151
|
Melatonin alleviates asphyxial cardiac arrest-induced cerebellar Purkinje cell death by attenuation of oxidative stress. Exp Neurol 2019; 320:112983. [PMID: 31251935 DOI: 10.1016/j.expneurol.2019.112983] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 06/07/2019] [Accepted: 06/22/2019] [Indexed: 12/21/2022]
Abstract
Although multiple reports using animal models have confirmed that melatonin appears to promote neuroprotective effects following ischemia/reperfusion-induced brain injury, the relationship between its protective effects and activation of autophagy in Purkinje cells following asphyxial cardiac arrest and cardiopulmonary resuscitation (CA/CPR) remains unclear. Rats used in this study were randomly assigned to 6 groups as follows; vehicle-treated sham operated group, vehicle-treated asphyxial CA/CPR operated group, melatonin-treated sham operated group, melatonin-treated asphyxial CA/CPR operated group, PDOT (a MT2 melatonin receptor antagonist) plus (+) melatonin-treated sham operated group and PDOT+melatonin-treated asphyxial CA/CPR operated group. Melatonin (20 mg/kg, i.p., 4 times before CA and 3 times after CA) treatment significantly improved survival rate and neurological deficit compared with the vehicle-treated asphyxial CA/CPR rats (survival rates ≥40% vs 10%), showing that melatonin treatment exhibited protective effect against asphyxial CA/CPR-induced Purkinje cell death. The protective effect of melatonin against CA/CPR-induced Purkinje cell death paralleled a remarkable attenuation of autophagy-like processes (Beclin-1, Atg7 and LC3), as well as a dramatic reduction in superoxide anion radical (O2·-), intense enhancements of CuZn superoxide dismutase (SOD1) and MnSOD (SOD2) expressions. Furthermore, the protective effect was notably reversed by treatment with PDOT, which is a selective MT2 antagonist. In brief, melatonin conferred neuroprotection against asphyxial CA/CPR-induced Purkinje cell death via inhibiting autophagic activation by reducing expressions of O2·- and increasing expressions of antioxidant enzymes, and suggests that MT2 is involved in neuroprotective effect of melatonin against Purkinje cell death caused by asphyxial CA/CPR.
Collapse
|
152
|
Thiebaut AM, Hedou E, Marciniak SJ, Vivien D, Roussel BD. Proteostasis During Cerebral Ischemia. Front Neurosci 2019; 13:637. [PMID: 31275110 PMCID: PMC6594416 DOI: 10.3389/fnins.2019.00637] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/03/2019] [Indexed: 12/21/2022] Open
Abstract
Cerebral ischemia is a complex pathology involving a cascade of cellular mechanisms, which deregulate proteostasis and lead to neuronal death. Proteostasis refers to the equilibrium between protein synthesis, folding, transport, and protein degradation. Within the brain proteostasis plays key roles in learning and memory by controlling protein synthesis and degradation. Two important pathways are implicated in the regulation of proteostasis: the unfolded protein response (UPR) and macroautophagy (called hereafter autophagy). Both are necessary for cell survival, however, their over-activation in duration or intensity can lead to cell death. Moreover, UPR and autophagy can activate and potentiate each other to worsen the issue of cerebral ischemia. A better understanding of autophagy and ER stress will allow the development of therapeutic strategies for stroke, both at the acute phase and during recovery. This review summarizes the latest therapeutic advances implicating ER stress or autophagy in cerebral ischemia. We argue that the processes governing proteostasis should be considered together in stroke, rather than focusing either on ER stress or autophagy in isolation.
Collapse
Affiliation(s)
- Audrey M Thiebaut
- INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, University of Caen Normandy, Caen, France
| | - Elodie Hedou
- INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, University of Caen Normandy, Caen, France
| | - Stefan J Marciniak
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom.,Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Denis Vivien
- INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, University of Caen Normandy, Caen, France.,Department of Clinical Research, University of Caen Normandy, Caen, France
| | - Benoit D Roussel
- INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, University of Caen Normandy, Caen, France
| |
Collapse
|
153
|
Lee JY, Song H, Dash O, Park M, Shin NE, McLane MW, Lei J, Hwang JY, Burd I. Administration of melatonin for prevention of preterm birth and fetal brain injury associated with premature birth in a mouse model. Am J Reprod Immunol 2019; 82:e13151. [PMID: 31131935 DOI: 10.1111/aji.13151] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/25/2019] [Accepted: 05/19/2019] [Indexed: 12/14/2022] Open
Abstract
PROBLEM Maternal inflammation leads to preterm birth and perinatal brain injury. Melatonin, through its anti-inflammatory effects, has been shown to be protective against inflammation-induced perinatal adverse effects. However, the immunomodulatory effects of melatonin on preterm birth and prematurity-related morbidity remain unknown. We wanted to investigate the effects of maternally administered melatonin on preterm birth and perinatal brain injury in a mouse model of maternal inflammation. METHOD OF STUDY A model of maternal inflammation employing lipopolysaccharide (LPS) was used to mimic the most common clinical scenario of preterm birth, that of maternal inflammation. Mice were randomly divided into the following groups: control, LPS, and LPS with melatonin pre-treatment. Doppler ultrasonography was used to obtain fetal and maternal hemodynamic measurements in utero. Placenta and fetal brains were harvested and analyzed for proinflammatory markers and signs of perinatal brain injury, respectively. Surviving offspring were assessed for neuromotor outcomes. RESULTS Melatonin pre-treatment lowered the level of proinflammatory cytokines in the uterus and the placenta, significantly improved LPS-induced acute fetal neuroinflammation and perinatal brain injury, as well as significantly upregulated the SIRT1/Nrf2 signaling pathway to reduce LPS-induced inflammation. Melatonin also prevented adverse neuromotor outcomes in offspring exposed to maternal inflammation. CONCLUSION Maternally administered melatonin modulated immune responses to maternal inflammation and decreased preterm birth and perinatal brain injury. These results suggest that melatonin, a safe treatment during pregnancy, may be used as an experimental therapeutic in clinical trials.
Collapse
Affiliation(s)
- Ji Yeon Lee
- Department of Obstetrics and Gynecology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea.,Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Haengseok Song
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Korea
| | - Oyunbileg Dash
- Department of Obstetrics and Gynecology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
| | - Mira Park
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Korea
| | - Na E Shin
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael W McLane
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jun Lei
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jong Yun Hwang
- Department of Obstetrics and Gynecology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Irina Burd
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
154
|
Nan D, Jin H, Deng J, Yu W, Liu R, Sun W, Huang Y. Cilostazol ameliorates ischemia/reperfusion-induced tight junction disruption in brain endothelial cells by inhibiting endoplasmic reticulum stress. FASEB J 2019; 33:10152-10164. [PMID: 31184927 DOI: 10.1096/fj.201900326r] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Endoplasmic reticulum (ER) stress is essential for brain ischemia/reperfusion (I/R) injury. However, whether it contributes to I/R-induced blood-brain barrier (BBB) injury remains unclear. cilostazol exerts protective effects toward I/R-induced BBB injury, with unclear mechanisms. This study explored the potential role of ER stress in I/R-induced endothelial cell damage and determined whether the therapeutic potential of cilostazol, with respect to I/R-induced endothelial cell damage, is related to inhibition of ER stress. We found that exposing brain endothelial cells (bEnd.3) to oxygen-glucose deprivation/reoxygenation (OGD/R) significantly activated ER stress and diminished the barrier function of cell monolayers; treatment with the ER stress inhibitor 4-phenylbutyric acid (4-PBA) or cilostazol prevented OGD/R-induced ER stress and preserved barrier function. Furthermore, OGD/R induced the expression and secretion of matrix metalloproteinase-9 and nuclear translocation of phosphorylated NF-κB. These changes were partially reversed by 4-PBA or cilostazol treatment. In vivo, 4-PBA or cilostazol significantly attenuated I/R-induced ER stress and ameliorated Evans blue leakage and tight junction loss. These results demonstrate that I/R-induced ER stress participates in BBB disruption. Targeting ER stress could be a useful strategy to protect the BBB from ischemic stroke, and cilostazol is a promising therapeutic agent for this process.-Nan, D., Jin, H., Deng, J., Yu, W., Liu, R., Sun, W., Huang, Y. Cilostazol ameliorates ischemia/reperfusion-induced tight junction disruption in brain endothelial cells by inhibiting endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Ding Nan
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Haiqiang Jin
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Jianwen Deng
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Weiwei Yu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Ran Liu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Weiping Sun
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Yining Huang
- Department of Neurology, Peking University First Hospital, Beijing, China
| |
Collapse
|
155
|
Chen C, Ling M, Lin F, Xu L, Lv ZM. Melatonin appears to protect against steroidogenic collapse in both mice fed with high‐fat diet and H
2
O
2
‐treated TM3 cells. Andrologia 2019; 51:e13323. [PMID: 31134680 DOI: 10.1111/and.13323] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 04/06/2019] [Accepted: 04/30/2019] [Indexed: 12/21/2022] Open
Affiliation(s)
- Chao Chen
- Department of Histology and Embryology Anhui Medical University Hefei China
- Department of Operating Room The First Affiliated Hospital of USTC (Anhui Provincial Hospital) Hefei China
| | - Meng‐yu Ling
- Department of Histology and Embryology Anhui Medical University Hefei China
| | - Fan‐hong Lin
- Department of Histology and Embryology Anhui Medical University Hefei China
| | - Ling Xu
- Department of Histology and Embryology Anhui Medical University Hefei China
| | - Zheng Mei Lv
- Department of Histology and Embryology Anhui Medical University Hefei China
| |
Collapse
|
156
|
Su Q, Liu Y, Lv XW, Ye ZL, Sun YH, Kong BH, Qin ZB. Inhibition of lncRNA TUG1 upregulates miR-142-3p to ameliorate myocardial injury during ischemia and reperfusion via targeting HMGB1- and Rac1-induced autophagy. J Mol Cell Cardiol 2019; 133:12-25. [PMID: 31145943 DOI: 10.1016/j.yjmcc.2019.05.021] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 05/07/2019] [Accepted: 05/25/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) play a central role in regulating heart diseases. In the present study, we examined the effects of lncRNA taurine up-regulated gene 1 (TUG1) in ischemia/reperfusion (I/R)- or hydrogen peroxide-challenged cardiomyocytes, with specific focus on autophagy-induced cell apoptosis. METHODS The expressions of miR-142-3p and TUG1 in H2O2-challenged cardiomyocytes and I/R-injured heart tissue were measured by RT-qPCR. Cell death was measured by trypan blue staining assay. Cell apoptosis was determined by Annexin V/PI staining and TUNEL assay. Autophagy was examined by quantifying cells or tissues containing LC3+ autophagic vacuoles by immunofluorescence, or by measuring the expressions of autophagy-related biomarkers by Western blot. The direct interaction between miR-142-3p and TUG1, high mobility group box 1 protein (HMGB1), or Ras-related C3 botulinum toxin substrate 1 (Rac1) was examined using luciferase reporter assay. The significance of miR-142-3p and TUG1 on cell apoptosis or autophagy was examined using both gain-of-function and loss-of-function approaches. The importance of HMGB1 or Rac1 was assessed using siRNA-mediated gene silencing. RESULTS miR-142-3p was down-regulated, while TUG1 up-regulated in H2O2-challenged cardiomyocytes in vitro and I/R-injured heart tissues in vivo. Functionally, inhibition of TUG1 and overexpression of miR-142-3p inhibited cell apoptosis and autophagy in cardiomyocytes. The function of TUG1 were achieved by sponging miR-142-3p and releasing the suppression of the putative targets of miR-142-3p, HMGB1 and Rac1. Both HMGB1 and Rac1 essentially mediated cell apoptosis and autophagy induced by TUG1. CONCLUSIONS TUG1, by targeting miR-142-3p and up-regulating HMGB1 and Rac1, plays a central role in stimulating autophagic cell apoptosis in ischemia/hypoxia-challenged cardiomyocytes. Down-regulating TUG1 or up-regulating miR-142-3p may ameliorate myocardial injury and protect against acute myocardial infarction.
Collapse
Affiliation(s)
- Qiang Su
- Department of Cardiology, The Affiliated Hospital of Guilin Medical University, Guilin 541001, PR China.
| | - Yang Liu
- Department of Cardiology, The Second People's Hospital of Nanning City, The Third Affiliated Hospital of Guangxi Medical University, Nanning 530031, PR China
| | - Xiang-Wei Lv
- Department of Cardiology, The Affiliated Hospital of Guilin Medical University, Guilin 541001, PR China
| | - Zi-Liang Ye
- Department of Cardiology, The Affiliated Hospital of Guilin Medical University, Guilin 541001, PR China
| | - Yu-Han Sun
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, PR China
| | - Bing-Hui Kong
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, PR China
| | - Zhen-Bai Qin
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, PR China
| |
Collapse
|
157
|
Wang MM, Feng YS, Yang SD, Xing Y, Zhang J, Dong F, Zhang F. The Relationship Between Autophagy and Brain Plasticity in Neurological Diseases. Front Cell Neurosci 2019; 13:228. [PMID: 31244604 PMCID: PMC6542992 DOI: 10.3389/fncel.2019.00228] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 05/07/2019] [Indexed: 11/17/2022] Open
Abstract
Autophagy, a catabolic degradation system, is utilized for destroying and recycling the damaged or unnecessary cellular components. Brain plasticity refers to the remarkable characteristics of brain neurons that change their structure and function according to previous experience. This review was performed by searching the relevant articles in databases of SCIENCEDIRECT, PUBMED, and Web of Science, from respective inception to January 2019. Here, we review the neuroprotective effect of autophagy in neurological diseases and the mechanism of autophagy in brain plasticity. Moreover, the mechanism of autophagy in the process of brain plasticity can provide the possibility for the development of new treatment methods in the future, thus benefiting patients with neurological diseases. In summary, autophagy and brain plasticity play important roles in neurological diseases.
