1
|
Xu R, Zhang L, Pan H, Zhang Y. Retinoid X receptor heterodimers in hepatic function: structural insights and therapeutic potential. Front Pharmacol 2024; 15:1464655. [PMID: 39478961 PMCID: PMC11521896 DOI: 10.3389/fphar.2024.1464655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/14/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
Nuclear receptors (NRs) are key regulators of multiple physiological functions and pathological changes in the liver in response to a variety of extracellular signaling changes. Retinoid X receptor (RXR) is a special member of the NRs, which not only responds to cellular signaling independently, but also regulates multiple signaling pathways by forming heterodimers with various other NR. Therefore, RXR is widely involved in hepatic glucose metabolism, lipid metabolism, cholesterol metabolism and bile acid homeostasis as well as hepatic fibrosis. Specific activation of particular dimers regulating physiological and pathological processes may serve as important pharmacological targets. So here we describe the basic information and structural features of the RXR protein and its heterodimers, focusing on the role of RXR heterodimers in a number of physiological processes and pathological imbalances in the liver, to provide a theoretical basis for RXR as a promising drug target.
Collapse
Affiliation(s)
- Renjie Xu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Linyue Zhang
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Pan
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Zhao Y, Fei Y, Zhao Y, Li M, Hu Y, Cai K, Yu SH, Luo Z. Biomineralization-Tuned Nanounits Reprogram the Signal Transducer and Activator of Transcription 3 Signaling for Ferroptosis-Immunotherapy in Cancer Stem Cells. ACS NANO 2024; 18:21268-21287. [PMID: 39083438 DOI: 10.1021/acsnano.4c05084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 08/02/2024]
Abstract
Cancer stem cells (CSCs) are promising targets for improving anticancer treatment outcomes while eliminating recurrence, but their treatment remains a major challenge. Here, we report a nanointegrative strategy to realize CSC-targeted ferroptosis-immunotherapy through spatiotemporally controlled reprogramming of STAT3-regulated signaling circuits. Specifically, STAT3 inhibitor niclosamide (Ni) and an experimental ferroptosis drug (1S, 3R)-RSL3 (RSL3) are integrated into hyaluronic acid-modified amorphous calcium phosphate (ACP) nanounits through biomineralization (CaP-PEG-HA@Ni/RSL3), which could be recognized by CD44-overexpressing CSCs and released in a synchronized manner. Ni inhibits the CSC-intrinsic STAT3-PD-L1 axis to stimulate adaptive immunity and enhance interferon gamma (IFNγ) secretion by CD8+ T cells to downregulate SLC7A11 and SLC3A2 for blocking glutathione biosynthesis. Meanwhile, Ni-dependent STAT3 inhibition also upregulates ACSL4 through downstream signaling and IFNγ feedback. These effects cooperate with RSL3-mediated GPX4 deactivation to induce pronounced ferroptosis. Furthermore, CaP-PEG-HA@Ni/RSL3 also impairs the immunosuppressive M2-like tumor-associated macrophages, while Ca2+ ions released from degraded ACP could chelate with lipid peroxides in ferroptotic CSCs to avoid CD8+ T-cell inhibition, thus boosting the effector function of activated CD8+ T cells. This study offers a cooperative ferroptosis-immunotherapeutic approach for the treatment of refractory cancer.
Collapse
Affiliation(s)
- Youbo Zhao
- Center for Tissue Engineering and Stem Cell Research, Key Laboratory for Autoimmune Disease Research, Department of Hepatic-Biliary-Pancreatic Surgery Affiliate Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang 550025, P. R. China
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yang Fei
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yang Zhao
- Division of Nanomaterials and Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, CAS Center for Excellence in Nanoscience, Collaborative Innovation Center of Suzhou Nano Science and Technology, Department of Chemistry, Hefei Science Center of CAS, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing 400044, P. R. China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing 400044, P. R. China
| | - Shu-Hong Yu
- Division of Nanomaterials and Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, CAS Center for Excellence in Nanoscience, Collaborative Innovation Center of Suzhou Nano Science and Technology, Department of Chemistry, Hefei Science Center of CAS, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| |
Collapse
|
3
|
Fogarty EF, Harch PG. Case report: Dementia sensitivity to altitude changes and effective treatment with hyperbaric air and glutathione precursors. Front Neurol 2024; 15:1356662. [PMID: 38978816 PMCID: PMC11229546 DOI: 10.3389/fneur.2024.1356662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/15/2023] [Accepted: 05/10/2024] [Indexed: 07/10/2024] Open
Abstract
A 78-year-old man with dementia experienced waxing and waning of symptoms with changes in altitude as he traveled from his home in the Rocky Mountains to lower elevations and back. To replicate the improvement in his symptoms with travel to lower elevations (higher pressure), the patient was treated with a near-identical repressurization in a hyperbaric chamber using compressed air. With four 1-h treatments at 1.3 Atmospheres Absolute (ATA) and concurrent administration of low-dose oral glutathione amino acid precursors, he recovered speech and showed improvement in activities of daily living. Regional broadcast media had documented his novel recovery. Nosocomial COVID-19 and withdrawal of hyperbaric air therapy led to patient demise 7 months after initiation of treatment. It is theorized that hyperbaric air therapy stimulated mitochondrial biochemical and physical changes, which led to clinical improvement.
Collapse
Affiliation(s)
| | - Paul G. Harch
- LSU Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
4
|
Wu Z, Wang Y, Gao R, Chen J, Chen Y, Li M, Gao Y. Potential therapeutic effects of traditional Chinese medicine in acute mountain sickness: pathogenesis, mechanisms and future directions. Front Pharmacol 2024; 15:1393209. [PMID: 38895636 PMCID: PMC11183292 DOI: 10.3389/fphar.2024.1393209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/28/2024] [Accepted: 05/06/2024] [Indexed: 06/21/2024] Open
Abstract
Background and objectives Acute mountain sickness (AMS) is a pathology with different symptoms in which the organism is not adapted to the environment that occurs under the special environment of high altitude. Its main mechanism is the organism's tissue damage caused by acute hypobaric hypoxia. Traditional Chinese medicine (TCM) theory focuses on the holistic concept. TCM has made remarkable achievements in the treatment of many mountain sicknesses. This review outlines the pathogenesis of AMS in modern and traditional medicine, the progress of animal models of AMS, and summarizes the therapeutic effects of TCM on AMS. Methods Using the keywords "traditional Chinese medicine," "herbal medicine," "acute mountain sickness," "high-altitude pulmonary edema," "high-altitude cerebral edema," "acute hypobaric hypoxia," and "high-altitude," all relevant TCM literature published up to November 2023 were collected from Scopus, Web of Science, PubMed, and China National Knowledge Infrastructure databases, and the key information was analyzed. Results We systematically summarised the effects of acute hypobaric hypoxia on the tissues of the organism, the study of the methodology for the establishment of an animal model of AMS, and retrieved 18 proprietary Chinese medicines for the clinical treatment of AMS. The therapeutic principle of medicines is mainly invigorating qi, activating blood and removing stasis. The components of botanical drugs mainly include salidroside, ginsenoside Rg1, and tetrahydrocurcumin. The mechanism of action of TCM in the treatment of AMS is mainly through the regulation of HIF-1α/NF-κB signaling pathway, inhibition of inflammatory response and oxidative stress, and enhancement of energy metabolism. Conclusion The main pathogenesis of AMS is unclear. Still, TCM formulas and components have been used to treat AMS through multifaceted interventions, such as compound danshen drip pills, Huangqi Baihe granules, salidroside, and ginsenoside Rg1. These components generally exert anti-AMS pharmacological effects by inhibiting the expression of VEGF, concentration of MDA and pro-inflammatory factors, down-regulating NF-κB/NLRP3 pathway, and promoting SOD and Na + -K + -ATPase activities, which attenuates acute hypobaric hypoxia-induced tissue injury. This review comprehensively analyses the application of TCM in AMS and makes suggestions for more in-depth studies in the future, aiming to provide some ideas and insights for subsequent studies.
