JP2004166590A - Method for screening inhibitor of oxidized purine nucleoside triphosphatase - Google Patents

Method for screening inhibitor of oxidized purine nucleoside triphosphatase Download PDF

Info

Publication number
JP2004166590A
JP2004166590A JP2002336181A JP2002336181A JP2004166590A JP 2004166590 A JP2004166590 A JP 2004166590A JP 2002336181 A JP2002336181 A JP 2002336181A JP 2002336181 A JP2002336181 A JP 2002336181A JP 2004166590 A JP2004166590 A JP 2004166590A
Authority
JP
Japan
Prior art keywords
mth1
cell
cells
purine nucleoside
inhibitor
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
JP2002336181A
Other languages
Japanese (ja)
Other versions
JP3910525B2 (en
Inventor
Yusaku Nakabeppu
雄作 中別府
Masato Furuichi
正人 古市
Kunihiko Sakumi
邦彦 作見
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.)
Japan Science and Technology Agency
Original Assignee
Japan Science and Technology Agency
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 Japan Science and Technology Agency filed Critical Japan Science and Technology Agency
Priority to JP2002336181A priority Critical patent/JP3910525B2/en
Publication of JP2004166590A publication Critical patent/JP2004166590A/en
Application granted granted Critical
Publication of JP3910525B2 publication Critical patent/JP3910525B2/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Images

Landscapes

  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

<P>PROBLEM TO BE SOLVED: To provide a new assaying system by which a chemical inhibiting an oxidized purine nucleoside triphosphatase (MTH1) is easily and precisely screened. <P>SOLUTION: The screening method comprises adding a test agent to a cell obtained by forcibly expressing human MTH1 in a fibroblast of a fetus of an MTH1-deficient knockout mouse, culturing the cell in the presence of hydrogen peroxide, and selecting the test agent as the MTH1 inhibitor when death of the cell is recognized. The problem that a chemical having no cell membrane permeability is selected, as shown in a cell-free screening system is eliminated by the screening method. <P>COPYRIGHT: (C)2004,JPO

