)。結果は、増加された化学発光システム(enhanced chemiluminescence system)を使用して視覚化し、濃度計(densitometric analysis)(Image J software、NIH)で定量した。全ての実験は、独立して少なくとも3回繰り返して行われた。
<1-12> Analysis of Western Blot The right and left hemispheres of H / I mice were dissected, and protease inhibitors [2 mM phenylmethylsulfonfluoride, 100 μgml −1 leueptin, 10 μgml −1 peptin, 1 μmml2 peptin, 1 μgml- 1 Homogenized by pellet pellet (Fisher) with ice-cooled RIPA buffer containing [EDTA]. The homogenized product was centrifuged at 12,000 rpm for 30 minutes at 4 ° C., and the upper layer liquid was collected. Samples were separated by SDS-polyacrylamide gel electrophoresis, transferred to nitrocellulose membranes and cultured with the following primary antibodies: goat anti-TIM-3 (R & D Systems, AF1529, 0.1 μgml −1 ), Mouse anti-PARP (Zymed, 33-3100, 2 μgml −1 ), rabbit anti-MPO (Dako, A0398, 2 μgml −1 ), goat anti-Iba-1 (Abcam, ab5076, 0.5 μgml −1 ), mouse anti-GFAP (Cell Signaling Technology, # 3670, 1: 1,000), mouse anti-NeuN (Millipore, # MAB377, 1 μgm) l −1 ), mouse anti-α-tubulin (Sigma, T5168, 1: 5,000), microtubule-associated protein 2 (Millipore, # MAB3418, 1 μgml −1 ), glutamate decalboxycylamine 110 μg, 1 , Peroxidase-conjugate goat anti-rabbit (Bio-Rad, # 170-6515, 1: 5,000), peroxidase-conjugated rabbit anti-goat (Zymed, R-21259, 1: 5,000), peroxydase crate anti-mouse (Bio-Rad, # 170-6516, 1: 5,000). Results were visualized using an enhanced chemiluminescence system and quantified with a densitometric analysis (Image J software, NIH). All experiments were performed independently and repeated at least three times.

<1−13>RT−PCR分析
Easy−Blue(iNtRON)を使用して総RNAを分離し、avian myeloblastosis virus reverse transcriptase(TaKaRa)を製造社の説明に従って使用してcDNAを合成した。25−30サイクルの連続反応でPCRを行った。全ての実験は、独立して少なくとも3回繰り返して行われ、PCR産物は、NIH Image Jを使用して定量してアクチンに対して標準化した。QuantiFast SYBR Green PCR kit(Qiagen)を使用してreal−time PCRを行った。Roche LightCycler 480 Real−Time PCR System(Roche Applied Science)及びLigthCycler 480 Quantification Software Version1.5を使用してreal−time PCRを行い分析した。
<1-13> RT-PCR analysis Total RNA was isolated using Easy-Blue (iNtRON), and cDNA was synthesized using avian myeloblastosis reverse transcriptase (TaKaRa) according to the manufacturer's instructions. PCR was performed in a continuous reaction of 25-30 cycles. All experiments were performed independently and repeated at least three times, and PCR products were quantified using NIH Image J and normalized to actin. Real-time PCR was performed using the QuantFast SYBR Green PCR kit (Qiagen). Real-time PCR was performed using the Roche LightCycler 480 Real-Time PCR System (Roche Applied Science) and the LightCycler 480 Quantification Software Version 1.5.

定量的(quantitative)PCRに使用されたプライマーは、以下の通りである:
IL−1βに対して(forward)5’−GGATGAGGACATGAGCACCT−3’(序列番号5)及び(reverse)5’−TCCATTGAGGTGGAGAGCTT−3’(序列番号6);
CXCL1に対して(forward)5’−TGCACCCAAACCGAAGTCAT−3’(序列番号7)及び(reverse)5’−TTGTCAGAAGCCAGCGTTCAC−3’(序列番号8);
HIF−1αに対して(forward)5’−CTCATCAGTTGCCACTTCC−3’(序列番号9)及び(reverse)5’−TCATCTTCACTGTCTAGACCAC−3’(序列番号10);
GAPDHに対して(forward)5’−TGTCGTGGAGTCTACTGGTGTCTTC−3’(序列番号11)及び(reverse)5’−CGTGGTTCACACCCATCACAA−3’(序列番号12)。
The primers used for quantitative PCR are as follows:
For IL-1β (forward) 5′-GGATGGAGACATGAGCACCT-3 ′ (order number 5) and (reverse) 5′-TCCATTGAGGTGGAGAGCTTT-3 ′ (order number 6);
(Forward) 5'-TGCACCCAAACCGAAGTCCAT-3 '(order number 7) and (reverse) 5'-TTGTCAGAAGCCAGGTTCAC-3' (order number 8) for CXCL1;
(Forward) 5′-CTCATCAGTTGCCACTTCC-3 ′ (order number 9) and (reverse) 5′-TCATCTTCACTGTTCTAGACCAC-3 ′ (order number 10) for HIF-1α;
(Forward) 5′-TGTCGTGGAGTCTACTGGTGTCTCTC-3 ′ (order number 11) and (reverse) 5′-CGTGGTTCACACCCATCACAA-3 ′ (order number 12) for GAPDH.

その他に使用されたPCRプライマー序列は、以下の通りである:
TIM−3に対して(forward)5’−CCCTGCAGTTACACTCTACC−3’(序列番号13)及び(reverse)5’−GTATCCTGCAGCAGTAGGTC−3’(序列番号14);
HIF1αに対して(forward)5’−AGCCTTAACCTGTCTGCCACTT−3’(序列番号15)及び(reverse)5’−GAAATCATTTAACATTGCATATATACTAGAACAT−3’(序列番号16);
MPOに対して(forward)5’−AGGATAGGACTGGATTTGCCTG−3’(序列番号17)及び(reverse)5’−GTGGTGATGCCAGTGTTGTCA−3’(序列番号18);
IL−1βに対して(forward)5’−TACAGGCTCCGAGATGAACAACAA−3’(序列番号19)及び(reverse)5’−TGGGGAAGGCATTAGAAACAGTCC−3’(序列番号20);
CXCL1に対して(forward)5’−CGCTCGCTTCTCTGTGCAGC−3’(序列番号21)及び(reverse)5’−GTGGCTATGACTTCGGTTTGG−3’(序列番号22);
Actinに対して(forward)5’−CATGTTTGAGACCTTCAACACCCC−3’(序列番号23)及び(reverse)5’−GCCATCTCCTGCTCGAAGTCTAG−3’(序列番号24)。
Other PCR primer sequences used are as follows:
(Forward) 5'-CCCTGCAGGTTACACTCTACC-3 '(order number 13) and (reverse) 5'-GTATCCTGCAGCAGTAGGGTC-3' (order number 14);
(Forward) 5′-AGCCCTTAACCCTTCTGCCACTT-3 ′ (order number 15) and (reverse) 5′-GAAATCATTATACATTGCATATATAGAGACAT-3 ′ (order number 16) for HIF1α;
(Forward) 5′-AGGATAGAGACTGGATTTGCCTG-3 ′ (order number 17) and (reverse) 5′-GTGGTGATGCCAGTGTGTCA-3 ′ (order number 18);
For IL-1β (forward) 5′-TACAGGCTCCCGAGATGAACAACAA-3 ′ (order number 19) and (reverse) 5′-TGGGGAAGCATCATAAAACAGTCC-3 ′ (order number 20);
(Forward) 5'-CGCTCGCTTCTCTGTGCAGC-3 '(order number 21) and (reverse) 5'-GTGGCTATGAACTTCGGTTTGG-3' (order number 22) for CXCL1;
(Forward) 5'-CATGTTTGAGCACTTCAACACCCC-3 '(order number 23) and (reverse) 5'-GCCATCTCCTCTCGAGATCTAG-3' (order number 24).

<1−14>フローサイトメトリー(Flow cytometry)
全ての染色段階は、闇中で行われ、BD Fc Blockで遮断された。新たに得た小膠細胞及び星状細胞を、次の抗体で染色した:rabbit anti−Iba−1(Wako、#019−19741、1μgml−1)後にAlexa 488−conjugated chick anti−rabbit(Invitrogen、A21441、2μgml−1)、及びPE−conjugated anti−mouse TIM−3(eBioscience、RMT−3−23、2μgml−1)またはisotype control Ab(eBioscience、2μgml−1)で4℃で30分間、GFAPの細胞内染色のために、IC fixation/permeabilizationバッファー(eBioscience)を使用して細胞を20分間固定及び透過化し、透過化(permeabilization)バッファーで二回洗浄し、抗GFAP(Cell Signaling Technology、#3672、1:500)と共に透過化バッファーで30分間培養し、Alexa 488−conjugated chick anti−mouse(Invitrogen、A21200、2μgml−1)で染色した。データは、Cell−Quest software(BD Bioscience)及びFlow Jo software(Treestar)パッケージで分析した。
<1-14> Flow cytometry
All staining steps were performed in the dark and blocked with BD Fc Block. Freshly obtained microglia and astrocytes were stained with the following antibodies: rabbit anti-Iba-1 (Wako, # 019-19714, 1 μgml −1 ) followed by Alexa 488-conjugated chicken anti-rabit (Invitrogen, A21441, 2 μgml −1 ), and PE-conjugated anti-mouse TIM-3 (eBioscience, RMT-3-23, 2 μgml −1 ) or isotype control Ab (eBioscience, 2 μgml −1 ) for 30 minutes at 4 ° C. For intracellular staining, cells were fixed and permeabilized for 20 minutes using IC fixation / permeabilization buffer (eBioscience) and permeabilized (pe rmeabilization) buffer twice, incubated with anti-GFAP (Cell Signaling Technology, # 3672, 1: 500) in permeabilization buffer for 30 minutes, Alexa 488-conjugated chicken anti-mouse (Invitrogen, A21200), 2 μgml- 1 Stained with Data were analyzed with Cell-Quest software (BD Bioscience) and Flow Jo software (Treestar) packages.

