1
|
Sadeghloo Z, Nabavi-Rad A, Zali MR, Klionsky DJ, Yadegar A. The interplay between probiotics and host autophagy: mechanisms of action and emerging insights. Autophagy 2024:1-23. [PMID: 39291740 DOI: 10.1080/15548627.2024.2403277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 09/19/2024] Open
Abstract
Autophagy, a lysosome-dependent protein degradation mechanism, is a highly conserved catabolic process seen in all eukaryotes. This cell protection system, which is present in all tissues and functions at a basic level, can be up- or downregulated in response to various stresses. A disruption in the natural route of the autophagy process is frequently followed by an interruption in the inherent operation of the body's cells and organs. Probiotics are live bacteria that protect the host through various mechanisms. One of the processes through which probiotics exert their beneficial effects on various cells and tissues is autophagy. Autophagy can assist in maintaining host homeostasis by stimulating the immune system and affecting numerous physiological and pathological responses. In this review, we particularly focus on autophagy impairments occurring in several human illnesses and investigate how probiotics affect the autophagy process under various circumstances.Abbreviation: AD: Alzheimer disease; AKT: AKT serine/threonine kinase; AMPK: 5'AMP-activated protein kinase; ATG: autophagy related; CCl4: carbon tetrachloride; CFS: cell-free supernatant; CMA: chaperone-mediated autophagy; CRC: colorectal cancer; EPS: L. plantarum H31 exopolysaccharide; HD: Huntington disease; HFD: high-fat diet; HPV: human papillomavirus; IFNG/IFN-γ: interferon gamma; IL6: interleukin 6; LGG: L. rhamnosus GG; LPS: lipopolysaccharide; MTOR: mechanistic target of rapamycin kinase; MTORC1: MTOR complex 1; NAFLD: non-alcoholic fatty liver disease; NASH: non-alcoholic steatohepatitis; PD: Parkinson disease; Pg3G: pelargonidin-3-O-glucoside; PI3K: phosphoinositide 3-kinase; PolyQ: polyglutamine; ROS: reactive oxygen species; SCFAs: short-chain fatty acids; SLAB51: a novel formulation of lactic acid bacteria and bifidobacteria; Slp: surface layer protein (of acidophilus NCFM); SNCA: synuclein alpha; ULK1: unc-51 like autophagy-activating kinase 1; YB: B. longum subsp. infantis YB0411; YFP: yeast fermentate prebiotic.
Collapse
Affiliation(s)
- Zahra Sadeghloo
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Nabavi-Rad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Daniel J Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Xu Z, Liu Q, Li J, Wang J, Yang Z, Wang J, Gao L, Cheng J, He J, Dong Y, Guo X, Cui J, Zhang W. AMPKα is active in autophagy of endothelial cells in arsenic-induced vascular endothelial dysfunction by regulating mTORC1/p70S6K/ULK1. Chem Biol Interact 2024; 388:110832. [PMID: 38101599 DOI: 10.1016/j.cbi.2023.110832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 12/01/2023] [Accepted: 12/11/2023] [Indexed: 12/17/2023]
Abstract
Cardiovascular disease (CVD) is the most common cause of death, environmental factors, such as arsenic, playing an important role in the progress of CVD. Vascular endothelial dysfunction (VED) is a crucial early feature for CVD, inorganic arsenic (iAs) can induce autophagy in various cells. However, the role of endothelial autophagy has rarely been studied in VED triggered by arsenic. Total of one hundred and twenty healthy male C57BL/6J mice weighing 18-22 g were randomly divided into an arsenic-exposure group and a control group for 3, 6, 9, and 12 weeks. The results showed that, independent of the exposure period, autophagy markers of p-ATG16L1 levels and Beclin 1 contents in the aortic arch endothelium increased significantly compared with those of the corresponding control group. And different exposure duration decreased NO contents in the serum significantly. Combined with the histological changes that endothelial injury aggravated gradually with the increasing exposure period, suggesting that under exposure to iAs over 9 weeks, VED was remarkably induced, and consistant high levels of endothelial autophagy may play an important role. Additionally, levels of p-AMPKα/AMPKα increased significantly and p-mTORC1/mTORC1 levels decreased remarkably in the aortic arch endothelium. Then, a NaAsO2-induced-VED in vitro model was used to explore the mechanism of arsenic-induced endothelial autophagy. Similarly, p-AMPKα/AMPKα level significantly increased, and p-mTORC1/mTORC1 level remarkably decreased induced by 30 μmol/L NaAsO2 in HUAECs. Further, an AMPK inhibitor (Compound C) pre-treatment prior to arsenic exposure reversed the increased autophagy level, and alleviated the endothelial dysfunction in HUVECs, as shown by the significant increase in the intracellular NO content and the cell vitality. Mechanistically, we revealed that AMPKα is active in autophagy of endothelial cells in arsenic-induced VED by regulating mTORC1/p70S6K/ULK1. The present study provide a new promising target for prevention and control arsenic-associated CVD.