Collapse
Affiliation(s)
- Man-Man Wang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ya-Shuo Feng
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Si-Dong Yang
- Department of Spine Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ying Xing
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jing Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Provincial Orthopedic Biomechanics Key Laboratory, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
158
|
Zhang B, Zhang HX, Shi ST, Bai YL, Zhe X, Zhang SJ, Li YJ. Interleukin-11 treatment protected against cerebral ischemia/reperfusion injury. Biomed Pharmacother 2019; 115:108816. [PMID: 31096144 DOI: 10.1016/j.biopha.2019.108816] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 03/19/2019] [Accepted: 03/26/2019] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Inflammation and immune responses are crucial factors associated with the onset and progression of stroke. Interleukin-11 (IL-11) is a hematopoietic IL-6 family cytokine that functions as an anti-inflammatory agent against various inflammatory diseases. However, its roles in stroke remain unknown. In this study, we investigated the effects of IL-11 on cerebral ischemia-reperfusion injury in a model of focal cerebral ischemia. METHODS Mice were randomly divided into five groups the vehicle group, the middle cerebral artery occlusion (MCAO) group, the MCAO plus adenosine monophosphate-activated protein kinase (AMPK) inhibitor compound C group, the MCAO plus IL-11 treatment group, and the MCAO plus IL-11 treatment and compound C group. Focal cerebral ischemia was induced by occluding the left middle cerebral artery, and reperfusion was achieved by withdrawing the suture 2 h after ischemia. The protein expression levels of IL-11 were measured using Western blot analysis, and its location was detected using immunohistochemistry and immunofluorescence staining. The infarct volume was examined using 2,3,5-triphenyl tetrazolium chloride (TTC) staining, and the neurobehavioral progression was assessed using the neurological scoring system. The expression of astrocytes and microglia was detected using immunochemistry, and real-time quantitative PCR was used for the gene quantification of inflammatory cytokines. The extent of cerebral ischemia-reperfusion injury was tested using Nissl staining and the terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) assay. The expression of the apoptotic proteins Bax, Bcl-2 and cleaved caspase-3 were detected using Western blot analysis, and the oxidative stress was also measured. RESULTS The expression of IL-11 mRNA and protein significantly decreased after cerebral ischemia. Immunohistochemical staining showed a large amount of IL-11 in the cerebral cortex of the mice in the vehicle group, whereas the immunoreactivity of IL-11 remained weak for 24 h in the MCAO group. Immunofluorescent staining further confirmed that IL-11 was mainly expressed in the neurons. It was suggested that IL-11 (20 μg/kg) treatment ameliorated infarction and reduced neurological scores. In addition, IL-11 proved to reduce neuropathic damage, glial activation, and the expression of proinflammatory cytokines and increase the expression of anti-inflammatory cytokines after cerebral ischemia. IL-11 was also able to alleviate oxidative stress caused by cerebral ischemia, and AMPK inhibition enhanced the alleviation. Moreover, IL-11 was found to inhibit apoptosis caused by cerebral ischemia, which could also be facilitated by AMPK inhibitors. SIGNIFICANCE Our research suggests that IL-11 is decreased during cerebral ischemia-reperfusion injury, but IL-11 treatment can improve neurological function and reduce the cerebral infarct volume, which can trigger stroke in mice. AMPK inhibition can further promote the protective effect of IL-11 in stroke. Overall, we demonstrate that IL-11 is of therapeutic interest in controlling stroke and managing cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Bei Zhang
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China
| | - Hai-Xiong Zhang
- Otolaryngology Department, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China
| | - Shao-Ting Shi
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China
| | - Yu-Lan Bai
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China
| | - Xiao Zhe
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China
| | - Shi-Jun Zhang
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China
| | - Ya-Jun Li
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China.
| |
Collapse
|
159
|
Luo R, Liao Z, Song Y, Yin H, Zhan S, Li G, Ma L, Lu S, Wang K, Li S, Zhang Y, Yang C. Berberine ameliorates oxidative stress-induced apoptosis by modulating ER stress and autophagy in human nucleus pulposus cells. Life Sci 2019; 228:85-97. [PMID: 31047897 DOI: 10.1016/j.lfs.2019.04.064] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/15/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023]
Abstract
AIM Nucleus pulposus (NP) cell apoptosis induced by oxidative stress is known to be closely involved in the pathogenesis of intervertebral disc (IVD) degeneration. Berberine, a small molecule derived from Rhizoma coptidis, has been found to exert antioxidative activity and preserve cell viability. The present study aims to investigate whether berberine can prevent NP cell apoptosis under oxidative damage and the potential underlying mechanisms. METHODS AND MATERIALS The effects of berberine on IVD degeneration were investigated both in vitro and in vivo. KEY FINDINGS Our results showed that berberine significantly mitigated oxidative stress-decreased cell viability as well as apoptosis in human NP cells. Berberine treatment could attenuate oxidative stress-induced ER stress and autophagy in a concentration-dependent manner. With 4-PBA (ER stress specific inhibitor) and 3-MA (autophagy specific inhibitor) administration, we demonstrated that berberine inhibited oxidative stress-induced apoptosis by modulating the ER stress and autophagy pathway. We also found that the IRE1/JNK pathway was involved in the induction of ER stress-dependent autophagy. With Ca2+ chelator BAPTA-AM utilization, we revealed that oxidative stress-mediated ER stress and autophagy repressed by berberine could be restored by inducing intracellular Ca2+ dysregulation. Furthermore, in vivo study provided evidence that berberine treatment could retard the process of puncture-induced IVD degeneration in a rat model. SIGNIFICANCE Our results indicate that berberine could prevent oxidative stress-induced apoptosis by modulating ER stress and autophagy, thus offering a novel potential pharmacological treatment strategy for IVD degeneration.
Collapse
Affiliation(s)
- Rongjin Luo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhiwei Liao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yu Song
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Huipeng Yin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shengfeng Zhan
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Gaocai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liang Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Saideng Lu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kun Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shuai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yukun Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
160
|
Shi Q, Zhang Q, Peng Y, Zhang X, Wang Y, Shi L. A natural diarylheptanoid protects cortical neurons against oxygen–glucose deprivation-induced autophagy and apoptosis. J Pharm Pharmacol 2019; 71:1110-1118. [DOI: 10.1111/jphp.13096] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 03/16/2019] [Indexed: 12/31/2022]
Abstract
Abstract
Objectives
This study aims to investigate the neuroprotective effects of curcumin analogues, 7-(4-Hydroxy-3-methoxyphenyl)-1-phenyl-4E-hepten-3-one (AO-2) on oxygen–glucose deprivation and re-oxygenation (OGD/R) induced injury in cortical neurons, which is a widely accepted in-vitro model for ischaemic reperfusion.
Methods
In this study, AO-2 was added to cortical neurons for 2 h as pretreatment, and then cortical neurons were subjected to OGD/R in the presence of AO-2 for 4 h. Cell viability was tested by 2′, 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide assay and apoptosis by flow cytometry and Live & Dead cell assay. Western blot analysis detected the change in AKT/mTOR (mammalian target of rapamycin) signalling pathway.
Key findings
Treatment of AO-2 increased cell survival of OGD/R-treated cortical neurons. Transient AKT/mTOR inhibition, induction of the autophagy marker LC3-II (microtubule-associated protein 1A/1B-light chain 3 phosphatidylethanolamine conjugate), and cleavage of the apoptosis marker Caspase-3 were observed at different stages of OGD/R, and AO-2 reversed all three events. Importantly, treatment of the mTOR inhibitor rapamycin blocked the neuroprotective effects of AO-2 on reducing LC3-II and cleaved Caspase-3 expression and cancelled AO-2-mediated neuronal survival.
Conclusions
These results demonstrate that AO-2 increases resistance of cortical neurons to OGD/R by decreasing autophagy and cell apoptosis, which involves an mTOR-dependent mechanism.
Collapse
Affiliation(s)
- Qiaoyun Shi
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, Jinan University, Guangzhou, Guangdong, China
| | - Qinghua Zhang
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, Jinan University, Guangzhou, Guangdong, China
| | - Yinghui Peng
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, Jinan University, Guangzhou, Guangdong, China
| | - Xiaoqi Zhang
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, Jinan University, Guangzhou, Guangdong, China
| | - Ying Wang
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, Jinan University, Guangzhou, Guangdong, China
| | - Lei Shi
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
161
|
Tang Y, Shen J, Zhang F, Yang FY, Liu M. Human serum albumin attenuates global cerebral ischemia/reperfusion-induced brain injury in a Wnt/β-Catenin/ROS signaling-dependent manner in rats. Biomed Pharmacother 2019; 115:108871. [PMID: 31026729 DOI: 10.1016/j.biopha.2019.108871] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/06/2019] [Accepted: 04/09/2019] [Indexed: 11/26/2022] Open
Abstract
This study sought to clarify the role and underlying mechanisms of human serum albumin (HSA) therapy in global cerebral ischemia/reperfusion (GCI/R)-induced brain damage in rats. Five groups of adult male Wistar rats (n = 12 per group) were created as follows: sham operation (Sham), global cerebral ischemia/reperfusion (GCI/R), HSA treatment (GCI/R + HSA), Dickkopf-1 (DDK1) treatment (GCI/R + DDK1), and DDK1 plus HSA treatment (GCI/R + DKK1 + HSA). The GCI/R injury model was created using the modified Pusinelli four-vessel occlusion method. After 24 h, rats were evaluated using neurological scoring, Nissl staining, and brain tissue water content. The mRNA expression of Wnt, GSK3β, and β-Catenin in the brain were detected by quantitative real time polymerase chain reaction. The protein expression of β-Catenin and GSK-3β were investigated by western blot and immunohistochemical analysis in the presence and absence of the Wnt/β-Catenin antagonist, DKK-1. Complex I activity and ROS content were also measured. After 24 h of reperfusion, the behavior score and brain tissue water content in the GCI/R + HSA group were lower than that in the GCI/R group. In addition, the degree of neuronal injury was significantly reduced in the GCI/R + HSA group (P < 0.05). The ROS content was significantly decreased and Complex I activity was markedly raised in the GCI/R + HSA group compared to the GCI/R group (P < 0.05). Further, GSK-3β expression in the GCI/R + HSA group was lower than that in the GCI/R group, while the Wnt and β-catenin expression were increased. These effects were reversed by DKK1. Taken together, we showed that HSA attenuates GCI/R-induced brain damage and may be neuroprotective via regulation of the Wnt/β-catenin/ROS signaling pathway.
Collapse
Affiliation(s)
- Yuedong Tang
- Department of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai, China; Medical Center of Chemical Injury, Emergency and Critical Care, Jinshan Hospital, Fudan University, Shanghai, China; Medical Research Centre for Chemical Injury, Emergency and Critical Care, Fudan University, Shanghai, China
| | - Jie Shen
- Department of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai, China; Medical Center of Chemical Injury, Emergency and Critical Care, Jinshan Hospital, Fudan University, Shanghai, China; Medical Research Centre for Chemical Injury, Emergency and Critical Care, Fudan University, Shanghai, China.
| | - Feng Zhang
- Department of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai, China; Medical Center of Chemical Injury, Emergency and Critical Care, Jinshan Hospital, Fudan University, Shanghai, China; Medical Research Centre for Chemical Injury, Emergency and Critical Care, Fudan University, Shanghai, China
| | - Fei-Yu Yang
- Department of Emergency and Critical Care Medicine, Jinshan Hospital, Fudan University, Shanghai, China; Medical Center of Chemical Injury, Emergency and Critical Care, Jinshan Hospital, Fudan University, Shanghai, China; Medical Research Centre for Chemical Injury, Emergency and Critical Care, Fudan University, Shanghai, China
| | - Ming Liu
- Department of Respiration, Shanghai Punan Hospital, Shanghai, China
| |
Collapse
|
162
|
Autophagy as a Homeostatic Mechanism in Response to Stress Conditions in the Central Nervous System. Mol Neurobiol 2019; 56:6594-6608. [DOI: 10.1007/s12035-019-1546-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 03/12/2019] [Indexed: 12/11/2022]
|
163
|
Wei J, Wu X, Luo P, Yue K, Yu Y, Pu J, Zhang L, Dai S, Han D, Fei Z. Homer1a Attenuates Endoplasmic Reticulum Stress-Induced Mitochondrial Stress After Ischemic Reperfusion Injury by Inhibiting the PERK Pathway. Front Cell Neurosci 2019; 13:101. [PMID: 30930751 PMCID: PMC6428733 DOI: 10.3389/fncel.2019.00101] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 02/27/2019] [Indexed: 12/17/2022] Open
Abstract
Homer1a is the short form of a scaffold protein that plays a protective role in many forms of stress. However, the role of Homer1a in cerebral ischemia/reperfusion (I/R) injury and its potential mechanism is still unknown. In this study, we found that Homer1a was upregulated by oxygen and glucose deprivation (OGD) and that overexpression of Homer1a alleviated OGD-induced lactate dehydrogenase (LDH) release and cell death in cultured cortical neurons. After OGD treatment, the overexpression of Homer1a preserved mitochondrial function, as evidenced by less cytochrome c release, less reactive oxygen species (ROS) production, less ATP and mitochondrial membrane potential (MMP) loss, less caspase-9 activation, and inhibition of endoplasmic reticulum (ER) stress confirmed by the decreased expression of phosphate-PKR-like ER Kinase (p-PERK)/PERK and phosphate- inositol-requiring enzyme 1 (p-IRE1)/IRE1 and immunofluorescence (IF) staining. In addition, mitochondrial protection of Homer1a was blocked by the ER stress activator Tunicamycin (TM) with a re-escalated ROS level, increasing ATP and MMP loss. Furthermore, Homer1a overexpression-induced mitochondrial stress attenuation was significantly reversed by activating the PERK pathway with TM and p-IRE1 inhibitor 3,5-dibromosalicylaldehyde (DBSA), as evidenced by increased cytochrome c release, increased ATP loss and a higher ROS level. However, activating the IRE1 pathway with TM and p-PERK inhibitor GSK2656157 showed little change in cytochrome c release and exhibited a moderate upgrade of ATP loss and ROS production in neurons. In summary, these findings demonstrated that Homer1a protects against OGD-induced injury by preserving mitochondrial function through inhibiting the PERK pathway. Our finding may reveal a promising target of protecting neurons from cerebral I/R injury.
Collapse
Affiliation(s)
- Jialiang Wei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Health Services, Fourth Military Medical University, Xi'an, China
| | - Xiuquan Wu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Kangyi Yue
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yang Yu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jingnan Pu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lei Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shuhui Dai
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Donghui Han
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
164
|
Epicatechin Gallate Protects HBMVECs from Ischemia/Reperfusion Injury through Ameliorating Apoptosis and Autophagy and Promoting Neovascularization. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7824684. [PMID: 30962864 PMCID: PMC6431361 DOI: 10.1155/2019/7824684] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 11/03/2018] [Accepted: 11/26/2018] [Indexed: 12/16/2022]
Abstract
Green tea is one of the most beverages with antioxidants and nutrients. As one of the major components of green tea, (-)-epicatechin gallate (ECG) was evaluated for its antioxidative properties in the present study. Cell proliferation assay, tube formation, cell migration, apoptosis, and autophagy were performed in human brain microvascular endothelial cells (HBMVECs) after oxygen-glucose deprivation/reoxygenation (OGD/R) to investigate potential anti-ischemia/reperfusion injury properties of ECG in vitro. Markers of oxidative stress as ROS, LDH, MDA, and SOD were further assayed in our study. Data indicated that ECG could affect neovascularization and promote cell proliferation, tube formation, and cell migration while inhibiting apoptosis and autophagy through affecting VEGF, Bcl-2, BAX, LC3B, caspase 3, mTOR, and Beclin-1 expression. All the data suggested that ECG may be protective for the brain against ischemia/reperfusion injury by promoting neovascularization, alleviating apoptosis and autophagy, and promoting cell proliferation in HBMVECs of OGD/R.