<1−15>レンチウイルス生産及び定位注射(stereotaxic injection)
TIM−3(GE Dharmacon)のコーディング序列をPLL3.7.EF1αプラスミド(Addgene、Inc.)に接合させてPLL3.7.EF1α−TIM3を製作した。前記プラスミドを使用して再組合レンチウイルスLV−TIM3−GFPを製作した。対照群としてGFPのみを発現するレンチウイルスベクター(LV−GFP)を作った。レンチウイルスをフローサイトメトリーを使用して滴定した(非特許文献10)。脳固定装置(stereotaxic instrument)を利用してLV−TIM3−GFPまたはLV−GFPを注射した。それぞれのマウスは、4回のレンチウイルス(5×10TUml−1を含有した20μリットルを右側半球に)頭蓋注射(intracranial injections)を打たれた。試験管内(in vitro)蛍光イメージングのため、収集された細胞をFACS及び抗GFP抗体(Santacruz、sc−9996、1:1,000)を使用したウエスタンブロッティングで分析した。Caliper Life Science’s Xenogen IVIS Spectrumを使用して全身の生体内(in vivo)イメージングを行った[励起(excitation)フィルターで445から490nm、放出(emission)フィルターで515から575nmで照射]。
<1-15> Lentivirus production and stereotaxic injection
The coding sequence of TIM-3 (GE Dharmacon) is PLL3.7. PLL3.7. Conjugated to EF1α plasmid (Addgene, Inc.). EF1α-TIM3 was produced. Recombinant lentivirus LV-TIM3-GFP was constructed using the plasmid. As a control group, a lentiviral vector (LV-GFP) expressing only GFP was prepared. Lentivirus was titrated using flow cytometry (Non-Patent Document 10). LV-TIM3-GFP or LV-GFP was injected using a stereotaxic instrument. Each mouse received four intracranial injections of lentivirus (20 μl containing 5 × 10 6 TUml −1 in the right hemisphere). Collected cells were analyzed by Western blotting using FACS and anti-GFP antibody (Santacruz, sc-9996, 1: 1,000) for in vitro fluorescence imaging. Whole body in vivo imaging was performed using Caliper Life Science's Xenogen IVIS Spectrum [excitation filters at 445 to 490 nm, emission filters at 515 to 575 nm].

<1−16>データの分析
全てのデータは、平均±s.dで表示した。SigmaPlot10.0を使用してPost−hoc comparisons(Student−Newman−Keuls test)を行った。神経学的点数(neurological scores)は非母数的(nonparametric)統計処理で評価した。二つのグループ(IgG vs anti−TIM−3、HIF−1α+f/+fマウス vs LysM−Hif−1α−/−マウス、LV−GFP注射 LysM−Hif−1α−/−マウス vs LV−TIM3−GFP注射LysM− Hif−1α−/−)間の比較は、Mann−hitney U−testsで分析した。
<1-16> Analysis of data All data are expressed as mean ± s. Displayed with d. Post-hoc comparisons (Student-Newman-Keuls test) were performed using SigmaPlot 10.0. Neurological scores were evaluated by nonparametric statistical processing. Two groups (IgG vs anti-TIM-3, HIF-1α + f / + f mice vs LysM-Hif-1α − / − mice, LV-GFP injection LysM-Hif-1α − / − mice vs LV-TIM3-GFP injection Comparison between LysM-Hif-1α − / − ) was analyzed by Mann-hitney U-tests.

低酸素半陰影(hypoxic penumbra)でのTIM−3発現の増加
虚血性脳損傷と炎症間の相互依存的関連性の基礎となる分子的機作を調べるために、本発明者らは、脳の低酸素虚血症(cerebral hypoxia−ischaemia、H/I)による病態生理学的炎症反応に主な役割をすることができる候補分子を調査した。このため、右側頚動脈の一方接合(unilateral ligation)後、全身的低酸素症(systemic hypoxia)を誘発した一時的一側脳虚血症(transient unilateral cerebral ischaemia)マウスモデルを利用した(非特許文献11)。H/Iの24時間後に対側性(contralateral)及び半陰影(penumbral)皮質領域から組織を得た後、多様な炎症関連分子の発現水準をRNA及びタンパク質水準で調査した。その結果、同側性半陰影(ipsilateral penumbra)でTIM−3(T−cell immunoglobulin and mucin domain−3)の転写水準が対側性領域(contralateral regions)ではるかに高く増加したことを発見した。また、同側性半陰影(ipsilateral penumbra)でTIM−3タンパク質も対側性領域より増加したことを確認した(図1a、b)。前記同側性半陰影領域は、低酸素下で陽性対照群(positive control)であるHIF−1の転写体及びタンパク質水準が高いと報告された(非特許文献12;及び非特許文献13)。
Increased TIM-3 expression in the hypoxic penumbra To investigate the molecular mechanism underlying the interdependent relationship between ischemic brain injury and inflammation, we Candidate molecules that could play a major role in the pathophysiological inflammatory response due to cerebral hypoxia-ischaemia (H / I) were investigated. For this reason, after unilateral ligation of the right carotid artery, a transient unilateral ischemia mouse model in which systemic hypoxia was induced (Non-patent Document 11) ). After obtaining tissue from the contralateral and penumbral cortex areas 24 hours after H / I, the expression levels of various inflammation-related molecules were investigated at the RNA and protein levels. As a result, it was found that the transcriptional level of TIM-3 (T-cell immunoglobulin and mucin domain-3) increased much higher in the contralateral regions in the ipsilateral penumbra. In addition, it was confirmed that TIM-3 protein was also increased from the contralateral region in the ipsilateral penumbra (FIGS. 1a and b). The ipsilateral semi-shaded area was reported to have a high transcript and protein level of HIF-1, which is a positive control group under hypoxia (Non-patent Document 12; and Non-patent Document 13).

上記結果を確証するために、TIM−3に対する抗体を利用してH/Iマウスの冠状面(coronal sections)に免疫組織化学法を行った(非特許文献14;及び非特許文献15)。その結果、上記結果と一致するように、同側性半陰影でTIM−3−陽性細胞が非常に増加したことが確認できた(図1c)。さらに低酸素症マーカーであるピモニダゾール(pimonidazole)(hypoxyprobe−1)を利用して、H/Iマウスのhypoxyprobe−1染色された低酸素半陰影でTIM−3が高く発現されたことを確認した(図1d)。   In order to confirm the above results, immunohistochemistry was performed on coronal sections of H / I mice using an antibody against TIM-3 (Non-Patent Document 14; and Non-Patent Document 15). As a result, it was confirmed that the number of TIM-3-positive cells was greatly increased in the ipsilateral semi-shade so as to agree with the above results (FIG. 1c). Furthermore, it was confirmed that TIM-3 was highly expressed in the hypoxia half-shade of H / I mice stained with hypoxyprobe-1 using pimonidazole (hypoxyprobe-1), which is a hypoxia marker ( FIG. 1d).

このような結果は、TIM−3発現が低酸素半陰影で上向き調節されるということを示し、TIM−3が脳虚血による病態生理学的変化に所定の役割ができることを示唆する。   These results indicate that TIM-3 expression is up-regulated in hypoxic half-shadows, suggesting that TIM-3 can play a role in pathophysiological changes due to cerebral ischemia.

