1
|
Cai W, Liu Y, Zhang T, Ji P, Tian C, Liu J, Zheng Z. GDNF facilitates the differentiation of ADSCs to Schwann cells and enhances nerve regeneration through GDNF/MTA1/Hes1 axis. Arch Biochem Biophys 2024; 753:109893. [PMID: 38309681 DOI: 10.1016/j.abb.2024.109893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 02/05/2024]
Abstract
Adipose tissue-derived stem cells (ADSCs) are a kind of stem cells with multi-directional differentiation potential, which mainly restore tissue repair function and promote cell regeneration. It can be directionally differentiated into Schwann-like cells to promote the repair of peripheral nerve injury. Glial cell line-derived neurotrophic factor (GDNF) plays an important role in the repair of nerve injury, but the underlying mechanism remains unclear, which seriously limits its further application.The study aimed to identify the molecular mechanism by which overexpression of glial cell line-derived neurotrophic factor (GDNF) facilitates the differentiation of ADSCs into Schwann cells, enhancing nerve regeneration after injury. In vitro, ADSCs overexpressing GDNF for 48 h exhibited changes in their morphology, with 80% of the cells having two or more prominences. Compared with that of ADSCs, GDNF-ADSCs exhibited increased expression of the Schwann cell marker S100, nerve damage repair-related factors.ADSC cells in normal culture and ADSC cells were overexpressing GDNF(GDNF-ADSCs) were analysed using TMT-Based Proteomic Analysis and revealed a significantly higher expression of MTA1 in GDNF-ADSCs than in control ADSCs. Hes1 expression was significantly higher in GDNF-ADSCs than in ADSCs and decreased by MTA1 silencing, along with a simultaneous decrease in the expression of S100 and nerve damage repair factors. These findings indicate that GDNF promotes the differentiation of ADSCs into Schwann cells and induces factors that accelerate peripheral nerve damage repair.
Collapse
Affiliation(s)
- Weixia Cai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Yang Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Ting Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Peng Ji
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Chenyang Tian
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China
| | - Jiaqi Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China.
| | - Zhao Zheng
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
2
|
Jia T, Wang M, Yan W, Wu W, Shen R. Upregulation of miR-489-3p attenuates cerebral ischemia/reperfusion injury by targeting histone deacetylase 2 (HDAC2). Neuroscience 2021; 484:16-25. [PMID: 34914969 DOI: 10.1016/j.neuroscience.2021.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 11/25/2021] [Accepted: 12/07/2021] [Indexed: 12/31/2022]
Abstract
Cerebral ischemia/reperfusion (I/R) injury is the continuation and deterioration of ischemic injury, and there are no effective treatment strategies for this condition. It has been reported that microRNAs (miRNAs) are considered as potential targets to protect the brain against I/R injury. Previous studies have shown that miR-489-3p plays a vital role in regulating apoptosis of neurons. miR-489-3p is considered as a potential target to protect the brain against I/R injury-induced neuron apoptosis. This study aimed to explore the molecular mechanism of miR-489-3p in protection against cerebral I/R injury. A rat model with cerebral I/R injury was established using the MCAO method. The cell model was constructed using the oxygen‑glucose deprivation (OGD) method. The expression of miR-489-3p was detected by qRT-PCR. The expression of HDAC2 was detected by western blot assay and immunofluorescence assay. Cell apoptosis was evaluated by flow cytometry and TUNEL staining assay. The relationship between miR-489-3p and HDAC2 was determined by bioinformatics analysis and luciferase reporter assay. Rescue experiments were performed to investigate the mechanism of the miR-489-3p/HDAC2 axis. miR-489-39 was significantly downregulated, while HDAC2 was upregulated during cerebral I/R injury both in vitro and in vivo. Upregulation of miR-489-3p obviously attenuated cerebral I/R injury by increasing PC12 cell viability, reducing LDH release, and inhibiting cell apoptosis. HDAC2 was identified as a direct target of miR-489-3p. Silencing of HDAC2 showed a neuroprotective effect against OGD/R injury in vitro. Overexpression of HDAC2 significantly attenuated the protective effects of miR-489-3p mimics on cell injury in vitro. Our results revealed that the upregulation of miR-489-3p attenuated cerebral I/R injury by negatively regulating HDAC2.