Collapse
|
165
|
Zhang Y, Wang Y, Xu J, Tian F, Hu S, Chen Y, Fu Z. Melatonin attenuates myocardial ischemia-reperfusion injury via improving mitochondrial fusion/mitophagy and activating the AMPK-OPA1 signaling pathways. J Pineal Res 2019; 66:e12542. [PMID: 30516280 DOI: 10.1111/jpi.12542] [Citation(s) in RCA: 261] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 11/24/2018] [Accepted: 11/29/2018] [Indexed: 12/14/2022]
Abstract
Optic atrophy 1 (OPA1)-related mitochondrial fusion and mitophagy are vital to sustain mitochondrial homeostasis under stress conditions. However, no study has confirmed whether OPA1-related mitochondrial fusion/mitophagy is activated by melatonin and, consequently, attenuates cardiomyocyte death and mitochondrial stress in the setting of cardiac ischemia-reperfusion (I/R) injury. Our results indicated that OPA1, mitochondrial fusion, and mitophagy were significantly repressed by I/R injury, accompanied by infarction area expansion, heart dysfunction, myocardial inflammation, and cardiomyocyte oxidative stress. However, melatonin treatment maintained myocardial function and cardiomyocyte viability, and these effects were highly dependent on OPA1-related mitochondrial fusion/mitophagy. At the molecular level, OPA1-related mitochondrial fusion/mitophagy, which was normalized by melatonin, substantially rectified the excessive mitochondrial fission, promoted mitochondria energy metabolism, sustained mitochondrial function, and blocked cardiomyocyte caspase-9-involved mitochondrial apoptosis. However, genetic approaches with a cardiac-specific knockout of OPA1 abolished the beneficial effects of melatonin on cardiomyocyte survival and mitochondrial homeostasis in vivo and in vitro. Furthermore, we demonstrated that melatonin affected OPA1 stabilization via the AMPK signaling pathway and that blockade of AMPK repressed OPA1 expression and compromised the cardioprotective action of melatonin. Overall, our results confirm that OPA1-related mitochondrial fusion/mitophagy is actually modulated by melatonin in the setting of cardiac I/R injury. Moreover, manipulation of the AMPK-OPA1-mitochondrial fusion/mitophagy axis via melatonin may be a novel therapeutic approach to reduce cardiac I/R injury.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Yue Wang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Junnan Xu
- Beijing Key Laboratory of Immunology Regulatory and Organ Transplantation, Organ Transplant Institute of People's Liberation Army, The 309th Hospital of People's Liberation Army, Beijing, China
| | - Feng Tian
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Shunying Hu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Zhenhong Fu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
166
|
Cao J, Wei R, Yao S. Matrine has pro-apoptotic effects on liver cancer by triggering mitochondrial fission and activating Mst1-JNK signalling pathways. J Physiol Sci 2019; 69:185-198. [PMID: 30155612 PMCID: PMC10717886 DOI: 10.1007/s12576-018-0634-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 08/16/2018] [Indexed: 02/08/2023]
Abstract
Mitochondrial homeostasis is closely associated with liver cancer progression via multiple mechanisms and is also a potential tumour-suppressive target in clinical practice. However, the role of mitochondrial fission in liver cancer cell viability has not been adequately investigated. Matrine, a type of alkaloid isolated from Sophoraflavescens, has been widely used to treat various types of cancer. However, the molecular effect of matrine on mitochondrial homeostasis is unclear. Therefore, the aim of the current study was to determine the role of mitochondrial fission in cell apoptosis, viability, migration and proliferation of HepG2 cells in vitro. The effect of matrine on mitochondrial fission and its mechanism were also explored. The results of our study showed that HepG2 cells treated with matrine had reduced viability, an increased apoptotic rate, a blunted migratory response, and impaired proliferation capacity. At the molecular level, matrine treatment activated mitochondrial fission, which promoted mitochondrial dysfunction, caused cellular oxidative stress, disrupted cellular energy metabolism and initiated cell apoptotic pathways. However, blockade of mitochondrial fission abolished the deleterious effects of matrine on HepG2 cells. Further, we demonstrated that the Mst1-JNK signalling axis was required for matrine-modulated mitochondrial fission. Matrine-mediated mitochondrial dysfunction was reversed by inhibiting Mst1-JNK pathways. Together, our results demonstrated that mitochondrial fission could be a potential upstream tumour-suppressive signal for liver cancer by modifying mitochondrial function and cell death. By contrast, matrine exerted an anticancer function in liver cancer by activating mitochondrial fission mediated by Mst1-JNK pathways.
Collapse
Affiliation(s)
- Jian Cao
- School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Runjie Wei
- Peking University China-Japan Friendship School of Clinical Medicine, No. 2 Yinghua East Road, Chaoyang District, Beijing, 100029, China
| | - Shukun Yao
- School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100191, China.
- Department of Gastroenterology, China-Japan Friendship Hospital, No. 2 Yinghua East Road, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
167
|
Ouyang H, Zhou E, Wang H. Mst1-Hippo pathway triggers breast cancer apoptosis via inducing mitochondrial fragmentation in a manner dependent on JNK-Drp1 axis. Onco Targets Ther 2019; 12:1147-1159. [PMID: 30809096 PMCID: PMC6376886 DOI: 10.2147/ott.s193787] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Mst1-Hippo pathway and mitochondrial fragmentation participate in the progression of several types of cancers. However, their roles in breast cancer requires investigation. The aim of our study is to determine whether Mst1 overexpression regulates the viability of breast cancer cells via modulating mitochondrial fragmentation. MATERIALS AND METHODS TUNEL staining, MTT assay and Western blotting were used to detect cancer cell death. Adenovirus-loaded Mst1 was transfected into cells to overexpress Mst1. Mitochondrial fragmentation was observed via immunofluorescence staining and Western blotting. Pathway blocker was used to detect whether Mst1 modulated cell death and mitochondrial fragmentation via JNK signaling pathway. RESULTS Our data showed that Mst1 overexpression promoted breast cancer cell death in a manner dependent on mitochondrial apoptosis. Mitochondrial oxidative stress, energy metabolism disorder, mitochondrial cyt-c liberation and mitochondrial apoptosis activation were observed after Mst1 overexpression. Furthermore, we demonstrated that Mst1 overexpression activated mitochondrial stress via triggering Drp1-related mitochondrial fragmentation, and that inhibition of Drp1-related mitochondrial fragmentation abrogated the proapoptotic effect of Mst1 overexpression on breast cancer cells. To this end, we found that Mst1 modulated Drp1 expression via the JNK signaling pathway, and that blockade of the JNK pathway attenuated mitochondrial stress and repressed apoptosis in Mst1-overexpressed cells. CONCLUSION Altogether, our results identified a tumor suppressive role for Mst1 overexpression in breast cancer via activation of the JNK-Drp1 axis and subsequent initiation of fatal mitochondrial fragmentation. Given these findings, strategies to enhance Mst1 activity and elevate the JNK-Drp1-mitochondrial fragmentation cascade have clinical benefits for patients with breast cancer.
Collapse
Affiliation(s)
- Hui Ouyang
- Department of Breast and Thyroid Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China,
| | - Enxiang Zhou
- Department of Breast and Thyroid Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China,
| | - Huan Wang
- Department of Breast and Thyroid Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China,
| |
Collapse
|
168
|
Li H, Zhang Y, Liu S, Li F, Wang B, Wang J, Cao L, Xia T, Yao Q, Chen H, Zhang Y, Zhu X, Li Y, Li G, Wang J, Li X, Ni S. Melatonin Enhances Proliferation and Modulates Differentiation of Neural Stem Cells Via Autophagy in Hyperglycemia. Stem Cells 2019; 37:504-515. [PMID: 30644149 DOI: 10.1002/stem.2968] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 11/07/2018] [Accepted: 12/11/2018] [Indexed: 12/19/2022]
Abstract
Dysfunction of neural stem cells (NSCs) has been linked to fetal neuropathy, one of the most devastating complications of gestational diabetes. Several studies have demonstrated that melatonin (Mel) exerted neuroprotective actions in various stresses. However, the role of autophagy and the involvement of Mel in NSCs in hyperglycemia (HG) have not yet been fully established. Here, we found that HG increased autophagy and autophagic flux of NSCs as evidenced by increasing LC3B II/I ratio, Beclin-1 expression, and autophagosomes. Moreover, Mel enhanced NSCs proliferation and self-renewal in HG with decreasing autophagy and activated mTOR signaling. Consistently, inhibition of autophagy by 3-Methyladenine (3-Ma) could assist Mel effects above, and induction of autophagy by Rapamycin (Rapa) could diminish Mel effects. Remarkably, HG induced premature differentiation of NSCs into neurons (Map2 positive cells) and astrocytes (GFAP positive cells). Furthermore, Mel diminished HG-induced premature differentiation and assisted NSCs in HG differentiation as that in normal condition. Coincidentally, inhibiting of NSCs autophagy by 3-Ma assisted Mel to modulate differentiation. However, increasing NSCs autophagy by Rapa disturbed the Mel effects and retarded NSCs differentiation. These findings suggested that Mel supplementation could contribute to mimicking normal NSCs proliferation and differentiation in fetal central nervous system by inhibiting autophagy in the context of gestational diabetes. Stem Cells 2019;37:504-515.
Collapse
Affiliation(s)
- Haoyuan Li
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Yanmin Zhang
- Brain Science Research Institute, Shandong University, Jinan, People's Republic of China.,Key Laboratory of the Ministry of Education for Experimental Teratology, Department of Histology and Embryology, Shandong University School of Basic Medical Sciences, Jinan, People's Republic of China
| | - Shangming Liu
- Key Laboratory of the Ministry of Education for Experimental Teratology, Department of Histology and Embryology, Shandong University School of Basic Medical Sciences, Jinan, People's Republic of China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, the State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Fengpeng Li
- Department of Neurosurgery, Yinan County People's Hospital, Linyi, People's Republic of China
| | - Benlin Wang
- Department of Neurosurgery, PLA No. 970 Hospital, Yantai, Shandong, People's Republic of China
| | - Jianjie Wang
- Shandong University School of Medicine, Jinan, People's Republic of China
| | - Lanfang Cao
- Department of Infection Management, The Second People's Hospital of Yunnan Province, Kunming, People's Republic of China
| | - Tongliang Xia
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Qingyu Yao
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, People's Republic of China
| | - Haijun Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Yulin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Xiaodong Zhu
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, People's Republic of China
| | - Yang Li
- Shandong University School of Medicine, Jinan, People's Republic of China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China.,Brain Science Research Institute, Shandong University, Jinan, People's Republic of China
| | - Jian Wang
- Brain Science Research Institute, Shandong University, Jinan, People's Republic of China.,KG Jebsen Brain Tumor Research Center, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China.,Brain Science Research Institute, Shandong University, Jinan, People's Republic of China
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China.,Brain Science Research Institute, Shandong University, Jinan, People's Republic of China
| |
Collapse
|
169
|
Boga JA, Caballero B, Potes Y, Perez-Martinez Z, Reiter RJ, Vega-Naredo I, Coto-Montes A. Therapeutic potential of melatonin related to its role as an autophagy regulator: A review. J Pineal Res 2019; 66:e12534. [PMID: 30329173 DOI: 10.1111/jpi.12534] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/01/2018] [Accepted: 10/08/2018] [Indexed: 12/16/2022]
Abstract
There are several pathologies, syndromes, and physiological processes in which autophagy is involved. This process of self-digestion that cells trigger as a survival mechanism is complex and tightly regulated, according to the homeostatic conditions of the organ. However, in all cases, its relationship with oxidative stress alterations is evident, following a pathway that suggests endoplasmic reticulum stress and/or mitochondrial changes. There is accumulating evidence of the beneficial role that melatonin has in the regulation and restoration of damaged autophagic processes. In this review, we focus on major physiological changes such as aging and essential pathologies including cancer, neurodegenerative diseases, viral infections and obesity, and document the essential role of melatonin in the regulation of autophagy in each of these different situations.
Collapse
Affiliation(s)
- Jose A Boga
- Service of Microbiology, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Beatriz Caballero
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Department of Morphology and Cell Biology, University of Oviedo, Oviedo, Spain
| | - Yaiza Potes
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Department of Morphology and Cell Biology, University of Oviedo, Oviedo, Spain
| | - Zulema Perez-Martinez
- Service of Microbiology, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health Science Center, San Antonio, Texas
| | - Ignacio Vega-Naredo
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Department of Morphology and Cell Biology, University of Oviedo, Oviedo, Spain
| | - Ana Coto-Montes
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Department of Morphology and Cell Biology, University of Oviedo, Oviedo, Spain
| |
Collapse
|
170
|
He M, Xiang Z, Xu L, Duan Y, Li F, Chen J. Lipopolysaccharide induces human olfactory ensheathing glial apoptosis by promoting mitochondrial dysfunction and activating the JNK-Bnip3-Bax pathway. Cell Stress Chaperones 2019; 24:91-104. [PMID: 30374881 PMCID: PMC6363633 DOI: 10.1007/s12192-018-0945-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 10/02/2018] [Accepted: 10/04/2018] [Indexed: 02/06/2023] Open
Abstract
Olfactory ensheathing glia (OEG) play an important role in regulating the regeneration of an injured nervous system. However, chronic inflammation damage reduces the viability of OEG via poorly understood mechanisms. We aimed to investigate the pathological responses of OEG in response to LPS-mediated inflammation stress in vitro. The results indicated that lipopolysaccharide (LPS) treatment significantly reduced the viability of OEG in a dose-dependent fashion. Mechanistically, LPS stimuli induced mitochondrial oxidative damage, mitochondrial fragmentation, mitochondrial metabolism disruption, and mitochondrial apoptosis activation. Furthermore, we verified that LPS modulated mitochondrial apoptosis by promoting Bax upregulation, and this process was regulated by the JNK-Bnip3 pathway. Inhibition of the JNK-Bnip3 pathway prevented LPS-mediated Bax activation, thus attenuating OEG apoptosis. Altogether, our data illustrated that LPS-mediated inflammation injury evoked mitochondrial abnormalities in OEG damage via the JNK-Bnip3-Bax pathway. This finding provides a potential target to protect OEG against chronic inflammation stress.