低酸素環境の神経膠細胞でTIM−3発現の上向き調節
本発明者は、H/I後に如何なる細胞がTIM−3の上向き調節(upregulation)を表すについて調査した。ウエスタンブロット分析の結果、H/I24時間後にH/Iマウスの同側性皮質で、活性化された小膠細胞マーカーであるIba−1(ionized calciumbinding adaptor molecule−1)及び活性化された星状膠細胞マーカーであるGFAP(glial fibrillary acidic protein)のタンパク質発現水準が対側性皮質よりさらに高かった。一方、NeuN(neuronal nuclei)、マイクロチューブル−連関タンパク質2(microtubule−associated protein 2)及びグルタメートデカルボキシラーゼ(glutamate decarboxylase)のようなニューロン細胞マーカーの発現水準は、半陰影皮質組織(penumbral cortex tissues)で減少した。
Upregulation of TIM-3 expression in hypoxic glial cells The inventors investigated that after H / I any cell exhibits TIM-3 upregulation. As a result of Western blot analysis, activated microglia marker Iba-1 (ionized calcium binding adapter molecule-1) and activated stars in the ipsilateral cortex of H / I mice 24 hours after H / I. The protein expression level of GFAP (glial fibrous acid protein), which is a glial cell marker, was even higher than that of the contralateral cortex. On the other hand, the expression levels of neuronal cell markers such as NeuN (neuronal nuclei), microtubule-associated protein 2 and glutamate decarboxylase are determined by the semi-shadow cortex tissue. Decreased.

従って、本発明者らは、H/I24時間後に小膠細胞(microglia)及び星状膠細胞(astrocytes)でTIM−3の発現水準を調査した。免疫組織化学の結果、H/Iマウスの同側性皮質でTIM−3−発現細胞の多くの領域は、Iba−1陽性で表れた。また、同側性皮質のGFAP−免疫活性(immunoreactive)星状膠細胞でTIM−3の強い発現も観察された。さらに、H/Iマウスから分離した脳細胞のFACS(Fluorescence−activated cell sorting)分析の結果、低酸素虚血症(hypoxia−ischaemia)は、小膠細胞及び星状膠細胞の活性をもたらし、これは、増加したTIM−3の発現を表す。高い水準のIba−1を発現する小膠細胞及び高い水準のGFAPを発現する星状膠細胞は、H/I24時間後に同側性半陰影(ipsilateral penumbra)で非常に増加し、これは、小膠細胞及び星状膠細胞が低酸素環境で活性化されたことを意味する。また、TIM−3の発現は、同側性皮質から分離したIba−1−陽性小膠細胞及びGFAP−陽性星状膠細胞において、対側性領域で分離したものより、有意味な水準で高く表れた(図1e、f)。   Therefore, the present inventors investigated the expression level of TIM-3 in microglia and astrocytes 24 hours after H / I. As a result of immunohistochemistry, many regions of TIM-3-expressing cells in the ipsilateral cortex of H / I mice appeared Iba-1 positive. Strong expression of TIM-3 was also observed in GFAP-immunoreactive astrocytes in the ipsilateral cortex. Furthermore, as a result of FACS (Fluorescence-activated cell sorting) analysis of brain cells isolated from H / I mice, hypoxia-ischaemia leads to microglial and astrocyte activity. Represents increased TIM-3 expression. Microglia that express high levels of Iba-1 and astrocytes that express high levels of GFAP greatly increase in ipsilateral penumbra after 24 hours of H / I, It means that glial cells and astrocytes were activated in a hypoxic environment. In addition, TIM-3 expression is significantly higher in Iba-1-positive microglia and GFAP-positive astrocytes isolated from the ipsilateral cortex than in the contralateral region. Appeared (FIGS. 1e, f).

このような結果は、低酸素下で活性化された小膠細胞及び星状膠細胞でTIM−3の発現が非常に増加するという事実を裏付ける。   These results support the fact that TIM-3 expression is greatly increased in microglia and astrocytes activated under hypoxia.

低酸素環境でTIM−3のHIF−1−依存的増加
前記実験結果に基づいて本発明者らは、神経膠細胞(glial cell)でTIM−3の発現が酸素分圧(oxygen tension)によって変更され得るか否かを、BV2小膠細胞及び1次培養された神経膠細胞を使用して実験した。BV2細胞は、正常酸素(normoxic)(20%のO)または低酸素(hypoxic)(1%のO)条件で24時間の間培養し、TIM−3の細胞表面水準は、FACS分析で測定した。興味深いことに、TIM−3発現は、低酸素条件で非常に増加した(図2a)。免疫細胞化学(immunocytochemistry)の分析結果も、マウス1次混合神経膠細胞(primary mixed glial cells)でTIM−3発現が正常酸素(normoxic)環境に比べて低酸素(hypoxic)環境で非常に増加するということを示した(図2b)。また、本発明者らは、低酸素環境でTIM−3の転写水準が1次混合神経膠細胞では増加したことに対し、1次ニューロン細胞(primary neuronal cells)では増加しないことを確認した(図2c、d)。このような結果は、神経膠細胞で低酸素症がTIM−3発現を誘導することを示す。
HIF-1-dependent increase of TIM-3 in hypoxic environment Based on the results of the experiment, the present inventors changed the expression of TIM-3 in glia cells by oxygen tension. Whether it could be done was studied using BV2 microglia and primary cultured glial cells. BV2 cells were cultured for 24 hours in normoxic (20% O 2 ) or hypoxic (1% O 2 ) conditions, and TIM-3 cell surface levels were determined by FACS analysis. It was measured. Interestingly, TIM-3 expression was greatly increased in hypoxic conditions (Figure 2a). Immunocytochemistry analysis results also show that TIM-3 expression in mouse primary mixed glial cells is greatly increased in a hypoxic environment compared to a normoxic environment (FIG. 2b). In addition, the present inventors have confirmed that the transcription level of TIM-3 was increased in primary mixed glial cells in a hypoxic environment, but not increased in primary neuronal cells (Fig. 2c, d). Such results indicate that hypoxia induces TIM-3 expression in glial cells.

HIF−1は、低酸素環境で多くの遺伝子の主な転写調節因子である。神経膠細胞で低酸素によって刺激されたTIM−3の上向き調節がHIF−1によって媒介されるかについて調べるために、本発明者らは、抗HIF−1α抗体及び潜在的HREコンセンサス序列(HIF−responsive element(HRE)consensus sequences)を含むTIM−3プロモータ領域(elements)を利用してChIPアッセイ(chromatin immunoprecipitation assay)を行った。図2eに示すように、低酸素環境の1次混合神経膠細胞(primary mixed glial cell)でHIF−1αは、HRE−含みTIM−3プロモータ領域に結合することができた。さらに、上記の結果を確認するために、本発明者らは、HIF−1α−欠乏神経膠細胞でTIM−3プロモータの活性を調査した。HIF−1αflox/flox(HIF−1α+f/+f)マウスから1次混合神経膠細胞を培養した後、アデノウイルス−Cre/GFP(Ad−Cre/GFP)または対照群GFP(緑蛍光タンパク質(GFP)を暗号化するアデノウイルス(Ad−GFP))で感染させた。FACSを利用してウイルス感染の効率を確認し、細胞をTIM−3ルシフェラーゼレポーター(−1,517/+1)でトランスフェクションした後、TIM−3プロモータ活性を測定した。予想どおり、低酸素環境でTIM−3プロモータ活性は、対照群Ad−GFP−感染された神経膠細胞(HIF1α+f/+f)では非常に増加したが、Ad−Cre/GFP−感染された、HIF−1α−欠乏神経膠細胞(HIF1αΔ/Δ)では非常に減少した(図2f)。TIM−3プロモータの潜在的HREsの部位特異的突然変異(site−directed mutagenesis)は、ルシフェラーゼ活性の低酸素−依存的増加を野生型レポーターに比べて非常に減少させた。また、Ad−Cre/GFP−感染されたHIF−1α−欠乏神経膠細胞でTIM−3転写体及びタンパク質の低酸素刺激による増加は非常に抑制された(図2g、h)。 HIF-1 is a major transcriptional regulator of many genes in a hypoxic environment. To investigate whether the up-regulation of TIM-3 stimulated by hypoxia in glial cells is mediated by HIF-1, we have identified anti-HIF-1α antibodies and potential HRE consensus sequences (HIF- A ChIP assay (chromatin immunoprecipitation assay) was performed using TIM-3 promoter regions (responsible elements (HRE) consensus sequences). As shown in FIG. 2e, HIF-1α was able to bind to the HRE-containing TIM-3 promoter region in primary mixed glial cells in a hypoxic environment. Furthermore, to confirm the above results, we investigated the activity of the TIM-3 promoter in HIF-1α-deficient glial cells. After culturing primary mixed glial cells from HIF-1α flox / flox (HIF-1α + f / + f ) mice, adenovirus-Cre / GFP (Ad-Cre / GFP) or control group GFP (green fluorescent protein (GFP) ) Was encoded with the adenovirus encoding (Ad-GFP)). The efficiency of virus infection was confirmed using FACS, and cells were transfected with TIM-3 luciferase reporter (-1,517 / + 1), and then TIM-3 promoter activity was measured. As expected, TIM-3 promoter activity in hypoxic environments was greatly increased in the control group Ad-GFP-infected glial cells (HIF1α + f / + f ), but Ad-Cre / GFP-infected, HIF -1α-deficient glial cells (HIF1α Δ / Δ ) were greatly reduced (FIG. 2f). Site-directed mutation of potential HREs of the TIM-3 promoter greatly reduced the hypoxia-dependent increase in luciferase activity compared to the wild-type reporter. In addition, the increase in TIM-3 transcript and protein by hypoxic stimulation was significantly suppressed in Ad-Cre / GFP-infected HIF-1α-deficient glial cells (FIGS. 2g and h).

