Role and Mechanism of Metformin in Oligodendrocyte Precursor Cell Differentiation

TAN Pei-lin,ZHANG Ying,ZHANG Jun,GAO Xiao,WANG Shu-kun,HOU Lin,YUAN Zeng-qiang

China Biotechnology ›› 2021, Vol. 41 ›› Issue (9) : 1-9.

PDF(1742 KB)
PDF(1742 KB)
China Biotechnology ›› 2021, Vol. 41 ›› Issue (9) : 1-9. DOI: 10.13523/j.cb.2103013

Role and Mechanism of Metformin in Oligodendrocyte Precursor Cell Differentiation

Author information +
History +

Abstract

Objective:To demonstrate the role of metformin in oligodendrocyte precursor cell (OPC) differentiation and preliminarily discuss the molecular mechanism.Methods:OPC was directly isolated and purified by immune adsorption from the brain and identified using immunofluorescence. Firstly, the concentration of metformin was decided through cell viability assay. Then, the effects of metformin on OPC-differentiation related positive cells, the mRNA or protein level were analyzed by immunofluorescence, flow cytometry, qRT-PCR, and western blot.Results:High purity of primary cells were obtained. CCK8 assay showed that there is no significant toxicity of metformin (<100 μmol/L) on cell viability. Moreover, the significant increasement of PDGFRα+OLIG2+ and MBP+ cells, up-regulation levels of Mag, Olig2, Mbp and Sox10 mRNA and OLIG2, MBP protein were detected in OPC after metformin treatment. Mechanically, compared to the control group, RAS, p-MEK and p-ERK proteins were significantly increased after metformin treatment for 5min in Oli-neu cells and OPC. Conclusion:Metformin promotes the differentiation of oligodendrocyte precursor cells through the RAS-MEK-ERK signaling pathway.

Key words

Metformin / Oligodendrocyte precursor cells(OPCs) / Demyelinating diseases

Cite this article

Download Citations
Pei-lin TAN, Ying ZHANG, Jun ZHANG, et al. Role and Mechanism of Metformin in Oligodendrocyte Precursor Cell Differentiation[J]. China Biotechnology, 2021, 41(9): 1-9 https://doi.org/10.13523/j.cb.2103013
目前多种神经系统疾病常伴有脱髓鞘或髓鞘再生缺陷。髓鞘包裹轴突形成绝缘膜,保证神经冲动沿轴突快速传导[1,2,3,4];并为神经元提供营养和代谢支持[5,6,7,8,9]。髓鞘缺失时,动作电位传导减弱甚至中断,从而引发脱髓鞘相关疾病。病患表现为肢体无力、共济失调、意识障碍等,有时可产生急性症状,如强直痉挛和疼痛不适等。
少突胶质前体细胞特化形成少突胶质细胞(oligodendrocyte, OL),OL的主要功能是形成髓鞘[10];神经活动通过影响OPC的增殖分化及髓鞘形成和再生而引发髓鞘变化[11,12,13]。少突胶质细胞是出生后中枢神经系统血管生成的关键调节因子之一,而OPC也可参与调节出生后髓鞘形成的起始和白质中血管系统的建立[14]
二甲双胍是一种双胍类药物,用于治疗2型糖尿病,可显著降低血糖[15,16],在抗炎、抗氧化、抗癌、神经保护等方面也发挥重要作用[16,17,18,19,20,21,22,23,24,25]。因其可激活脑中内源性干细胞,促进神经再生,二甲双胍在神经退行性疾病研究中得到广泛运用。
二甲双胍可影响髓鞘再生,实现神经修复。研究证实,在EAE动物模型中,二甲双胍可通过激活mTOR/AMPK信号通路,使神经营养因子水平升高,诱导并加速髓鞘再生[26,27]。Neumann等[28]、Cosgrove等[29]和Cantuti-Castelvetri等[30]发现,二甲双胍可使老年鼠OPC恢复活力,促进脱髓鞘损伤后髓鞘再生。但是,二甲双胍如何影响OPC的分化及其内在机制,目前还不清楚。
因此,我们在构建少突胶质前体细胞体外直接分离培养体系的基础上[31],利用生化与分子生物学等技术手段,研究二甲双胍在OPC分化中的作用,并初步探讨其调控机制,这将为神经系统中髓鞘相关疾病的临床治疗提供理论依据和新思路。