Description

【0001】
【発明の属する技術分野】
本発明は、薬剤を開発するための技術分野に属し、特に、抗癌剤となり得る酸化プリンヌクレオシド三リン酸分解酵素阻害剤をスクリーニングする方法に関する。
【0002】
【従来の技術】
がん治療において化学療法は重要なアプローチの一つであり、現在臨床で用いられている抗癌剤は50種以上にものぼるが、未だ充分ではなく、新しい抗癌剤の開発が日夜続けられている。抗癌剤の効率的な開発のためには、本格的な薬理学的試験や臨床試験の前に、学術的検討から明らかにされた知見に基づき、抗癌剤の候補となり得る各種の生理活性物質を予めスクリーニングすることのできる系を確立することが必要である。
【0003】
酸化プリンヌクレオシド三リン酸分解酵素(一般に、MTH1またはNUDT1と略称される。本明細書においてもMTH1と略称することがある)は、酸化されたプリンヌクレオシド三リン酸(8−oxo−dGTP,2−OH−dATP,2−OH−ATP等)を一リン酸に分解する活性を有することから、酸化核酸前駆体が複製や転写の際にゲノムDNAやRNAに取り込まれる結果引き起こされる突然変異や翻訳エラーを回避することにより遺伝情報の維持に重要な役割を持つと考えられている。事実、Mth1ノックアウトマウスでは肝細胞がんを始めとする複数の臓器の自然発癌頻度の増加が認められている(T. Tsuzuki, Y. Nakabeppu他、Proc. Natl. Acad. Sci. USA 98, 11456−11461 (2001)(非特許文献1)。一方、MTH1は、脳腫瘍細胞において8−オキソデオキシグアノシン(8−oxo−dG)の細胞内蓄積の上昇とともに高発現することも認められている(T. Iida, Y. Nakabeppu他、Neuro−Oncology, April, 73−81 (2001))。これらの事実から、MTH1は遺伝情報の維持から腫瘍発生を抑制するのみならず、酸化ストレスを受けた細胞の機能障害や細胞死の回避に関わる可能性が示唆される。
【0004】
したがって、MTH1を阻害する薬剤は癌細胞の持つ酸化ストレス抵抗性を抑制することが予想され、他の抗癌剤との併用によりその抗癌剤の用量の軽減や、放射線療法時の照射線量の軽減、さらにこれらの療法に抵抗性の癌の化学療法及び放射線療法に応用できるものと期待されるが、そのような薬剤を簡便且つ確実にスクリーニングすることができる系は未だ見出されていない。特に、MTH1は、試験管内では酸化プリンヌクレオシド二リン酸のような物質により効率よく阻害されることは知られているが、酸化プリンヌクレオシド二リン酸は細胞膜を透過できないために薬剤としては使用できない。すなわち、MTH1を阻害する薬剤を判別するにあたり、無細胞スクリーニング系を用いることは、細胞膜透過性を有しない薬剤が得られるという問題がある。
【非特許文献1】T. Tsuzuki, Y. Nakabeppu他、Proc. Natl. Acad. Sci.USA 98, 11456−11461 (2001)
【非特許文献2】T. Iida, Y. Nakabeppu他、Neuro−Oncology, April, 73−81 (2001)
【0005】
【発明が解決しようとする課題】
本発明の目的は、酸化プリンヌクレオシド三リン酸分解酵素(MTH1)を阻害する薬剤を簡便且つ確実にスクリーニングすることのできる新しいアッセイ系を提供することにある。
【0006】
【課題を解決するための手段】
本発明者は、研究を重ねた結果、MTH1が発現しないと細胞死に陥る細胞系に着目し、これを利用することにより上述の目的を達成し得る本発明を案出したものである。
かくして、本発明に従えば、酸化プリンヌクレオシド三リン酸分解酵素(MTH1)を阻害する薬剤のスクリーニングが可能となり、Mth1欠損ノックアウトマウス胎仔の線維芽細胞にヒトMTH1を強制発現させた細胞に被験物質を添加し、過酸化水素の存在下に細胞を培養して、その細胞死が認められたときに被験物質をMTH1阻害剤として選択することを特徴とする方法が提供される。
【0007】
【発明の実施の形態】
本発明が対象とする酸化プリンヌクレオシド三リン酸分解酵素MTH1は、哺乳動物における核酸の酸化損傷の防御機構に関与する酵素の1種であり、そのcDNAと遺伝子は本発明者らによりクローニングされ、大腸菌のMutT蛋白質の配列とホモロジーを有することからMTH1(mutT homolog 1)と命名された(H. Oda, Y. Nakabeppu他、Nucleic Acids Res., 27, 4335 (1999))。
【0008】
本発明者らは、このMth1遺伝子が欠損したノックアウトマウスを用いて各種臓器の自然発癌性に関する研究を行なう(前記の非特許文献1)とともに、MTH1欠損ノックアウトマウスの胎仔の線維芽細胞にヒトMTH1(hMTH1)を強制発現させた細胞株を樹立し、過酸化水素負荷条件下の細胞の機能障害や細胞死に注目して解析を行なった。その結果、hMTH1強制発現株は非発現株と比較して有意に高い生存率を示すことを見出し、この現象がMTH1(ヒトMTH1)を阻害する薬剤の当否判別に利用できるものと考え、本発明を導き出したものである。
【0009】
以下、本発明に従うMTH1阻害剤のスクリーニング法において用いられる細胞株の由来するMTH1欠損ノックアウトマウスの作製法、その胎児線維芽細胞株およびhMTH1過剰発現株の樹立法、ならびに、該細胞株を用いるスクリーニング法に沿って本発明の実施の形態を説明する。但し、以下に示す具体的な実施の形態は例示のためのものであり、本発明の実施の形態はこれに限定されるものではなく、当業者であれば同等の材料と手法を用いて同等のものを作製できることは勿論である。
【0010】
MTH1 欠損マウスの作製
MTH1が欠損したノックアウトマウスは、ES細胞と遺伝子ターゲッティングを用いる常法に従って作製することができ、その1例は本発明者らによる上記文献(非特許文献1)にも記載されているが、以下のとおりである:
マウス129SV系由来の遺伝子ライブラリー(Stratagene社より購入、 Cat. No.946313)よりMth1遺伝子を単離しその構造を解析した。Mth1遺伝子は約 7 kbp の大きさで 5 つのエクソンより成り、第1,第2の2つのエクソンは 5’ 側非翻訳領域で、開始コドンは第3エクソンに含まれていた。ヒト、マウス及び大腸菌を含む3種の細菌由来のMutT様蛋白質間でアミノ酸配列がよく保存されている領域が認められるが、その部分はマウスの遺伝子上でエクソン3と4に分かれてコードされていた。第3エクソンをG418耐性遺伝子カセット(Deng, C., Thomas, K. R.およびCapecchi, M. R., Mol. Cell. Biol. 13: 2134−2140 (1993)参照)と置換したターゲティングベクターを構築した。ベクターの両端には、ネガティブ選別用にHSVのチミジンキナーゼ遺伝子(Rancourt, D. E., Tsuzuki, T. and Capecchi, M. R., Genes Dev. : 108−122 (1995))を配置した。
【0011】
このターゲティングベクターをエレクトロポレーション法によりES細胞に導入し、G418・ガンシクロビル耐性クローンの中からMth1の対立遺伝子の片方にG418耐性遺伝子カセットが挿入されたMth1ヘテロ欠損細胞クローン(Mth1+/−)を選択した。なお、用いたES細胞は、CCE細胞株(E. J. Robertson博士より分与:Robertson, E. J. Tetracarcinomas and Embrionic Stem Cells: A Practical Approach, IRL Press, NY (1987)参照)であり、このCCE細胞は研究目的であれば無償配布されている。同等のES細胞は、例えば、ATCCよりCRL−11632としても入手することができる。
上述のMth1+/− ES細胞をマウスの胚盤胞に導入して得られたキメラマウスをBDF1マウス(日本クレア社より購入)と交配し、Mth1+/− マウスを樹立した。Mth1+/− マウスはC57BL6マウスと12世代戻し交配を行い、その遺伝的背景を純化した。
【0012】
MTH1 ホモ欠損マウス胎仔線維芽細胞株( Mth1 −/− MEF )の樹立
MTH1欠損ノックアウトマウスからその胎仔線維芽細胞株の樹立化は、当該分野で既知の細胞培養の手法に従い次のように実施することができる:
12世代戻し交配したMth1+/− マウスの雄と雌を交配し、妊娠17.5日目のマウスを開腹、無菌的に子宮から1個体づつ胎仔を摘出した。Mth1−/−マウス胎仔より約8mm×8mmの皮膚を剥離、切除し、培養シャーレに移す。少量の培地中(DMEM (低グルコース)、10%ウシ胎仔血清)で組織片をはさみでミンチし、シャーレの培養面に広げ、COインキュベーター中で静置する。
1時間後、組織片が剥がれないように注意深く培地を添加し、数日間培養する。組織片より移行して増殖してきた繊維芽細胞を0.05%トリプシン−EDTAで剥がして回収し、新しいディッシュに移して3日おきに継代培養を継続する。30世代以上の継代の後にコロニー形成率(plating efficiency)が30%を超えた段階で株化したものとみなした。T2およびT5の2つの独立の株を樹立した。
【0013】
ヒト MTH1 を過剰発現する Mth1 −/− MEF の樹立
以上のようにして得られるMTH1ホモ欠損マウス胎仔線維芽細胞株Mth1−/−MEF(T2およびT5)に、ヒトMTH1 cDNAをサブクローニングした発現ベクター(pcDEBΔ:hMTH1)をリポフェクション法により導入し、ハイグロマイシンB耐性細胞クローンを複数分離した。単一細胞由来のコロニー形成を2回繰り返し、純化したクローンについて抗MTH1抗体を用いたウエスタンブロッティング法によりヒトMTH1の発現を確認し、2つのクローン(T2:hMTH1およびT5:hMTH1)を樹立した。コントロールとしてpcDEBΔのみを導入したT2:vectorおよびT5:vectorを樹立した。なお、上記の発現ベクター(pcDEBΔ:hMTH1)や抗MTH1抗体については、文献(Kang, D.,およびTakeshige, K.他、J. Biol. Chem. 270, 14659−14665 (1995))に詳述されている。また、ヒトMTH1 cDNAはTHE I.M.A.G.E. CONSORTIUMから入手できる。
【0014】
MTH 1阻害剤のスクリーニング法
本発明に従えば、以上のようにして樹立したMTH1欠損ノックアウトマウス胎仔線維芽細胞(Mth1−/−MEF)を利用して、MTH1を阻害する薬剤をスクリーニングすることができる。すなわち、Mth1−/−MEFにヒトMTH1を強制発現させた細胞に、MTH1阻害剤の候補となる被験物質を添加し、過酸化水素の存在下に細胞を培養して、その細胞死が認められたときに被験物質をMTH1阻害剤として選択する。