Collapse
Affiliation(s)
- Tianxia Jia
- Department of neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of science and Technology, Luoyang City, Henan Province,471003, PR. China
| | - Mengjie Wang
- Department of neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of science and Technology, Luoyang City, Henan Province,471003, PR. China
| | - Wenjun Yan
- Department of neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of science and Technology, Luoyang City, Henan Province,471003, PR. China
| | - Wenjuan Wu
- Department of neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of science and Technology, Luoyang City, Henan Province,471003, PR. China
| | - Ruile Shen
- Department of neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of science and Technology, Luoyang City, Henan Province,471003, PR. China.
| |
Collapse
|
3
|
Nuclear Receptors in Myocardial and Cerebral Ischemia-Mechanisms of Action and Therapeutic Strategies. Int J Mol Sci 2021; 22:ijms222212326. [PMID: 34830207 PMCID: PMC8617737 DOI: 10.3390/ijms222212326] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Nearly 18 million people died from cardiovascular diseases in 2019, of these 85% were due to heart attack and stroke. The available therapies although efficacious, have narrow therapeutic window and long list of contraindications. Therefore, there is still an urgent need to find novel molecular targets that could protect the brain and heart against ischemia without evoking major side effects. Nuclear receptors are one of the promising targets for anti-ischemic drugs. Modulation of estrogen receptors (ERs) and peroxisome proliferator-activated receptors (PPARs) by their ligands is known to exert neuro-, and cardioprotective effects through anti-apoptotic, anti-inflammatory or anti-oxidant action. Recently, it has been shown that the expression of aryl hydrocarbon receptor (AhR) is strongly increased after brain or heart ischemia and evokes an activation of apoptosis or inflammation in injury site. We hypothesize that activation of ERs and PPARs and inhibition of AhR signaling pathways could be a promising strategy to protect the heart and the brain against ischemia. In this Review, we will discuss currently available knowledge on the mechanisms of action of ERs, PPARs and AhR in experimental models of stroke and myocardial infarction and future perspectives to use them as novel targets in cardiovascular diseases.
Collapse
|
4
|
Zang M, Zhao Y, Gao L, Zhong F, Qin Z, Tong R, Ai L, Petersen L, Yan Y, Gao Y, Zhu C, Pu J. The circadian nuclear receptor RORα negatively regulates cerebral ischemia-reperfusion injury and mediates the neuroprotective effects of melatonin. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165890. [PMID: 32599143 DOI: 10.1016/j.bbadis.2020.165890] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/13/2020] [Accepted: 06/22/2020] [Indexed: 12/24/2022]
Abstract
Disruptions of the circadian rhythm and reduced circulating levels of the circadian hormone melatonin predispose to ischemic stroke. Although the nuclear receptor RORα is considered as a circadian rhythm regulator and a mediator of certain melatonin effects, its potential role in cerebral ischemia-reperfusion (CI/R) injury and in the neuroprotective effects of melatonin remain undefined. Here, we observed that CI/R injury in RORα-deficient mice was associated with greater cerebral infarct size, brain edema, and cerebral apoptosis compared with wild-type model. In contrast, transgenic mice with brain-specific overexpression of RORα versus non-transgenic controls exerted significantly reduced infarct volume, brain edema and apoptotic response induced by CI/R. Mechanistically, RORα deficiency was found to exacerbate apoptosis pathways mediated by endoplasmic-reticulum stress and mitochondria and aggravate oxidative/nitrative stress after CI/R. Further studies revealed that RORα deficiency intensified the activation of nuclear factor-κB signaling induced by CI/R. Given the emerging evidence of RORα as an essential melatonin activity mediator, we further investigated the RORα roles in melatonin-exerted neuroprotection against acute ischemic stroke. Melatonin treatment significantly decreased infarct volume and cerebral apoptosis; mitigated endoplasmic reticulum stress and mitochondrial dysfunction; and inhibited CI/R injury-induced oxidative/nitrative stress and nuclear factor-κB activation, which was eradicated in RORα-deficient mice. Collectively, current findings suggest that RORα is a novel endogenous neuroprotective receptor, and a pivotal mediator of melatonin's suppressive effects against CI/R injury.
Collapse
Affiliation(s)
- Minhua Zang
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai, China
| | - Yichao Zhao
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai, China
| | - Lingchen Gao
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai, China
| | - Fangyuan Zhong
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai, China
| | - Zihan Qin
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai, China
| | - Renyang Tong
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai, China
| | - Lulu Ai
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lauren Petersen
- Department of Anesthesiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Yang Yan
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai, China
| | - Yu Gao
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai, China
| | - Cansheng Zhu
- Department of Neurology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Jun Pu
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai, China.