1 材料与方法

1.1 材料

(1)二甲双胍购自Selleck公司(中国);细胞培养因子N2、B27,DMEM/F12培养基购自Gibco公司(美国);胎牛血清、0.25%/0.05%胰蛋白酶购自BI公司(以色列);生物素、胰岛素、多聚赖氨酸、谷氨酰胺、青霉素-链霉素、Triton X-100、三碘甲状腺氨酸购自Sigma公司(美国);牛血清白蛋白购自上海翊圣公司(中国);PDGFRα-AA、bFGF等细胞添加物购自Peprotech公司(美国)。
(2)一抗:OLIG2购自Millipore公司(美国);PDGFRα购自BD公司(美国);MBP、ALDH1L1购自Abcam公司(英国);MAG购自Proteintech公司(美国);GFAP购自Dako公司(丹麦);ACTIN购自Abclone公司(美国);PDGFRα、p-ERK、ERK、p-MEK、MEK、RAS、p-FoxO3a、FoxO3a购自CST公司(美国);荧光标记二抗购自Jackson公司(美国)、Trizol购自Invitrogen公司(美国);抗荧光淬灭封片剂购自上海碧云天公司(中国)。反转录试剂盒购自全式金公司(中国)、qRT-PCR试剂盒购自康为世纪公司(中国)等。
(3)细胞培养箱购自Thermo fisher公司(美国,型号SeriesⅡ WJ);制冰机购自Panasonic公司(日本,型号SIM-F140ADL);Milli-Q纯水仪购自Merck公司(德国,型号Milli-Q Biocel);qRT-PCR仪购自Agilent公司(美国,型号Stratagene Mx3005P);PCR仪购自杭州博日公司[中国,型号TC-96/G/H(b)B];蛋白质电泳系统购自Bio-Rad公司(美国,型号MyCycler);多功能酶标仪购自Tecan公司(瑞士,型号IM200);全自动洗片机购自Eastman Kodak公司(美国,型号Medical X-ray processor 102);倒置激光共聚焦显微镜购自PerkinElmer公司(美国,型号UltraVIEW VOX);分光光度计购自Implen公司(德国,型号Nanophotometer P330);BD流式细胞仪(美国,型号FACSCalibur)等。

1.2 方法

1.2.1 分离和培养少突胶质前体细胞
分别用IgG、IgG/IgM包被细胞培养皿,置于4℃孵育;第二天去除二抗,洗涤后分别用GalC、O4孵育培养皿,室温3 h后备用。剥离C57BL/6新生小鼠(P3)的大脑皮层,洗涤后剪碎组织置于papain溶液中消化20 min,而后获得单细胞悬液依次置于抗体GalC、O4包被的培养皿中孵育1~2 h。培养皿上的细胞经PBS洗涤、胰蛋白酶消化后重悬平铺于细胞板(多聚赖氨酸包被)中,37℃培养箱孵育1~2 h后替换为增殖培养基继续培养,隔天半换液。或次日换为分化培养基,进行后续化合物处理。
1.2.2 免疫荧光染色
OPC经二甲双胍处理48 h后,弃去培养基后用缓冲液洗涤3次,加入2%多聚甲醛固定5~10 min,洗涤3次后加入封闭液孵育1 h,吸走上清,加入一抗溶液,放入4℃冰箱孵育;次日洗涤3次后,加荧光二抗溶液,1~2 h后用PBS洗涤3次,而后加Hoechst溶液染色10 min左右;洗涤后可将细胞爬片取出,倒扣于抗荧光淬灭剂上,封片后使用激光共聚焦显微镜分析。一抗稀释比例:PDGFRα(BD, 1∶1 000)、OLIG2(1∶1 000)、MBP(1∶1 000)。
1.2.3 RNA提取和qRT-PCR检测mRNA表达量
细胞经二甲双胍处理后,加入Trizol溶液(500 μL/孔)后震荡裂解细胞,而后加三氯甲烷(100 μL/孔),充分混匀后静置10~15 min,13 000 r/min低温离心10~15 min,上层水相转移至含异丙醇(500 μL)的新EP管中,混匀后静置10~30 min,13 000 r/min低温离心15 min,沉淀用75%乙醇洗涤后于4℃下13 000 r/min离心15 min,倒去上清后室温静置3~5 min晾干,加入10~20 μL 无酶无菌水溶解。检测样品RNA浓度和纯度后,取0.5~1 μg进行反转录合成cDNA(操作参考全式金公司反转录试剂盒说明书)。实时荧光定量检测步骤参照试剂盒说明书(康为世纪),引物序列见表1
Table 1 qRT-PCR primers

表1 qRT-PCR引物序列

Name Sequence (5'-3')
Mag-Fp CTGCCGCTGTTTTGGATAATGA
Mag-Rp CATCGGGGAAGTCGAAACGG
Mbp-Fp GACCATCCAAGAAGACCCCAC
Mbp-Rp GCCATAATGGGTAGTTCTCGTGT
Olig2-Fp GGGAGGTCATGCCTTACGC
Olig2-Rp CTCCAGCGAGTTGGTGAGC
Sox10-Fp ACACCTTGGGACACGGTTTTC
Sox10-Rp TAGGTCTTGTTCCTCGGCCAT
Gfap-Fp CGGAGACGCATCACCTCTG
Gfap-Rp AGGGAGTGGAGGAGTCATTCG
β-actin-Fp GGCTGTATTCCCCTCCATCG
β-actin-Rp CCAGTTGGTAACAATGCCATGT
1.2.4 Western blot检测
200 μL细胞裂解液中添加蛋白酶抑制剂、磷酸酶抑制剂(100×),加入细胞中裂解,低温静置30 min后转移至EP管中,震荡离心,BCA法测定蛋白质浓度后加loading buffer置于99℃金属浴10 min。样品进行蛋白质电泳和转膜,NC膜置于封闭液(5%牛奶)中1~2 h,洗膜后加一抗(OLIG2等)孵育,第二天NC膜经TBST洗涤后加HRP标记的二抗孵育1~2 h,TBST洗涤3次后显影定影检测。一抗稀释比例:OLIG2(1∶1 000)、MBP(1∶2 000)、GFAP(1∶20 000)、MAG(1∶2 000)、ALDH1L1(1∶1 000)、ACTIN(1∶2 000)、pERK(1∶500)、ERK(1∶1 000)、pMEK(1∶400)、MEK(1∶1 000)、RAS(1∶1 000)、pFoxO3a(1∶1 000)、FoxO3a(1∶1 000)。
1.2.5 细胞活力检测
按CCK8试剂盒说明书操作,原代少突胶质细胞培养基中加不同浓度二甲双胍处理48 h后,加入CCK8试剂孵育约10 min,使用酶标仪检测450 nm下的OD值变化。
1.2.6 流式细胞分析
细胞经二甲双胍处理后,转移至流式管中,2 000 r/min离心5 min,弃上清,加入2%多聚甲醛室温固定10 min,2 000 r/min离心5 min后弃上清,PBS重悬清洗一次,加入一抗室温孵育1 h,加入PBS洗涤,2 000 r/min离心5 min,加入荧光二抗溶液,室温孵育30 min,加入PBS洗涤,2 000 r/min离心5 min弃上清,最后加入300 μL PBS重悬细胞,细胞悬液进行流式细胞分析。一抗稀释比例:PDGFRα(CST, 1∶500)。
1.2.7 数据分析
采用Excel软件和GraphPad Prism 8.0分析,差异检验使用双尾学生t检验,标注平均值±标准误(SEM),统计显著性阈值标记用* P<0.05、 ** P<0.01、*** P<0.001标注。