Collapse
Affiliation(s)
- Maowei He
- Bengbu Medical College, Affiliated Fuzhou General Hospital of Nanjing Military Area Command of Chinese PLA, Fuzhou, 350025, China
| | - Zimin Xiang
- Department of Orthopedics, Fuzhou General Hospital of Nanjing Military Area Command of Chinese PLA, Fuzhou, 350025, China
| | - Libin Xu
- Department of Orthopedics, Fuzhou General Hospital of Nanjing Military Area Command of Chinese PLA, Fuzhou, 350025, China
| | - Yanting Duan
- Bengbu Medical College, Affiliated Fuzhou General Hospital of Nanjing Military Area Command of Chinese PLA, Fuzhou, 350025, China
| | - Fangqin Li
- Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Jianmei Chen
- Department of Orthopedics, Fuzhou General Hospital of Nanjing Military Area Command of Chinese PLA, Fuzhou, 350025, China.
| |
Collapse
|
171
|
Luo C, Yang Q, Liu Y, Zhou S, Jiang J, Reiter RJ, Bhattacharya P, Cui Y, Yang H, Ma H, Yao J, Lawler SE, Zhang X, Fu J, Rozental R, Aly H, Johnson MD, Chiocca EA, Wang X. The multiple protective roles and molecular mechanisms of melatonin and its precursor N-acetylserotonin in targeting brain injury and liver damage and in maintaining bone health. Free Radic Biol Med 2019; 130:215-233. [PMID: 30315933 DOI: 10.1016/j.freeradbiomed.2018.10.402] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/01/2018] [Accepted: 10/02/2018] [Indexed: 12/20/2022]
Abstract
Melatonin is a neurohormone associated with sleep and wakefulness and is mainly produced by the pineal gland. Numerous physiological functions of melatonin have been demonstrated including anti-inflammation, suppressing neoplastic growth, circadian and endocrine rhythm regulation, and its potent antioxidant activity as well as its role in regeneration of various tissues including the nervous system, liver, bone, kidney, bladder, skin, and muscle, among others. In this review, we summarize the recent advances related to the multiple protective roles of melatonin receptor agonists, melatonin and N-acetylserotonin (NAS), in brain injury, liver damage, and bone health. Brain injury, including traumatic brain injury, ischemic stroke, intracerebral hemorrhage, subarachnoid hemorrhage, and newborn perinatal hypoxia-ischemia encephalopathy, is a major cause of mortality and disability. Liver disease causes serious public health problems and various factors including alcohol, chemical pollutants, and drugs induce hepatic damage. Osteoporosis is the most common bone disease in humans. Due in part to an aging population, both the cost of care of fracture patients and the annual fracture rate have increased steadily. Despite the discrepancy in the pathophysiological processes of these disorders, time frames and severity, they may share several common molecular mechanisms. Oxidative stress is considered to be a critical factor in these pathogeneses. We update the current state of knowledge related to the molecular processes, mainly including anti-oxidative stress, anti-apoptosis, autophagy dysfunction, and anti-inflammation as well as other properties of melatonin and NAS. Particularly, the abilities of melatonin and NAS to directly scavenge oxygen-centered radicals and toxic reactive oxygen species, and indirectly act through antioxidant enzymes are disscussed. In this review, we summarize the similarities and differences in the protection provided by melatonin and/or NAS in brain, liver and bone damage. We analyze the involvement of melatonin receptor 1A (MT1), melatonin receptor 1B (MT2), and melatonin receptor 1C (MT3) in the protection of melatonin and/or NAS. Additionally, we evaluate their potential clinical applications. The multiple mechanisms of action and multiple organ-targeted properties of melatonin and NAS may contribute to development of promising therapies for clinical trials.
Collapse
Affiliation(s)
- Chengliang Luo
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Qiang Yang
- Hubei Provincial Key Lab for Quality and Safety of Traditional Chinese Medicine Health Food, Jing Brand Research Institute, Daye, Hubei, China
| | - Yuancai Liu
- Hubei Provincial Key Lab for Quality and Safety of Traditional Chinese Medicine Health Food, Jing Brand Research Institute, Daye, Hubei, China
| | - Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jiying Jiang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University Texas Health Science Center, San Antonio, TX, USA
| | - Pallab Bhattacharya
- National Institute of Pharmaceutical Education and Research, Ahmedabad, India
| | - Yongchun Cui
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hongwei Yang
- Department of Neurosurgery, University of Massachusetts Medical School, Worcester, MA, USA
| | - He Ma
- Third Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Jiemin Yao
- Third Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Sean E Lawler
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xinmu Zhang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jianfang Fu
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Renato Rozental
- Lab Neuroproteção & Estratégias Regenerativas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Hany Aly
- Department of Neonatology, Cleveland Clinic Children's Hospital, Cleveland, OH, USA
| | - Mark D Johnson
- Department of Neurosurgery, University of Massachusetts Medical School, Worcester, MA, USA
| | - E Antonio Chiocca
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
172
|
Xu N, Lu Y, Hou J, Liu C, Sun Y. A Polysaccharide Purified from Morchella conica Pers. Prevents Oxidative Stress Induced by H₂O₂ in Human Embryonic Kidney (HEK) 293T Cells. Int J Mol Sci 2018; 19:ijms19124027. [PMID: 30551572 PMCID: PMC6320779 DOI: 10.3390/ijms19124027] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 02/07/2023] Open
Abstract
Morchella conica Pers. (M. conica) has been used both as a medical and edible mushroom and possesses antimicrobial properties and antioxidant activities. However, the antioxidant properties of polysaccharides purified from M. conica have not been studied. The aim of this study was to investigate the in vitro antioxidant properties of a polysaccharide NMCP-2 (neutral M. conica polysaccharides-2) purified from M. conica, as determined by radical scavenging assay and H₂O₂-induced oxidative stress in HEK 293T cells. Results showed that NMCP-2 with an average molecular weight of 48.3 kDa possessed a much stronger chelating ability on ferrous ions and a higher ability to scavenge radical scavenging 2,2-diphenyl-1-picrylhydrazyl (DPPH) than the other purified fraction of NMCP-1 from M. conica. Moreover, 3-(4, 5-Dimethylthiazol-2-yl)-2, 5-diphenyltetra-zolium bromide (MTT) assay showed that NMCP-2 dose-dependently preserved cell viability of H₂O₂-induced cells. The NMCP-2 pretreated group reduced the generation of reactive oxygen species (ROS) content and increased the mitochondria membrane potential (MMP) levels. In addition, Hoechst 33342 staining revealed cells treated with NMCP-2 declined nuclear condensation. Ultrastructural observation revealed that NMCP-2 pretreatment alleviated the ruptured mitochondria when exposed to H₂O₂. Furthermore, western blot analysis showed that NMCP-2 prevented significant downregulation of the protein expression of Bax, cleaved caspases 3, and upregulated Bcl-2 levels. These results suggest the protective effects of NMCP-2 against H₂O₂-induced injury in HEK 293T cells. NMCP-2 could be used as a natural antioxidant of functional foods and natural drugs.
Collapse
Affiliation(s)
- Na Xu
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Yi Lu
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, Jilin University, Changchun 130062, China.
| | - Jumin Hou
- College of Food Science and Engineering, Changchun University, Changchun 130028, China.
| | - Chao Liu
- School of Food Engineering, Jilin Agriculture Science and Technology College, Jilin 132101, China.
| | - Yonghai Sun
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| |
Collapse
|
173
|
Yao S, Yan W. Overexpression of Mst1 reduces gastric cancer cell viability by repressing the AMPK-Sirt3 pathway and activating mitochondrial fission. Onco Targets Ther 2018; 11:8465-8479. [PMID: 30555239 PMCID: PMC6278716 DOI: 10.2147/ott.s180851] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Mammalian sterile 20-like kinase 1 (Mst1) plays a critical role in regulating cell survival and apoptosis. However, its influence on gastric cancer cell viability is not understood. Our study aims to explore the specific role of Mst1 in gastric cancer. MATERIALS AND METHODS Cellular viability was measured via TUNEL staining, MTT assays, and Western blotting. Immunofluorescence was performed to observe mitochondrial fission. Mst1 overexpression assays were conducted to observe the regulatory mechanisms of Mst1 in mitochondrial fission and cell apoptosis. RESULTS The results demonstrated that Mst1 was downregulated in AGS cells when compared with GES-1 cells. However, overexpression of Mst1 reduced cell viability and increased apoptosis in AGS cells. Molecular experiments showed that Mst1 overexpression mediated mitochondrial damage, as evidenced by decreased ATP production, increased ROS generation, more cyt-c translocation from the mitochondria into the cytoplasm and nucleus, and activated the caspase-9-related apoptotic pathway. Furthermore, we found that mitochondrial fission was required for Mst1-induced mitochondrial dysfunction; inhibition of mitochondrial fission sustained mitochondrial homeostasis in response to Mst1 overexpression. In addition, our data revealed that Mst1 controlled mitochondrial fission via repressing the AMPK-Sirt3 pathway. Activation of the AMPK-Sirt3 pathway negated the promoting effect of Mst1 overexpression on mitochondrial fission. CONCLUSION Altogether, our data identified Mst1 as a novel tumor-suppressive factor in promoting cell death in gastric cancer cells by triggering mitochondrial fission and blocking the AMPK-Sirt3 axis.
Collapse
Affiliation(s)
- Shiwei Yao
- Department of Gastroenterology, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, China,
| | - Wei Yan
- Department of Gastroenterology, The First Hospital of Tsinghua University, Beijing, China
| |
Collapse
|
174
|
Lee JH, Yoon YM, Han YS, Jung SK, Lee SH. Melatonin protects mesenchymal stem cells from autophagy-mediated death under ischaemic ER-stress conditions by increasing prion protein expression. Cell Prolif 2018; 52:e12545. [PMID: 30430685 PMCID: PMC6495509 DOI: 10.1111/cpr.12545] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/06/2018] [Accepted: 09/24/2018] [Indexed: 12/30/2022] Open
Abstract
Object The purpose of this study was to explore whether melatonin could protect mesenchymal stem cells (MSCs) against ischaemic injury, by inhibiting endoplasmic reticulum (ER) stress and autophagy both in vivo and in vitro. Materials and Methods To confirm the protective effect of melatonin against ER stress in MSCs, markers of cell viability, apoptosis and autophagy were analysed. To further investigate the regenerative effect of melatonin‐treated MSCs in ischaemic tissues, a murine hindlimb ischaemic model was established. Results Under oxidative stress conditions, treatment with melatonin suppressed the activation of ER stress–associated proteins and autophagy‐associated proteins acting through upregulation of cellular prion protein (PrPC) expression. Consequently, inhibition of apoptotic cell death occurred. Melatonin also promoted the activation of MnSOD and catalase activities in MSCs. In a murine hindlimb ischaemia model, melatonin‐treated MSCs also enhanced the functional limb recovery as well as neovascularization. These beneficial effects of melatonin were all blocked by knock‐down of PrPC expression. Conclusion Melatonin protects against ER stress/autophagy‐induced apoptotic cell death by augmenting PrPC expression. Thus, melatonin‐treated MSCs could be a potential cell‐based therapeutic agent for ER stress–induced ischaemic diseases, and melatonin‐induced PrPC might be a key molecule in ameliorating ER stress and autophagy.
Collapse
Affiliation(s)
- Jun Hee Lee
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Yeo Min Yoon
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Yong-Seok Han
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Seo Kyung Jung
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea.,Departments of Biochemistry, Soonchunhyang University College of Medicine, Cheonan, Korea
| |
Collapse
|
175
|
Wei R, Cao J, Yao S. Matrine promotes liver cancer cell apoptosis by inhibiting mitophagy and PINK1/Parkin pathways. Cell Stress Chaperones 2018; 23:1295-1309. [PMID: 30209783 PMCID: PMC6237690 DOI: 10.1007/s12192-018-0937-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/23/2018] [Accepted: 09/02/2018] [Indexed: 02/05/2023] Open
Abstract
Matrine is a natural alkaloid isolated from the root and stem of the legume plant Sophora. Its anti-proliferative and pro-apoptotic effects on several types of cancer have been well-documented. However, the role of matrine in regulating mitochondrial homeostasis, particularly mitophagy in liver cancer apoptosis, remains uncertain. The aim of our study was to explore whether matrine promotes liver cancer cell apoptosis by modifying mitophagy. HepG2 cells were used in the study and treated with different doses of matrine. Cell viability and apoptosis were determined by MTT assay, TUNEL staining, western blotting, and LDH release assay. Mitophagy was monitored by immunofluorescence assay and western blotting. Mitochondrial function was assessed by immunofluorescence assay, ELISA, and western blotting. The results of our study indicated that matrine treatment dose-dependently reduced cell viability and increased the apoptotic rate of HepG2 cells. Functional studies demonstrated that matrine treatment induced mitochondrial dysfunction and activated mitochondrial apoptosis by inhibiting protective mitophagy. Re-activation of mitophagy abolished the pro-apoptotic effects of matrine on HepG2 cells. Molecular investigations further confirmed that matrine regulated mitophagy via the PINK1/Parkin pathways. Matrine blocked the PINK1/Parkin pathways and repressed mitophagy, whereas activation of the PINK1/Parkin pathways increased mitophagy activity and promoted HepG2 cell survival in the presence of matrine. Together, our data indicated that matrine promoted HepG2 cell apoptosis through a novel mechanism that acted via inhibiting mitophagy and the PINK1/Parkin pathways. This finding provides new insight into the molecular mechanism of matrine for treating liver cancer and offers a potential target to repress liver cancer progression by modulating mitophagy and the PINK1/Parkin pathways.
Collapse
Affiliation(s)
- Runjie Wei
- Peking University China-Japan Friendship School of Clinical Medicine, No. 2 Yinghua East Road, Chaoyang District, 100029, Beijing, China
| | - Jian Cao
- School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, 100191, Beijing, China
| | - Shukun Yao
- Peking University China-Japan Friendship School of Clinical Medicine, No. 2 Yinghua East Road, Chaoyang District, 100029, Beijing, China.
- Department of Gastroenterology, China-Japan Friendship Hospital, No. 2 Yinghua East Road, Chaoyang District, 100029, Beijing, China.
| |
Collapse
|
176
|
Yang L, Guan G, Lei L, Lv Q, Liu S, Zhan X, Jiang Z, Gu X. Palmitic acid induces human osteoblast-like Saos-2 cell apoptosis via endoplasmic reticulum stress and autophagy. Cell Stress Chaperones 2018; 23:1283-1294. [PMID: 30194633 PMCID: PMC6237680 DOI: 10.1007/s12192-018-0936-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/27/2018] [Accepted: 09/01/2018] [Indexed: 12/30/2022] Open
Abstract
Palmitic acid (PA) is the most common saturated long-chain fatty acid in food that causes cell apoptosis. However, little is known about the molecular mechanisms of PA toxicity. In this study, we explore the effects of PA on proliferation and apoptosis in human osteoblast-like Saos-2 cells and uncover the signaling pathways involved in the process. Our study showed that endoplasmic reticulum (ER) stress and autophagy are involved in PA-induced Saos-2 cell apoptosis. We found that PA inhibited the viability of Saos-2 cells in a dose- and time-dependent manner. At the same time, PA induced the expression of ER stress marker genes (glucose-regulated protein 78 (GRP78) and CCAAT/enhancer binding protein homologous protein (CHOP)), altered autophagy-related gene expression (microtubule-associated protein 1 light chain 3 (LC3), ATG5, p62, and Beclin), promoted apoptosis-related gene expression (Caspase 3 and BAX), and affected autophagic flux. Inhibiting ER stress with 4-PBA diminished the PA-induced cell apoptosis, activated autophagy, and increased the expression of Caspase 3 and BAX. Inhibiting autophagy with 3-MA attenuated the PA and ER stress-induced cell apoptosis and the apoptosis-related gene expression (Caspase 3 and BAX), but seemed to have no obvious effects on ER stress, although the CHOP expression was downregulated. Taken together, our results suggest that PA-induced Saos-2 cell apoptosis is activated via ER stress and autophagy, and the activation of autophagy depends on the ER stress during this process.