Collapse
Affiliation(s)
- Zhenhui Wu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
- Department of Hematology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yihao Wang
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Rong Gao
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Junru Chen
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Yingfan Chen
- Department of Traditional Chinese Medicine, The Sixth Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Maoxing Li
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Yue Gao
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
- Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
5
|
Zhao R, Wang X, Zhou X, Jiang S, Zhang L, Yu Z. Metabolites and gene expression in the myocardium of fasting rats in an acute hypoxic environment. BMC Genomics 2023; 24:251. [PMID: 37165337 PMCID: PMC10173486 DOI: 10.1186/s12864-023-09309-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/31/2022] [Accepted: 04/12/2023] [Indexed: 05/12/2023] Open
Abstract
With the rising demand for entry to extremely high altitudes (HAs), rapid adaptability to extremely hypoxic environments is a challenge that we need to explore. Fasting was used to evaluate acute hypoxia tolerance at HA and was proven to be an effective method for improving the survival rate at extreme HA. Our experiments also showed that fasting pretreatment for 72 h significantly increased the 24 h survival rate of rats at 7620 m from 10 to 85% and protected the myocardium cells of rats. Here, we compared the metabolites and gene expression in the myocardium of SD rats pretreated with fasting and nonfasting at normal altitude and extreme HA. Our findings demonstrated that the dynamic contents of detected differential metabolites (DMs) between different rat groups were consistent with the expression of differentially expressed genes (DEGs), and DM clusters also showed strong correlations with DEG clusters. DM clusters related to amino acids and lipids were significantly lower in the fasting groups, and the correlated DEG clusters were enriched in mitotic pathways, including CDK1, CDC7, NUF2, and MCM6, suggesting that fasting can attenuate mitotic processes in cardiac tissues and reduce the synthesis of amino acids and lipids. L-Glutamine-related metabolites were particularly low at extreme HA without pretreatment but were normal in the fasting groups. The DEGs in the cluster related to L-glutamine-related metabolites were enriched for T-cell receptor V(D)J recombination, the Hippo signaling pathway, the Wnt signaling pathway, the cGMP-PKG signaling pathway, and the mTOR signaling pathway and were significantly downregulated, indicating that the content of L-glutamine decreased at extreme HA, while fasting increased it to adapt to the environment. Moreover, abundant fatty acids were detected when rats were exposed to extreme HA without pretreatment. Our study revealed the fasting and hypoxic environment-related factors in SD rats and provided new insights into the genetic and molecular characteristics in the myocardium, which is critical to developing more potential rapid adaptation methods to extreme HA.
Collapse
Affiliation(s)
- Ruzhou Zhao
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences (AMMS), Beijing, China
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Xiaobo Wang
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Xiang Zhou
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
- Department of Nuclear Medicine, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Shuai Jiang
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Lin Zhang
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Zhibin Yu
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China.
| |
Collapse
|
6
|
Mallet RT, Burtscher J, Pialoux V, Pasha Q, Ahmad Y, Millet GP, Burtscher M. Molecular Mechanisms of High-Altitude Acclimatization. Int J Mol Sci 2023; 24:ijms24021698. [PMID: 36675214 PMCID: PMC9866500 DOI: 10.3390/ijms24021698] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/30/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
High-altitude illnesses (HAIs) result from acute exposure to high altitude/hypoxia. Numerous molecular mechanisms affect appropriate acclimatization to hypobaric and/or normobaric hypoxia and curtail the development of HAIs. The understanding of these mechanisms is essential to optimize hypoxic acclimatization for efficient prophylaxis and treatment of HAIs. This review aims to link outcomes of molecular mechanisms to either adverse effects of acute high-altitude/hypoxia exposure or the developing tolerance with acclimatization. After summarizing systemic physiological responses to acute high-altitude exposure, the associated acclimatization, and the epidemiology and pathophysiology of various HAIs, the article focuses on molecular adjustments and maladjustments during acute exposure and acclimatization to high altitude/hypoxia. Pivotal modifying mechanisms include molecular responses orchestrated by transcription factors, most notably hypoxia inducible factors, and reciprocal effects on mitochondrial functions and REDOX homeostasis. In addition, discussed are genetic factors and the resultant proteomic profiles determining these hypoxia-modifying mechanisms culminating in successful high-altitude acclimatization. Lastly, the article discusses practical considerations related to the molecular aspects of acclimatization and altitude training strategies.
Collapse
Affiliation(s)
- Robert T. Mallet
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Johannes Burtscher
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
- Institute of Sport Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Vincent Pialoux
- Inter-University Laboratory of Human Movement Biology EA7424, University Claude Bernard Lyon 1, University of Lyon, FR-69008 Lyon, France
| | - Qadar Pasha
- Institute of Hypoxia Research, New Delhi 110067, India
| | - Yasmin Ahmad
- Defense Institute of Physiology & Allied Sciences (DIPAS), Defense Research & Development Organization(DRDO), New Delhi 110054, India
| | - Grégoire P. Millet
- Department of Biomedical Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
- Institute of Sport Sciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, A-6020 Innsbruck, Austria
- Austrian Society for Alpine and High-Altitude Medicine, A-6020 Innsbruck, Austria
- Correspondence:
| |
Collapse
|
7
|
Zhao R, Xu Y, Wang X, Zhou X, Liu Y, Jiang S, Zhang L, Yu Z. Withaferin A Enhances Mitochondrial Biogenesis and BNIP3-Mediated Mitophagy to Promote Rapid Adaptation to Extreme Hypoxia. Cells 2022; 12:cells12010085. [PMID: 36611879 PMCID: PMC9818179 DOI: 10.3390/cells12010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/01/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Rapid adaptation to extreme hypoxia is a challenging problem, and there is no effective scheme to achieve rapid adaptation to extreme hypoxia. In this study, we found that withaferin A (WA) can significantly reduce myocardial damage, maintain cardiac function, and improve survival in rats in extremely hypoxic environments. Mechanistically, WA protects against extreme hypoxia by affecting BCL2-interacting protein 3 (BNIP3)-mediated mitophagy and the peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α)-mediated mitochondrial biogenesis pathway among mitochondrial quality control mechanisms. On the one hand, enhanced mitophagy eliminates hypoxia-damaged mitochondria and prevents the induction of apoptosis; on the other hand, enhanced mitochondrial biogenesis can supplement functional mitochondria and maintain mitochondrial respiration to ensure mitochondrial ATP production under acute extreme hypoxia. Our study shows that WA can be used as an effective drug to improve tolerance to extreme hypoxia.
Collapse
Affiliation(s)
- Ruzhou Zhao
- Department of Aerospace Physiology, Air Force Medical University, 169# Changle West Road, Xi’an 710032, China
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences (AMMS), Beijing 100071, China
| | - Yixin Xu
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences (AMMS), Beijing 100071, China
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xiaobo Wang
- Department of Aerospace Physiology, Air Force Medical University, 169# Changle West Road, Xi’an 710032, China
| | - Xiang Zhou
- Department of Aerospace Physiology, Air Force Medical University, 169# Changle West Road, Xi’an 710032, China
| | - Yanqi Liu
- Department of Aerospace Physiology, Air Force Medical University, 169# Changle West Road, Xi’an 710032, China
| | - Shuai Jiang
- Department of Aerospace Physiology, Air Force Medical University, 169# Changle West Road, Xi’an 710032, China
| | - Lin Zhang
- Department of Aerospace Physiology, Air Force Medical University, 169# Changle West Road, Xi’an 710032, China
| | - Zhibin Yu
- Department of Aerospace Physiology, Air Force Medical University, 169# Changle West Road, Xi’an 710032, China
- Correspondence:
| |
Collapse
|
8
|
Yang J, Jia Z, Song X, Shi J, Wang X, Zhao X, He K. Proteomic and clinical biomarkers for acute mountain sickness in a longitudinal cohort. Commun Biol 2022; 5:548. [PMID: 35668171 PMCID: PMC9170681 DOI: 10.1038/s42003-022-03514-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/12/2021] [Accepted: 05/18/2022] [Indexed: 11/17/2022] Open
Abstract
Ascending to high-altitude by non-high-altitude natives is a well-suited model for studying acclimatization to extreme environments. Acute mountain sickness (AMS) is frequently experienced by visitors. The diagnosis of AMS mainly depends on a self-questionnaire, revealing the need for reliable biomarkers for AMS. Here, we profiled 22 AMS symptom phenotypes, 65 clinical indexes, and plasma proteomic profiles of AMS via a combination of proximity extension assay and multiple reaction monitoring of a longitudinal cohort of 53 individuals. We quantified 1069 proteins and validated 102 proteins. Via differential analysis, machine learning, and functional association analyses. We found and validated that RET played an important role in the pathogenesis of AMS. With high-accuracies (AUCs > 0.9) of XGBoost-based models, we prioritized ADAM15, PHGDH, and TRAF2 as protective, predictive, and diagnostic biomarkers, respectively. Our findings shed light on the precision medicine for AMS and the understanding of acclimatization to high-altitude environments.