実際の操作に当たっては、ヒトMTH1を過剰発現するMth1−/−MEFを対数増殖期から適当な条件で培養(一般的には、37℃、5%CO存在下に24時間程度)した後、被験物質を添加して適当時間(例えば1〜5時間)培養し、その後、異なる濃度の過酸化水素(H)を添加し、さらに培養(一般的には24時間程度)し、細胞の生存率を測定または観察し、その細胞死が有意に認められたときに被験物質をMTH1阻害剤として選択する。用いる培地は、特殊なものではなく標準的なMEM培地でよい。また、細胞毒性の効果を軽減するためにピルビン酸を加えてもよいが、本発明のアッセイ系は一般にピルビン酸を添加しなくても測定できる。
【0015】
本発明のスクリーニング法において細胞の生存率を測定または観察する手段は、特に限定されるものではなく、細胞死を判定するための各種の手法、例えば、代謝酵素活性測定法、コロニー形成法、細胞分染法による顕微鏡観察、生合成能測定法などを用いることができる。このうち、代謝酵素活性測定法は、96穴マイクロプレートなどを用いて多数の検体を比較的簡単に測定できることから好ましい。代謝酵素活性測定法の例としては、Cell count kit WST−8 (Dojindo:株式会社同仁化学研究所製)が挙げられ、これは、テトラゾリウム塩が細胞内脱水素酵素により還元されて生じる水溶性のホルマザンの吸光度を測定することにより生細胞数を検出するタイプの測定法の1種である。
【0016】
【実施例】
本発明の特徴を更に具体的に示すために以下に実施例を記す。実施例1は、本発明のスクリーニング法の背景となる現象を明らかにするものであり、MTH1ノックアウトマウス胎仔線維芽細胞にヒトMTH1を強制発現させた細胞株は非発現株と比較して過酸化水素負荷後に有意に高い細胞生存率を有し、この系においてはMTH1が存在しないと細胞死が起こることを示すものである。実施例2は、MTH1ノックアウトマウス胎仔線維芽細胞にヒトMTH1を強制発現させた細胞株は、そのMTH1活性が、特定の物質の存在により阻害され、したがって、そのような阻害物質を選別し得るのに使用できることを示すものである。
【0017】
実施例1:過酸化水素感受性試験
対数増殖期のT5:vector(ヒトMTH1非発現株:既述)とT5:hMTH細胞(ヒトMTH1強制発現株)を96穴マイクロプレートに50%飽和になるように播種し(3x10 cells/100μl/well)、37℃、5% CO存在下に24時間培養した。用いた培地はMEM (Invitrogen 11095−080)である。次に、異なる濃度のH(10μl/well)を培地に添加し、さらに24時間培養した。Cell count Kit WST−8 (Dojindo)を10 μl/well添加し、90分から120分後に培養液の2つの吸光度の差(A450nm−A620nm)を測定した。未処理細胞の吸光度の値を1.0として相対生存率を求めた。
その結果を図1に示す。この系では、MTH1が存在しないと細胞死が起こることが理解される。
【0018】
実施例2: MTH 1阻害剤
MTH1阻害剤として硫酸銅(CuSO)を用いた。試験管内ではMTH1の8−oxo−dGTP分解活性は17μMのCu2+の存在下で50%阻害されることが知られており(Porter, D. W., Nakabeppu他、Carcinogenesis 18 (9), 1785−1791 (1997))、本発明で用いるMTH1ノックアウトマウス胎仔線維芽細胞にヒトMTH1を強制発現させた細胞のMTH1活性が、そのような阻害物質によって阻害されるか否かを調べた。
対数増殖期のT5:hMTH細胞を96穴マイクロプレートに50%飽和になるように播種し(3x10 cells/100μl/well)、37℃、5% CO存在下に24時間培養した。培地はMEM (Invitrogen 11095−080)である。次に、MTH1阻害剤(CuSO)を最終濃度50μMで培地に添加し2時間培養した。その後、異なる濃度のH(10μl/well)を培地に添加し、さらに24時間培養した。Cell count Kit WST−8 (Dojindo)を10μl/well添加し、90分から120分後に培養液の2つの吸光度の差(A450nm−A620nm)を測定した。H未処理細胞の吸光度の値を1.0として相対生存率を求めた。測定結果を図2に示す。図2に示されるように、MTH1阻害剤の存在により細胞死が認められる。
【0019】
【発明の効果】
以上の説明から明らかなように、本発明の方法を用いれば、MTH1を阻害する薬剤を簡便且つ確実にスクリーニングすることができ、無細胞スクリーニング系におけるような細胞膜透過性を有しない薬剤を選別するという問題も回避される。かくして、本発明は、MTH1阻害剤の効率的なスクリーニングを通じて抗癌剤や癌治療法の開発に資するものである。
【図面の簡単な説明】
【図1】本発明で用いられるMTH1ノックアウトマウス胎仔線維芽細胞にヒトMTH1を強制発現させた細胞株が非発現株と比較して過酸化水素負荷条件下に有意に高い細胞生存率を有することを示す。
【図2】本発明で用いられるMTH1ノックアウトマウス胎仔線維芽細胞にヒトMTH1を強制発現させた細胞株がMTH1阻害剤により細胞死を起こすことを示す。
[0001]
TECHNICAL FIELD OF THE INVENTION
The present invention belongs to the technical field for developing a drug, and particularly relates to a method for screening an oxidized purine nucleoside triphosphate degrading enzyme inhibitor that can be an anticancer agent.
[0002]
[Prior art]
Chemotherapy is one of the important approaches in cancer treatment. Currently, more than 50 kinds of anticancer drugs are being used in the clinic, but they are still insufficient, and new anticancer drugs are being developed day and night. For efficient development of anticancer drugs, various bioactive substances that can be candidates for anticancer drugs are screened in advance based on the findings revealed from academic studies before full-scale pharmacological and clinical trials It is necessary to establish a system that can do this.
[0003]
Oxidized purine nucleoside triphosphate degrading enzyme (generally abbreviated as MTH1 or NUDT1; sometimes also abbreviated as MTH1 herein) is an oxidized purine nucleoside triphosphate (8-oxo-dGTP, 2). -OH-dATP, 2-OH-ATP, etc.) into monophosphate, so that oxidized nucleic acid precursors are incorporated into genomic DNA or RNA during replication or transcription, resulting in mutation or translation. Avoiding errors is thought to play an important role in maintaining genetic information. In fact, an increase in spontaneous carcinogenesis frequency of multiple organs including hepatocellular carcinoma has been observed in Mth1 knockout mice (T. Tsuzuki, Y. Nakabeppu et al., Proc. Natl. Acad. Sci. USA 98 , 11456). -11461 (2001) (Non-Patent Document 1) On the other hand, it has also been recognized that MTH1 is highly expressed in brain tumor cells with an increase in intracellular accumulation of 8-oxodeoxyguanosine (8-oxo-dG) (T Iida, Y. Nakabeppu, et al., Neuro-Oncology, April, 73-81 (2001). These facts indicate that MTH1 not only suppresses tumor development from maintaining genetic information, but also inhibits oxidative stress in cells. Possible involvement in avoiding dysfunction and cell death
[0004]
Therefore, an agent that inhibits MTH1 is expected to suppress the oxidative stress resistance of cancer cells, and the dose of the anticancer agent can be reduced by using it in combination with other anticancer agents, and the irradiation dose during radiation therapy can be further reduced. Is expected to be applicable to chemotherapy and radiation therapy for cancer resistant to the treatment of cancer, but a system that can easily and reliably screen such a drug has not been found yet. In particular, MTH1 is known to be efficiently inhibited in vitro by substances such as oxidized purine nucleoside diphosphate, but cannot be used as a drug because oxidized purine nucleoside diphosphate cannot penetrate cell membranes. . That is, when a drug that inhibits MTH1 is determined, using a cell-free screening system has a problem that a drug that does not have cell membrane permeability can be obtained.