このような結果は、低酸素環境でTIM−3の発現がHIF−1−依存的方式で調節されることを示す。   These results indicate that TIM-3 expression is regulated in a HIF-1-dependent manner in a hypoxic environment.

マウスH/IモデルでTIM−3抑制による脳損傷の減少
H/Iマウスモデルの神経膠細胞でTIM−3が上向き調節されたので、本発明者らは、大脳(cerebral)H/I後に脳で低酸素誘導されたTIM−3の役割を調査した。このため、H/I24時間後にTIM−3−抑制抗体が脳損傷に及ぼす影響をTTC(2,3,5−triphenyltetrazolium)染色を利用して調査した。図3aに示すように、対照群IgG−注射マウスに比べて100μgのTIM−3−抑制抗体を静脈注射したマウスでTTC−陰性領域が非常に減少したことを確認することができた。このような結果は、低酸素環境でTIM−3−抑制抗体が脳損傷を減少させることができることを示す。
Reduction of brain damage due to TIM-3 inhibition in mouse H / I model Since TIM-3 was up-regulated in glial cells of the H / I mouse model, the inventors post-cerebral H / I brain The role of TIM-3 induced by hypoxia was investigated. Therefore, the effect of TIM-3-inhibitory antibody on brain damage 24 hours after H / I was investigated using TTC (2,3,5-triphenyltetrazolium) staining. As shown in FIG. 3a, it was confirmed that the TTC-negative region was greatly reduced in the mouse intravenously injected with 100 μg of TIM-3-suppressing antibody compared to the control group IgG-injected mouse. Such results indicate that TIM-3-suppressing antibodies can reduce brain damage in a hypoxic environment.

脳梗塞の生命を脅威する結果である浮腫は、炎症と虚血性脳損傷に伴って表れる(非特許文献16)。従って、本発明者らは、TIM−3−抑制がH/Iによる浮腫の形成に及ぼす影響を調査した。梗塞(infarct)領域と浮腫の形成を観察するために、H/Iの1日から7日までT2−加重(weighted)磁気共鳴映像を得た。TTC染色から得た結果と同様に、H/Iの1日目、TIM−3−抗体−注入マウスの同側性半球(ipsilateral hemispheres)で梗塞と浮腫の形成は、IgG−注入マウスに比べて非常に減少し(図3b−d)、このような浮腫の形成と梗塞の減少は、3、5及び7日目にも持続した(図3c、d)。   Edema, which is a life threatening result of cerebral infarction, appears with inflammation and ischemic brain injury (Non-patent Document 16). Therefore, the inventors investigated the effect of TIM-3-suppression on H / I-induced edema formation. To observe infarct area and edema formation, T2-weighted magnetic resonance images were acquired from day 1 to day 7 of H / I. Similar to the results obtained from TTC staining, infarct and edema formation in the ipsilateral hemisphere of TIM-3-antibody-injected mice on day 1 of H / I compared to IgG-injected mice The edema formation and infarct reduction persisted on days 3, 5, and 7 (FIGS. 3c, d).

H/I後の脳損傷とTIM−3の関連性を追加で調査するために、TIM−3−抑制抗体がニューロン細胞の死滅に及ぼす影響を、脳虚血症に重要な役割をする細胞死滅エフェクタープロテアーゼ(cell death effector protease)であるカスパーゼ(caspase)−3の発現を測定することで調査した(非特許文献17;及び非特許文献18)。免疫組織化学の結果、IgG−処理H/Iマウスの同側性皮質領域のニューロン細胞でカスパーゼ−3の発現は非常に増加したことに対し、TIM−3抑制抗体処理マウスでこのような増加は非常に減少した(図3e)。次に、対照群IgGまたはTIM−3−抑制抗体を処理したH/Iマウスの同側性及び対側性皮質において、カスパーゼ−3によって切断するカスパーゼ−3活性のマーカーで、虚血性細胞の死滅と関連のあるPARP(poly(ADP−ribose)polymerase)の水準を測定した(非特許文献19)。図3fに示すように、対照群IgG−注射H/Iマウスの同側性皮質組織で全長PARPの発現は非常に減少したが、TIM−3−抑制抗体−注射H/Iマウスでは減少しなかった。   To further investigate the relationship between post-H / I brain injury and TIM-3, the effect of TIM-3-inhibitory antibodies on neuronal cell death is considered to be an important role in cerebral ischemia. It investigated by measuring the expression of caspase-3 which is an effector protease (cell non-patent literature 17; and non-patent literature 18). As a result of immunohistochemistry, the expression of caspase-3 was greatly increased in neuronal cells in the ipsilateral cortical region of IgG-treated H / I mice, whereas such increase was observed in mice treated with TIM-3 inhibitory antibody. It was greatly reduced (Fig. 3e). Next, ischemic cell killing with a marker of caspase-3 activity cleaved by caspase-3 in the ipsilateral and contralateral cortex of H / I mice treated with control group IgG or TIM-3-suppressor antibody The level of PARP (poly (ADP-ribose) polymerase) related to the above was measured (Non-patent Document 19). As shown in FIG. 3f, expression of full length PARP was greatly reduced in the ipsilateral cortical tissue of control IgG-injected H / I mice, but not in TIM-3-suppressing antibody-injected H / I mice. It was.

このような結果は、TIM−3の抑制がマウスで脳虚血症後の梗塞部位とニューロン細胞の死滅を非常に減少させることができることを示す。   Such results indicate that suppression of TIM-3 can greatly reduce infarct sites and neuronal cell death after cerebral ischemia in mice.

TIM−3抑制による好中球(neutrophil)の浸潤の減少
様々な研究によると、好中球は、虚血性脳で数時間中に速く浸潤されて、炎症反応と脳損傷の発生に関与する(非特許文献20;及び非特許文献21)。神経膠細胞は、虚血症発生後の数分内に関連した活性を表す脳損傷に1次的に反応する細胞のうち一つであるので、本発明者らは、神経膠細胞でTIM−3のHIF−1−依存的増加が好中球の虚血半陰影(ischaemic penumbra)への浸潤に影響を及ぼし、TIM−3が好中球を集める能力の下向き調節(downregulation)は、脳虚血後の脳損傷を減少させることができるという仮説を立てた。これにより、先ず代表的な二つの好中球マーカーであるMPO(myeloperoxidase)及びGr−1(granulocyte receptor−1)の発現を測定し、H/I後、24時間になった時、対側性領域に比べて半陰影皮質(penumbral cortex)及び線条体(striatum)で前記マーカーに陽性である細胞(MPOGr−1)が大きく増加することを確認した。次に、本発明者らは、神経膠細胞(glial cells)が低酸素環境でGr−1highCD11bhigh好中球を集めることができるか否かについて調査した。C57BL/6マウスから脾臓細胞(splenocytes)を分離し、1次混合神経膠細胞または兔疫細胞を損傷部位に集めると知られたマウス胎仔線維芽細胞(murine embryonic fibroblast)の対照群細胞を含むか含まないトランスウェル(Transwell)システムにおいて、1または20%の酸素条件で24時間の間培養した(非特許文献22)。神経膠細胞またはマウス胎仔線維芽細胞の存在下で、Gr−1highCD11bhigh細胞は、低酸素環境では下側チャンバに非常に多く移動したが、正常(normoxic)環境では数個の細胞のみが移動した。しかし、このようなGr−1highCD11bhigh細胞の移動の低酸素依存的増加は、神経膠細胞のない状態では非常に減少した。このような結果は、神経膠細胞が低酸素環境でGr−1highCD11bhigh細胞を集めることに関与し得ることを示唆する。
Reduction of Neutrophil Infiltration by TIM-3 Inhibition According to various studies, neutrophils are rapidly infiltrated in the ischemic brain within hours and are involved in the development of inflammatory responses and brain damage ( Non-patent document 20; and Non-patent document 21). Since glial cells are one of the cells that respond primarily to brain injury that exhibits associated activity within minutes after the onset of ischemia, we have identified TIM- HIF-1-dependent increase of 3 affects neutrophil infiltration into the ischemic penumbra, and downregulation of TIM-3's ability to collect neutrophils We hypothesized that brain damage after blood could be reduced. Thus, first, the expression of two typical neutrophil markers, MPO (myeloperoxidase) and Gr-1 (granulocyte receptor-1), was measured, and at 24 hours after H / I, It was confirmed that cells positive for the marker (MPO + Gr-1 + ) greatly increased in the penumbral cortex and striatum compared to the region. Next, the present inventors investigated whether glial cells (gial cells) can collect Gr-1 high CD11b high neutrophils in a hypoxic environment. Does it contain control group cells of mouse embryonic fibroblasts known to isolate spleen cells from C57BL / 6 mice and collect primary mixed glial cells or epithelial cells at the site of injury? In a Transwell system not containing the cells, the cells were cultured for 24 hours under oxygen conditions of 1 or 20% (Non-patent Document 22). In the presence of glial cells or mouse fetal fibroblasts, Gr-1 high CD11b high cells migrated very much into the lower chamber in a hypoxic environment, but only a few cells in a normic environment moved. However, such hypoxia-dependent increase in Gr-1 high CD11b high cell migration was greatly reduced in the absence of glial cells. Such results suggest that glial cells may be involved in collecting Gr-1 high CD11b high cells in a hypoxic environment.