Collapse
Affiliation(s)
- Jing Yang
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
- Research Center for Medical Big Data, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- Key Laboratory of Biomedical Engineering and Translational Medicine, Ministry of Industry and Information Technology, Chinese PLA General Hospital, Beijing, China
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Chinese PLA General Hospital, Beijing, China
| | - Zhilong Jia
- Key Laboratory of Biomedical Engineering and Translational Medicine, Ministry of Industry and Information Technology, Chinese PLA General Hospital, Beijing, China.
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Chinese PLA General Hospital, Beijing, China.
- Center for Artificial Intelligence in Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China.
| | - Xinyu Song
- Key Laboratory of Biomedical Engineering and Translational Medicine, Ministry of Industry and Information Technology, Chinese PLA General Hospital, Beijing, China
- Center for Artificial Intelligence in Medicine, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Jinlong Shi
- Research Center for Medical Big Data, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
- Key Laboratory of Biomedical Engineering and Translational Medicine, Ministry of Industry and Information Technology, Chinese PLA General Hospital, Beijing, China
| | - Xiaoreng Wang
- Laboratory of Radiation Injury Treatment, Medical Innovation Research Division, PLA General Hospital, Beijing, China
| | - Xiaojing Zhao
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Chinese PLA General Hospital, Beijing, China
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Kunlun He
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China.
- Research Center for Medical Big Data, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China.
- Key Laboratory of Biomedical Engineering and Translational Medicine, Ministry of Industry and Information Technology, Chinese PLA General Hospital, Beijing, China.
- Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
9
|
Zhao R, Zhao X, Wang X, Liu Y, Yang J, Jiang S, Zhou X, Jiao B, Zhang L, Liu Y, Yu Z. Fasting promotes acute hypoxic adaptation by suppressing mTOR-mediated pathways. Cell Death Dis 2021; 12:1045. [PMID: 34732698 PMCID: PMC8566556 DOI: 10.1038/s41419-021-04351-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/13/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 11/09/2022]
Abstract
Rapid adaptation to a hypoxic environment is an unanswered question that we are committed to exploring. At present, there is no suitable strategy to achieve rapid hypoxic adaptation. Here, we demonstrate that fasting preconditioning for 72 h reduces tissue injuries and maintains cardiac function, consequently significantly improving the survival rates of rats under extreme hypoxia, and this strategy can be used for rapid hypoxic adaptation. Mechanistically, fasting reduces blood glucose and further suppresses tissue mTOR activity. On the one hand, fasting-induced mTOR inhibition reduces unnecessary ATP consumption and increases ATP reserves under acute hypoxia as a result of decreased protein synthesis and lipogenesis; on the other hand, fasting-induced mTOR inhibition improves mitochondrial oxygen utilization efficiency to ensure ATP production under acute hypoxia, which is due to the significant decrease in ROS generation induced by enhanced mitophagy. Our findings highlight the important role of mTOR in acute hypoxic adaptation, and targeted regulation of mTOR could be a new strategy to improve acute hypoxic tolerance in the body.
Collapse
Affiliation(s)
- Ruzhou Zhao
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Xingcheng Zhao
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Xiaobo Wang
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Yanqi Liu
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Jie Yang
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Shuai Jiang
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Xiang Zhou
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
- Department of Nuclear Medicine, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Bo Jiao
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Lin Zhang
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Yong Liu
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China.
| | - Zhibin Yu
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China.
| |
Collapse
|
10
|
Yue XY, Wang XB, Zhao RZ, Jiang S, Zhou X, Jiao B, Zhang L, Yu ZB. Fasting improves tolerance to acute hypoxia in rats. Biochem Biophys Res Commun 2021; 569:161-166. [PMID: 34252588 DOI: 10.1016/j.bbrc.2021.06.099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/17/2021] [Revised: 06/22/2021] [Accepted: 06/29/2021] [Indexed: 11/29/2022]
Abstract
Acute high-altitude illness seriously threatens the health and lives of people who rapidly ascend to high altitudes, but there is currently no particularly effective method for the prevention or treatment of acute high-altitude illness. In the present study, we found that fasting preconditioning effectively improved the survival rate of rats exposed to a simulated altitude of 7620 m for 24 h, and a novel animal model of rapid adaptation to acute hypoxia was established. Compared with control treatment, fasting preconditioning activated AMPK, induced autophagy, decreased ROS levels, and inhibited NF-κB signaling in the cardiac tissues of rats. Our results suggested that fasting effectively improved the acute hypoxia tolerance of rats, which was gradually enhanced with prolongation of fasting. In addition, the acute hypoxia tolerance of young rats was significantly higher than that of adult rats. These experimental results lay the foundation for achieving rapid adaptation to acute hypoxia in humans.
Collapse
Affiliation(s)
- Xiao-Ya Yue
- Department of Aerospace Physiology, Fourth Military Medical University, 169(#) Changle West Road, Xi'an, 710032, China; Department of Internal Medicine, Affiliated Hospital of Xizang Minzu University, Xianyang, 712082, China.
| | - Xiao-Bo Wang
- Department of Aerospace Physiology, Fourth Military Medical University, 169(#) Changle West Road, Xi'an, 710032, China.
| | - Ru-Zhou Zhao
- Department of Aerospace Physiology, Fourth Military Medical University, 169(#) Changle West Road, Xi'an, 710032, China.
| | - Shuai Jiang
- Department of Aerospace Physiology, Fourth Military Medical University, 169(#) Changle West Road, Xi'an, 710032, China.
| | - Xiang Zhou
- Department of Aerospace Physiology, Fourth Military Medical University, 169(#) Changle West Road, Xi'an, 710032, China.
| | - Bo Jiao
- Department of Aerospace Physiology, Fourth Military Medical University, 169(#) Changle West Road, Xi'an, 710032, China.
| | - Lin Zhang
- Department of Aerospace Physiology, Fourth Military Medical University, 169(#) Changle West Road, Xi'an, 710032, China.
| | - Zhi-Bin Yu
- Department of Aerospace Physiology, Fourth Military Medical University, 169(#) Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
11
|
Ferrer B, Suresh H, Santamaria A, Rocha JB, Bowman AB, Aschner M. The antioxidant role of STAT3 in methylmercury-induced toxicity in mouse hypothalamic neuronal GT1-7 cell line. Free Radic Biol Med 2021; 171:245-259. [PMID: 34010664 PMCID: PMC8217327 DOI: 10.1016/j.freeradbiomed.2021.05.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 02/26/2021] [Revised: 05/04/2021] [Accepted: 05/13/2021] [Indexed: 12/27/2022]
Abstract
Oxidative stress, impairment of antioxidant defenses, and disruption of calcium homeostasis are associated with the toxicity of methylmercury (MeHg). Yet, the relative contribution and interdependence of these effects and other molecular mechanisms that mediate MeHg-induced neurotoxicity remain uncertain. The signal transducer and activator of transcription 3 (STAT3) is a transcription factor that regulates the expression of anti-apoptotic and cell cycle progression genes. In addition to its role in cell growth and survival, STAT3 regulates redox homeostasis and prevents oxidative stress by the modulation of nuclear genes that encode for electron transport complexes (ETC) and antioxidant enzymes. Here we tested the hypothesis that STAT3 contributes to the orchestration of the antioxidant defense response against MeHg injury. We show that MeHg (>1 μM) exposure induced STAT3 activation within 1 h and beyond in mouse hypothalamic neuronal GT1-7 cells in a concentration-and time-dependent manner. Pharmacological inhibition of STAT3 phosphorylation exacerbated MeHg-induced reactive oxygen species (ROS) production and antioxidant responses. Finally, treatment with the antioxidant Trolox demonstrated that MeHg-induced STAT3 activation is mediated, at least in part, by MeHg-induced ROS generation. Combined, our results demonstrated a role for the STAT3 signaling pathway as an early response to MeHg-induced oxidative stress.