Collapse
Affiliation(s)
- Lei Yang
- College of Basic Medical, Jiujiang University, Jiujiang, 332000, Jiangxi, China.
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, Jiangxi, China.
| | - Gaopeng Guan
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, Jiangxi, China
- Affiliated Hospital of Jiujiang University, Jiujiang University, Jiujiang, 332000, Jiangxi, China
- Medicine Graduate School, Nanchang University, Nanchang, 330006, China
| | - Lanjie Lei
- Affiliated Hospital of Jiujiang University, Jiujiang University, Jiujiang, 332000, Jiangxi, China
| | - Qizhuang Lv
- College of Biology & Pharmacy, Yulin Normal University, Yulin, 537000, Guangxi, China
| | - Shengyuan Liu
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, Jiangxi, China
- Affiliated Hospital of Jiujiang University, Jiujiang University, Jiujiang, 332000, Jiangxi, China
- Medicine Graduate School, Nanchang University, Nanchang, 330006, China
| | - Xiuwen Zhan
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, Jiangxi, China
- Affiliated Hospital of Jiujiang University, Jiujiang University, Jiujiang, 332000, Jiangxi, China
| | - Zhenzhen Jiang
- Medicine Graduate School, Nanchang University, Nanchang, 330006, China
| | - Xiang Gu
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, Jiangxi, China.
- Affiliated Hospital of Jiujiang University, Jiujiang University, Jiujiang, 332000, Jiangxi, China.
| |
Collapse
|
177
|
Wan J, Cui J, Wang L, Wu K, Hong X, Zou Y, Zhao S, Ke H. Excessive mitochondrial fragmentation triggered by erlotinib promotes pancreatic cancer PANC-1 cell apoptosis via activating the mROS-HtrA2/Omi pathways. Cancer Cell Int 2018; 18:165. [PMID: 30377412 PMCID: PMC6196464 DOI: 10.1186/s12935-018-0665-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/16/2018] [Indexed: 12/19/2022] Open
Abstract
Background Mitochondrial fragmentation drastically regulates the viability of pancreatic cancer through a poorly understood mechanism. The present study used erlotinib to activate mitochondrial fragmentation and then investigated the downstream events that occurred in response to mitochondrial fragmentation. Methods Cell viability and apoptosis were determined via MTT assay, TUNEL staining and ELISA. Mitochondrial fragmentation was measured via an immunofluorescence assay and qPCR. siRNA transfection and pathway blockers were used to perform the loss-of-function assays. Results The results of our study demonstrated that erlotinib treatment mediated cell apoptosis in the PANC-1 pancreatic cancer cell line via evoking mitochondrial fragmentation. Mechanistically, erlotinib application increased mitochondrial fission and reduced mitochondrial fusion, triggering mitochondrial fragmentation. Subsequently, mitochondrial fragmentation caused the overproduction of mitochondrial ROS (mROS). Interestingly, excessive mROS induced cardiolipin oxidation and mPTP opening, finally facilitating HtrA2/Omi liberation from the mitochondria into the cytoplasm, where HtrA2/Omi activated caspase-9-dependent cell apoptosis. Notably, neutralization of mROS or knockdown of HtrA2/Omi attenuated erlotinib-mediated mitochondrial fragmentation and favored cancer cell survival. Conclusions Together, our results identified the mROS-HtrA2/Omi axis as a novel signaling pathway that is activated by mitochondrial fragmentation and that promotes PANC-1 pancreatic cancer cell mitochondrial apoptosis in the presence of erlotinib. Electronic supplementary material The online version of this article (10.1186/s12935-018-0665-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jun Wan
- 1Department of Pharmacy, Third Clinical Medical College, Three Gorges University, Gezhouba Group Central Hospital, Yichang, 443002 Hubei China
| | - Jie Cui
- 1Department of Pharmacy, Third Clinical Medical College, Three Gorges University, Gezhouba Group Central Hospital, Yichang, 443002 Hubei China
| | - Lei Wang
- 2Department of Pathogenic Biology, School of Medicine, China Three Gorges University, Yichang, 443002 Hubei China
| | - Kunpeng Wu
- 1Department of Pharmacy, Third Clinical Medical College, Three Gorges University, Gezhouba Group Central Hospital, Yichang, 443002 Hubei China
| | - Xiaoping Hong
- 1Department of Pharmacy, Third Clinical Medical College, Three Gorges University, Gezhouba Group Central Hospital, Yichang, 443002 Hubei China
| | - Yulin Zou
- 1Department of Pharmacy, Third Clinical Medical College, Three Gorges University, Gezhouba Group Central Hospital, Yichang, 443002 Hubei China
| | - Shuang Zhao
- 1Department of Pharmacy, Third Clinical Medical College, Three Gorges University, Gezhouba Group Central Hospital, Yichang, 443002 Hubei China
| | - Hong Ke
- 3Department of Oncology, Third Clinical Medical College, Three Gorges University, Gezhouba Group Central Hospital, No. 60 Qiaohu Lake Road, Xiling District, Yichang, 443002 Hubei China
| |
Collapse
|
178
|
Li H, Feng J, Zhang Y, Feng J, Wang Q, Zhao S, Meng P, Li J. Mst1 deletion attenuates renal ischaemia-reperfusion injury: The role of microtubule cytoskeleton dynamics, mitochondrial fission and the GSK3β-p53 signalling pathway. Redox Biol 2018; 20:261-274. [PMID: 30384260 PMCID: PMC6205415 DOI: 10.1016/j.redox.2018.10.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 10/15/2018] [Indexed: 11/17/2022] Open
Abstract
Despite extensive research that has been carried out over the past three decades in the field of renal ischaemia-reperfusion (I/R) injury, the pathogenic role of mitochondrial fission in renal I/R injury is poorly understood. The aim of our study is to investigate the molecular mechanism by which mammalian STE20-like kinase 1 (Mst1) participates in renal I/R injury through modifying mitochondrial fission, microtubule cytoskeleton dynamics, and the GSK3β-p53 signalling pathway. Our data demonstrated that genetic ablation of Mst1 improved renal function, alleviated reperfusion-mediated tubular epithelial cell apoptosis, and attenuated the vulnerability of kidney to I/R injury. At the molecular level, Mst1 upregulation exacerbated mitochondrial damage, as evidenced by reduced mitochondrial potential, increased ROS generation, more cyt-c liberation from mitochondria into the cytoplasm, and an activated mitochondrial apoptotic pathway. Furthermore, we demonstrated that I/R-mediated mitochondrial damage resulted from mitochondrial fission, and the blockade of mitochondrial fission preserved mitochondrial homeostasis in the I/R setting. Functional studies have discovered that Mst1 regulated mitochondrial fission through two mechanisms: induction of Drp1 phosphorylation and enhancement of F-actin assembly. Activated Mst1 promoted Drp1 phosphorylation at Ser616, contributing to Drp1 translocation from the cytoplasm to the surface of the mitochondria. Additionally, Mst1 facilitated F-actin polymerization, contributing to mitochondrial contraction. Finally, we confirmed that Mst1 regulated Drp1 post-transcriptional modification and F-actin stabilization via the GSK3β-p53 signalling pathway. Inhibition of GSK3β-p53 signalling provided a survival advantage for the tubular epithelial cell in the context of renal I/R injury by repressing mitochondrial fission. Collectively, our study identified Mst1 as the primary pathogenesis for the development and progression of renal I/R injury via activation of fatal mitochondrial fission by modulating Drp1 phosphorylation, microtubule cytoskeleton dynamics, and the GSK3β-p53 signalling pathway. Mst1 deletion sustains renal function after I/R injury. Excessive mitochondrial fission accounts for Mst1-mediated mitochondrial apoptosis. Mst1 enhances reperfusion-mediated mitochondrial fission via Drp1 phosphorylation and F-actin polymerization. Mst1 regulates Drp1 phosphorylation and F-actin polymerization by activating the GSK3β-p53 axis.
Collapse
Affiliation(s)
- Hongyan Li
- Department of Nephrology, Huadu District People's Hospital of Guangzhou, Southern Medical University, Guangzhou 510800, China.
| | - Jianxun Feng
- Department of Nephorology, Xuhui DIstrict Centeral Hospital of Shanghai, Shanghai 20031, China
| | - Yunfang Zhang
- Department of Nephrology, Huadu District People's Hospital of Guangzhou, Southern Medical University, Guangzhou 510800, China
| | - Junxia Feng
- Department of Nephrology, Huadu District People's Hospital of Guangzhou, Southern Medical University, Guangzhou 510800, China
| | - Qi Wang
- Department of Nephrology, Huadu District People's Hospital of Guangzhou, Southern Medical University, Guangzhou 510800, China
| | - Shili Zhao
- Department of Nephrology, Huadu District People's Hospital of Guangzhou, Southern Medical University, Guangzhou 510800, China
| | - Ping Meng
- Department of Nephrology, Huadu District People's Hospital of Guangzhou, Southern Medical University, Guangzhou 510800, China
| | - Jingchun Li
- Department of Nephrology, Huadu District People's Hospital of Guangzhou, Southern Medical University, Guangzhou 510800, China
| |
Collapse
|
179
|
Tanshinone IIA promotes IL2-mediated SW480 colorectal cancer cell apoptosis by triggering INF2-related mitochondrial fission and activating the Mst1-Hippo pathway. Biomed Pharmacother 2018; 108:1658-1669. [PMID: 30372868 DOI: 10.1016/j.biopha.2018.09.170] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 09/27/2018] [Accepted: 09/28/2018] [Indexed: 01/02/2023] Open
Abstract
IL-2-based therapy is a promising tool to treat colorectal cancer, but drug resistance always occurs in clinical practice. Mitochondrial fission is a novel target to modulate cancer development and progression. The aim of our study is to explore the effect of IL-2 combined with Tan IIA on SW480 colorectal cancer cell apoptosis in vitro and to determine whether IL-2/Tan IIA cotreatment could reduce SW480 cell viability via activating mitochondrial fission. The results indicated that Tan IIA increased IL-2-mediated cell death in SW480 colorectal cancer cells, and this effect was also accompanied with a reduction in cell proliferation. Functional investigations demonstrated that Tan IIA/IL-2 cotreatment enhanced INF2-related mitochondrial fission. Excessive mitochondrial division induced mitochondrial oxidative stress, mitochondrial energy metabolism disorder and mitochondrial apoptosis in SW480 cells. Inhibition of mitochondrial fission attenuated the antitumor effect of Tan IIA/IL-2 cotreatment on SW480 cell apoptosis. Further, we demonstrated that Tan IIA/IL-2 combination therapy controlled INF2-related mitochondrial fission via the Mst1-Hippo pathway. Moreover, Mst1 knockdown abrogated Tan IIA/IL-2-activated mitochondrial fission. Altogether, our results demonstrated that Tan IIA enhances the therapeutic efficiency of IL-2-mediated SW480 colorectal cancer cell apoptosis via promoting INF2-related mitochondrial fission and activating the Mst1-Hippo pathway.
Collapse
|
180
|
Liu J, Xu Y, Wu Q, Ding Q, Fan W. Sirtuin‑1 protects hair follicle stem cells from TNFα-mediated inflammatory stress via activating the MAPK-ERK-Mfn2 pathway. Life Sci 2018; 212:213-224. [PMID: 30292830 DOI: 10.1016/j.lfs.2018.10.003] [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: 08/26/2018] [Revised: 10/01/2018] [Accepted: 10/03/2018] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Stem cell transplantation is a promising tool to treat burn injuries. However, the inflammatory microenvironment in damaged skin limits the efficiency of stem cell-based therapy via poorly understood mechanisms. The aim of our study is to explore the contribution and mechanism of Sirtuin-1 (Sirt1) in TNFα-mediated inflammatory stress in hair follicle stem cells (HFSCs). METHODS Cellular viability was determined using the MTT assay, TUNEL staining, western blot analysis and LDH release assay. Adenovirus-loaded Sirt1 was transduced into HFSCs to overexpress Sirt1 in the presence of TNFα. Mitochondrial function was determined using JC-1 staining, mitochondrial ROS staining, immunofluorescence staining and western blotting. RESULTS Sirt1 was downregulated in response to the TNFα treatment. Additionally, TNFα stress reduced the viability, mobility and proliferation of HFSCs, and these effects were reversed by the overexpression of Sirt1. At the molecular level, Sirt1 overexpression attenuated TNFα-mediated mitochondrial damage, as evidenced by increased mitochondrial energy metabolism, decreased mitochondrial ROS generation, stabilized mitochondrial potential and blockage of the mitochondrial apoptotic pathway. Furthermore, Sirt1 modulated mitochondrial homeostasis by activating the MAPK-ERK-Mfn2 axis; inhibition of this pathway abrogated the protective effects of Sirt1 on HFSC survival, migration and proliferation. SIGNIFICANCE Based on our results, the inflammatory stress-mediated HFSC injury may be associated with a decrease in Sirt1 expression and subsequent mitochondrial dysfunction. Accordingly, strategies designed to enhance Sirt1 expression would be an effective approach to enhance the survival of HFSCs in the inflammatory microenvironment.
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Dermatology and Venereology, Jiangsu Provincial People's Hospital, First Affiliated Hospital of Nanjing Medical University, 210029, China
| | - Yuxuan Xu
- Department of Dermatology and Venereology, Jiangsu Provincial People's Hospital, First Affiliated Hospital of Nanjing Medical University, 210029, China
| | - Qiaofang Wu
- Department of Dermatology and Venereology, Jiangsu Provincial People's Hospital, First Affiliated Hospital of Nanjing Medical University, 210029, China
| | - Qi Ding
- Department of Dermatology and Venereology, Jiangsu Provincial People's Hospital, First Affiliated Hospital of Nanjing Medical University, 210029, China
| | - Weixin Fan
- Department of Dermatology and Venereology, Jiangsu Provincial People's Hospital, First Affiliated Hospital of Nanjing Medical University, 210029, China.
| |
Collapse
|
181
|
Ji MH, Xia DG, Zhu LY, Zhu X, Zhou XY, Xia JY, Yang JJ. Short- and Long-Term Protective Effects of Melatonin in a Mouse Model of Sepsis-Associated Encephalopathy. Inflammation 2018; 41:515-529. [PMID: 29198013 DOI: 10.1007/s10753-017-0708-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Brain dysfunction is a common complication after sepsis and is an independent risk factor for a poor prognosis, which is partly attributed to the dysregulated inflammatory response and oxidative damage. Melatonin regulates the sleep-wake cycle and also has potent anti-inflammatory and antioxidant properties, yet the protective effects of melatonin on sepsis-induced neurobehavioral dysfunction remain to be elucidated. In the present study, melatonin was administered intraperitoneally daily at a dose of 10 mg/kg for three consecutive days immediately (early treatment) or 7 days (delayed treatment) after sham operation or cecal ligation and puncture (CLP), followed by an additional treatment in drinking water until the end of behavioral tests. The concentrations of pro-inflammatory cytokines (tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), IL-6, IL-10), malondialdehyde (MDA), superoxide dismutase (SOD), reactive oxygen species (ROS), brain-derived neurotrophic factor (BDNF), and glial cell line-derived neurotrophic factor (GDNF) were determined at the indicated time points. Compared with the CLP + vehicle group, we found that early melatonin treatment resulted in increased survival rate but not improvement in measures of neurobehavioral outcomes, which was accompanied by significantly lower plasma level of IL-1β. Intriguingly, delayed melatonin treatment improved neurobehavioral dysfunction by normalization of hippocampal BDNF and GDNF expressions. In conclusion, our study suggests the beneficial effects of both early and delayed melatonin treatment after sepsis development, which implicates melatonin has a potential therapeutic value in sepsis-associated organ damage including brain dysfunction.