次に、本発明者らは、H/I後の24時間になった時、TIM−3−抑制が好中球の同側性半球(ipsilateral hemispheres)への浸潤に及ぼす効果を実験した。H/Iマウスの皮質組織に対する逆転写−PCR(RT−PCR)及びウエスタンブロット分析の結果は、対照群IgG−処理マウスに比べてTIM−3−抑制抗体−処理マウスでMPO発現の水準が非常に減少することを示した(図4a、b)。同側性皮質の冠状面(coronal section)に対する免疫組織化学の実験結果も、TIM−3−抑制抗体処理によってMPOGr−1細胞が非常に減少することを示す(図4c)。このような結果は、抗好中球及び抗MPO抗体を使用した免疫組織化学実験によっても確認された。また、H/I脳(bregma−2から+2)の多くの同側性領域の冠状面を使用して、TIM−3抑制が好中球の浸潤に及ぼす影響を様々な時点で測定した。図4d、eに示すように、全ての観察時点(1〜7日)でTIM−3を抑制させたマウスの半陰影皮質及び線条体(striatum)でさらに少ない数のMPOGr−1細胞が観察された。 Next, we tested the effect of TIM-3-suppression on the infiltration of neutrophils into ipsilateral hemispheres at 24 hours after H / I. The results of reverse transcription-PCR (RT-PCR) and Western blot analysis on cortical tissue of H / I mice showed that the level of MPO expression was much higher in TIM-3-suppressing antibody-treated mice compared to control group IgG-treated mice (FIGS. 4a and 4b). Experimental results of immunohistochemistry on the coronal surface of the ipsilateral cortex also show that MPO + Gr-1 + cells are greatly reduced by TIM-3-suppressing antibody treatment (FIG. 4c). Such a result was also confirmed by an immunohistochemical experiment using an anti-neutrophil and an anti-MPO antibody. Also, the effect of TIM-3 inhibition on neutrophil infiltration was measured at various time points using the coronal plane of many ipsilateral regions of the H / I brain (bregma-2 to +2). As shown in FIGS. 4d, e, even fewer numbers of MPO + Gr-1 + in the semi-shadow cortex and striatum of mice that suppressed TIM-3 at all observation time points (1-7 days). Cells were observed.

上記の結果は、低酸素環境でTIM−3が好中球の損傷された脳への浸潤と関連していることを強く示唆する。   The above results strongly suggest that TIM-3 is associated with infiltration of neutrophils into the damaged brain in a hypoxic environment.

TIM−3の遮断による好中球補充(recruitment)の減少
膠細胞TIM−3が好中球の移動に及ぼす影響をさらに特異的に測定するために、低酸素環境で膠細胞が好中球を補充する能力がTIM−3の遮断によって影響を受けるか否かを調査した。トランスウェル(Transwell)システムを利用して、1次膠細胞(primary glial cells)を下側チャンバにプレーティングし、TIM−3−抑制抗体または対照群IgGで前処理した後、上側チャンバに脾臓細胞(splenocytes)をローディングした。1%の酸素条件で24時間の間細胞を培養し、下側チャンバにあるGr−1highCD11bhigh細胞の割合をFACS分析で測定した。その結果、低酸素環境で下側チャンバにあるGr−1highCD11bhigh細胞が、対照群IgGに比べて、10mgのTIM−3−抑制抗体によって非常に減少したことを確認した(図5a)。
上記の結果をさらに検証するために、低酸素環境で骨髄(BM)由来のGr−1highCD11bhigh細胞の移動を調査した。Gr−1highCD11bhigh
細胞をBM細胞から分離して上側チャンバにプレーティングし、下側チャンバには、1%の酸素条件でTIM−3−抑制抗体または対照群IgG−処理された1次混合神経膠細胞(primary mixed glial cells)をローディングした。上記の結果と一致するように、BM由来のGr−1highCD11bhigh細胞の下側チャンバへの移動は、対照群IgG処理に比べてTIM−3−抑制抗体処理によって非常に減少した(図5b)。このような結果は、脳虚血後、低酸素領域に好中球が補充されるにあたって膠細胞TIM−3の役割を明確に示す。
Reduction of neutrophil recruitment due to TIM-3 blockade To further specifically measure the effect of TIM-3 on neutrophil migration, glial cells in the hypoxic environment It was investigated whether the ability to replenish was affected by TIM-3 blockade. Using a Transwell system, primary glial cells are plated in the lower chamber, pretreated with TIM-3-suppressing antibody or control IgG, and then spleen cells in the upper chamber (Splenocytes) was loaded. Cells were cultured for 24 hours under 1% oxygen conditions, and the percentage of Gr-1 high CD11b high cells in the lower chamber was determined by FACS analysis. As a result, it was confirmed that Gr-1 high CD11b high cells in the lower chamber in a hypoxic environment were greatly reduced by 10 mg of TIM-3-suppressor antibody compared to control group IgG (FIG. 5a).
In order to further verify the above results, migration of bone marrow (BM) -derived Gr-1 high CD11b high cells in a hypoxic environment was investigated. Gr-1 high CD11b high
Cells were separated from BM cells and plated in the upper chamber, and the lower chamber contained TIM-3-suppressor antibody or control group IgG-treated primary mixed glia in 1% oxygen conditions. glial cells) was loaded. Consistent with the above results, migration of BM-derived Gr-1 high CD11b high cells to the lower chamber was greatly reduced by TIM-3-suppressor antibody treatment compared to control IgG treatment (FIG. 5b). ). These results clearly show the role of glial TIM-3 in neutrophil recruitment to the hypoxic region after cerebral ischemia.

TIM−3抑制による好中球走化因子(chemoattractants)の減少
好中球の炎症または損傷部位への浸潤は、化学走性因子(chemoattractants)によって調節され、これらは虚血後脳の好中球浸潤に先立って上向き調節される(非特許文献23)。従って、本発明者らは、TIM−3抑制が虚血状態の脳で好中球化学走性因子として作用するIL−1β及びCXCL1の水準に及ぼす影響を調査した(非特許文献24)。H/I後、30分になった時、マウスに100mgのTIM−3−抑制抗体または対照群IgGを静脈注射した。24時間後に、同側性及び対側性皮質組織でIL−1β及びCXCL1転写水準を調査した。図5c、dに示すように、対照群IgGを注射したH/Iマウスの同側性皮質領域でIL−1β及びCXCL1の転写体水準が全て非常に増加したが、このような効果は、TIM−3−抑制抗体を注射したマウスでは非常に減少した。
Decrease of neutrophil chemotactics by TIM-3 suppression Neutrophil infiltration or infiltration of the site of injury is regulated by chemoattractants, which are neutrophils in the postischemic brain Prior to infiltration, it is adjusted upward (Non-patent Document 23). Therefore, the present inventors investigated the influence of TIM-3 inhibition on the levels of IL-1β and CXCL1 that act as neutrophil chemotactic factors in ischemic brain (Non-patent Document 24). At 30 minutes after H / I, mice were injected intravenously with 100 mg TIM-3-suppressor antibody or control group IgG. After 24 hours, IL-1β and CXCL1 transcription levels were examined in ipsilateral and contralateral cortical tissues. As shown in FIGS. 5c, d, all transcript levels of IL-1β and CXCL1 were greatly increased in the ipsilateral cortical regions of H / I mice injected with control IgG, but this effect is -3- Much decreased in mice injected with inhibitory antibodies.

膠細胞TIM−3の役割をさらに調べるために、TIM−3の遮断がIL−1β及びCXCL1発現水準に及ぼす影響を調査した。前記細胞にTIM−3−抑制抗体または対照群IgGを処理し、1%の酸素または20%の酸素条件下で24時間の間培養した。上記の結果と一致するように、20%の酸素条件に比べて1%の酸素条件で培養したIgG−処理対照群細胞でIL−1β及びCXCL1転写体の水準は増加したが、このような増加は、TIM−3−抑制抗体を処理した細胞で非常に減少した(図5e、f)。   To further investigate the role of glial TIM-3, the effect of TIM-3 blockade on IL-1β and CXCL1 expression levels was investigated. The cells were treated with TIM-3-suppressor antibody or control group IgG and cultured for 24 hours under 1% oxygen or 20% oxygen conditions. Consistent with the above results, the levels of IL-1β and CXCL1 transcripts increased in IgG-treated control cells cultured in 1% oxygen compared to 20% oxygen, but this increase Was greatly reduced in cells treated with TIM-3-suppressor antibody (FIGS. 5e, f).