Collapse
Affiliation(s)
- Beatriz Ferrer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, 10461, Bronx, NY, USA.
| | - Harshini Suresh
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, 10461, Bronx, NY, USA.
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, 14269, Mexico City, Mexico.
| | - João Batista Rocha
- Department of Biochemistry and Molecular Biology, Universidade Federal de Santa Maria, Santa Maria, Brazil.
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, United States.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, 10461, Bronx, NY, USA; IM Sechenov First Moscow State Medical University, Moscow, Russia, Institute of Cellular and Intracellular Symbiosis, Russian Academy of Sciences, Orenburg, Russia.
| |
Collapse
|
12
|
Modulation of lung cytoskeletal remodeling, RXR based metabolic cascades and inflammation to achieve redox homeostasis during extended exposures to lowered pO 2. Apoptosis 2021; 26:431-446. [PMID: 34002323 DOI: 10.1007/s10495-021-01679-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 05/10/2021] [Indexed: 10/21/2022]
Abstract
Extended exposure to low pO2 has multiple effects on signaling cascades. Despite multiple exploratory studies, omics studies elucidating the signaling cascades essential for surviving extended low pO2 exposures are lacking. In this study, we simulated low pO2 (PB = 40 kPa; 7620 m) exposure in male Sprague-Dawley rats for 3, 7 and 14 days. Redox stress assays and proteomics based network biology were performed using lungs and plasma. We observed that redox homeostasis was achieved after day 3 of exposure. We investigated the causative events for this. Proteo-bioinformatics analysis revealed STAT3 to be upstream of lung cytoskeletal processes and systemic lipid metabolism (RXR) derived inflammatory processes, which were the key events. Thus, during prolonged low pO2 exposure, particularly those involving slowly decreasing pressures, redox homeostasis is achieved but energy metabolism is perturbed and this leads to an immune/inflammatory signaling impetus after third day of exposure. We found that an interplay of lung cytoskeletal elements, systemic energy metabolism and inflammatory proteins aid in achieving redox homeostasis and surviving extended low pO2 exposures. Qualitative perturbations to cytoskeletal stability and innate immunity/inflammation were also observed during extended low pO2 exposure in humans exposed to 14,000 ft for 7, 14 and 21 days.
Collapse
|
13
|
Mirzaei S, Mohammadi AT, Gholami MH, Hashemi F, Zarrabi A, Zabolian A, Hushmandi K, Makvandi P, Samec M, Liskova A, Kubatka P, Nabavi N, Aref AR, Ashrafizadeh M, Khan H, Najafi M. Nrf2 signaling pathway in cisplatin chemotherapy: Potential involvement in organ protection and chemoresistance. Pharmacol Res 2021; 167:105575. [PMID: 33771701 DOI: 10.1016/j.phrs.2021.105575] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/22/2021] [Revised: 03/20/2021] [Accepted: 03/21/2021] [Indexed: 12/14/2022]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a vital transcription factor and its induction is of significant importance for protecting against oxidative damage. Increased levels of Reactive Oxygen Species (ROS) stimulate Nrf2 signaling, enhancing the activity of antioxidant enzymes such as catalase, superoxide dismutase and glutathione peroxidase. These enzymes are associated with retarding oxidative stress. On the other hand, Nrf2 activation in cancer cells is responsible for the development of chemoresistance due to disrupting oxidative mediated-cell death by reducing ROS levels. Cisplatin (CP), cis-diamminedichloroplatinum(II), is a potent anti-tumor agent extensively used in cancer therapy, but its frequent application leads to the development of chemoresistance as well. In the present study, association of Nrf2 signaling with chemoresistance to CP and protection against its deleterious effects is discussed. Anti-tumor compounds, mainly phytochemicals, retard chemoresistance by suppressing Nrf2 signaling. Upstream mediators such as microRNAs can regulate Nrf2 expression during CP chemotherapy regimens. Protection against side effects of CP is mediated via activating Nrf2 signaling and its downstream targets activating antioxidant defense system. Protective agents that activate Nrf2 signaling, can ameliorate CP-mediated ototoxicity, nephrotoxicity and neurotoxicity. Reducing ROS levels and preventing cell death are the most important factors involved in alleviating CP toxicity upon Nrf2 activation. As pre-clinical experiments advocate the role of Nrf2 in chemoprotection and CP resistance, translating these findings to the clinic can provide a significant progress in treatment of cancer patients.
Collapse
Affiliation(s)
- Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Aliasghar Tabatabaei Mohammadi
- Asu Vanda Gene Research Company, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Science Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956 Istanbul, Turkey
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Pooyan Makvandi
- Centre for Materials Interface, Istituto Italiano di Tecnologia, viale Rinaldo Piaggio 34, 56025 Pisa, Pontedera, Italy
| | - Marek Samec
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Alena Liskova
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Noushin Nabavi
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6 Canada
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Translational Sciences, Xsphera Biosciences Inc., Boston, MA, USA
| | - Milad Ashrafizadeh
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956 Istanbul, Turkey; Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan.
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanashah University of Medical Sciences, Kermanshah 6715847141, Iran; Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
14
|
Paul S, Gangwar A, Bhargava K, Ahmad Y. D4F prophylaxis enables redox and energy homeostasis while preventing inflammation during hypoxia exposure. Biomed Pharmacother 2021; 133:111083. [PMID: 33378979 DOI: 10.1016/j.biopha.2020.111083] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/23/2020] [Revised: 11/18/2020] [Accepted: 11/28/2020] [Indexed: 02/02/2023] Open
Abstract
Apo-A1 is correlated with conditions like hyperlipidemia, cardiovascular diseases, high altitude pulmonary edema and etc. where hypoxia constitutes an important facet.Hypoxia causes oxidative stress, vaso-destructive and inflammatory outcomes.Apo-A1 is reported to have vasoprotective, anti-oxidative, anti-apoptotic, and anti-inflammatory effects. However, effects of Apo-A1 augmentation during hypoxia exposure are unknown.In this study, we investigated the effects of exogenously supplementing Apo-A1-mimetic peptide on SD rats during hypoxia exposure. For easing the processes of delivery, absorption and bio-availability, Apo-A1 mimetic peptide D4F was used. The rats were given 10 mg/kg BW dose (i.p.) of D4F for 7 days and then exposed to hypoxia. D4F was observed to attenuate both oxidative stress and inflammation during hypoxic exposure. D4F improved energy homeostasis during hypoxic exposure. D4F did not affect HIF-1a levels during hypoxia but increased MnSOD levels while decreasing CRP and Apo-B levels. D4F showed promise as a prophylactic against hypoxia exposure.
Collapse
Affiliation(s)
- Subhojit Paul
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi, 110054, India
| | - Anamika Gangwar
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi, 110054, India
| | - Kalpana Bhargava
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi, 110054, India
| | - Yasmin Ahmad
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi, 110054, India.
| |
Collapse
|
15
|
Liu Q, He J, Zhou X, Han M, Li J, Liu C, Yuan H. ACP-5862 suppresses esophageal squamous cell carcinoma growth through inducing apoptosis via activation of endoplasmic reticulum stress and ROS production. Biochem Biophys Res Commun 2021; 534:995-1002. [PMID: 33168189 DOI: 10.1016/j.bbrc.2020.10.052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/23/2020] [Accepted: 10/17/2020] [Indexed: 01/14/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a common type of human oral malignancy with poor survival. Presently, it is necessary to find new and effective drugs for clinical therapy. This study aimed to identify the potential anti-tumor effects of ACP-5862, a major metabolite of acalabrutinib, on human ESCC progression, and to reveal the underlying mechanisms. Our findings suggested that ACP-5862 treatments markedly reduced the cell proliferation of ESCC cell lines in a time- and dose-dependent manner, while had no significant cytotoxicity to normal cells. Cell cycle arrest in G2/M phase was markedly induced by ACP-5862 in ESCC cells. Furthermore, apoptosis and endoplasmic reticulum (ER) stress were detected in ESCC cells treated with ACP-5862. Intriguingly, ACP-5862-induced apoptotic cell death was partly dependent on ER stress. Moreover, reactive oxygen species (ROS) was greatly triggered in ACP-5862-incubated ESCC cells, which was closely involved in apoptosis and ER stress mediated by ACP-5862. In addition, we showed that the expression of nuclear factor-erythroid 2-related factor-2 (Nrf-2) was considerably reduced in ACP-5862-treated cells. Importantly, ACP-5862 combined with Nrf-2 knockdown could further induce apoptosis and ER stress in ESCC cells compared with the ACP-5862 single group. Animal studies confirmed that repressing Nrf-2 promoted the anti-tumor effect of ACP-5862 on ESCC growth. Taken together, these findings demonstrated that ACP-5862 exerted anti-cancer effects on ESCC through inducing ER stress-mediated apoptosis via the ROS production. Meanwhile, ACP-5862 co-treated with Nrf-2 inhibitors may supply new and effective therapeutic strategies for ESCC treatment in future.