Collapse
Affiliation(s)
- Mu-Huo Ji
- Department of Anesthesiology, Jinling Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - De-Guo Xia
- Department of Anesthesiology, Clinical Medical College of Yangzhou University (Subei People's Hospital of Jiangsu Province), Yangzhou, Jiangsu Province, China
| | - Lan-Yue Zhu
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Xia Zhu
- Department of Anesthesiology, Jinling Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Xiao-Yan Zhou
- Department of Anesthesiology, Jinling Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Jiang-Yan Xia
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Jian-Jun Yang
- Department of Anesthesiology, Jinling Clinical Medical College of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
182
|
Du Z, Xu S, Hu S, Yang H, Zhou Z, Sidhu K, Miao Y, Liu Z, Shen W, Reiter RJ, Hua J, Peng S. Melatonin attenuates detrimental effects of diabetes on the niche of mouse spermatogonial stem cells by maintaining Leydig cells. Cell Death Dis 2018; 9:968. [PMID: 30237484 PMCID: PMC6148071 DOI: 10.1038/s41419-018-0956-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/10/2018] [Accepted: 08/20/2018] [Indexed: 01/04/2023]
Abstract
Diabetes mellitus affects a large number of men of reproductive age and it usually leads to serious reproductive disorders. However, the underlying mechanisms and specific therapies still remain largely unknown. We observed Leydig cell loss in the testes of diabetic mice. Continuous high glycemic status of testes stimulated expression of Caspase12, Grp78, and Chop, the three ERS response factors; this might induce cell cycle arrest and apoptosis of Leydig cells in response to ERS. In these diabetic mouse models, melatonin alleviated apoptosis of testicular stromal cell induced by ERS, and promoted SSCs self-renewal by recovering Leydig cells secretion of CSF1 after 8 weeks of treatment. To explore the relationship between CSF-1 and ERS in Leydig cells, we treated Leydig tumor cell line with an activator Tuniamycin and an inhibitor 4-Phenylbutyrate of ERS. Our data showed that the CSF-1 expression in mouse Leydig cell lines decreased six-fold while reversely increasing five-fold in the 4-Phenylbutyrate-treated group. Thus, melatonin likely alleviates the loss of Leydig cells in diabetic testes and provides a healthier niche for SSCs to self-renew and continually provide healthy sperm for male fertility.
Collapse
Affiliation(s)
- Zhaoyu Du
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Shuanshuan Xu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Shuxian Hu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Hong Yang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zhe Zhou
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Kuldip Sidhu
- Centre for Healthy Brain Ageing, UNSW Medicine, Randwick, NSW, 2031, Australia
| | - Yiliang Miao
- College of Animal Science & Technology, College of Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, China
| | - Zhonghua Liu
- College of Life Science, Northeast Agricultural University, 150036, Harbin, China
| | - Wei Shen
- College of life sciences, Institute of Reproductive Sciences, Qingdao Agriculture University, 266109, Qingdao, China
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX78229-3900, USA
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Sha Peng
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
183
|
Zhou H, Wang J, Hu S, Zhu H, Toanc S, Ren J. BI1 alleviates cardiac microvascular ischemia-reperfusion injury via modifying mitochondrial fission and inhibiting XO/ROS/F-actin pathways. J Cell Physiol 2018; 234:5056-5069. [PMID: 30256421 DOI: 10.1002/jcp.27308] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 08/01/2018] [Indexed: 12/23/2022]
Abstract
Pathogenesis of cardiac microvascular ischemia-reperfusion (IR) injury is associated with excessive mitochondrial fission. However, the upstream mediator of mitochondrial fission remains obscure. Bax inhibitor 1 (BI1) is linked to multiple mitochondrial functions, and there have been no studies investigating the contribution of BI1 on mitochondrial fission in the setting of cardiac microvascular IR injury. This study was undertaken to establish the action of BI1 on the cardiac microvascular reperfusion injury and figure out whether BI1 sustained endothelial viability via inhibiting mitochondrial fission. Our observation indicated that BI1 was downregulated in reperfused hearts and overexpression of BI1 attenuated microvascular IR injury. Mechanistically, reperfusion injury elevated the levels of xanthine oxidase (XO), an effect that was followed by increased reactive oxygen species (ROS) production. Subsequently, oxidative stress mediated F-actin depolymerization and the latter promoted mitochondrial fission. Aberrant fission caused mitochondrial dysfunction and ultimately activated mitochondrial apoptosis in cardiac microvascular endothelial cells. By comparison, BI1 overexpression repressed XO expression and thus neutralized ROS, interrupting F-actin-mediated mitochondrial fission. The inhibitory effect of BI1 on mitochondrial fission sustained endothelial viability, reversed endothelial barrier integrity, attenuated the microvascular inflammation response, and maintained microcirculation patency. Altogether, we conclude that BI1 is essential in maintaining mitochondrial homeostasis and alleviating cardiac microvascular IR injury. Deregulated BI1 via the XO/ROS/F-actin pathways plays a causative role in the development of cardiac microvascular reperfusion injury.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Cardiology, Medical School of Chinese PLA Hospital, Beijing, China.,Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, Wyoming
| | - Jin Wang
- Department of Cardiology, Medical School of Chinese PLA Hospital, Beijing, China
| | - Shunying Hu
- Department of Cardiology, Medical School of Chinese PLA Hospital, Beijing, China
| | - Hong Zhu
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, Wyoming
| | - Sam Toanc
- Department of Chemical and Environmental Engineering, University of California, Riverside, California
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, Wyoming
| |
Collapse
|
184
|
Wang Y, Sun X, Ji K, Du L, Xu C, He N, Wang J, Liu Y, Liu Q. Sirt3-mediated mitochondrial fission regulates the colorectal cancer stress response by modulating the Akt/PTEN signalling pathway. Biomed Pharmacother 2018; 105:1172-1182. [DOI: 10.1016/j.biopha.2018.06.071] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/11/2018] [Accepted: 06/13/2018] [Indexed: 02/07/2023] Open
|
185
|
Wang H, Wu Y, Han W, Li J, Xu K, Li Z, Wang Q, Xu K, Liu Y, Xie L, Wu J, He H, Xu H, Xiao J. Hydrogen Sulfide Ameliorates Blood-Spinal Cord Barrier Disruption and Improves Functional Recovery by Inhibiting Endoplasmic Reticulum Stress-Dependent Autophagy. Front Pharmacol 2018; 9:858. [PMID: 30210332 PMCID: PMC6121111 DOI: 10.3389/fphar.2018.00858] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/16/2018] [Indexed: 12/11/2022] Open
Abstract
Spinal cord injury (SCI) induces the disruption of blood-spinal cord barrier (BSCB), which elicits neurological deficits by triggering secondary injuries. Hydrogen sulfide (H2S) is a gaseous mediator that has been reported to have neuroprotective effect in the central nervous system. However, the relationship between H2S and BSCB disruption during SCI remains unknown. Therefore, it is interesting to evaluate whether the administration of NaHS, a H2S donor, can protect BSCB integrity against SCI and investigate the potential mechanisms underlying it. In present study, we found that SCI markedly activated endoplasmic reticulum (ER) stress and autophagy in a rat model of complete crushing injury to the spinal cord at T9 level. NaHS treatment prevented the loss of tight junction (TJ) and adherens junction (AJ) proteins both in vivo and in vitro. However, the protective effect of NaHS on BSCB restoration was significantly reduced by an ER stress activator (tunicamycin, TM) and an autophagy activator (rapamycin, Rapa). Moreover, SCI-induced autophagy was remarkably blocked by the ER stress inhibitor (4-phenylbutyric acid, 4-PBA). But the autophagy inhibitor (3-Methyladenine, 3-MA) only inhibited autophagy without obvious effects on ER stress. Finally, we had revealed that NaHS significantly alleviated BSCB permeability and improved functional recovery after SCI, and these effects were markedly reversed by TM and Rapa. In conclusion, our present study has demonstrated that NaHS treatment is beneficial for SCI recovery, indicating that H2S treatment is a potential therapeutic strategy for promoting SCI recovery.
Collapse
Affiliation(s)
- Haoli Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Yanqing Wu
- The Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Wen Han
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Jiawei Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Kebin Xu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Zhengmao Li
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Qingqing Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Ke Xu
- The Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Yanlong Liu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Ling Xie
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Jiang Wu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Huacheng He
- The Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Huazi Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian Xiao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
186
|
Zhang Z, Yu J. NR4A1 Promotes Cerebral Ischemia Reperfusion Injury by Repressing Mfn2-Mediated Mitophagy and Inactivating the MAPK-ERK-CREB Signaling Pathway. Neurochem Res 2018; 43:1963-1977. [PMID: 30136162 DOI: 10.1007/s11064-018-2618-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/09/2018] [Accepted: 08/17/2018] [Indexed: 12/31/2022]
Abstract
Mitochondrial dysfunction has been acknowledged as the key pathogenic mechanism in cerebral ischemia-reperfusion (IR) injury. Mitophagy is the protective system used to sustain mitochondrial homeostasis. However, the upstream regulator of mitophagy in response to brain IR injury is not completely understood. Nuclear receptor subfamily 4 group A member 1 (NR4A1) has been found to be associated with mitochondrial protection in a number of diseases. The aim of our study is to explore the functional role of NR4A1 in cerebral IR injury, with a particular focus on its influence on mitophagy. Wild-type mice and NR4A1-knockout mice were used to generate cerebral IR injury in vivo. Mitochondrial function and mitophagy were detected via immunofluorescence assays and western blotting. Cellular apoptosis was determined via MTT assays, caspase-3 activity and western blotting. Our data revealed that NR4A1 was significantly increased in the reperfused brain tissues. Genetic ablation of NR4A1 reduced the cerebral infarction area and repressed neuronal apoptosis. The functional study demonstrated that NR4A1 modulated cerebral IR injury by inducing mitochondrial damage. Higher NR4A1 promoted mitochondrial potential reduction, evoked cellular oxidative stress, interrupted ATP generation, and initiated caspase-9-dependent apoptosis. Mechanistically, NR4A1 induced mitochondrial damage by disrupting Mfn2-mediated mitophagy. Knockdown of NR4A1 elevated Mfn2 expression and therefore reversed mitophagic activity, sending a prosurvival signal for mitochondria in the setting of cerebral IR injury. Further, we demonstrated that NR4A1 modulated Mfn2 expression via the MAPK-ERK-CREB signaling pathway. Blockade of the ERK pathway could abrogate the permissive effect of NR4A1 deletion on mitophagic activation, contributing to neuronal mitochondrial apoptosis. Overall, our results demonstrate that the pathogenesis of cerebral IR injury is closely associated with a drop in protective mitophagy due to increased NR4A1 through the MAPK-ERK-CREB signaling pathway.
Collapse
Affiliation(s)
- Zhanwei Zhang
- Department of Neurosurgery, First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Jianbai Yu
- Department of Neurosurgery, First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China.
| |
Collapse
|
187
|
Reiter RJ, Tan DX, Rosales-Corral S, Galano A, Jou MJ, Acuna-Castroviejo D. Melatonin Mitigates Mitochondrial Meltdown: Interactions with SIRT3. Int J Mol Sci 2018; 19:E2439. [PMID: 30126181 PMCID: PMC6121285 DOI: 10.3390/ijms19082439] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/03/2018] [Accepted: 08/08/2018] [Indexed: 02/07/2023] Open
Abstract
Melatonin exhibits extraordinary diversity in terms of its functions and distribution. When discovered, it was thought to be uniquely of pineal gland origin. Subsequently, melatonin synthesis was identified in a variety of organs and recently it was shown to be produced in the mitochondria. Since mitochondria exist in every cell, with a few exceptions, it means that every vertebrate, invertebrate, and plant cell produces melatonin. The mitochondrial synthesis of melatonin is not photoperiod-dependent, but it may be inducible under conditions of stress. Mitochondria-produced melatonin is not released into the systemic circulation, but rather is used primarily in its cell of origin. Melatonin's functions in the mitochondria are highly diverse, not unlike those of sirtuin 3 (SIRT3). SIRT3 is an NAD+-dependent deacetylase which regulates, among many functions, the redox state of the mitochondria. Recent data proves that melatonin and SIRT3 post-translationally collaborate in regulating free radical generation and removal from mitochondria. Since melatonin and SIRT3 have cohabitated in the mitochondria for many eons, we predict that these molecules interact in many other ways to control mitochondrial physiology. It is predicted that these mutual functions will be intensely investigated in the next decade and importantly, we assume that the findings will have significant applications for preventing/delaying some age-related diseases and aging itself.
Collapse
Affiliation(s)
- Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX 78229, USA.
| | - Dun Xian Tan
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX 78229, USA.
| | - Sergio Rosales-Corral
- Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guardalajara, 4436 Jalisco, Mexico.
| | - Annia Galano
- Departamento de Quimica, Universidad Antonoma Metropolitana-Unidad Iztapalapa, San Rafael Atlixco 186, Col. Vicentina, Iztapalapa, C.P. 09340 Mexico D.F., Mexico.
| | - Mei-Jie Jou
- Department of Physiology and Pharmacology, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan 333, Taiwan.
| | - Dario Acuna-Castroviejo
- Departamento de Fisiologia, Instituto de Biotecnologia, Universidad de Granada, Avenida de Conocimiento S/U, 18016 Granada, Spain.
| |
Collapse
|
188
|
Guo ZN, Jin H, Sun H, Zhao Y, Liu J, Ma H, Sun X, Yang Y. Antioxidant Melatonin: Potential Functions in Improving Cerebral Autoregulation After Subarachnoid Hemorrhage. Front Physiol 2018; 9:1146. [PMID: 30174621 PMCID: PMC6108098 DOI: 10.3389/fphys.2018.01146] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 07/30/2018] [Indexed: 12/30/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is a subtype of stroke with high mortality and morbidity. Impaired cerebral autoregulation following SAH has been reported owing to effects on sympathetic control, endothelial function, myogenic response, and cerebral metabolism. Impaired cerebral autoregulation is associated with early brain injury, cerebral vasospasm/delayed cerebral ischemia, and SAH prognosis. However, few drugs have been reported to improve cerebral autoregulation after SAH. Melatonin is a powerful antioxidant that is effective (easily crosses the blood brain barrier) and safe (tolerated in large doses without toxicity). Theoretically, melatonin may impact the control mechanisms of cerebral autoregulation via antioxidative effects, protection of endothelial cell integrity, suppression of sympathetic nerve activity, increase in nitric oxide bioavailability, mediation of the myogenic response, and amelioration of hypoxemia. Furthermore, melatonin may have a comprehensive effect on cerebral autoregulation. This review discusses the potential effects of melatonin on cerebral autoregulation following SAH, in terms of the association between pharmacological activities and the mechanisms of cerebral autoregulation.