2 结果

2.1 构建少突胶质前体细胞体外分离培养体系

密度梯度法、震荡分离纯化等常用方法获取的OPC纯度不高且周期长,本文采用抗体免疫吸附法直接分离新生鼠OPC(图1a),加增殖培养基48 h后,利用免疫荧光染色检测细胞纯度和形态,用PDGFRα、OLIG2双标指示OPC,结果显示双阳性细胞比例高达90%以上,原代OPC纯度较高(图1b)。
Fig.1 Isolation and cultivation of oligodendrocyte precursor cells

(a) Flowchart showing the isolation and cultivation of OPC in vitro (b) Immunofluorescence labeling of PDGFRα (red) and OLIG2 (green) in cultured cells

图1 少突胶质前体细胞体外分离培养

Full size|PPT slide

2.2 二甲双胍对OPC的影响

为确定药物处理浓度,使用少突胶质细胞系Oli-neu或OPC检测不同浓度二甲双胍对细胞活性的影响。Western blot结果显示,300 μmol/L、900 μmol/L高浓度二甲双胍处理,细胞状态不佳,OLIG2、ALDH1L1、GFAP等蛋白质量明显降低,药物有毒性(图2a、b)。CCK8检测结果显示,二甲双胍浓度在100 μmol/L左右对细胞活性无显著差异,可作为细胞处理浓度(图2c)。
Fig.2 Effect of metformin on cell viability and protein expression

(a) Western blot analysis of MAG, ALDH1L using indicated antibodies in Oli-neu cells after metformin (0, 100 μmol/L, 300 μmol/L, 900 μmol/L) treatment for 48 h (b) Western blot analysis of OLIG2, MBP, ALDH1L1, GFAP in OPC after metformin treatment for 48 h (c) CCK8 assay showed relative cell viability in OPC after metformin treatment

图2 二甲双胍对细胞活力和蛋白表达的影响

Full size|PPT slide

2.3 二甲双胍促进OPC分化

选用100 μmol/L二甲双胍处理原代细胞以检测其对OPC分化的影响。免疫荧光染色结果显示:二甲双胍处理后,PDGFRα+OLIG2+双阳性的OPC数量较对照组显著增加(图3a、b)。流式细胞分析结果显示:二甲双胍处理原代细胞后, PDGFRα阳性细胞数显著高于对照组(图3c~e)。免疫荧光染色发现,分化条件下二甲双胍处理原代细胞后,MBP标记的OL数量相较于对照组显著增加(图3f、g)。收集细胞提取RNA、准备蛋白质样品分别进行qRT-PCR、Western blot检测,结果表明,经二甲双胍处理后,少突胶质细胞谱系分化相关基因MagMbpOlig2Sox10的mRNA水平显著增加,星形胶质细胞标记基因Gfap mRNA水平变化不明显(图3h);OPC分化相关蛋白质OLIG2、MBP的量逐渐增加,星形胶质细胞蛋白ALDH1L1和GFAP无变化(图3i)。这表明二甲双胍促进OPC分化。
Fig.3 Metformin modulates OPC differentiation

(a) Immunofluorescence labeling of PDGFRα (green) and OLIG2 (red) in cells after 100 μmol/L metformin treatment for 48 h (b) Quantification of the percentage of PDGFRα+OLIG2+ cells *** P< 0.001 (c),(d) Flow cytometry analysis of PDGFRα in living cells after 100 μmol/L metformin treatment for 48 h (e) Quantification of the percentage of PDGFRα+ cells *** P<0.001 (f) Immunostainings of MBP (green) in cells treated with metformin for 48 h (g) Quantification of MBP+ cells *** P<0.001 (h) The mRNA levels of Mag, Mbp, Olig2, Sox10 and Gfap in primary cells after 100 μmol/L metformin treatment n=4 each group, * P< 0.05, ** P<0.01 (i) Western blot analysis of protein levels (OLIG2, MBP, ALDH1L1, GFAP) in OPCs treated with metformin for 48 h