[Non-Patent Document 1] Tsuzuki, Y .; Nakabeppu et al., Proc. Natl. Acad. Sci. USA 98 , 11456-11461 (2001).
[Non-Patent Document 2] Iida, Y .; Nakabeppu et al., Neuro-Oncology, April, 73-81 (2001).
[0005]
[Problems to be solved by the invention]
An object of the present invention is to provide a new assay system that can easily and reliably screen for a drug that inhibits oxidized purine nucleoside triphosphate degrading enzyme (MTH1).
[0006]
[Means for Solving the Problems]
As a result of repeated studies, the present inventor has focused on a cell line that will undergo cell death if MTH1 is not expressed, and has devised the present invention that can achieve the above object by utilizing this.
Thus, according to the present invention, it is possible to screen for a drug that inhibits oxidized purine nucleoside triphosphate degrading enzyme (MTH1), and a test substance can be added to cells in which human MTH1 is forcibly expressed in fibroblasts of Mth1-deficient knockout mouse embryos. And culturing the cells in the presence of hydrogen peroxide, and when the cell death is observed, selecting a test substance as an MTH1 inhibitor.
[0007]
BEST MODE FOR CARRYING OUT THE INVENTION
The oxidized purine nucleoside triphosphate degrading enzyme MTH1 targeted by the present invention is one of enzymes involved in the defense mechanism against oxidative damage of nucleic acids in mammals, and its cDNA and gene have been cloned by the present inventors, It was named MTH1 (mutT homolog 1) because of its homology with the MutT protein sequence of Escherichia coli (H. Oda, Y. Nakabeppu et al., Nucleic Acids Res., 27 , 4335 (1999)).
[0008]
The present inventors conduct studies on the spontaneous carcinogenicity of various organs using the knockout mouse lacking the Mth1 gene (Non-Patent Document 1 described above), and also show that human MTH1 is expressed in the fibroblasts of the fetus of the knockout mouse lacking MTH1. A cell line in which (hMTH1) was forcibly expressed was established, and the analysis was carried out focusing on cell dysfunction and cell death under hydrogen peroxide loading conditions. As a result, it was found that the strain forcedly expressing hMTH1 exhibited a significantly higher survival rate as compared with the non-expressing strain, and it was considered that this phenomenon could be used for discriminating the presence or absence of a drug that inhibits MTH1 (human MTH1). Is derived.
[0009]
Hereinafter, a method for producing an MTH1-deficient knockout mouse derived from a cell line used in the method for screening for an MTH1 inhibitor according to the present invention, a method for establishing a fetal fibroblast cell line and an hMTH1-overexpressing cell line, and screening using the cell line An embodiment of the present invention will be described in accordance with the law. However, the specific embodiments described below are for the purpose of illustration, and the embodiments of the present invention are not limited thereto, and those skilled in the art may use the same materials and techniques to make the same. Of course can be manufactured.
[0010]
Preparation of MTH1- deficient mouse Knockout mice deficient in MTH1 can be prepared according to a conventional method using ES cells and gene targeting, and one example is described in the above-mentioned document (Non-patent Document 1) by the present inventors. But as follows:
The Mth1 gene was isolated from a mouse 129SV-derived gene library (purchased from Stratagene, Cat. No. 946313) and its structure was analyzed. The Mth1 gene was about 7 kbp in size and consisted of five exons, the first and second two exons were 5 ′ untranslated regions, and the start codon was included in the third exon. A region in which the amino acid sequence is well conserved among MutT-like proteins derived from three kinds of bacteria including human, mouse and Escherichia coli is recognized, and this region is encoded separately in exons 3 and 4 on the mouse gene. Was. A targeting vector in which the third exon was replaced with a G418 resistance gene cassette (see Deng, C., Thomas, K. R. and Capecchi, M. R., Mol. Cell. Biol. 13 : 2134-2140 (1993)). It was constructed. The thymidine kinase gene of HSV (Rancourt, DE, Tsuzuki, T and capecchi, MR, Genes Dev. 9 : 108-122 (1995)) was placed at both ends of the vector for negative selection. .