Collapse
Affiliation(s)
- Zhen-Ni Guo
- Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Hang Jin
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Huijie Sun
- Cadre Ward, The First Hospital of Jilin University, Changchun, China
| | - Yingkai Zhao
- Cadre Ward, The First Hospital of Jilin University, Changchun, China
| | - Jia Liu
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hongyin Ma
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Xin Sun
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yi Yang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
189
|
Ji K, Lin K, Wang Y, Du L, Xu C, He N, Wang J, Liu Y, Liu Q. TAZ inhibition promotes IL-2-induced apoptosis of hepatocellular carcinoma cells by activating the JNK/F-actin/mitochondrial fission pathway. Cancer Cell Int 2018; 18:117. [PMID: 30127666 PMCID: PMC6092825 DOI: 10.1186/s12935-018-0615-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 08/09/2018] [Indexed: 01/31/2023] Open
Abstract
Background Cytokine-based cancer therapies have attracted a great deal of attention in recent years. Unfortunately, resistance to treatment limits the efficacy of these therapeutics. Therefore, the aim of our study was to explore the mechanism of IL-2-based therapy for hepatocellular carcinoma in an attempt to increase the efficiency of this treatment option. Methods HepG2 cells were treated with IL-2. Then, siRNA against TZA was used to transfected into HepG2 cells. Cellular apoptosis was measured via MTT assay, TUNEL assay and caspase-3 activity. Cellular proliferation was evaluated via EdU assay and western blotting. Cellular migration was detected via Transwell assay. Mitochondrial function was monitored by mitochondrial potential analysis, ROS staining, immunofluorescence and western blotting. Pathway blocker and activator were used to establish the role of JNK/F-actin/mitochondrial fission signaling pathway in HepG2 cells stress response. Results Our study found that IL-2 treatment significantly reduced the viability, mobility and proliferation of HepG2 cells in vitro. We also demonstrated that IL-2 treatment was accompanied by an increase in the expression of transcriptional co-activator with PDZ-binding motif (TAZ). Interestingly, genetic ablation of TAZ in the presence of IL-2 further promoted apoptosis, inhibited mobility, and arrested proliferation in HepG2 cells. At the molecular level, IL-2 administration activated excessive mitochondrial fission via the JNK/F-actin pathway; these effects were further enhanced by TAZ deletion. Mechanistically, TAZ knockdown further increased the expression of mitochondrial fission-related proteins such as Drp1, Mff and Fis. The augmented mitochondrial fission stimulated ROS overproduction, mediated redox imbalance, interrupted mitochondrial energy generation, reduced mitochondrial membrane potential, promoted leakage of the pro-apoptotic molecule cyt-c into the nucleus, and initiated caspase-9-related mitochondrial death. Further, we demonstrated that the anti-proliferative and anti-metastatic effects of IL-2 in HepG2 cells were enhanced by TAZ deletion, suggesting that IL-2 sensitizes HepG2 cells to IL-2-based cytokine therapy. However, JNK/F-actin pathway blockade could abrogate the inhibitory effects of TAZ deletion on HepG2 migration, proliferation and survival. Conclusions Taken together, our data indicate that the anti-tumor effects of IL-2-based therapies may be enhanced by TAZ deletion in a JNK/F-actin pathway-dependent manner. This finding provides a novel combinatorial therapeutic approach for treating hepatocellular carcinoma that might significantly increase the efficacy of cytokine-based therapies in a clinical setting. Electronic supplementary material The online version of this article (10.1186/s12935-018-0615-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kaihua Ji
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin, 300192 China
| | - Kaili Lin
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin, 300192 China
| | - Yan Wang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin, 300192 China
| | - Liqing Du
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin, 300192 China
| | - Chang Xu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin, 300192 China
| | - Ningning He
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin, 300192 China
| | - Jinhan Wang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin, 300192 China
| | - Yang Liu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin, 300192 China
| | - Qiang Liu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin, 300192 China
| |
Collapse
|
190
|
Tao J, Yang M, Wu H, Ma T, He C, Chai M, Zhang X, Zhang J, Ding F, Wang S, Deng S, Zhu K, Song Y, Ji P, Liu H, Lian Z, Liu G. Effects of AANAT overexpression on the inflammatory responses and autophagy activity in the cellular and transgenic animal levels. Autophagy 2018; 14:1850-1869. [PMID: 29985091 DOI: 10.1080/15548627.2018.1490852] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
To explore the anti-inflammatory activity of endogenous produced melatonin, a melatonin-enriched animal model (goat) with AANAT transfer was successfully generated with somatic cell nuclear transfer (SCNT) technology. Basically, a pIRES2-EGFP-AANAT expression vector was constructed and was transferred into the female fetal fibroblast cells (FFCs) via electrotransfection and then the nuclear of the transgenic FFC was transferred to the eggs of the donor goats. The peripheral blood mononuclear cells (PBMCs) of the transgenic offspring expressed significantly higher levels of AANAT and melatonin synthetic function than those PBMCs from the wild-type (WT) animals. After challenge with lipopolysaccharide (LPS), the transgenic PBMCs had increased autophagosomes and LC3B expression while they exhibited suppressed production of the proinflammatory cytokines, IL1B and IL12 (IL12A-IL12B/p70), compared to their WT. The mechanistic analysis indicated that the anti-inflammatory activity of endogenous melatonin was mediated by MTNR1B (melatonin receptor 1B). MTNR1B stimulation activated the MAPK14 signaling pathway to promote cellular macroautophagy/autophagy, thus, suppressing the excessive inflammatory response of cellular. However, when the intact animals challenged with LPS, the serum proinflammatory cytokines were significantly higher in the transgenic goats than that in the WT. The results indicated that endogenous melatonin inhibited the MAPK1/3 signaling pathway and ROS production, subsequently downregulated gene expression of BECN1, ATG5 in PMBCs and then suppressed the autophagy activity of PBMCs and finally elevated levels of serum proinflammatory cytokines in transgenic animals, Herein we provided a novel melatonin-enriched animal model to study the potential effects of endogenously produced melatonin on inflammatory responses and autophagy activity.
Collapse
Affiliation(s)
- Jingli Tao
- a National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology , China Agricultural University , Beijing , China
| | - Minghui Yang
- a National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology , China Agricultural University , Beijing , China
| | - Hao Wu
- a National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology , China Agricultural University , Beijing , China
| | - Teng Ma
- a National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology , China Agricultural University , Beijing , China
| | - Changjiu He
- a National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology , China Agricultural University , Beijing , China.,b College of Animal Science and Technology , Huazhong Agricultural University , Wuhan , China
| | - Menglong Chai
- a National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology , China Agricultural University , Beijing , China
| | - Xiaosheng Zhang
- c Institute of Animal Husbandry and Veterinary , Academy of Agricultural Sciences of Tianjin , Tianjin , China
| | - Jinlong Zhang
- c Institute of Animal Husbandry and Veterinary , Academy of Agricultural Sciences of Tianjin , Tianjin , China
| | - Fangrong Ding
- d State Key Laboratory of Agrobiotechnology, College of Biological Sciences , China Agricultural University , Beijing , China
| | - Sutian Wang
- a National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology , China Agricultural University , Beijing , China
| | - Shoulong Deng
- e State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology , Chinese Academy of Sciences , Beijing , China
| | - Kuanfeng Zhu
- a National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology , China Agricultural University , Beijing , China
| | - Yukun Song
- a National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology , China Agricultural University , Beijing , China
| | - Pengyun Ji
- a National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology , China Agricultural University , Beijing , China
| | - Haijun Liu
- c Institute of Animal Husbandry and Veterinary , Academy of Agricultural Sciences of Tianjin , Tianjin , China
| | - Zhengxing Lian
- a National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology , China Agricultural University , Beijing , China
| | - Guoshi Liu
- a National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology , China Agricultural University , Beijing , China
| |
Collapse
|
191
|
Li R, Xin T, Li D, Wang C, Zhu H, Zhou H. Therapeutic effect of Sirtuin 3 on ameliorating nonalcoholic fatty liver disease: The role of the ERK-CREB pathway and Bnip3-mediated mitophagy. Redox Biol 2018; 18:229-243. [PMID: 30056271 PMCID: PMC6079484 DOI: 10.1016/j.redox.2018.07.011] [Citation(s) in RCA: 247] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 07/18/2018] [Accepted: 07/19/2018] [Indexed: 12/11/2022] Open
Abstract
Increased mitochondrial damage is related to the progression of a diet-induced nonalcoholic fatty liver disease. The aim of our study is to investigate the role of Sirtuin 3 (Sirt3) in treating nonalcoholic fatty liver disease with a focus on mitophagy and the ERK-CREB pathway. Our data indicated that Sirt3 was downregulated in liver tissue in response to chronic HFD treatment. Interestingly, re-introduction of Sirt3 protected hepatic function, attenuated liver fibrosis, alleviated the inflammatory response, and prevented hepatocyte apoptosis. Molecular investigations demonstrated that lipotoxicity was associated with an increase in mitochondrial apoptosis as evidenced by reduced mitochondrial potential, augmented ROS production, increased cyt-c leakage into the nucleus, and activated caspase-9 apoptotic signalling. Additionally, Sirt3 overexpression protected hepatocytes against mitochondrial apoptosis via promoting Bnip3-required mitophagy. Functional studies showed that Sirt3 reversed Bnip3 expression and mitophagy activity via the ERK-CREB signalling pathway. Blockade of the ERK-CREB axis repressed the promotive effects of Sirt3 on Bnip3 activation and mitophagy augmentation, finally negating the anti-apoptotic influences of Sirt3 on hepatocytes in the setting of high-fat-stress. Collectively, our data show that high-fat-mediated liver damage is associated with Sirt3 downregulation, which is followed by ERK-CREB pathway inactivation and Bnip3-mediated inhibition of mitophagy, causing hepatocytes to undergo mitochondria-dependent cell death. Based on this, strategies for enhancing Sirt3 activity and activating the ERK-CREB-Bnip3-mitophagy pathways could be used to treat nonalcoholic fatty liver disease. Sirt3 overexpression prevents diet-mediated fatty liver disease. Sirt3 blocks hepatocyte mitochondrial apoptosis in the setting of high-fat injury. Bnip3-mediated mitophagy protects mitochondria against high-fat-mediated damage. Sirt3 controls Bnip3-mediated mitophagy via the ERK-CREB signalling pathway.
Collapse
Affiliation(s)
- Ruibing Li
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, PR China
| | - Ting Xin
- Department of Cardiology, Tianjin First Central Hospital, Tianjin 300192, PR China
| | - Dandan Li
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, PR China
| | - Chengbin Wang
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, PR China.
| | - Hang Zhu
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, PR China.
| | - Hao Zhou
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, PR China; Center for Cardiovascular Research and Alternative Medicine, Wyoming University, Laramie, WY 82071, USA.
| |
Collapse
|
192
|
Roohbakhsh A, Shamsizadeh A, Hayes A, Reiter RJ, Karimi G. Melatonin as an endogenous regulator of diseases: The role of autophagy. Pharmacol Res 2018; 133:265-276. [DOI: 10.1016/j.phrs.2018.01.022] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/17/2017] [Accepted: 01/29/2018] [Indexed: 12/22/2022]
|
193
|
Liu J, Wang Q, Yang S, Huang J, Feng X, Peng J, Lin Z, Liu W, Tao J, Chen L. Electroacupuncture Inhibits Apoptosis of Peri-Ischemic Regions via Modulating p38, Extracellular Signal-Regulated Kinase (ERK1/2), and c-Jun N Terminal Kinases (JNK) in Cerebral Ischemia-Reperfusion-Injured Rats. Med Sci Monit 2018; 24:4395-4404. [PMID: 29943755 PMCID: PMC6048997 DOI: 10.12659/msm.908473] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 02/23/2018] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Previous studies suggested that inhibition of apoptosis prevents the dysfunction of ischemia-reperfusion injury. In the pathogenesis of ischemia-reperfusion injury, JNK/ERK1/2 and p38 play an essential role in regulation of cell apoptosis. Electroacupuncture (EA), a form of acupuncture, has demonstrated superiority in preventing ischemia-reperfusion injury, but the underlying mechanism is unclear. In the present study, we explored the effects of electroacupuncture at Shenting (GV24) and Baihui (GV20) acupoints on focal cerebral ischemia-reperfusion (MCAO) rats, and explored whether JNK/ERK1/2- and p38-mediated cell apoptosis are involved. MATERIAL AND METHODS The rats were divided into a sham operation control group, an ischemia group, and an electroacupuncture group with acupuncture applied for 10 days (30 min per day). TTC staining was used to calculate the ischemic brain volume. TUNEL staining and transmission electron microscopy were used to detect cell apoptosis. Western blot analysis and Bio-Plex were used to detect JNK, p38, ERK1/2, Bcl-2, and Bax protein expression. RESULTS We found that electroacupuncture at day 10 significantly reduced cerebral infarction. In addition, electroacupuncture suppressed activation of JNK and p38, while enhancing the activation of ERK1/2 in the peri-ischemic regions. Consequently, the effect of electroacupuncture on these pathways resulted in the inhibition of apoptosis, which was demonstrated by TUNEL and transmission electron microscopy. We found that electroacupuncture upregulated the anti-apoptotic Bcl-2/Bax ratio in peri-ischemic regions. CONCLUSIONS Our findings suggest that inhibition of cell apoptosis via regulating multiple signaling pathways might be a mechanism whereby electroacupuncture has a positive therapeutic effect on post-stroke impairment.