このような結果は、細胞TIM−3が好中球の浸潤の調節を通じて脳虚血症の発病に重要な役割をする因子であることを示す。   These results indicate that cell TIM-3 is a factor that plays an important role in the pathogenesis of cerebral ischemia through the regulation of neutrophil infiltration.

HIF−1欠乏による好中球の移動及び梗塞(infarct)の減少
低酸素環境の神経膠細胞でHIF−1αがTIM−3の発現を調節するという発見に基づいて、本発明者らは、HIF−1αが低酸素環境で神経膠細胞の好中球の補充能力に影響を及ぼすか否かを調査した。HIF−1α+f/+fマウスから培養した1次混合神経膠細胞をAd−GFPまたはAd−GFP/Creで感染させ、トランスウェル(Transwell)システムで脾臓細胞(splenocytes)と共に1%または20%の酸素条件で24時間の間培養した。低酸素環境で下側チャンバのGr−1highCD11bhigh細胞の割合は、脾臓細胞をAd−GFP/Cre感染されたHIF−1α−欠乏神経膠細胞と共に培養した時、対照群Ad−GFP−感染細胞に比べて非常に減少した。一方、20%の酸素条件で移動したGr−1highCD11bhigh細胞の数は、HIF−1α−欠乏及び正常細胞の間に大きな差がなかった(図6a)。次に、本発明者らは、移動したBM−由来Gr−1highCD11bhigh細胞の数が、1%の酸素条件でHIF−1α−欠乏神経膠細胞と共に培養することによって非常に減少したことを発見した(図6b)。また、対照群Ad−GFP−感染細胞に比べて、TIM−3の低酸素−依存的増加が表れないAd−GFP/Cre−感染されたHIF−1α−欠乏神経膠細胞でIL−1β及びCXCL1の低酸素−依存的増加は非常に減少した(図6c、d)。
HIF-1 deficiency reduces neutrophil migration and infarcts Based on the discovery that HIF-1α regulates TIM-3 expression in hypoxic glial cells, we have developed HIF-1 It was investigated whether -1α affects neutrophil recruitment ability of glial cells in a hypoxic environment. Primary mixed glial cells cultured from HIF-1α + f / + f mice were infected with Ad-GFP or Ad-GFP / Cre and 1% or 20% oxygen with splenocytes in a transwell system The culture was performed for 24 hours under the above conditions. The percentage of Gr-1 high CD11b high cells in the lower chamber in a hypoxic environment was determined by the control group Ad-GFP-infection when spleen cells were cultured with Ad-GFP / Cre-infected HIF-1α-deficient glial cells. It was greatly reduced compared to cells. On the other hand, the number of Gr-1 high CD11b high cells that migrated under 20% oxygen conditions was not significantly different between HIF-1α-deficient and normal cells (FIG. 6a). Next, we found that the number of migrated BM-derived Gr-1 high CD11b high cells was greatly reduced by culturing with HIF-1α-deficient glial cells in 1% oxygen condition. Discovered (Figure 6b). Also, IL-1β and CXCL1 in Ad-GFP / Cre-infected HIF-1α-deficient glial cells that do not show hypoxia-dependent increase in TIM-3 compared to control group Ad-GFP-infected cells. The hypoxia-dependent increase in was greatly reduced (FIGS. 6c, d).

小膠細胞(microglia)は、脳で常在骨髄細胞(residentmyeloid cells)となることが知られている(非特許文献25)。膠細胞HIF−1αの役割を確認するために、本発明者らは、骨髄細胞で特異的にHIF−1αが欠けたLysMCre−HIF−1α+f/+f(LysM−Hif−1α−/−)マウスでH/I後の脳損傷の程度を調査した。先ず、本発明者らは、LysM−Hif−1α−/−マウスの1次小膠細胞でHIF−1αの水準を測定した。図7aに示すように、HIF−1α転写体の水準は、HIF−1α+f/+fに比べてLysM−Hif−1α−/−マウスの小膠細胞で非常に低かった。H/I後、24時間になった時、TIM−3転写体の水準もLysM−Hif−1α−/−マウスの東側皮質領域でさらに低かった(図7b)。本発明者らは、HIF−1α+f/+fマウスに比べてLysM−Hif−1α−/−マウスでTTC染色−陰性領域が非常に減少したことを発見し、これは、H/Iの24時間後、脳損傷で小膠細胞HIF−1αの役割を表す(図7c)。HIF−1α+f/+fマウスに比べてLysM−Hif−1α−/−マウスのニューロン細胞でカスパーゼ(caspase)−3の発現も非常に減少した(図7d)。さらに、H/Iの24時間後、LysM−Hif−1α−/−マウスの同側性皮質でIL−1β及びCXCL1発現の有意味な増加は検出されなかった。 It is known that microglia become resident myeloid cells in the brain (Non-patent Document 25). In order to confirm the role of glial cell HIF-1α, the inventors of the present invention used LysMCre-HIF-1α + f / + f (LysM-Hif-1α − / − ) mice specifically lacking HIF-1α in bone marrow cells. The degree of brain damage after H / I was investigated. First, the present inventors measured the level of HIF-1α in primary microglia of LysM-Hif-1α − / − mice. As shown in FIG. 7a, the levels of HIF-1α transcripts were very low in LysM-Hif-1α − / − mouse microglia compared to HIF-1α + f / + f . At 24 hours after H / I, the level of TIM-3 transcripts was also lower in the eastern cortical region of LysM-Hif-1α − / − mice (FIG. 7b). We found that the TTC staining-negative region was greatly reduced in LysM-Hif-1α − / − mice compared to HIF-1α + f / + f mice, which was 24 hours of H / I. Later, it represents the role of microglia HIF-1α in brain injury (FIG. 7c). The expression of caspase-3 was also greatly reduced in LysM-Hif-1α − / − mouse neuron cells compared to HIF-1α + f / + f mice (FIG. 7d). Furthermore, no significant increase in IL-1β and CXCL1 expression was detected in the ipsilateral cortex of LysM-Hif-1α − / − mice 24 hours after H / I.

このような結果は、低酸素症でHIF−1αがTIM−3−関連の好中球の浸潤及び繋がる脳損傷と密接な関連があることを示す。   These results indicate that in hypoxia, HIF-1α is closely associated with TIM-3-related neutrophil infiltration and linked brain damage.

TIM−3の遮断及びHIF−1αの欠乏がNDSに及ぼす影響
減少された梗塞(infarct)の体積及びニューロン細胞の死滅が神経機能の改善と連関するか否かを調べるために、公知の方法を使用してH/IモデルでNDS(neurological deficit score)を測定した(非特許文献26;及び非特許文献27)。神経学的後遺症(neurological deficits)は、対側性胴体(contralateral torso)と前肢の屈折(flexion)、対側への回転(circling to the contralateral side)、停止期の対側への偏向(leaning to the contralateral side at rest)、及び自発的運動活動(spontaneous motor activity)によって測定した。H/Iによる神経学的後遺症は、IgG−処理マウスに比べて、TIM−3−抑制抗体を処理したマウスで減少した。H/Iの20時間後に、IgG処理マウスに対するNDSは2.8±0.8(±s.d.)であったことに対し、TIM−3−抑制抗体処理マウスに対するNDSは0.8±0.8であった(表1;P=0.012;Mann−Whitney U−test)。
Effects of TIM-3 blockade and HIF-1α deficiency on NDS In order to investigate whether reduced infarct volume and neuronal cell death are associated with improved neurological function, known methods are used. The NDS (neurologic defect score) was measured using the H / I model (Non-Patent Document 26; and Non-Patent Document 27). Neurological sequelae are contralateral torso and forelimb flexion, circling to the contralateral side, leaning to the contralateral side. It was measured by the lateral side at rest, and spontaneous motor activity (spontaneous motor activity). Neurological sequelae due to H / I were reduced in mice treated with TIM-3-suppressing antibodies compared to IgG-treated mice. NDS for IgG treated mice was 2.8 ± 0.8 (± sd) 20 hours after H / I, whereas NDS for TIM-3-suppressed antibody treated mice was 0.8 ± 0.8. 0.8 (Table 1; P = 0.012; Mann-Whitney U-test).