[0011]
This targeting vector was introduced into ES cells by electroporation, and an Mth1 hetero-deficient cell clone (Mth1 +/− ) in which a G418 resistance gene cassette was inserted into one of the Mth1 alleles from among the G418 · gancyclovir-resistant clones. Selected. The ES cells used were CCE cell lines (distributed by Dr. JJ Robertson: Robertson, EJ Tetraccarinomas and Embronic Stem Cells: A Practical Approach, IRL Press, IRL Press 87). These CCE cells are distributed free of charge for research purposes. Equivalent ES cells can also be obtained, for example, from the ATCC as CRL-11632.
Chimeric mice obtained by introducing the above-described Mth1 +/− ES cells into mouse blastocysts were crossed with BDF1 mice (purchased from CLEA Japan) to establish Mth1 +/− mice. Mth1 +/− mice were backcrossed to C57BL6 mice for 12 generations to purify their genetic background.
[0012]
Establishment of MTH1 homo-deficient mouse fetal fibroblast cell line ( Mth1 − / − MEF ) Establishment of fetal fibroblast cell line from MTH1-deficient knockout mouse is performed according to a cell culture technique known in the art as follows. be able to:
Males and females of the Mth1 +/− mice backcrossed for 12 generations were bred, and the mice on day 17.5 of gestation were laparotomized and aseptically excised one fetus from the uterus. Approximately 8 mm × 8 mm skin is peeled off from the Mth1 − / − mouse embryo, excised, and transferred to a culture dish. The tissue pieces are minced with scissors in a small amount of medium (DMEM (low glucose), 10% fetal bovine serum), spread on the culture surface of a petri dish, and allowed to stand in a CO 2 incubator.
After 1 hour, the medium is carefully added so that the tissue pieces do not peel off, and the cells are cultured for several days. The fibroblasts that have migrated and proliferated from the tissue piece are detached and collected with 0.05% trypsin-EDTA, transferred to a new dish, and subcultured every three days. After 30 or more passages, the strain was considered to have established when the plating efficiency exceeded 30%. Two independent strains, T2 and T5, were established.
[0013]
Overexpressing human MTH1 Mth1 - / - MTH1 homo-deficient mouse embryonic fibers obtained as establishment <br/> more MEF blast cell lines Mth1 - / - in MEF (T2 and T5), subcloned into human MTH1 cDNA The obtained expression vector (pcDEBΔ: hMTH1) was introduced by a lipofection method, and a plurality of hygromycin B-resistant cell clones were separated. The formation of colonies derived from single cells was repeated twice, and the expression of human MTH1 was confirmed for the purified clones by Western blotting using an anti-MTH1 antibody, and two clones (T2: hMTH1 and T5: hMTH1) were established. As controls, T2: vector and T5: vector into which only pcDEBΔ was introduced were established. The expression vector (pcDEBΔ: hMTH1) and the anti-MTH1 antibody are described in detail in the literature (Kang, D., and Takeshige, K., et al., J. Biol. Chem. 270, 14659-14665 (1995)). Have been. In addition, human MTH1 cDNA was obtained from THEI. M. A. G. FIG. E. FIG. Available from CONSORTIUM.
[0014]
According to the screening method <br/> present invention MTH 1 inhibitors, or more way MTH1 was established by deficient knockout mouse embryonic fibroblasts (Mth1 - / - MEF) using a, an agent that inhibits MTH1 Can be screened. That is, a test substance that is a candidate for an MTH1 inhibitor is added to cells in which human MTH1 is forcibly expressed in Mth1 − / − MEF, and the cells are cultured in the presence of hydrogen peroxide. The test substance is selected as an MTH1 inhibitor when the test is completed.
In actual operation, Mth1 − / − MEFs that overexpress human MTH1 are cultured under appropriate conditions from the logarithmic growth phase (generally, at 37 ° C. for about 24 hours in the presence of 5% CO 2 ). A test substance is added and cultured for an appropriate time (for example, 1 to 5 hours), and then hydrogen peroxide (H 2 O 2 ) of a different concentration is added, and further cultured (generally for about 24 hours). Is measured or observed, and the test substance is selected as an MTH1 inhibitor when the cell death is significantly observed. The medium used may be a standard MEM medium, not a special one. Further, pyruvate may be added to reduce the cytotoxic effect, but the assay system of the present invention can generally measure without adding pyruvate.
[0015]