Collapse
Affiliation(s)
- Jiao Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, P.R. China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, P.R. China
| | - Qin Wang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, P.R. China
| | - Shanli Yang
- Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, P.R. China
| | - Jia Huang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, P.R. China
| | - Xiaodong Feng
- First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, P.R. China
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, P.R. China
| | - Zhengkun Lin
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, P.R. China
| | - Weilin Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, P.R. China
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, P.R. China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, P.R. China
- Fujian Key Laboratory of Exercise Rehabilitation, Fuzhou, Fujian, P.R. China
| | - Lidian Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, P.R. China
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, P.R. China
- Fujian Key Laboratory of Exercise Rehabilitation, Fuzhou, Fujian, P.R. China
| |
Collapse
|
194
|
Gao Z, Wang H, Zhang B, Wu X, Zhang Y, Ge P, Chi G, Liang J. Trehalose inhibits H 2O 2-induced autophagic death in dopaminergic SH-SY5Y cells via mitigation of ROS-dependent endoplasmic reticulum stress and AMPK activation. Int J Med Sci 2018; 15:1014-1024. [PMID: 30013443 PMCID: PMC6036158 DOI: 10.7150/ijms.25656] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 05/21/2018] [Indexed: 01/30/2023] Open
Abstract
Autophagy is a catabolic process to maintain intracellular homeostasis via removal of cytoplasmic macromolecules and damaged cellular organelles through lysosome-mediated degradation. Trehalose is often regarded as an autophagy inducer, but we reported previously that it could prevent ischemic insults-induced autophagic death in neurons. Thus, we further investigated in this study whether trehalose could protect human dopaminergic SH-SY5Y cells against H2O2-induced lethal autophagy. We found pretreatment with trehalose not only prevented H2O2-induced death in SH-SY5Y cells, but also reversed H2O2-induced upregulation of LC3II, Beclin1 and ATG5 and downregulation of p62. Then, we proved that either autophagy inhibitor 3MA or genetic knockdown of ATG5 prevented H2O2-triggered death in SH-SY5Y cells. These indicated that trehalose could inhibit H2O2-induced autophagic death in SH-SY5Y cells. Further, we found that trehalose inhibited H2O2-induced AMPK activation and endoplasmic reticulum (ER) stress. Moreover, inhibition of AMPK activation with compound C or alleviation of ER stress with chemical chaperone 4-PBA obviously attenuated H2O2-induced changes in autophagy-related proteins. Notably, we found that trehalose inhibited H2O2-induced increase of intracellular ROS and reduction in the activities of CAT and SOD. Consistently, our data revealed as well that mitigation of intracellular ROS levels with antioxidant NAC markedly attenuated H2O2-induced AMPK activation and ER stress. Therefore, we demonstrated in this study that trehalose prevented H2O2-induced autophagic death in SH-SY5Y cells via mitigation of ROS-dependent endoplasmic reticulum stress and AMPK activation.
Collapse
Affiliation(s)
- Zhijie Gao
- Department of Neurosurgery, First hospital of Jilin University, Changchun 130021, China
| | - Helei Wang
- Department of Gastrointestinal Surgery, First hospital of Jilin University, Changchun 130021, China
| | - Bo Zhang
- Department of Pediatric Neurology, First hospital of Jilin University, Changchun 130021, China
| | - Xuemei Wu
- Department of Pediatric Neurology, First hospital of Jilin University, Changchun 130021, China
| | - Yanfeng Zhang
- Department of Pediatric Neurology, First hospital of Jilin University, Changchun 130021, China
| | - Pengfei Ge
- Department of Neurosurgery, First hospital of Jilin University, Changchun 130021, China
- Research center of neuroscience, First hospital of Jilin University, Changchun 130021, China
| | - Guangfan Chi
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Jianmin Liang
- Department of Pediatric Neurology, First hospital of Jilin University, Changchun 130021, China
- Research center of neuroscience, First hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
195
|
Wang H, Zhao X, Ni C, Dai Y, Guo Y. Zearalenone regulates endometrial stromal cell apoptosis and migration via the promotion of mitochondrial fission by activation of the JNK/Drp1 pathway. Mol Med Rep 2018; 17:7797-7806. [PMID: 29620184 DOI: 10.3892/mmr.2018.8823] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 12/12/2017] [Indexed: 11/05/2022] Open
Abstract
Increased endometrial stromal cell (ESC) survival and migration is responsible for the development and progression of endometriosis. However, little is known about the mechanisms underlying ESC survival and migration, and limited therapeutic strategies that are able to reverse these abnormalities are available. The present study investigated the effects of zearalenone (ZEA) on ESC survival and migration, particularly focusing on mitochondrial fission and the c‑Jun N‑terminal kinase (JNK)/dynamin‑related protein 1 (Drp1) pathway. The results revealed that ZEA induced ESC apoptosis in a dose‑dependent manner. Furthermore, ZEA treatment triggered excessive mitochondrial fission resulting in structural and functional mitochondrial damage, leading to the collapse of the mitochondrial membrane potential and subsequent leakage of cytochrome c into the cytoplasm. This triggered the mitochondrial pathway of apoptosis. Additionally, ZEA‑induced mitochondrial fission decreased ESC migration through F‑actin/G‑actin homeostasis dysregulation. ZEA also increased JNK phosphorylation and subsequently Drp1 phosphorylation at the serine 616 position, resulting in Drp1 activation. JNK/Drp1 pathway inhibition abolished the inhibitory effects of ZEA on ESC survival and migration. In summary, the present study demonstrated that ZEA reduced ESC survival and migration through the stimulation of mitochondrial fission by activation of the JNK/Drp1 pathway.
Collapse
Affiliation(s)
- Huixiang Wang
- Department of Gynecology and Obstetrics, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Xiaoli Zhao
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Chengxiang Ni
- Department of Gynecology and Obstetrics, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Yuyang Dai
- Department of National Institute for Drug Clinical Trial, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Yan Guo
- Department of Gynecology and Obstetrics, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| |
Collapse
|
196
|
Ni H, Rui Q, Xu Y, Zhu J, Gao F, Dang B, Li D, Gao R, Chen G. RACK1 upregulation induces neuroprotection by activating the IRE1-XBP1 signaling pathway following traumatic brain injury in rats. Exp Neurol 2018. [DOI: 10.1016/j.expneurol.2018.03.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
197
|
Zhou Z, Lu J, Liu WW, Manaenko A, Hou X, Mei Q, Huang JL, Tang J, Zhang JH, Yao H, Hu Q. Advances in stroke pharmacology. Pharmacol Ther 2018; 191:23-42. [PMID: 29807056 DOI: 10.1016/j.pharmthera.2018.05.012] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Stroke occurs when a cerebral blood vessel is blocked or ruptured, and it is the major cause of death and adult disability worldwide. Various pharmacological agents have been developed for the treatment of stroke either through interrupting the molecular pathways leading to neuronal death or enhancing neuronal survival and regeneration. Except for rtPA, few of these agents have succeeded in clinical trials. Recently, with the understanding of the pathophysiological process of stroke, there is a resurrection of research on developing neuroprotective agents for stroke treatment, and novel molecular targets for neuroprotection and neurorestoration have been discovered to predict or offer clinical benefits. Here we review the latest major progress of pharmacological studies in stroke, especially in ischemic stroke; summarize emerging potential therapeutic mechanisms; and highlight recent clinical trials. The aim of this review is to provide a panorama of pharmacological interventions for stroke and bridge basic and translational research to guide the clinical management of stroke therapy.
Collapse
Affiliation(s)
- Zhenhua Zhou
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA; Department of Neurology, Southwest Hospital, the Third Military Medical University, Chongqing 400038, China
| | - Jianfei Lu
- Discipline of Neuroscience, Department of Physiology and Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wen-Wu Liu
- Department of Diving and Hyperbaric Medicine, the Second Military Medical University, Shanghai 200433, China
| | - Anatol Manaenko
- Department of Neurology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Xianhua Hou
- Department of Neurology, Southwest Hospital, the Third Military Medical University, Chongqing 400038, China
| | - Qiyong Mei
- Department of Neurosurgery, Changzheng Hospital, the Second Military Medical University, Shanghai 200003, China
| | - Jun-Long Huang
- Discipline of Neuroscience, Department of Physiology and Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Honghong Yao
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, China.
| | - Qin Hu
- Discipline of Neuroscience, Department of Physiology and Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
198
|
Kim KA, Shin D, Kim JH, Shin YJ, Rajanikant GK, Majid A, Baek SH, Bae ON. Role of Autophagy in Endothelial Damage and Blood-Brain Barrier Disruption in Ischemic Stroke. Stroke 2018; 49:1571-1579. [PMID: 29724893 DOI: 10.1161/strokeaha.117.017287] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Kyeong-A Kim
- From the College of Pharmacy Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea (K.-A.K., D.S., J.-H.K., Y.-J.S., O.-N.B.)
| | - Donggeun Shin
- From the College of Pharmacy Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea (K.-A.K., D.S., J.-H.K., Y.-J.S., O.-N.B.)
| | - Jeong-Hyeon Kim
- From the College of Pharmacy Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea (K.-A.K., D.S., J.-H.K., Y.-J.S., O.-N.B.)
| | - Young-Jun Shin
- From the College of Pharmacy Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea (K.-A.K., D.S., J.-H.K., Y.-J.S., O.-N.B.)
| | - G K Rajanikant
- School of Biotechnology, National Institute of Technology Calicut, Kerala, India (G.K.R.)
| | - Arshad Majid
- Sheffield Institute for Translational Neuroscience, University of Sheffield, England (A.M.)
| | - Seung-Hoon Baek
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon, Republic of Korea (S.-H.B.)
| | - Ok-Nam Bae
- From the College of Pharmacy Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea (K.-A.K., D.S., J.-H.K., Y.-J.S., O.-N.B.)
| |
Collapse
|
199
|
Dong T, Chen N, Ma X, Wang J, Wen J, Xie Q, Ma R. The protective roles of L-borneolum, D-borneolum and synthetic borneol in cerebral ischaemia via modulation of the neurovascular unit. Biomed Pharmacother 2018; 102:874-883. [PMID: 29728011 DOI: 10.1016/j.biopha.2018.03.087] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 03/14/2018] [Accepted: 03/14/2018] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE Borneol has been used to treat stroke in China since ancient times. In our previous research, we demonstrated the effect of borneol on cerebral ischaemia injury via meta-analysis. The neurovascular unit (NVU) is the structural basis of the preservation of the brain microenvironment and is believed to be a promising target in treating stroke. In this research, we explored the roles of three kinds of borneol, namely, L-borneolum (B1), D-borneolum (B2) and synthetic borneol (B3), in the NVU with permanent middle cerebral artery occluded (pMCAO) rats. METHODS The Longa scoring method was used to evaluate nerve function deficits in the pMCAO rats. Awakening time, brain water content, brain index and brain edema rate were also measured. TTC staining was used to calculate the cerebral infarction rate. The morphology of the ischaemia penumbra brain tissue was observed via HE staining, and the neuronal denatured cell index (DCI) was calculated. An enzyme-linked immunosorbent assay (ELISA) was used to determine the levels of vascular endothelial growth factor VEGF and TNF-α in the serum. Moreover, the ultrastructures of the neurons and of the blood-brain barrier (BBB) were observed using transmission electron microscopy. The expression levels of Claudin-5, Bcl-2 and Bax in the ischaemia penumbra of pMCAO rats were detected using real-time PCR and immunohistochemistry. RESULTS Pretreatment with B1, B2 and B3 delayed the recovery time (P < 0.01). B1 remarkably ameliorated neurological deficits 24 h after cerebral ischaemia (P < 0.05). Moreover, B1 and B3 were both able to ameliorate brain edema and the area of cerebral infarction. In addition, B1, B2 and B3 all increased serum VEGF levels and decreased serum TNF-α levels (P < 0.01). For the ultrastructure determination, the BBB and the nerve centre were significantly improved by B1, B2 and B3. The mechanistic exploration revealed that B2 and B3 protected the brain by reducing the Bax/Bcl-2 ratio (P < 0.05, P < 0.01, respectively). Immunohistochemistry suggested that B1, B2 and B3 could also enhance the expression of Claudin-5 (P < 0.01). CONCLUSION The three kinds of borneol demonstrated different protective effects on cerebral ischaemia injury. L-Borneolum displayed the most prominent anti-cerebral ischaemia effect among them. The mechanism was most likely executed via anti-apoptosis and anti-inflammation effects and maintenance of the stability of the BBB and TJs to comprehensively improve NVU function.
Collapse
Affiliation(s)
- Taiwei Dong
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Nian Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiao Ma
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jian Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Jing Wen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qian Xie
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Rong Ma
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
200
|
Wang M, Wang J, Liu Z, Guo X, Wang N, Jia N, Zhang Y, Yuan J. Effects of intermedin on autophagy in cerebral ischemia/reperfusion injury. Neuropeptides 2018; 68:15-21. [PMID: 29128104 DOI: 10.1016/j.npep.2017.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 09/30/2017] [Accepted: 10/31/2017] [Indexed: 12/23/2022]
Abstract
OBJECTIVE This study aimed to evaluate the effects of intermedin (IMD) on autophagy in cerebral ischemia/reperfusion (I/R) injury (CIRI). METHODS Sixty rats were randomly averaged into four groups: sham, ischemia/reperfusion (I/R), IMD, and 3-methyladenine (3-MA). In the sham group, the right common carotid artery, external carotid artery, and internal carotid artery were detached, and no monofilament was inserted. In the other groups, two hours after cerebral ischemia, the rats were injected through the lateral ventricle with normal saline for I/R group, IMD for the IMD group, and 3-MA for the 3-MA group for 24h. The cerebral injury was assessed by evaluation of neurological function, hematoxylin and eosin (H&E) staining, and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. The expressions of autophagy associated proteins, such as microtubule-associated protein 1 light chain 3 (LC3), Beclin1, and sequestosome 1 (P62) were analyzed using immunohistochemistry staining and western blot. Meanwhile, transmission electron microscopy was used to investigate the ultrastructure of the brains. RESULTS IMD could reduce neuron cell damage and infarction formation and has a protective effect against CIRI as 3-MA. The levels of LC3II/LC3I and Beclin1 were significantly decreased and the P62 level was significantly higher in the IMD group compared with I/R group, which is similar to the effect of 3-MA on CIRI. CONCLUSIONS IMD has a similar effect as 3-MA, can reduce pathological neuronal injury and protect the brain against CIRI in rats by attenuating the effects of autophagy. Our findings provide evidence for IMD's protective effects in relation to ischemic cerebrovascular diseases.
Collapse
Affiliation(s)
- Meng Wang
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei Province, China
| | - Jing Wang
- Department of Clinical Medicine, Tangshan Vocational and Technical College, Tangshan 063000, Hebei Province, China; Department of Internal Medicine, Tangshan Union Medical College Hospital, Tangshan 063000, Hebei Province, China
| | - Zhengang Liu
- Department of Neurosurgery, The Second People's Hospital of Liaocheng, Liaocheng 252600, Shandong province, China
| | - Xin Guo
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, Tangshan 063000, Hebei Province, China
| | - Ning Wang
- College of Psychology, North China University of Science and Technology, Tangshan 063000, Hebei Province, China
| | - Nana Jia
- College of Psychology, North China University of Science and Technology, Tangshan 063000, Hebei Province, China
| | - Yanshu Zhang
- School of Public Health, North China University of Science and Technology, Tangshan 063000, Hebei Province, China
| | - Jie Yuan
- Institute of Mental Health, North China University of Science and Technology, Tangshan 063000, Hebei Province, China.
| |
Collapse
|