| |
Collapse
|
5
|
Xu Z, Zou C, Guo M, Bian H, Zhao W, Wang J. Metastasis-associated protein 1 (MTA1) regulates the catecholamine production homeostasis via transcriptional repression of aromatic l-amino acid decarboxylase (Aadc) in the interstitial cells of Cajal of mouse prostate. Biochem Biophys Res Commun 2020; 528:732-739. [PMID: 32522342 DOI: 10.1016/j.bbrc.2020.05.125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 05/17/2020] [Indexed: 12/30/2022]
Abstract
Based on the lately identified role for the interstitial cells of Cajal (ICCs) of mouse prostate in catecholamine production, as well as the well-established role for the master coregulator metastasis-associated protein 1 (MTA1) in inflammation, we probed into the functional link between aberrant MTA1 expression and pathogenesis of chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) using both a MTA1-/- mouse model of experimental autoimmune prostatitis (EAP) and an in vitro chronic prostatitis model in cultured murine ICCs. EAP-induced MTA1 expression was enriched in ICCs of mouse prostate. EAP resulted in a higher increase in the pelvic pain response in MTA1-/- mice compared to WT mice. Consistently, the ICCs from MTA1-/- mice produced higher levels of catecholamines upon induction of in vitro chronic prostatitis. Mechanistically, MTA1 could directly suppress the transcription of Aadc, a rate-limiting enzyme during catecholamine synthesis, in a HDAC2-depdendent manner. Importantly, treatment with AADC inhibitor NSD-1015 significantly ameliorated EAP-elicited pain response and catecholamine overactivity in MTA1-/- mice. Taken together, our findings reveal an inherent regulatory role of the MTA1/AADC pathway in the maintenance of catecholamine production homeostasis in prostate ICCs, and also point to a potential use of HDAC inhibitors and/or AADC inhibitors to treat CP/CPPS.
Collapse
Affiliation(s)
- Zhibin Xu
- Department of Urology, Taizhou People's Hospital, Taizhou, 225300, Jiangsu Province, PR China
| | - Chunbo Zou
- Department of Urology, Taizhou People's Hospital, Taizhou, 225300, Jiangsu Province, PR China
| | - MaoMao Guo
- Department of Urology, Taizhou People's Hospital, Taizhou, 225300, Jiangsu Province, PR China
| | - Hao Bian
- Department of Urology, Taizhou People's Hospital, Taizhou, 225300, Jiangsu Province, PR China
| | - Wenchao Zhao
- Department of Urology, Taizhou People's Hospital, Taizhou, 225300, Jiangsu Province, PR China
| | - Jiangping Wang
- Department of Urology, Taizhou People's Hospital, Taizhou, 225300, Jiangsu Province, PR China.
| |
Collapse
|
6
|
Liu W, Guo Q, Zhao H. Oxidative stress-elicited YY1 potentiates antioxidative response via enhancement of NRF2-driven transcriptional activity: A potential neuronal defensive mechanism against ischemia/reperfusion cerebral injury. Biomed Pharmacother 2018; 108:698-706. [DOI: 10.1016/j.biopha.2018.09.082] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 09/10/2018] [Accepted: 09/14/2018] [Indexed: 12/21/2022] Open
|
7
|
Wnuk A, Rzemieniec J, Litwa E, Lasoń W, Kajta M. Prenatal exposure to benzophenone-3 (BP-3) induces apoptosis, disrupts estrogen receptor expression and alters the epigenetic status of mouse neurons. J Steroid Biochem Mol Biol 2018; 182:106-118. [PMID: 29704544 DOI: 10.1016/j.jsbmb.2018.04.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/17/2018] [Accepted: 04/24/2018] [Indexed: 12/31/2022]
Abstract
Current evidence indicates that benzophenone-3 (BP-3) can pass through the placental and blood-brain barriers and thus can likely affect infant neurodevelopment. Despite widespread exposure, data showing the effects of BP-3 on the developing nervous system are scarce. This study revealed for the first time that prenatal exposure to BP-3 led to apoptosis and neurotoxicity, altered the levels of estrogen receptors (ERs) and changed the epigenetic status of mouse neurons. In the present study, subcutaneous injections of pregnant mice with BP-3 at 50 mg/kg, which is an environmentally relevant dose, evoked activation of caspase-3 and lactate dehydrogenase (LDH) release as well as substantial loss of mitochondrial membrane potential in neocortical cells of their embryonic offspring. Apoptosis-focused microarray analysis of neocortical cells revealed up-regulation of 22 genes involved in apoptotic cell death. This effect was supported by increased BAX and CASP3 mRNA and protein levels, as evidenced by qPCR, ELISAs and western blots. BP-3-induced apoptosis and neurotoxicity were accompanied by decreases in the mRNA and protein expression levels of ESR1 and ESR2 (also known as ERα and ERβ), with a simultaneous increase in GPER1 (also known as GPR30) expression. In addition to the demonstration that treatment of pregnant mice with BP-3 induced apoptosis, caused neurotoxicity and altered ERs expression levels in neocortical cells of their embryonic offspring, we showed that prenatal administration of BP-3 inhibited global DNA methylation as well as reduced DNMTs activity. BP-3 also caused specific hypomethylation of the genes Gper1 and Bax, an effect that was accompanied by increased mRNA and protein expression levels. In addition, BP-3 caused hypermethylation of the genes Esr1, Esr2 and Bcl2, which could explain the reduced mRNA and protein levels of the estrogen receptors. This study demonstrated for the first time that prenatal exposure to BP-3 caused severe neuronal apoptosis that was accompanied by impaired ESR1/ESR2 expression, enhanced GPER1 expression, global DNA hypomethylation and altered methylation statuses of apoptosis-related and ERs genes. We suggest that the effects of BP-3 in embryonic neurons may be the fetal basis of the adult onset of nervous system disease.