Means for measuring or observing cell viability in the screening method of the present invention is not particularly limited, and various methods for determining cell death, for example, a metabolic enzyme activity measuring method, a colony forming method, and a cell Microscopic observation by a staining method, a biosynthesis ability measuring method, and the like can be used. Among them, the metabolic enzyme activity measurement method is preferable because a large number of samples can be measured relatively easily using a 96-well microplate or the like. As an example of the method for measuring metabolic enzyme activity, Cell count kit WST-8 (Dojindo: manufactured by Dojindo Laboratories Inc.) may be mentioned, which is a water-soluble compound produced by reducing tetrazolium salts by intracellular dehydrogenase. This is a type of measuring method that detects the number of living cells by measuring the absorbance of formazan.
[0016]
【Example】
Examples are described below to more specifically illustrate the features of the present invention. Example 1 clarifies the phenomenon behind the screening method of the present invention. The cell line in which human MTH1 was forcibly expressed in MTH1 knockout mouse fetal fibroblasts was compared with a non-expressing cell line in comparison with a non-expressing cell line. It has significantly higher cell viability after hydrogen loading, indicating that cell death occurs in the absence of MTH1 in this system. Example 2 shows that a cell line in which human MTH1 was forcibly expressed in MTH1 knockout mouse fetal fibroblasts was inhibited in its MTH1 activity by the presence of a specific substance, and therefore, such an inhibitory substance could be selected. It shows that it can be used for
[0017]
Example 1: Sensitivity test of hydrogen peroxide T5: vector (human MTH1 non-expressing strain: described above) and T5: hMTH cells (human MTH1 forced expressing strain) in logarithmic growth phase were placed in a 96-well microplate at 50%. The cells were seeded so as to be saturated (3 × 10 3 cells / 100 μl / well) and cultured at 37 ° C. in the presence of 5% CO 2 for 24 hours. The medium used was MEM (Invitrogen 11095-080). Next, different concentrations of H 2 O 2 (10 μl / well) were added to the medium, and the cells were further cultured for 24 hours. Cell count Kit WST-8 (Dojindo) was added at 10 μl / well, and after 90 minutes to 120 minutes, the difference between the two absorbances of the culture solution (A450 nm-A620 nm) was measured. The relative viability was determined by setting the absorbance value of the untreated cells to 1.0.
The result is shown in FIG. In this system, it is understood that cell death occurs in the absence of MTH1.
[0018]
Example 2: MTH 1 inhibitor Copper sulfate (CuSO 4 ) was used as the MTH 1 inhibitor . It is known that the 8-oxo-dGTP degradation activity of MTH1 in vitro is inhibited by 50% in the presence of 17 μM Cu 2+ (Porter, DW, Nakabeppu et al., Carcinogenesis 18 (9), 1785). -1791 (1997)), it was examined whether or not the MTH1 activity of cells in which human MTH1 was forcibly expressed in MTH1 knockout mouse embryo fibroblasts used in the present invention was inhibited by such an inhibitor.
T5: hMTH cells in the logarithmic growth phase were seeded on a 96-well microplate at 50% saturation (3 × 10 3 cells / 100 μl / well), and cultured at 37 ° C. in the presence of 5% CO 2 for 24 hours. The medium is MEM (Invitrogen 11095-080). Next, an MTH1 inhibitor (CuSO 4 ) was added to the medium at a final concentration of 50 μM, and the cells were cultured for 2 hours. Thereafter, different concentrations of H 2 O 2 (10 μl / well) were added to the medium, and the cells were further cultured for 24 hours. 10 μl / well of Cell count Kit WST-8 (Dojindo) was added, and after 90 to 120 minutes, the difference between the two absorbances of the culture solution (A450 nm-A620 nm) was measured. The relative viability was determined by setting the absorbance value of the H 2 O 2 untreated cells to 1.0. FIG. 2 shows the measurement results. As shown in FIG. 2, cell death is observed due to the presence of the MTH1 inhibitor.
[0019]
【The invention's effect】
As is apparent from the above description, the use of the method of the present invention enables a simple and reliable screening for a drug that inhibits MTH1, and selects a drug that does not have cell membrane permeability as in a cell-free screening system. That problem is also avoided. Thus, the present invention contributes to the development of anticancer agents and cancer treatments through efficient screening of MTH1 inhibitors.
[Brief description of the drawings]
FIG. 1 shows that the cell line in which human MTH1 is forcibly expressed in MTH1 knockout mouse fetal fibroblasts used in the present invention has a significantly higher cell viability under hydrogen peroxide loading conditions compared to a non-expressing cell line. Is shown.
FIG. 2 shows that a cell line in which human MTH1 is forcibly expressed in MTH1 knockout mouse fetal fibroblasts used in the present invention causes cell death by an MTH1 inhibitor.