Collapse
Affiliation(s)
- Qiong Liu
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing, 100853, China; National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
| | - Jingjing He
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing, 100853, China; National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
| | - Xuejun Zhou
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing, 100853, China; National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
| | - Mingkun Han
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing, 100853, China; National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
| | - Jianhui Li
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing, 100853, China; National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China; Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital Sanya 572000, China
| | - Chenqing Liu
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing, 100853, China; National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China
| | - Hu Yuan
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing, 100853, China; National Clinical Research Center for Otolaryngologic Diseases, Beijing 100853, China.
| |
Collapse
|
16
|
Tian Y, Song H, Qin W, Ding Z, Zhang Y, Shan W, Jin D. Mammalian STE20-Like Kinase 2 Promotes Lipopolysaccharides-Mediated Cardiomyocyte Inflammation and Apoptosis by Enhancing Mitochondrial Fission. Front Physiol 2020; 11:897. [PMID: 32848850 PMCID: PMC7424023 DOI: 10.3389/fphys.2020.00897] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/28/2020] [Accepted: 07/02/2020] [Indexed: 12/16/2022] Open
Abstract
In this study, we analyzed the role of mammalian STE20-like protein kinase 2 (Mst2), a serine-threonine protein kinase, in Lipopolysaccharides (LPS)-mediated inflammation and apoptosis in the H9C2 cardiomyocytes. Mst2 mRNA and protein levels were significantly upregulated in the LPS-treated H9C2 cardiomyocytes. LPS treatment induced expression of IL-2, IL-8, and MMP9 mRNA and proteins in the H9C2 cardiomyocytes, and this was accompanied by increased caspase-3/9 mediating H9C2 cardiomyocyte apoptosis. LPS treatment also increased mitochondrial reactive oxygen species (ROS) and the levels of antioxidant enzymes, such as GSH, SOD, and GPX, in the H9C2 cardiomyocytes. The LPS-treated H9C2 cardiomyocytes showed lower cellular ATP levels and mitochondrial state-3/4 respiration but increased mitochondrial fragmentation, including upregulation of the mitochondrial fission genes Drp1, Mff, and Fis1. LPS-induced inflammation, mitochondrial ROS, mitochondrial fission, and apoptosis were all significantly suppressed by pre-treating the H9C2 cardiomyocytes with the Mst2 inhibitor, XMU-MP1. However, the beneficial effects of Mst2 inhibition by XMU-MP1 were abolished by carbonyl cyanide-4-(trifluoromethoxy) phenylhydrazone (FCCP), a potent activator of mitochondrial fission. These findings demonstrate that Mst2 mediates LPS-induced cardiomyocyte inflammation and apoptosis by increasing mitochondrial fission.
Collapse
Affiliation(s)
- Yanan Tian
- Department of Cardiology, The Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Haijiu Song
- The First Department of Medicine, Chengde City Hospital of traditional Chinese Medicine, Chengde, China
| | - Wei Qin
- Department of Cardiology, The Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Zhenjiang Ding
- Department of Cardiology, The Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Ying Zhang
- Department of Cardiology, The Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Weichao Shan
- Department of Cardiology, The Affiliated Hospital of Chengde Medical College, Chengde, China
| | - Dapeng Jin
- Department of Cardiology, The Affiliated Hospital of Chengde Medical College, Chengde, China
| |
Collapse
|
17
|
Deng B, Liu W, Pu L, Wang X, Duan R, Wang T, Wang Z, Du L, Gao Z, Chen Z. Quantitative Proteomics Reveals the Effects of Resveratrol on High-Altitude Polycythemia Treatment. Proteomics 2020; 20:e1900423. [PMID: 32468662 DOI: 10.1002/pmic.201900423] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/30/2019] [Revised: 05/18/2020] [Indexed: 12/17/2022]
Abstract
High-altitude polycythemia (HAPC) is a common plateau chronic disease in which red blood cells are compensatory hyperproliferative due to high altitude hypoxic environment. HAPC severely affects the physical and mental health of populations on the plateau. However, the pathogenesis and treatment of HAPC has been rarely investigated. Here, the hypoxia-induced HAPC model of rat is established, in which hemoglobin concentration significantly increases and platelets clearly decrease. The effect of resveratrol upon hypoxia enables HAPC remission and makes hemoglobin and platelet tend to a normal level. Furthermore, quantitative proteomics is applied to investigate the plasma proteome variation and the underlying molecular regulation during HAPC occurrence and treatment with resveratrol. Hypoxia promotes erythrocyte developing and differentiating and disrupts cytoskeleton organization. Notably, the resveratrol administration reverses the proteome change pattern due to hypoxia and contributes to plateau adaption. Quantitative verification of differentially expressed proteins confirms the roles of resveratrol in HAPC. Resveratrol is expected to be useful for HAPC treatment.
Collapse
Affiliation(s)
- Bingnan Deng
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Weili Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Lingling Pu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Xinxing Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Ruifeng Duan
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Tianhui Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Zirou Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Lianqun Du
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Zhixian Gao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Zhaoli Chen
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| |
Collapse
|
18
|
Ma G, Liu Y, Wang Y, Wen Z, Li X, Zhai H, Miao L, Luo J. Liraglutide reduces hyperglycemia-induced cardiomyocyte death through activating glucagon-like peptide 1 receptor and targeting AMPK pathway. J Recept Signal Transduct Res 2020; 40:133-140. [PMID: 32013667 DOI: 10.1080/10799893.2020.1719517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/11/2022]
Abstract
Objective: Hyperglycemia-mediated cardiomyocyte damage is associated with inflammation and AMPK inactivation.Aim: The aim of our study is to explore the protective effects exerted by liraglutide on AMPK pathway and glucagon-like peptide 1 receptor in diabetic cardiomyopathy.Methods: Cardiomyocytes were treated with high-glucose stress and cardiomyocyte viability was determined via (3-(4,5-dimethylthiazol-2-yl)-2,5diphenyltetrazolium bromide assay. Besides, LDH release, immunofluorescence, and qPCR were used to verify the influence of liraglutide on hyperglycemia-treated cardiomyocytes.Results: Hyperglycemia treatment caused inflammation response and oxidative stress were significantly elevated in cardiomyocytes. This alteration could be reversed by liraglutide. Besides, cell viability was reduced whereas apoptosis was increased after exposure to high glucose treatment. However, liraglutide treatment could attenuate apoptosis and reverse cell viability in cardiomyocyte. Further, we found that AMPK pathway was also activated and glucagon-like peptide 1 receptor expression was increased in response to liraglutide treatment.Conclusions: Liraglutide could attenuate hyperglycemia-mediated cardiomyocyte damage through reversing AMPK pathway and upregulating glucagon-like peptide 1 receptor.