图3 二甲双胍促进OPC分化

Full size|PPT slide

2.4 二甲双胍促进OPC分化的分子机制

为解析二甲双胍促进OPC分化的分子机制,我们使用少突胶质细胞系Oli-neu开展实验。二甲双胍处理细胞0 min、5 min、10 min和30 min后,收集蛋白质样品进行Western blot检测,细胞经二甲双胍处理5 min后, RAS-MEK-ERK信号通路中关键蛋白质p-MEK、p-ERK及RAS表达量骤增(图4a)。进一步在原代OPC中验证,结果显示,经二甲双胍处理5 min后,关键蛋白质p-MEK和p-ERK表达量增加, p-MEK/MEK和p-ERK/ERK值升高,与Oli-neu的实验结果一致(图4b、c)。这提示,二甲双胍可能通过RAS-MEK-ERK信号通路促进OPC分化(图4d)。
Fig.4 The mechanism of metformin modulating OPC differentiation

(a) Immunoblotting analysis of RAS, p-MEK, MEK, p-ERK, ERK protein levels in Oli-neu cells after metformin treatment for 0, 5, 10 and 30 min (b) Immunoblotting analysis of the protein expression in OPC after metformin treatment for 5 min (c) Quantification of protein expression * P<0.05, *** P<0.001 (d) The model depicts the role and mechanism of metformin in oligodendrocyte precursor cell differentiation

图4 二甲双胍促进OPC分化的机制

Full size|PPT slide

3 讨论

髓鞘缺失引发脱髓鞘相关疾病,如视神经脊髓炎(NMOSD)、多发性硬化症(MS)、急性播散性脑脊髓炎(ADEM)、阿尔茨海默病(AD)和亨延顿病(HD)等[31,32,33,34,35]
但是,目前此类疾病还没有行之有效的治疗措施和药物。因此,研究如何增加成髓鞘细胞的数量促进髓鞘发生,对脱髓鞘相关疾病的治疗和药物研发具有重要的研究意义。
OL是中枢神经系统中一群特化的胶质细胞,其主要功能是形成髓鞘,传递神经信号。OL是由少突胶质前体细胞分化成熟而来的。以往研究中,常使用密度梯度法、震荡差速贴壁法、神经干细胞诱导分化等方法获取OPC,但往往存在细胞纯度不高、培养周期长、与体内细胞状态差异较大等缺点。本文通过抗体免疫吸附法构建OPC体外分离培养体系,可获得纯度较高、更接近体内正常生理状态的原代细胞,且明显缩短了细胞培养周期。
研究证实,二甲双胍在多发性硬化症(MS)等神经系统疾病的动物模型中具有神经保护和抗炎作用[16, 36-38]。二甲双胍参与细胞分化的信号通路有AMPK、mTOR等。据报道,AMPK信号通路调控细胞增殖和能量代谢,提高细胞抗氧化防御[39]。Houshmand等[27]发现,二甲双胍可能通过神经营养因子BDNF、CNTF和NGF的表达,下调NogoA的表达,诱导活化AMPK,促进成熟少突胶质细胞在胼胝体中的增殖。mTOR信号通路调控细胞生长,二甲双胍降低Th17数量,通过mTOR/HIF-1α促进成熟OL增殖和髓鞘再生,对少突胶质细胞损伤起保护作用[40]。本文机制研究发现,二甲双胍通过RAS-MEK-ERK信号通路促进OPC分化。此外,我们还发现Oli-neu经二甲双胍处理后,p-FoxO3a表达在5 min、10 min时表达量增加,而FoxO3a无明显变化。这提示了p-FoxO3a也可能参与少突胶质细胞分化调控(图5)。
Fig.5 Effect of metformin on oligodendrocytes

Western blot analysis of p-FoxO3a, FoxO3a expression in cells after 100μmol/L metformin treatment for 0, 5, 10 and 30 min

图5 二甲双胍调控少突胶质细胞分化

Full size|PPT slide

虽然已发现二甲双胍在改善MS病人脱髓鞘病症方面的作用,但其具体机制还不清楚,且二甲双胍在其他髓鞘相关疾病中的作用也不明确。为此,本文在构建少突胶质前体细胞体外分离培养体系的基础上,运用生化与分子生物学、细胞生物学等技术手段,阐明二甲双胍对少突胶质前体细胞分化的影响,并初步探讨其分子机制,为二甲双胍在神经系统髓鞘相关疾病临床治疗中的运用提供理论依据,并为药物研发提供新思路。