Claims (1)

酸化プリンヌクレオシド三リン酸分解酵素(MTH1)を阻害する薬剤をスクリーニングする方法であって、MTH1欠損ノックアウトマウス胎仔の線維芽細胞にヒトMTH1を強制発現させた細胞に被験物質を添加し、過酸化水素の存在下に細胞を培養して、その細胞死が認められたときに被験物質をMTH1阻害剤として選択することを特徴とする方法。A method for screening for a drug that inhibits oxidized purine nucleoside triphosphate degrading enzyme (MTH1), comprising adding a test substance to cells in which human MTH1 is forcibly expressed in fibroblasts of an MTH1-deficient knockout mouse embryo, and A method comprising culturing cells in the presence of hydrogen and selecting a test substance as an MTH1 inhibitor when cell death is observed.
JP2002336181A 2002-11-20 2002-11-20 Screening method for oxidized purine nucleoside triphosphate degrading enzyme inhibitors Expired - Fee Related JP3910525B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2002336181A JP3910525B2 (en) 2002-11-20 2002-11-20 Screening method for oxidized purine nucleoside triphosphate degrading enzyme inhibitors

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
JP2002336181A JP3910525B2 (en) 2002-11-20 2002-11-20 Screening method for oxidized purine nucleoside triphosphate degrading enzyme inhibitors

