
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.
Role and Mechanism of Metformin in Oligodendrocyte Precursor Cell Differentiation
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.
Metformin / Oligodendrocyte precursor cells(OPCs) / Demyelinating diseases {{custom_keyword}} /
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 |
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 |
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 |
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 |
[1] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[2] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[3] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[4] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[5] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[6] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[7] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[8] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[9] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[10] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[11] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[12] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[13] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[14] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[15] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[16] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[17] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[18] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[19] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[20] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[21] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[22] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[23] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[24] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[25] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[26] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[27] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[28] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[29] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[30] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[32] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[33] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[34] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[35] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[36] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[37] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[38] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[39] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[40] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
{{custom_ref.label}} |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
/
〈 |
|
〉 |