References

[1]
Donkels C, Peters M, Fariña Núñez M T, et al. Oligodendrocyte lineage and myelination are compromised in the gray matter of focal cortical dysplasia type IIa. Epilepsia, 2020, 61(1):171-184.
[2]
Benamer N, Vidal M, Balia M, et al. Myelination of parvalbumin interneurons shapes the function of cortical sensory inhibitory circuits. Nature Communications, 2020, 11:5151.
[3]
Bradl M, Lassmann H. Oligodendrocytes: biology and pathology. Acta Neuropathologica, 2010, 119(1):37-53.
[4]
Groves A K, Barnett S C, Franklin R J M, et al. Repair of demyelinated lesions by transplantation of purified 0-2A progenitor cells. Nature, 1993, 362(6419):453-455.
[5]
Fünfschilling U, Supplie L M, Mahad D, et al. Glycolytic oligodendrocytes maintain myelin and long-term axonal integrity. Nature, 2012, 485(7399):517-521.
[6]
Harris J J, Attwell D. The energetics of CNS white matter. The Journal of Neuroscience: the Official Journal of the Society for Neuroscience, 2012, 32(1):356-371.
[7]
Lee Y, Morrison B M, Li Y, et al. Oligodendroglia metabolically support axons and contribute to neurodegeneration. Nature, 2012, 487(7408):443-448.
[8]
Rinholm J E, Hamilton N B, Kessaris N, et al. Regulation of oligodendrocyte development and myelination by glucose and lactate. The Journal of Neuroscience: the Official Journal of the Society for Neuroscience, 2011, 31(2):538-548.
[9]
Oluich L J, Stratton J A S, Xing Y L, et al. Targeted ablation of oligodendrocytes induces axonal pathology independent of overt demyelination. The Journal of Neuroscience, 2012, 32(24):8317-8330.
[10]
Dubois-Dalcq M, Armstrong R. The cellular and molecular events of central nervous system remyelination. BioEssays, 1990, 12(12):569-576.
Central nervous system (CNS)* regeneration is a subject of great interest, particularly in diseases causing a dramatic loss of neurons. However, some CNS diseases do not affect neurons but damage other cells, such as the myelin-forming cells--called oligodendrocytes--which are also crucial to the harmonious function of the nervous system. Diseases in which oligodendrocytes and myelin are attacked can cause devastating neurological dysfunction which is sometimes followed by recovery and myelin repair or remyelination. The question of the regeneration potential of oligodendrocytes in experimental and human demyelinating diseases such as multiple sclerosis has been debated for a long time. Present evidence suggests that oligodendrocyte precursor cells persist in the adult CNS and that oligodendrocyte regeneration can occur but may be limited by ongoing disease processes. Here we will briefly review recent advances which have broadened our understanding of the cellular and molecular events of CNS remyelination.
[11]
Snaidero N, Möbius W, Czopka T, et al. Myelin membrane wrapping of CNS axons by PI(3, 4, 5)P3-dependent polarized growth at the inner tongue. Cell, 2014, 156(1-2):277-290.
Central nervous system myelin is a multilayered membrane sheath generated by oligodendrocytes for rapid impulse propagation. However, the underlying mechanisms of myelin wrapping have remained unclear. Using an integrative approach of live imaging, electron microscopy, and genetics, we show that new myelin membranes are incorporated adjacent to the axon at the innermost tongue. Simultaneously, newly formed layers extend laterally, ultimately leading to the formation of a set of closely apposed paranodal loops. An elaborated system of cytoplasmic channels within the growing myelin sheath enables membrane trafficking to the leading edge. Most of these channels close with ongoing development but can be reopened in adults by experimentally raising phosphatidylinositol-(3,4,5)-triphosphate levels, which reinitiates myelin growth. Our model can explain assembly of myelin as a multilayered structure, abnormal myelin outfoldings in neurological disease, and plasticity of myelin biogenesis observed in adult life.Copyright © 2014 Elsevier Inc. All rights reserved.
[12]
Almeida R G, Lyons D A. On myelinated axon plasticity and neuronal circuit formation and function. The Journal of Neuroscience, 2017, 37(42):10023-10034.
[13]
Fields R D. A new mechanism of nervous system plasticity: activity-dependent myelination. Nature Reviews Neuroscience, 2015, 16(12):756-767.
[14]
Yuen T J, Silbereis J C, Griveau A, et al. Oligodendrocyte-encoded HIF function couples postnatal myelination and white matter angiogenesis. Cell, 2014, 158(2):383-396.
[15]
Tanaka Y, Konishi A, Obinata H, et al. Metformin activates KDM2A to reduce rRNA transcription and cell proliferation by dual regulation of AMPK activity and intracellular succinate level. Scientific Reports, 2019, 9:18694.
Metformin is used to treat type 2 diabetes. Metformin activates AMP-activated kinase (AMPK), which may contribute to the action of metformin. Metformin also shows anti-proliferation activity. However, the mechanism is remained unknown. We found that treatment of MCF-7 cells with metformin induced the demethylase activity of KDM2A in the rDNA promoter, which resulted in reductions of rRNA transcription and cell proliferation. AMPK activity was required for activation of KDM2A by metformin. Because demethylase activities of JmjC-type enzymes require a side reaction converting α-ketoglutarate to succinate, these organic acids may affect their demethylase activities. We found that metformin did not induce KDM2A demethylase activity in conditions of a reduced level of α-ketoglutarate. A four-hour treatment of metformin specifically reduced succinate, and the replenishment of succinate inhibited the activation of KDM2A by metformin, but did not inhibit the activation of AMPK. Metformin reduced succinate even in the conditions suppressing AMPK activity. These results indicate that metformin activates AMPK and reduces the intracellular succinate level, both of which are required for the activation of KDM2A to reduce rRNA transcription. The results presented here uncover a novel factor of metformin actions, reduction of the intracellular succinate, which contributes to the anti-proliferation activity of metformin.