Publications (2)

Publication Number Publication Date
JP2004166590A true JP2004166590A (en) 2004-06-17
JP3910525B2 JP3910525B2 (en) 2007-04-25

Family

ID=32700094

Family Applications (1)

Application Number Title Priority Date Filing Date
JP2002336181A Expired - Fee Related JP3910525B2 (en) 2002-11-20 2002-11-20 Screening method for oxidized purine nucleoside triphosphate degrading enzyme inhibitors

Country Status (1)

Country Link
JP (1) JP3910525B2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2015529665A (en) * 2012-08-27 2015-10-08 ツェーエムエム−フォルシュングスツェントルム フュア モレクラレ メディツィン ゲーエムベーハー Aminoheteroaryl compounds as MTH1 inhibitors

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2015529665A (en) * 2012-08-27 2015-10-08 ツェーエムエム−フォルシュングスツェントルム フュア モレクラレ メディツィン ゲーエムベーハー Aminoheteroaryl compounds as MTH1 inhibitors

Also Published As

Publication number Publication date
JP3910525B2 (en) 2007-04-25

Similar Documents

Publication Publication Date Title
El-Naggar et al. Translational activation of HIF1α by YB-1 promotes sarcoma metastasis
Vicinanza et al. Kit cre knock-in mice fail to fate-map cardiac stem cells
Ventura et al. Turning on stem cell cardiogenesis with extremely low frequency magnetic fields
Pan et al. MicroRNA-378 controls classical brown fat expansion to counteract obesity
Doupnik et al. Profile of RGS expression in single rat atrial myocytes
Rasmussen et al. Smyd1 facilitates heart development by antagonizing oxidative and ER stress responses
Liu et al. Overexpression of miR-142-3p improves mitochondrial function in cardiac hypertrophy
JPH09507021A (en) Method for producing subtraction cDNA library and use of the produced library
Sekiguchi et al. Induction of growth arrest and cell death by overexpression of the cyclin-Cdk inhibitor p21 in hamster BHK21 cells
Kalaiselvi Sivalingam et al. Zebrafish fin‐derived fibroblast cell line: A model for in vitro wound healing
ES2553332T3 (en) Methods and compositions for the treatment of diseases
US9091696B2 (en) Modulation of NR2F6 and methods and uses thereof
Liu et al. Ca2+ concentration–dependent premature death of igfbp5a−/− fish reveals a critical role of IGF signaling in adaptive epithelial growth
CA2487427A1 (en) Methods and compositions for treating neoplasia relating to hnrnp a1 and a2 nucleic acid molecules
US20100310532A1 (en) Gene targets in anti-aging therapy and tissue repair
Van Berlo et al. van Berlo et al. reply
Varga et al. Tissue-specific requirement for the GINS complex during Zebrafish development
JP3910525B2 (en) Screening method for oxidized purine nucleoside triphosphate degrading enzyme inhibitors
WO2009142271A1 (en) Cancer stem cell having high level of sld5 expression therein
JP4268169B2 (en) Determinants of self-renewal of embryonic stem cells
Gulyaeva The Study of Adipose Tissue Differentiation and Development
Lupu et al. SRSF3 is a key regulator of epicardial formation
Frank et al. Trabid patient mutations impede the axonal trafficking of adenomatous polyposis coli to disrupt neurite growth
WO2019088164A1 (en) Novel polynucleotide and utilization thereof
Iida et al. MECOM promotes leukemia progression and inhibits mast cell differentiation through functional competition with GATA2

Legal Events

Date Code Title Description
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: 20070109

A61 First payment of annual fees (during grant procedure)

Free format text: JAPANESE INTERMEDIATE CODE: A61

Effective date: 20070124

R150 Certificate of patent (=grant) or registration of utility model

Free format text: JAPANESE INTERMEDIATE CODE: R150

FPAY Renewal fee payment (prs date is renewal date of database)

Year of fee payment: 4

Free format text: PAYMENT UNTIL: 20110202

LAPS Cancellation because of no payment of annual fees