Collapse
Affiliation(s)
- Agnieszka Wnuk
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - Joanna Rzemieniec
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - Ewa Litwa
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - Władysław Lasoń
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland
| | - Małgorzata Kajta
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, 31-343 Krakow, Smetna Street 12, Poland.
| |
Collapse
|
8
|
Guo Y, Wang LP, Li C, Xiong YX, Yan YT, Zhao LQ, Li SD, Sun J, Luo HY, Xian CJ. Effects of Ginsenoside Rb1 on Expressions of Phosphorylation Akt/Phosphorylation mTOR/Phosphorylation PTEN in Artificial Abnormal Hippocampal Microenvironment in Rats. Neurochem Res 2018; 43:1927-1937. [PMID: 30167941 DOI: 10.1007/s11064-018-2612-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 07/31/2018] [Accepted: 08/09/2018] [Indexed: 02/07/2023]
Abstract
Artificial abnormal microenvironment caused by microperfusion of L-glutamate (Glu) and Ca2+ in the hippocampus results in neuron damage, which is closely related to cerebral ischemia. Ginsenoside Rb1, a compound from Panax notoginseng, was previously used to counter the artificial abnormal hippocampal environment in a microperfusion model. In addition, while the Akt/mTOR/PTEN signaling pathway has been shown to mediate neuronprotection in cerebral ischemia, whether this pathway is involved in the neuroprotection of ginsenoside Rb1 is unknown. Here SH-SY5Y cells exposed to OGD/R injury in treated with LY294002, ginsenoside Rb1, ginsenoside Rb1+ LY294002. Expressions of phosphorylation (P-)Akt/P-mTOR/P-PTEN (24 h after OGD/R) were detected by Western blotting. Effects were examined via the memory function of rats (by Morris water maze test), morphological changes in pyramidal cell (by histology), and mRNA expression (by qRT-PCR) and phosphorylation (P-) (by Western blotting and immunohistochemical staining) of Akt, P-mTOR, and P-PTEN in the hippocampus. The memory deficit of rats and pyramidal cellular necrosis and apoptosis in the CA1 region of hippocampus after microperfusion of Glu and Ca2+ were dose dependently alleviated by ginsenoside Rb1.Moreover,Western blot showed that ginsenoside Rb1 increased the expressions of P-Akt, P-mTOR and reduced P-PTEN in vivo and vitro. Thus, the potent neuroprotection of ginsenoside Rb1 in artificial abnormal microenvironment is, at least partially, related to the activation of P-AKT/P-mTOR signaling pathway and inhibition of P-PTEN protein.
Collapse
Affiliation(s)
- Ying Guo
- Department of Pharmacology, College of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Li-Ping Wang
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, 5001, Australia
| | - Chen Li
- Department of Pharmacology, College of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Yun-Xia Xiong
- Department of Pharmacology, College of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Yi-Tian Yan
- Department of Pharmacology, College of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Li-Qin Zhao
- Department of Pharmacology, College of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Shu-De Li
- Department of Biochemistry, College of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Jun Sun
- Department of Anatomy, College of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.
| | - Hai-Yun Luo
- Department of Pharmacology, College of Basic Medicine, Kunming Medical University, Kunming, 650500, Yunnan, China.