[16]
Largani S H H, Borhani-Haghighi M, Pasbakhsh P, et al. Oligoprotective effect of metformin through the AMPK-dependent on restoration of mitochondrial hemostasis in the cuprizone-induced multiple sclerosis model. Journal of Molecular Histology, 2019, 50(3):263-271.
[17]
Pernicova I, Korbonits M. Metformin:mode of action and clinical implications for diabetes and cancer. Nature Reviews Endocrinology, 2014, 10(3):143-156.
Metformin has been the mainstay of therapy for diabetes mellitus for many years; however, the mechanistic aspects of metformin action remained ill-defined. Recent advances revealed that this drug, in addition to its glucose-lowering action, might be promising for specifically targeting metabolic differences between normal and abnormal metabolic signalling. The knowledge gained from dissecting the principal mechanisms by which metformin works can help us to develop novel treatments. The centre of metformin's mechanism of action is the alteration of the energy metabolism of the cell. Metformin exerts its prevailing, glucose-lowering effect by inhibiting hepatic gluconeogenesis and opposing the action of glucagon. The inhibition of mitochondrial complex I results in defective cAMP and protein kinase A signalling in response to glucagon. Stimulation of 5'-AMP-activated protein kinase, although dispensable for the glucose-lowering effect of metformin, confers insulin sensitivity, mainly by modulating lipid metabolism. Metformin might influence tumourigenesis, both indirectly, through the systemic reduction of insulin levels, and directly, via the induction of energetic stress; however, these effects require further investigation. Here, we discuss the updated understanding of the antigluconeogenic action of metformin in the liver and the implications of the discoveries of metformin targets for the treatment of diabetes mellitus and cancer.
[18]
Zakikhani M, Dowling R, Fantus I G, et al. Metformin is an AMP kinase-dependent growth inhibitor for breast cancer cells. Cancer Research, 2006, 66(21):10269-10273.
Recent population studies provide clues that the use of metformin may be associated with reduced incidence and improved prognosis of certain cancers. This drug is widely used in the treatment of type 2 diabetes, where it is often referred to as an "insulin sensitizer" because it not only lowers blood glucose but also reduces the hyperinsulinemia associated with insulin resistance. As insulin and insulin-like growth factors stimulate proliferation of many normal and transformed cell types, agents that facilitate signaling through these receptors would be expected to enhance proliferation. We show here that metformin acts as a growth inhibitor rather than an insulin sensitizer for epithelial cells. Breast cancer cells can be protected against metformin-induced growth inhibition by small interfering RNA against AMP kinase. This shows that AMP kinase pathway activation by metformin, recently shown to be necessary for metformin inhibition of gluconeogenesis in hepatocytes, is also involved in metformin-induced growth inhibition of epithelial cells. The growth inhibition was associated with decreased mammalian target of rapamycin and S6 kinase activation and a general decrease in mRNA translation. These results provide evidence for a mechanism that may contribute to the antineoplastic effects of metformin suggested by recent population studies and justify further work to explore potential roles for activators of AMP kinase in cancer prevention and treatment.
[19]
Barzilai N, Crandall J P, Kritchevsky S B, et al. Metformin as a tool to target aging. Cell Metabolism, 2016, 23(6):1060-1065.
Aging has been targeted by genetic and dietary manipulation and by drugs in order to increase lifespan and health span in numerous models. Metformin, which has demonstrated protective effects against several age-related diseases in humans, will be tested in the TAME (Targeting Aging with Metformin) trial, as the initial step in the development of increasingly effective next-generation drugs.Copyright © 2016. Published by Elsevier Inc.
[20]
Zhang H H, Guo X L. Combinational strategies of metformin and chemotherapy in cancers. Cancer Chemotherapy and Pharmacology, 2016, 78(1):13-26.
[21]
Qi B X, Hu L B, Zhu L, et al. Metformin attenuates cognitive impairments in hypoxia-ischemia neonatal rats via improving remyelination. Cellular and Molecular Neurobiology, 2017, 37(7):1269-1278.
[22]
Abd-Elsameea A A, Moustaf A A, Mohamed A M. Modulation of the oxidative stress by metformin in the Cerebrum of rats exposed to global cerebral ischemia and ischemia/reperfusion. European Review for Medical and Pharmacological Sciences, 2014, 18(16):2387-2392.
Oxidative stress plays a major role in the pathogenesis of ischemic and reperfusion injury to many organs, including the brain. Chronic metformin treatment is associated with a lower risk of stroke in clinical populations. The aim of the present study was to investigate the effect of metformin on the oxidative stress induced in experimental model of incomplete global cerebral ischemia and ischemia/reperfusion in adult male Wistar rats.Metformin was administered to rats orally by gavage 500 mg/kg once daily for one week before induction of cerebral ischemia (rats were subjected to 30 min of ischemia before decapitation) and ischemia/reperfusion (rats were subjected to 30 min of ischemia then 60 minutes of reperfusion before decapitation). The selected parameters for oxidative stress were the activities of the antioxidant enzymes: glutathione peroxidase (GSHPx), superoxide dismutase (SOD), and catalase as well as malondialdehyde (MDA) levels.Metformin reduced the elevated activites of GSHPx, SOD and catalase as well as MDA levels in cerebrum of rats exposed to ischemia and ischemia/reperfusion injures.Metformin improved the oxidative stress induced by ischemia and ischemia/reperfusion injuries. This may be a mechanism that explains the cerebroprotective effect of the drug.