Collapse
Affiliation(s)
- Guanqun Ma
- Department of Cardiology, The Third Central Hospital of Tianjin, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Yingwu Liu
- Department of Cardiology, The Third Central Hospital of Tianjin, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Yu Wang
- Department of Cardiology, The Third Central Hospital of Tianjin, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Zhinan Wen
- Department of Cardiology, The Third Central Hospital of Tianjin, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Xin Li
- Department of Cardiology, The Third Central Hospital of Tianjin, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Hu Zhai
- Department of Cardiology, The Third Central Hospital of Tianjin, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Li Miao
- Department of Cardiology, The Third Central Hospital of Tianjin, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Jieying Luo
- Department of Cardiology, The Third Central Hospital of Tianjin, Tianjin, China.,Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases.,Artificial Cell Engineering Technology Research Center, Tianjin, China.,Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| |
Collapse
|
19
|
Zhang Y, Zhang H, Shi W, Wang W. Mief1 augments thyroid cell dysfunction and apoptosis through inhibiting AMPK-PTEN signaling pathway. J Recept Signal Transduct Res 2020; 40:15-23. [PMID: 31960779 DOI: 10.1080/10799893.2020.1716799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/25/2022]
Abstract
Objective: Inflammation-mediated thyroid cell dysfunction and apoptosis increases the like-hood of hypothyroidism.Aim: Our aim in the present study is to explore the role of mitochondrial elongation factor 1 (Mief1) in thyroid cell dysfunction induced by TNFα.Materials and methods: Different doses of TNFα were used to incubate with thyroid cells in vitro. The survival rate, apoptotic index and proliferation capacity of thyroid cells were measured. Cellular energy metabolism and endoplasmic reticulum function related to protein synthesis were detected.Results: In response to TNFα treatment, the levels of Mief1 were increased, coinciding with a drop in the viability of thyroid cells in vitro. Loss of Mief1 attenuates TNFα-induced cell death through reducing the ratio of cell apoptosis. Further, we found that Mief1 deletion reversed cell energy metabolism and this effect was attributable to mitochondrial protection. Mief1 knockdown sustained mitochondrial membrane potential and reduced mitochondrial ROS overproduction. In addition, Mief1 knockdown also reduced endoplasmic reticulum stress, as evidenced by decreased levels of Chop and Caspase-12. Finally, our data verified that TNFα treatment inhibited the activity of AMPK-PTEN pathway whereas Mief1 deletion reversed the activity of AMPK and thus promoted the upregulation of PTEN. However, inhibition of AMPK-PTEN pathways could abolish the beneficial effects exerted by Mief1 deletion on thyroid cells damage and dysfunction.Conclusions: Altogether, our data indicate that immune abnormality-mediated thyroid cell dysfunction and death are alleviated by Mief1 deletion possible driven through reversing the activity of AMPK-PTEN pathways.
Collapse
Affiliation(s)
- Yonglan Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, Otorhinolaryngology Research Institute of Tianjin, Tianjin Key Laboratory of Auditory Speech Balance Medicine, Tianjin, People's Republic of China
| | - Haichao Zhang
- Department of Thyroid and Breast Surgery, Tianjin Fourth Central Hospital, Tianjin, People's Republic of China
| | - Wenjie Shi
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, Otorhinolaryngology Research Institute of Tianjin, Tianjin Key Laboratory of Auditory Speech Balance Medicine, Tianjin, People's Republic of China
| | - Wei Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, Otorhinolaryngology Research Institute of Tianjin, Tianjin Key Laboratory of Auditory Speech Balance Medicine, Tianjin, People's Republic of China
| |
Collapse
|
20
|
Wang Y, Zhang X, Wang P, Shen Y, Yuan K, Li M, Liang W, Que H. Sirt3 overexpression alleviates hyperglycemia-induced vascular inflammation through regulating redox balance, cell survival, and AMPK-mediated mitochondrial homeostasis. J Recept Signal Transduct Res 2019; 39:341-349. [PMID: 31680596 DOI: 10.1080/10799893.2019.1684521] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/20/2022]
Abstract
Context: Sirtuin-3 (Sirt3), a NAD-dependent deacetylase, has been reported to be involved in many biological processes.Objective: The present study aimed to investigate the effect and mechanism of Sirt3 on diabetic mice and human umbilical vein endothelial cells (HUVECs) under high glucose (HG) condition.Materials and methods: HUVECs were cultured under HG and inflammation pathway was determined via qPCR, western blots, and immunofluorescence.Results: Sirt3 expression was reduced in the progression of diabetic nephropathy. Overexpression of Sirt3 sustains renal function and retard the development of diabetic nephropathy. Mechanistically, Sirt3 overexpression attenuated hyperglycemia-mediated endothelial cells apoptosis in kidney. Besides, Sirt3 overexpression repressed oxidative injury and blocked caspase-9-related apoptosis pathway. Moreover, we found that Sirt3 overexpression was associated with AMPK activation and the latter elevates PGC1α-related mitochondrial protective system, especially mitochondrial autophagy. Loss of opa1 and/or inhibition of AMPK could depress mitochondrial autophagy and exacerbates mitochondrial function, finally contributing to the death of human renal mesangial cells.Conclusions: Our results demonstrated the beneficial effects of Sirt3 in the progression of diabetic nephropathy. Increased Sirt3-activated AMPK pathway, augments PGC1α-related mitochondrial protective system, sustained redox balance and closed caspase-9-involved apoptosis pathway in the setting of diabetic nephropathy.
Collapse
Affiliation(s)
- Yunfei Wang
- Department of Traditional Chinese Medicine Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xue Zhang
- Department of Vascular Surgery, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Peng Wang
- Department of Vascular Surgery, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yiting Shen
- Department of Traditional Chinese Medicine Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kai Yuan
- Department of Vascular Surgery, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Maoran Li
- Department of Vascular Surgery, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Liang
- Department of Vascular Surgery, South Campus, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huafa Que
- Department of Traditional Chinese Medicine Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
21
|
Fan L, Wang J, Ma C. miR125a attenuates BMSCs apoptosis via the MAPK‐ERK pathways in the setting of craniofacial defect reconstruction. J Cell Physiol 2019; 235:2857-2865. [PMID: 31578723 DOI: 10.1002/jcp.29191] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/30/2019] [Accepted: 09/03/2019] [Indexed: 12/30/2022]
Affiliation(s)
- Longkun Fan
- Department of Medical Plastic Surgery, Cangzhou Central Hospital, Hebei, China
| | - Jingxian Wang
- Department of Medical Plastic Surgery, Cangzhou Central Hospital, Hebei, China
| | - Chao Ma
- Department of Medical Plastic Surgery, Cangzhou Central Hospital, Hebei, China
| |
Collapse
|
22
|
Si ZP, Wang G, Han SS, Jin Y, Hu YX, He MY, Brand-Saberi B, Yang X, Liu GS. CNTF and Nrf2 Are Coordinately Involved in Regulating Self-Renewal and Differentiation of Neural Stem Cell during Embryonic Development. iScience 2019; 19:303-315. [PMID: 31404831 PMCID: PMC6700439 DOI: 10.1016/j.isci.2019.07.038] [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] [Academic Contribution Register] [Received: 01/16/2019] [Revised: 06/04/2019] [Accepted: 07/24/2019] [Indexed: 01/10/2023] Open
Abstract
There is high risk of fetal neurodevelopmental defects in pregestational diabetes mellitus (PGDM). However, the effective mechanism of hyperglycemia-induced neurodevelopmental negative effects, including neural stem cell self-renewal and differentiation, still remains obscure. Neuropoietic cytokines have been shown to play a vital part during nervous system development and in the coordination of neurons and gliocytes. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) dysfunction might be related to a reduction of self-protective response in brain malformation induced by hyperglycemia. We therefore evaluated the role of Nrf2 and neuropoietic cytokines in fetal neurodevelopmental defects induced by PGDM and determined the mechanisms involved. Our data reveal that PGDM dramatically impairs the developmental switch of neural stem cells from neurogenesis to gliogenesis, principally under the cooperative mediation of neuropoietic cytokine CNTF and Nrf2 antioxidative signaling. This indicates that CNTF and Nrf2 could be potentially used in the prevention or therapy of neurodevelopmental defects of PGDM offspring.