| | - Cory J Xian
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, 5001, Australia
| |
Collapse
|
9
|
Wnuk A, Kajta M. Steroid and Xenobiotic Receptor Signalling in Apoptosis and Autophagy of the Nervous System. Int J Mol Sci 2017; 18:ijms18112394. [PMID: 29137141 PMCID: PMC5713362 DOI: 10.3390/ijms18112394] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/06/2017] [Accepted: 11/09/2017] [Indexed: 12/15/2022] Open
Abstract
Apoptosis and autophagy are involved in neural development and in the response of the nervous system to a variety of insults. Apoptosis is responsible for cell elimination, whereas autophagy can eliminate the cells or keep them alive, even in conditions lacking trophic factors. Therefore, both processes may function synergistically or antagonistically. Steroid and xenobiotic receptors are regulators of apoptosis and autophagy; however, their actions in various pathologies are complex. In general, the estrogen (ER), progesterone (PR), and mineralocorticoid (MR) receptors mediate anti-apoptotic signalling, whereas the androgen (AR) and glucocorticoid (GR) receptors participate in pro-apoptotic pathways. ER-mediated neuroprotection is attributed to estrogen and selective ER modulators in apoptosis- and autophagy-related neurodegenerative diseases, such as Alzheimer’s and Parkinson’s diseases, stroke, multiple sclerosis, and retinopathies. PR activation appeared particularly effective in treating traumatic brain and spinal cord injuries and ischemic stroke. Except for in the retina, activated GR is engaged in neuronal cell death, whereas MR signalling appeared to be associated with neuroprotection. In addition to steroid receptors, the aryl hydrocarbon receptor (AHR) mediates the induction and propagation of apoptosis, whereas the peroxisome proliferator-activated receptors (PPARs) inhibit this programmed cell death. Most of the retinoid X receptor-related xenobiotic receptors stimulate apoptotic processes that accompany neural pathologies. Among the possible therapeutic strategies based on targeting apoptosis via steroid and xenobiotic receptors, the most promising are the selective modulators of the ER, AR, AHR, PPARγ agonists, flavonoids, and miRNAs. The prospective therapies to overcome neuronal cell death by targeting autophagy via steroid and xenobiotic receptors are much less recognized.
Collapse
Affiliation(s)
- Agnieszka Wnuk
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Smetna Street 12, 31-343 Krakow, Poland.
| | - Małgorzata Kajta
- Institute of Pharmacology, Polish Academy of Sciences, Department of Experimental Neuroendocrinology, Smetna Street 12, 31-343 Krakow, Poland.
| |
Collapse
|
10
|
Gao ZJ, Min J, Wu XC, Yang T, Yan CY, Dong BH, Zhang T. Repression of neuronal nitric oxide (nNOS) synthesis by MTA1 is involved in oxidative stress-induced neuronal damage. Biochem Biophys Res Commun 2016; 479:40-7. [PMID: 27603575 DOI: 10.1016/j.bbrc.2016.09.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 09/03/2016] [Indexed: 12/14/2022]
Abstract
The Metastasis-associated protein 1 (MTA1) coregulator, an essential component of the nucleosome remodeling and deacetylase (NuRD) complex, potentiates neuroprotective effects against ischemia/reperfusion (I/R) injury. But the underlying mechanism(s) remain largely unknown. Here, we discovered that neuronal MTA1 was a target of oxidative stress, and stimulation of neurons with oxygen glucose deprivation (OGD) treatment significantly inhibited MTA1 expression. Additionally, MTA1 depletion augmented ischemic oxidative stress and thus promoted oxidative stress-induced neuronal cell death by OGD. While studying the impact of MTA1 status on global neuronal gene expression, we unexpectedly discovered that MTA1 may modulate OGD-induced neuronal damage via regulation of distinct nitric oxide synthase (NOS) (namely neuronal NOS, nNOS) signaling. We provided in vitro evidence that NOS1 is a chromatin target of MTA1 in OGD-insulted neurons. Mechanistically, neuronal ischemia-mediated repression of NOS1 expression is accompanied by the enhanced recruitment of MTA1 along with histone deacetylases (HDACs) to the NOS1 promoter, which could be effectively blocked by a pharmacological inhibitor of the HDACs. These findings collectively reveal a previously unrecognized, critical homeostatic role of MTA1, both as a target and as a component of the neuronal oxidative stress, in the regulation of acute neuronal responses against brain I/R damage. Our study also provides a molecular mechanistic explanation for the previously reported neurovascular protection by selective nNOS inhibitors.
Collapse
Affiliation(s)
- Zi-Jun Gao
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China
| | - Jie Min
- Department of Ophthalmology, Xi'an No. 4 Hospital, Guangren Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xu-Cai Wu
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China
| | - Tian Yang
- The 1st Cadet Brigade, Fourth Military Medical University, Xi'an 710032, China
| | - Chang-You Yan
- Xi'an Health Management Service Center, Xi'an 710032, China
| | - Bu-Huai Dong
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China.
| | - Tao Zhang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China.
| |
Collapse
|