Figure 2017057173
Figure 2017057173

このような結果は、TIM−3が低酸素環境で神経機能と関連があることを示す。次に、本発明者らは、HIF−1α+f/+fマウス(n=10)及びLysM−Hif−1α−/−マウス(n=11)に対して、H/I後24時間になった時、NDSを測定した。HIF−1α+f/+fマウスでは偏向(leaning)行動と自発的運動機能の不在が観察されたが、LysM−Hif−1α−/−マウスでは観察されなかった。LysM−Hif−1α−/−マウスにおける平均NDSは、HIF−1α+f/+fマウスより非常に低かった(表2;1.2±0.6 vs.2.6±1.1、P=0.0008) Such results indicate that TIM-3 is associated with neural function in a hypoxic environment. Next, the present inventors, for HIF-1α + f / + f mice (n = 10) and LysM-Hif-1α − / − mice (n = 11), when 24 hours after H / I NDS was measured. HIF-1α + f / + f mice were observed to have leaning behavior and absence of spontaneous motor function, but not to LysM-Hif-1α − / − mice. Mean NDS in LysM-Hif-1α − / − mice was much lower than in HIF-1α + f / + f mice (Table 2; 1.2 ± 0.6 vs. 2.6 ± 1.1, P = 0) .0008)

Figure 2017057173
Figure 2017057173

このような結果は、HIF−1α/TIM−3軸(axis)が脳梗塞体積及び病態生理学的炎症反応だけでなく、神経機能とも密接に関連していることを示す。   These results indicate that the HIF-1α / TIM-3 axis (axis) is closely related not only to cerebral infarct volume and pathophysiological inflammatory response, but also to neurological function.

HIF−1α−欠乏マウスでTIM−3による神経損傷の増加
本発明者らは、TIM−3がH/I後にHIF−1α−欠乏マウスの形質に影響を及ぼし得るか否かを実験した。このため、TIM−3及びGFPを発現するレンチウイルスベクター(LV−TIM3−GFP)を製作した。先ず、レンチウイルスが神経膠細胞を感染できるか否かを調査した後、レンチウイルス−注射マウスのGFP−陽性−CD11bhighCD45low神経膠細胞でTIM−3の発現が非常に増加したことを観察した。脳固定装置(stereotaxic instrument)を利用してウイルスをLysM−Hif−1α−/−マウスの右側半球に注射した。対照群マウスには、GFPのみを発現するLV−GFPを注射した。それぞれのマウスの右側半球に4回の頭蓋内注射(intracranial injection)を行った(図8a)。H/Iは、LysM−Hif−1α−/−マウスにLV−TIM3−GFPまたはLV−GFPを注射し、5日後に誘導し、梗塞大きさ(infarct size)及び神経学的結果は、24時間後に調査した。図8b、cに示すように、対照群LV−GFP−注射マウス(n=6)に比べて、LV−TIM3−GFP注射マウス(n=5)でTTC−染色−陰性領域が非常に増加した。また、LV−TIM3−GFPを注射したLysM−Hif−1α−/−マウスに対する平均NDSは、LV−GFP−注射対照群マウスより高かった(図8d)(1.1±0.7 vs.2.3±0.8、P=0.046)。このような結果は、低酸素環境でHIF−1/TIM−3軸と脳損傷の関連性を再度示す結果である。
Increased nerve damage by TIM-3 in HIF-1α-deficient mice We investigated whether TIM-3 could affect the traits of HIF-1α-deficient mice after H / I. Therefore, a lentiviral vector (LV-TIM3-GFP) expressing TIM-3 and GFP was produced. First, after investigating whether lentivirus can infect glial cells, we observed that TIM-3 expression was greatly increased in GFP-positive-CD11b high CD45 low glial cells of lentivirus-injected mice did. The virus was injected into the right hemisphere of LysM-Hif-1α − / − mice using a stereotaxic instrument. Control group mice were injected with LV-GFP expressing only GFP. Four intracranial injections were made in the right hemisphere of each mouse (Figure 8a). H / I was injected into LysM-Hif-1α − / − mice with LV-TIM3-GFP or LV-GFP and induced 5 days later, the infarct size and neurological results were 24 hours I investigated later. As shown in FIGS. 8b and c, the TTC-stained-negative region was greatly increased in the LV-TIM3-GFP-injected mice (n = 5) compared to the control group LV-GFP-injected mice (n = 6). . In addition, the mean NDS for LysM-Hif-1α − / − mice injected with LV-TIM3-GFP was higher than that of LV-GFP-injected control group mice (FIG. 8d) (1.1 ± 0.7 vs. 2). .3 ± 0.8, P = 0.046). Such a result is a result showing again the relationship between the HIF-1 / TIM-3 axis and brain damage in a hypoxic environment.

TIM−3に対するshRNAを利用したTIM−3抑制活性の分析
上記で行った実施例における実験は、TIM−3に対する抗体を利用して行い、さらに、本発明者らは、TIM−3を抑制することができるまた他の方法として、TIM−3に対するshRNAの使用可能性を確認した。このため、先ず一次培養膠細胞(図10A)またはV2小膠細胞(図10B)にTIM−3に対するshRNAを発現するレンチウイルスまたは対照群レンジウイルスを製品生産会社(Santacruz #sc−72015−V)から提供された説明書に従って細胞内に感染させた。以後、感染された細胞を24時間の間1%または20%の酸素条件で培養し、逆転写重合酵素連鎖反応分析法、免疫細胞化学法及び流細胞分析法を利用してTIM−3の発現を確認し、このような実験は、3回の独立した繰返し実験から結果を得て、meanSDとして示した。
Analysis of TIM-3 inhibitory activity using shRNA against TIM-3 The experiments in the above examples were carried out using an antibody against TIM-3, and the inventors further suppressed TIM-3. As another method that could be used, the availability of shRNA against TIM-3 was confirmed. For this reason, a lentivirus expressing a shRNA against TIM-3 or a control range virus was first produced in a primary cultured glial cell (FIG. 10A) or V2 microglia (FIG. 10B) (Santacruz # sc-72015-V). Cells were infected according to the instructions provided by Thereafter, the infected cells are cultured for 24 hours under 1% or 20% oxygen condition, and TIM-3 is expressed using reverse transcriptase chain reaction analysis, immunocytochemistry and flow cytometry. These experiments were obtained as results from three independent repeated experiments and expressed as meanSD.

分析の結果、図10に示すように、本発明の実験で使用したTIM−3に対するshRNAは、対照群と比べてみると、効果的にTim−3の発現を要請することが表れ、また、低酸素条件でTIM−3に対するshRNAを処理した群の場合、対照群を処理した群に比べてTIM−3の発現増加が阻害することが表れた。   As a result of the analysis, as shown in FIG. 10, the shRNA against TIM-3 used in the experiment of the present invention was effectively required to express Tim-3 when compared with the control group, In the group treated with shRNA against TIM-3 under hypoxic conditions, it was shown that the increase in TIM-3 expression was inhibited compared to the group treated with the control group.

従って、このような結果からみると、TIM−3の発現または活性を阻害し得るTIM−3に対する抗体またはshRNAを含むTIM−3阻害剤は、TIM−3の発現または活性を効果的に阻害することができることが表れ、よって、このような阻害剤を脳損傷疾患の予防または治療のための製剤として使用可能なことが分かった。   Therefore, in view of these results, a TIM-3 inhibitor comprising an antibody or shRNA against TIM-3 that can inhibit TIM-3 expression or activity effectively inhibits TIM-3 expression or activity. Thus, it has been found that such inhibitors can be used as formulations for the prevention or treatment of brain injury diseases.

これまで本発明についてその好ましい実施例を中心として検討した。本発明が属する技術分野で通常の知識を持った者は、本発明が本発明の本質的な特性から逸脱しない範囲で変形された形態で具現可能なことが理解できるであろう。従って、開示された実施例は、限定的な観点ではなく、説明的な観点で考慮されるべきである。本発明の範囲は、前述した説明ではなく、特許請求の範囲に表れており、それと同等な範囲内にある全ての差異点は、本発明に含まれると解釈されなければならない。   So far, the present invention has been studied focusing on its preferred embodiments. Those skilled in the art to which the present invention pertains will understand that the present invention can be embodied in a modified form without departing from the essential characteristics of the present invention. Accordingly, the disclosed embodiments are to be considered in an illustrative rather than a restrictive perspective. The scope of the present invention is shown not in the above description but in the claims, and all differences within the equivalent scope should be construed as being included in the present invention.