[23]
Venna V R, Li J, Hammond M D, et al. Chronic metformin treatment improves post-stroke angiogenesis and recovery after experimental stroke. The European Journal of Neuroscience, 2014, 39(12):2129-2138.
[24]
Liu Y Q, Tang G H, Li Y N, et al. Metformin attenuates blood-brain barrier disruption in mice following middle cerebral artery occlusion. Journal of Neuroinflammation, 2014, 11:177.
[25]
Tao L, Li D, Liu H X, et al. Neuroprotective effects of metformin on traumatic brain injury in rats associated with NF-κB and MAPK signaling pathway. Brain Research Bulletin, 2018, 140:154-161.
Traumatic brain injury (TBI) triggers a complex sequence of inflammatory responses that contribute to secondary injury. Metformin, a first-line drug used to treat type 2 diabetes, is reported to exhibit potent anti-inflammatory activity on diseases associated with the central nervous system (CNS). The aim of this study is to investigate the potential neuroprotective effects of metformin on acute brain injury after TBI and explore the underlying mechanisms. Male Sprague-Dawley (SD) rats were divided into four groups: sham group, TBI group, TBI + saline (NS) group and TBI + metformin group. A weight-dropping model was employed to induce TBI in rats. Modified neurological severity scores (mNSS) were employed to assess the short-term neurological deficits, neuronal degeneration and apoptosis in the brain tissues were assayed with Fluoro-Jade B and TUNEL staining, immunofluorescence was designed to investigate microglial activation. The mRNA and protein expression levels of pro-inflammatory cytokines such as necrosis factor-alpha (TNF-α), interleukin-beta (IL-1β) and nterleukin-6 (IL-6) were evaluated by real-time quantitative reverse transcriptase polymerase chain reaction (QPCR) and enzyme-linked immunosorbent assay (ELISA). Western blotting analysis was engaged to examine the expression of NF-κB p65 and phosphorylation of ERK1/2 and p38 MAPK. Our results showed that metformin significantly ameliorated neurological deficit, cerebral edema and neuronal apoptosis in rats following TBI. Moreover, metformin administration inhibited microglial activation and decreased the production of pro-inflammatory cytokines including TNF-α, IL-1β and IL-6. In addition, metformin inhibited the translocation of NF-κB p65 from cytoplasm into the nucleus, as well as the phosphorylation of ERK1/2 and p38 MAPK. This study suggests that metformin administration inhibits microglia activation-mediated inflammation via NF-κB and MAPK signaling pathway to improve neurobehavioral function following TBI, which provide a potential therapeutic benefit in treating brain injury.Copyright © 2018 Elsevier Inc. All rights reserved.
[26]
Sanadgol N, Barati M, Houshmand F, et al. Metformin accelerates myelin recovery and ameliorates behavioral deficits in the animal model of multiple sclerosis via adjustment of AMPK/Nrf2/mTOR signaling and maintenance of endogenous oligodendrogenesis during brain self-repairing period. Pharmacological Reports, 2020, 72(3):641-658.
Multiple sclerosis (MS) is a devastating autoimmune disorder characterized by oligodendrocytes (OLGs) loss and demyelination. In this study, we have examined the effects of metformin (MET) on the oligodendrogenesis, redox signaling, apoptosis, and glial responses during a self-repairing period (1-week) in the animal model of MS.For induction of demyelination, C57BL/6 J mice were fed a 0.2% cuprizone (CPZ) for 5 weeks. Thereafter, CPZ was removed for 1-week and molecular and behavioral changes were monitored in the presence or absence of MET (50 mg/kg body weight/day).MET remarkably increased the localization of precursor OLGs (NG2/O4 cells) and subsequently the renewal of mature OLGs (MOG cells) in the corpus callosum via AMPK/mammalian target of rapamycin (mTOR) pathway. Moreover, we observed a significant elevation in the antioxidant responses, especially in mature OLGs (MOG/nuclear factor erythroid 2-related factor 2 (Nrf2) cells) after MET intervention. MET also reduced brain apoptosis markers and lessened motor dysfunction in the open-field test. While MET was unable to decrease active astrogliosis (GFAP mRNA), it reduced microgliosis by down-regulation of Mac-3 mRNA a marker of pro-inflammatory microglia/macrophages. Molecular modeling studies, likewise, confirmed that MET exerts its effects via direct interaction with AMPK.Altogether, our study reveals that MET effectively induces lesion reduction and elevated molecular processes that support myelin recovery via direct activation of AMPK and indirect regulation of AMPK/Nrf2/mTOR pathway in OLGs. These findings facilitate the development of new therapeutic strategies based on AMPK activation for MS in the near future.
[27]
Houshmand F, Barati M, Golab F, et al. Metformin-induced AMPK activation stimulates remyelination through induction of neurotrophic factors, downregulation of NogoA and recruitment of Olig2+ precursor cells in the cuprizone murine model of multiple sclerosis. DARU Journal of Pharmaceutical Sciences, 2019, 27(2):583-592.
[28]
Neumann B, Baror R, Zhao C, et al. Metformin restores CNS remyelination capacity by rejuvenating aged stem cells. Cell Stem Cell, 2019, 25(4): 473-485.e8.