Collapse
Affiliation(s)
- Zhen-Peng Si
- Department of Pediatrics, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Sha-Sha Han
- Department of Pediatrics, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Ya Jin
- Department of Pediatrics, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Yu-Xuan Hu
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Mei-Yao He
- Department of Pediatrics, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany
| | - Xuesong Yang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou 510632, China; Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China.
| | - Guo-Sheng Liu
- Department of Pediatrics, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China.
| |
Collapse
|
23
|
Fan L, Wang J, Ma C. Pretreatment of bone mesenchymal stem cells with miR181-c facilitates craniofacial defect reconstruction via activating AMPK-Mfn1 signaling pathways. J Recept Signal Transduct Res 2019; 39:199-207. [PMID: 31466503 DOI: 10.1080/10799893.2019.1652649] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/26/2022]
Abstract
Context: Bone mesenchymal stem cells (BMSC)-based regenerative therapy is critical for the craniofacial defect reconstruction. However, oxidative stress micro-environment after transplantation limits the therapeutic efficiency of BMSC. The miR-181c has been found to be associated with cell survival and proliferation. Objective: Herein, we investigated whether prior miR-181c treatment promoted BMSC proliferation and survival under oxidative stress injury. Materials and methods: Cells were treated with hydrogen peroxide (H2O2) and then cell viability was determined via MTT assay, TUNEL staining and ELISA. Western blotting and immunofluorescence assay were used to detect those alterations of mitochondrial function. Results: H2O2 treatment reduced BMSC viability and this effect could be reversed via additional supplementation of miR181-c. Mechanistically, oxidative stress increased cell apoptosis, augmented caspase-3 activity, promoted reactive oxygen species (ROS) synthesis, impaired mitochondrial potential, and induced mitochondrial dynamics imbalance. However, miR-181c pretreatment reversed these effects of oxidative stress on BMSC. Moreover, miR-181c treatment improved BMSC proliferation, migration and paracrine, which are very important for craniofacial reconstruction. In addition, we identified that AMPK-Mfn1 axis was the direct targets of miR-181c in BMSC. Mfn1 silencing impaired the protective effects miR-181c on BMSC viability and proliferation under oxidative stress environment. Conclusions: Collectively, our results indicate that miR-181c participates in oxidative stress-mediated BMSC damage by modulating the AMPK-Mfn1 signaling pathway, suggesting miR-181c-AMPK-Mfn1 axis may serves as novel therapeutic targets to facilitate craniofacial defect reconstruction.
Collapse
Affiliation(s)
- Longkun Fan
- Department of Medical Plastic Surgery, Cangzhou Central Hospital , Cangzhou , China
| | - Jingxian Wang
- Department of Medical Plastic Surgery, Cangzhou Central Hospital , Cangzhou , China
| | - Chao Ma
- Department of Medical Plastic Surgery, Cangzhou Central Hospital , Cangzhou , China
| |
Collapse
|
24
|
Paul S, Gangwar A, Bhargava K, Khan N, Khurana P, Ahmad Y. Plasma protein(s)-based conceptual diagnostic tool for assessing high-altitude acclimation in humans. Funct Integr Genomics 2019; 20:191-200. [PMID: 31444657 DOI: 10.1007/s10142-019-00712-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/12/2019] [Revised: 07/30/2019] [Accepted: 08/20/2019] [Indexed: 11/24/2022]
Abstract
Exposure to high altitude above 3000 m leads to two outcomes-acclimation or high-altitude maladies. To reach a particular outcome, the plasma proteome is modified differentially, either in context of an acclimation response or mal-acclimation response leading to disease. This ensures that hypoxia-responsive plasma protein trends reflect acclimation in acclimated individuals when compared with their levels prior to acclimation. Such protein trends could be used to assess acclimation in an individual and any significant deviation from this trend may indicate non-acclimation, thereby preventing high-altitude illnesses before they manifest. In this study, we investigate and statistically evaluate the trendlines of various hypoxia-responsive plasma protein levels, reported significantly perturbed in our previous studies, in individuals (male; n = 20) exposed to 3520 m at high-altitude day 1 (HAD1), HAD4, and HAD7L and to 4420 m at HAD7H, HAD30, and HAD120. We observe that thioredoxin (Trx), glutathione peroxidase 3 (GPx-3), and apolipoprotein AI (Apo-AI) are statistically robust markers to assess acclimation across the exposure duration while sulfotransferase 1A1 (ST1A1) is a capable negative control whose levels increase only in cases of HAPE. We also observe exposure day-specific and resident altitude-specific proteins capable of accurately assessing acclimation when compared with baseline levels or the lower altitude zone.
Collapse
Affiliation(s)
- Subhojit Paul
- Defence R&D Organization (DRDO), Defence Institute of Physiology & Allied Sciences (DIPAS), Lucknow Road, Timarpur, New Delhi, 110054, India
| | - Anamika Gangwar
- Defence R&D Organization (DRDO), Defence Institute of Physiology & Allied Sciences (DIPAS), Lucknow Road, Timarpur, New Delhi, 110054, India
| | - Kalpana Bhargava
- Defence R&D Organization (DRDO), Defence Institute of Physiology & Allied Sciences (DIPAS), Lucknow Road, Timarpur, New Delhi, 110054, India
| | - Nilofar Khan
- Defence R&D Organization (DRDO), Defence Institute of Physiology & Allied Sciences (DIPAS), Lucknow Road, Timarpur, New Delhi, 110054, India
| | - Pankaj Khurana
- Defence R&D Organization (DRDO), Defence Institute of Physiology & Allied Sciences (DIPAS), Lucknow Road, Timarpur, New Delhi, 110054, India
| | - Yasmin Ahmad
- Defence R&D Organization (DRDO), Defence Institute of Physiology & Allied Sciences (DIPAS), Lucknow Road, Timarpur, New Delhi, 110054, India.
| |
Collapse
|
25
|
Tian H, Wang K, Jin M, Li J, Yu Y. Proinflammation effect of Mst1 promotes BV-2 cell death via augmenting Drp1-mediated mitochondrial fragmentation and activating the JNK pathway. J Cell Physiol 2019; 235:1504-1514. [PMID: 31283035 DOI: 10.1002/jcp.29070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/04/2019] [Accepted: 06/21/2019] [Indexed: 12/18/2022]
Abstract
Inflammation has been increasingly studied as part of the pathophysiology of neurodegenerative diseases. Mammalian Ste20-like kinase 1 (Mst1), a key factor of the Hippo pathway, is connected to cell death. Unfortunately, little study has been performed to detect the impact of Mst1 in neuroninflammation. The results indicated that Mst1 expression was upregulated because of LPS treatment. However, the loss of Mst1 sustained BV-2 cell viability and promoted cell survival in the presence of LPS treatment. Molecular investigation assay demonstrated that Mst1 deletion was followed by a drop in the levels of mitochondrial fission via repressing Drp1 expression. However, Drp1 adenovirus transfection reduced the protective impacts of Mst1 knockdown on mitochondrial stress and neuronal dysfunction. Finally, our results illuminated that Mst1 affected Drp1 content and mitochondrial fission in a JNK-dependent mechanism. Reactivation of the JNK axis inhibited Mst1 knockdown-mediated neuronal protection and mitochondrial homeostasis. Altogether, our results indicated that Mst1 upregulation and the activation of JNK-Drp1-mitochondrial fission pathway could be considered as the novel mechanism regulating the progression of neuroninflammation. This finding would pave a new road for the treatment of neurodegenerative diseases via modulating the Mst1-JNK-Drp1-mitochondrial fission axis.