Claims (13)

TIM−3(T−cell immunoglobulin and mucin domain protein 3)抑制剤を有効成分として含む脳損傷疾患の予防または治療用薬学的組成物。   A pharmaceutical composition for preventing or treating a brain injury disease comprising a TIM-3 (T-cell immunoglobulin and mucin domain protein 3) inhibitor as an active ingredient. 前記TIM−3抑制剤は、TIM−3の発現または活性を特異的に抑制または減少させることができる、化合物、タンパク質、抗体、ペプチドまたは核酸であることを特徴とする請求項1に記載の組成物。   The composition according to claim 1, wherein the TIM-3 inhibitor is a compound, protein, antibody, peptide, or nucleic acid capable of specifically suppressing or decreasing TIM-3 expression or activity. object. 前記抗体は、序列番号1のアミノ酸序列で表されるTIM−3に結合するか、これと反応して、TIM−3の活性を特異的に抑制または減少させる拮抗抗体またはその断片であることを特徴とする請求項2に記載の組成物。   The antibody is an antagonistic antibody or a fragment thereof that binds to or reacts with TIM-3 represented by the amino acid sequence of SEQ ID NO: 1 and specifically suppresses or decreases the activity of TIM-3. A composition according to claim 2 characterized. 前記核酸は、序列番号2の塩基序列で表されるTIM−3遺伝子またはその断片に対するアンチセンスヌクレオチド、siRNA、shRNA、またはmiRNAであることを特徴とする請求項2に記載の組成物。   The composition according to claim 2, wherein the nucleic acid is an antisense nucleotide, siRNA, shRNA, or miRNA against the TIM-3 gene represented by the base sequence of sequence number 2 or a fragment thereof. 前記TIM−3抑制剤は、HIF−1(hypoxia−inducible factor−1)の発現または活性を抑制させることを特徴とする請求項1に記載の組成物。   The composition according to claim 1, wherein the TIM-3 inhibitor suppresses the expression or activity of HIF-1 (hypoxia-inducible factor-1). 前記TIM−3抑制剤は、好中球走化因子(neutrophil chemotactic factor)の発現または活性を減少させることを特徴とする請求項1に記載の組成物。   2. The composition of claim 1, wherein the TIM-3 inhibitor decreases the expression or activity of a neutrophil chemotactic factor. 前記脳損傷疾患は、脳梗塞、脳卒中、低酸素性脳損傷、虚血性脳疾患及び中風からなる群から選択されることを特徴とする請求項1ないし6のいずれか一項に記載の組成物。   The composition according to any one of claims 1 to 6, wherein the brain injury disease is selected from the group consisting of cerebral infarction, stroke, hypoxic brain injury, ischemic brain disease and medium wind. . (a)TIM−3が発現される細胞または動物モデルに候補物質を処理する段階と、
(b)前記候補物質処理後、TIM−3の発現または活性程度を測定する段階と、
(c)前記TIM−3の発現または活性程度が候補物質を処理しない対照群に比べて減少した候補物質を選別する段階とを含む脳損傷疾患治療剤のスクリーニング方法。
(A) treating a candidate substance in a cell or animal model in which TIM-3 is expressed;
(B) measuring the expression or activity level of TIM-3 after the candidate substance treatment;
(C) screening a candidate substance for treating a brain injury disease, comprising selecting a candidate substance having a decreased expression or activity level of TIM-3 as compared to a control group not treating the candidate substance.
前記(b)段階の測定は、免疫組織化学染色、PCR、RT−PCR、ウエスタンブロット、ELISA、またはタンパク質チップで構成された群から選択される方法で行うことを特徴とする請求項8に記載の方法。   The measurement in the step (b) is performed by a method selected from the group consisting of immunohistochemical staining, PCR, RT-PCR, Western blot, ELISA, or protein chip. the method of. 前記選別した候補物質が対照群に比べてHIF−1の発現または活性を抑制させるか否かを追加で分析する段階をさらに含むことを特徴とする請求項8に記載の方法。   9. The method according to claim 8, further comprising the step of additionally analyzing whether the selected candidate substance suppresses the expression or activity of HIF-1 compared to the control group. 前記細胞は、神経膠細胞(glial cell)であることを特徴とする請求項8に記載の方法。   9. The method of claim 8, wherein the cell is a glial cell. 前記動物モデルは、低酸素虚血性(hypoxia−ischemia)脳損傷疾患モデルであることを特徴とする請求項8に記載の方法。   9. The method of claim 8, wherein the animal model is a hypoxia-ischemia brain injury disease model. 前記脳損傷疾患は、脳梗塞、脳卒中、低酸素性脳損傷、虚血性脳疾患及び中風からなる群から選択されることを特徴とする請求項8ないし12のいずれか一項に記載の方法。   13. The method according to any one of claims 8 to 12, wherein the brain injury disease is selected from the group consisting of cerebral infarction, stroke, hypoxic brain injury, ischemic brain disease and medium wind.
JP2015184356A 2015-09-17 2015-09-17 Composition for treating brain injury disease targeting TIM-3 and screening method thereof Active JP6653054B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2015184356A JP6653054B2 (en) 2015-09-17 2015-09-17 Composition for treating brain injury disease targeting TIM-3 and screening method thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
JP2015184356A JP6653054B2 (en) 2015-09-17 2015-09-17 Composition for treating brain injury disease targeting TIM-3 and screening method thereof

Publications (2)

Publication Number Publication Date
JP2017057173A true JP2017057173A (en) 2017-03-23
JP6653054B2 JP6653054B2 (en) 2020-02-26

Family

ID=58391074

Family Applications (1)

Application Number Title Priority Date Filing Date
JP2015184356A Active JP6653054B2 (en) 2015-09-17 2015-09-17 Composition for treating brain injury disease targeting TIM-3 and screening method thereof

Country Status (1)

Country Link
JP (1) JP6653054B2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021177448A1 (en) * 2020-03-06 2021-09-10 公益財団法人神戸医療産業都市推進機構 Screening method

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011155607A1 (en) * 2010-06-11 2011-12-15 協和発酵キリン株式会社 Anti-tim-3 antibody

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011155607A1 (en) * 2010-06-11 2011-12-15 協和発酵キリン株式会社 Anti-tim-3 antibody

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021177448A1 (en) * 2020-03-06 2021-09-10 公益財団法人神戸医療産業都市推進機構 Screening method

Also Published As

Publication number Publication date
JP6653054B2 (en) 2020-02-26

Similar Documents

Publication Publication Date Title
Koh et al. The HIF-1/glial TIM-3 axis controls inflammation-associated brain damage under hypoxia
Bellelli et al. Polε instability drives replication stress, abnormal development, and tumorigenesis
US9725719B2 (en) Compositions and methods for inhibiting NF-κB and SOD-1 to treat amyotrophic lateral sclerosis
Gross et al. Loss of slug compromises DNA damage repair and accelerates stem cell aging in mammary epithelium
US11779600B2 (en) Methods of treating or preventing alzheimer&#39;s disease and associated conditions
JP2015529685A (en) Compositions and methods for the treatment of amyotrophic lateral sclerosis
WO2014111876A2 (en) Modulation of mitophagy and use thereof
Zhan et al. A DEAD‐box RNA helicase Ddx54 protein in oligodendrocytes is indispensable for myelination in the central nervous system
Wang et al. SIRPα deficiency accelerates the pathologic process in models of Parkinson disease
WO2016172955A1 (en) Method for screening drug and therapeutic target used for treating alzheimer&#39;s disease
US20190111111A1 (en) Treatment of Cerebral Cavernous Malformations
KR101785155B1 (en) Composition for the treatment of brain injury targeting TIM-3 and screening method thereof
CA2733775A1 (en) Agent for promoting neuronal differentiation and method therefor
US11939633B2 (en) COTL1 protein involved in maintaining homeostasis of hematopoietic stem cell, and use thereof
JP6653054B2 (en) Composition for treating brain injury disease targeting TIM-3 and screening method thereof
EP3622958B1 (en) Use of potassium ion channel inhibitor for treatment of depression and pharmaceutical composition
CA2975101A1 (en) Compounds, compositions, and methods for using hla-f
WO2017168390A2 (en) Attenuation of neurodegeneration associated with parkinson&#39;s disease by inhibition of the dopamine d3 receptor in cd4+ t cells
WO2022186089A1 (en) Polynucleotide for treatment of neurodegenerative disease, vector, cell, pharmaceutical composition, and screening method
WO2022032592A1 (en) Interleukin-17d and cd93 as a new cytokine-receptor pair in the immune system
Jensen et al. Astrocytic expression of ALS-causative mutant FUS leads to TNFα-dependent neurodegeneration in vivo
US20160304881A1 (en) Ddr1 antagonist or an inhibitor of ddr1 gene expression for use in the prevention or treatment of crescentic glomerulonephritis
US20210163555A1 (en) TARGETING P18 FOR mTOR-RELATED DISORDERS
ALRASHDI CONTRIBUTION OF THE VOLTAGE-GATED SODIUM CHANNEL NAV1. 6 AND TOLL LIKE RECEPTOR 2 IN THE PATHOPHYSIOLOGY OF EAE
Gallizioli Identity and functions of dendritic cell subsets in ischaemia-induced neuroinflammation

Legal Events

Date Code Title Description
A80 Written request to apply exceptions to lack of novelty of invention

Free format text: JAPANESE INTERMEDIATE CODE: A80

Effective date: 20151013

A621 Written request for application examination

Free format text: JAPANESE INTERMEDIATE CODE: A621

Effective date: 20180905

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A131

Effective date: 20190716

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20191016

TRDD Decision of grant or rejection written
A01 Written decision to grant a patent or to grant a registration (utility model)

Free format text: JAPANESE INTERMEDIATE CODE: A01

Effective date: 20191210

A61 First payment of annual fees (during grant procedure)

Free format text: JAPANESE INTERMEDIATE CODE: A61

Effective date: 20200107

R150 Certificate of patent or registration of utility model

Ref document number: 6653054

Country of ref document: JP

Free format text: JAPANESE INTERMEDIATE CODE: R150

R250 Receipt of annual fees

Free format text: JAPANESE INTERMEDIATE CODE: R250

R250 Receipt of annual fees

Free format text: JAPANESE INTERMEDIATE CODE: R250