The age-related failure to produce oligodendrocytes from oligodendrocyte progenitor cells (OPCs) is associated with irreversible neurodegeneration in multiple sclerosis (MS). Consequently, regenerative approaches have significant potential for treating chronic demyelinating diseases. Here, we show that the differentiation potential of adult rodent OPCs decreases with age. Aged OPCs become unresponsive to pro-differentiation signals, suggesting intrinsic constraints on therapeutic approaches aimed at enhancing OPC differentiation. This decline in functional capacity is associated with hallmarks of cellular aging, including decreased metabolic function and increased DNA damage. Fasting or treatment with metformin can reverse these changes and restore the regenerative capacity of aged OPCs, improving remyelination in aged animals following focal demyelination. Aged OPCs treated with metformin regain responsiveness to pro-differentiation signals, suggesting synergistic effects of rejuvenation and pro-differentiation therapies. These findings provide insight into aging-associated remyelination failure and suggest therapeutic interventions for reversing such declines in chronic disease.Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.
[29]
Cosgrove B D, Gilbert P M, Porpiglia E, et al. Rejuvenation of the muscle stem cell population restores strength to injured aged muscles. Nature Medicine, 2014, 20(3):255-264.
[30]
Cantuti-Castelvetri L, Fitzner D, Bosch-Queralt M, et al. Defective cholesterol clearance limits remyelination in the aged central nervous system. Science, 2018, 359(6376):684-688.
Age-associated decline in regeneration capacity limits the restoration of nervous system functionality after injury. In a model for demyelination, we found that old mice fail to resolve the inflammatory response initiated after myelin damage. Aged phagocytes accumulated excessive amounts of myelin debris, which triggered cholesterol crystal formation and phagolysosomal membrane rupture and stimulated inflammasomes. Myelin debris clearance required cholesterol transporters, including apolipoprotein E. Stimulation of reverse cholesterol transport was sufficient to restore the capacity of old mice to remyelinate lesioned tissue. Thus, cholesterol-rich myelin debris can overwhelm the efflux capacity of phagocytes, resulting in a phase transition of cholesterol into crystals and thereby inducing a maladaptive immune response that impedes tissue regeneration.Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.
[31]
张莹, 孔祥熙, 侯琳, 等. Ozanimod(RPC1063)在少突胶质前体细胞分化中的作用和机制. 中国生物工程杂志, 2020, 40(6):10-19.
Zhang Y, Kong X X, Hou L, et al. Role and mechanism of ozanimod(RPC1063) in oligodendrocyte precursor cell differentiation. China Biotechnology, 2020, 40(6):10-19.
[32]
Wang J, He X L, Meng H Y, et al. Robust myelination of regenerated axons induced by combined manipulations of GPR17 and microglia. Neuron, 2020, 108(5): 876-886.e4.
[33]
Mathys H, Davila-Velderrain J, Peng Z Y, et al. Single-cell transcriptomic analysis of Alzheimer's disease. Nature, 2019, 570(7761):332-337.
[34]
Ma T C, Buescher J L, Oatis B, et al. Metformin therapy in a transgenic mouse model of Huntington's disease. Neuroscience Letters, 2007, 411(2):98-103.
[35]
Sridhar G R. Emerging links between type 2 diabetes and Alzheimer's disease. World Journal of Diabetes, 2015, 6(5):744.
[36]
Khallaghi B, Safarian F, Nasoohi S, et al. Metformin-induced protection against oxidative stress is associated with AKT/mTOR restoration in PC12 cells. Life Sciences, 2016, 148:286-292.
Reactive oxygen species have been recognized to impair cell function through suppressing Akt the well-known pro-survival molecule. Pile of concrete evidence imply metformin as an Insulin sensitizer may enhance Akt/mTOR activity however the significance of Akt/mTOR recruitment has not yet been revealed in metformin induced neuroprotection against oxidative stress.In the current study using H2O2 induced injury in PC12 cells; we first examined metformin impact on cell death by MTT assay and visual assessment. Metformin pretreated cells were then subjected to immunoblotting as well as real time PCR to find PI3K, Akt, mTOR and S6K concurrent transcriptional and post-transcriptional changes. The proportions of phosphorylated to non-phosphorylated constituents of PI3K/Akt/mTOR/S6K were determined to address their activation upon metformin treatment.According to cells morphology and MTT data metformin led to significant protection against H2O2 induced injury in 0.1 and 0.5mM concentrations. Metformin induced protection concurred with elevated PI3K/Akt/mTOR/S6K activity as well as enhanced GSH levels. These changes paralleled with a profound decline in the corresponding transcripts as determined by real time PCR.Taken together our experimentation supports the hypothesis that Akt/mTOR/S6K cascade may contribute to metformin alleviating effect. The present work while highlighting metformin anti-oxidant characteristics, concludes that Akt/mTOR signaling might be central to the drug's alleviating effects.Copyright © 2016 Elsevier Inc. All rights reserved.
[37]
Nath N, Khan M, Paintlia M K, et al. Metformin attenuated the autoimmune disease of the central nervous system in animal models of multiple sclerosis. Journal of Immunology, 2009, 182(12):8005-8014.
[38]
S Paintlia A. Combinatorial effect of metformin and lovastatin impedes T-cell autoimmunity and neurodegeneration in experimental autoimmune encephalomyelitis. Journal of Clinical & Cellular Immunology, 2013, 4(3). DOI: 10.4172/2155-9899.1000149.
[39]
Yun H E, Park S, Kim M J, et al. AMP-activated protein kinase mediates the antioxidant effects of resveratrol through regulation of the transcription factor FoxO1. The FEBS Journal, 2014, 281(19):4421-4438.
[40]
Sun Y F, Tian T, Gao J, et al. Metformin ameliorates the development of experimental autoimmune encephalomyelitis by regulating T helper 17 and regulatory T cells in mice. Journal of Neuroimmunology, 2016, 292:58-67.

RIGHTS & PERMISSIONS

Copyright reserved © 2021. Office of China Biotechnology.
PDF(1742 KB)

2276

Accesses

0

Citation

Detail

Sections
Recommended

/