Collapse
Affiliation(s)
- Hong Tian
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Kang Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Miao Jin
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Jingtao Li
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| | - Yanbing Yu
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
26
|
Yao W, Zhu S, Li P, Zhang S. Large tumor suppressor kinase 2 overexpression attenuates 5-FU-resistance in colorectal cancer via activating the JNK-MIEF1-mitochondrial division pathway. Cancer Cell Int 2019; 19:97. [PMID: 31011291 PMCID: PMC6460675 DOI: 10.1186/s12935-019-0812-3] [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] [Academic Contribution Register] [Received: 02/17/2019] [Accepted: 04/01/2019] [Indexed: 02/06/2023] Open
Abstract
Background 5-Fluorouracil (5-FU) is a standard treatment for colorectal cancer, but most patients develop 5-FU resistance. Here, we conducted experiments to identify an effective approach to augment 5-FU-based treatment in colorectal cancer in vitro. Methods SW480 cells were in the present study and treated with 5-FU. Besides, LATS2 adenovirus vectors were infected into SW480 cells. Western blotting, immunofluorescence and ELISA were used to evaluate cell death and mitochondrial function. Pathway blocker was used to verify the role of MAPK-JNK pathway in SW480 cell death. Results An obvious drop in large tumor suppressor kinase 2 (LATS2) expression was observed in SW480 cells after treatment with 5-FU. In addition, upregulation of LATS2 expression through infection with LATS2 adenovirus further increased the reduction of SW480 cell viability induced by 5-FU. Functional exploration showed that 5-FU treatment suppressed mitochondrial membrane potential, enhanced cyt-c release into the nucleus, induced an oxidative injury environment by promoting ROS production, and eventually upregulated Bax-related mitochondrial apoptosis. Besides, LATS2 overexpression in combination with 5-FU treatment further perturbed mitochondrial homeostasis, and this effect was achieved by elevating mitochondrial division. Mechanistically, LATS2 overexpression and 5-FU co-treatment amplified mitochondrial division by upregulating MIEF1 expression in a manner dependent on MAPK-JNK axis. Knockdown of MIEF1 using an siRNA-mediated loss of function assay and/or inhibition of the MAPK-JNK pathway using the specific inhibitor SP600125 abolished LATS2/5-FU-mediated deleterious effects on mitochondrial performance and SW480 cell viability. Conclusions In light of the above findings, LATS2 downregulation could be a potential mechanism of low response to 5-FU treatment. Overexpression of LATS2 to further disrupt mitochondrial function via the JNK-MIEF1 signalling pathway might be a method to optimize 5-FU-based chemotherapy.
Collapse
Affiliation(s)
- Weilong Yao
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, No. 95, Yong'an Road, Xicheng District, Beijing, 100050 People's Republic of China
| | - Shengtao Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, No. 95, Yong'an Road, Xicheng District, Beijing, 100050 People's Republic of China
| | - Peng Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, No. 95, Yong'an Road, Xicheng District, Beijing, 100050 People's Republic of China
| | - Shutian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, No. 95, Yong'an Road, Xicheng District, Beijing, 100050 People's Republic of China
| |
Collapse
|
27
|
Dual blockage of STAT3 and ERK1/2 eliminates radioresistant GBM cells. Redox Biol 2019; 24:101189. [PMID: 30986607 PMCID: PMC6463934 DOI: 10.1016/j.redox.2019.101189] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/18/2019] [Revised: 03/17/2019] [Accepted: 04/01/2019] [Indexed: 01/05/2023] Open
Abstract
Radiotherapy (RT) is the major modality for control of glioblastoma multiforme (GBM), the most aggressive brain tumor in adults with poor prognosis and low patient survival rate. To improve the RT efficacy on GBM, the mechanism causing tumor adaptive radioresistance which leads to the failure of tumor control and lethal progression needs to be further elucidated. Here, we conducted a comparative analysis of RT-treated recurrent tumors versus primary counterparts in GBM patients, RT-treated orthotopic GBM tumors xenografts versus untreated tumors and radioresistant GBM cells versus wild type cells. The results reveal that activation of STAT3, a well-defined redox-sensitive transcriptional factor, is causally linked with GBM adaptive radioresistance. Database analysis also agrees with the worse prognosis in GBM patients due to the STAT3 expression-associated low RT responsiveness. However, although the radioresistant GBM cells can be resensitized by inhibition of STAT3, a fraction of radioresistant cells can still survive the RT combined with STAT3 inhibition or CRISPR/Cas9-mediated STAT3 knockout. A complementally enhanced activation of ERK1/2 by STAT3 inhibition is identified responsible for the survival of the remaining resistant tumor cells. Dual inhibition of ERK1/2 and STAT3 remarkably eliminates resistant GBM cells and inhibits tumor regrowth. These findings demonstrate a previously unknown feature ofSTAT3-mediated ERK1/2 regulation and an effective combination of two targets in resensitizing GBM to RT.
Collapse
|
28
|
Song J, Zhao W, Lu C, Shao X. LATS2 overexpression attenuates the therapeutic resistance of liver cancer HepG2 cells to sorafenib-mediated death via inhibiting the AMPK-Mfn2 signaling pathway. Cancer Cell Int 2019; 19:60. [PMID: 30923462 PMCID: PMC6423758 DOI: 10.1186/s12935-019-0778-1] [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] [Academic Contribution Register] [Received: 02/08/2019] [Accepted: 03/08/2019] [Indexed: 12/21/2022] Open
Abstract
Background Effective therapy for hepatocellular carcinoma (HCC) is currently an imperative issue, and sorafenib is a first-line drug for the treatment of HCC. However, the clinical benefit of sorafenib is often impaired by drug resistance. Accordingly, the present study was conducted to investigate the molecular mechanisms involving sorafenib resistance, with a focus on large tumor suppressor 2 (LATS2) and mitophagy. Methods HepG2 liver cancer cells were treated with sorafenib and infected with adenovirus-loaded LATS2 (Ad-LATS2). Cell death, proliferation and migration were measured via western blotting analysis, immunofluorescence and qPCR. Mitochondrial function and mitophagy were determined via western blotting and immunofluorescence. Results Our data indicated that LATS2 expression was repressed by sorafenib treatment, and overexpression of LATS2 could further enhance sorafenib-mediated apoptosis in HepG2 liver cancer cells. At the molecular level, mitochondrial stress was triggered by sorafenib treatment, as evidenced by decreased mitochondrial membrane potential, increased mitochondrial ROS production, more cyc-c release into the nucleus, and elevated mitochondrial pro-apoptotic proteins. However, in response to mitochondrial damage, mitophagy was activated by sorafenib treatment, whereas LATS2 overexpression effectively inhibited mitophagy activity and thus augmented sorafenib-mediated mitochondrial stress. Subsequently, we also demonstrated that the AMPK–MFN2 signaling pathway was involved in mitophagy regulation after exposure to sorafenib treatment and/or LATS2 overexpression. Inhibition of the AMPK pathway interrupted mitophagy and thus enhanced the antitumor property of sorafenib, similar to the results obtained via overexpression of LATS2. Conclusions Altogether, our findings revealed the importance of the LATS2/AMPK/MFN2/mitophagy axis in understanding sorafenib resistance mechanisms, with a potential application to increase the sensitivity response of sorafenib in the treatment of liver cancer.
Collapse
Affiliation(s)
- Jie Song
- 1Department of Hepatopancreatobiliary Medicine, The Second Hospital of Jilin University, Changchun, 130000 China
| | - Wei Zhao
- 2Department of Pharmacy, The Second Hospital of Jilin University, Changchun, 130000 China
| | - Chang Lu
- 3Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, 130000 China
| | - Xue Shao
- 1Department of Hepatopancreatobiliary Medicine, The Second Hospital of Jilin University, Changchun, 130000 China
| |
Collapse
|
29
|
Paul S, Gangwar A, Bhargava K, Khurana P, Ahmad Y. Diagnosis and prophylaxis for high-altitude acclimatization: Adherence to molecular rationale to evade high-altitude illnesses. Life Sci 2018; 203:171-176. [PMID: 29698652 DOI: 10.1016/j.lfs.2018.04.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/11/2018] [Revised: 04/13/2018] [Accepted: 04/21/2018] [Indexed: 11/25/2022]
Abstract
Lack of zero side-effect, prescription-less prophylactics and diagnostic markers of acclimatization status lead to many suffering from high altitude illnesses. Although not fully translated to the clinical setting, many strategies and interventions are being developed that are aimed at providing an objective and tangible answer regarding the acclimatization status of an individual as well as zero side-effect prophylaxis that is cost-effective and does not require medical supervision. This short review brings together the twin problems associated with high-altitude acclimatization, i.e. acclimatization status and zero side-effect, easy-to-use prophylaxis, for the reader to comprehend as cogs of the same phenomenon. We describe current research aimed at preventing all the high-altitude illnesses by considering them an assault on redox and energy homeostasis at the molecular level. This review also entails some proteins capable of diagnosing either acclimatization or high-altitude illnesses. The future strategies based on bioinformatics and systems biology is also discussed.
Collapse
Affiliation(s)
- Subhojit Paul
- Peptide & Proteomics Division, Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India
| | - Anamika Gangwar
- Peptide & Proteomics Division, Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India
| | - Kalpana Bhargava
- Peptide & Proteomics Division, Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India
| | - Pankaj Khurana
- Peptide & Proteomics Division, Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India
| | - Yasmin Ahmad
- Peptide & Proteomics Division, Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India.
| |
Collapse
|