Collapse
Affiliation(s)
- Ziqi Xu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, 150081, China
| | - Qiaoling Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, 150081, China
| | - Jinyu Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, 150081, China
| | - Jingqiu Wang
- Institute for Prevention and Treatment of Sexually Transmitted Disease and AIDS, Center for Disease Control and Prevention of Hebei Province, Shijiazhuang, 050021, China
| | - Zhihan Yang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, 150081, China
| | - Juan Wang
- Department of Reproductive Medicine, Affiliated Hospital of Jining Medical College, Jining, 272000, China
| | - Lin Gao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, 150081, China
| | - Jin Cheng
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, 150081, China
| | - Jing He
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, 150081, China
| | - Yishan Dong
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, 150081, China
| | - Xiangnan Guo
- Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Jing Cui
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, 150081, China
| | - Wei Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, 150081, China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin, 150081, China.
| |
Collapse
|
3
|
Zhang R, Yang Y, He C, Zhang X, Torraca V, Wang S, Liu N, Yang J, Liu S, Yuan J, Gou D, Li S, Dong X, Xie Y, He J, Bai H, Hu M, Liao Z, Huang Y, Lyu H, Xiao S, Guo D, Ali DW, Michalak M, Ma C, Chen XZ, Tang J, Zhou C. RUNDC1 inhibits autolysosome formation and survival of zebrafish via clasping ATG14-STX17-SNAP29 complex. Cell Death Differ 2023; 30:2231-2248. [PMID: 37684417 PMCID: PMC10589263 DOI: 10.1038/s41418-023-01215-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 07/28/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
Autophagy serves as a pro-survival mechanism for a cell or a whole organism to cope with nutrient stress. Our understanding of the molecular regulation of this fusion event remains incomplete. Here, we identified RUNDC1 as a novel ATG14-interacting protein, which is highly conserved across vertebrates, including zebrafish and humans. By gain and loss of function studies, we demonstrate that RUNDC1 negatively modulates autophagy by blocking fusion between autophagosomes and lysosomes via inhibiting the assembly of the STX17-SNAP29-VAMP8 complex both in human cells and the zebrafish model. Moreover, RUNDC1 clasps the ATG14-STX17-SNAP29 complex via stimulating ATG14 homo-oligomerization to inhibit ATG14 dissociation. This also prevents VAMP8 from binding to STX17-SNAP29. We further identified that phosphorylation of RUNDC1 Ser379 is crucial to inhibit the assembly of the STX17-SNAP29-VAMP8 complex via promoting ATG14 homo-oligomerization. In line with our findings, RunDC1 is crucial for zebrafish in their response to nutrient-deficient conditions. Taken together, our findings demonstrate that RUNDC1 is a negative regulator of autophagy that restricts autophagosome fusion with lysosomes by clasping the ATG14-STX17-SNAP29 complex to hinder VAMP8 binding.
Collapse
Affiliation(s)
- Rui Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Yuyan Yang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Chao He
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Xin Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Vincenzo Torraca
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK
- School of Life Sciences, University of Westminster, London, UK
| | - Shen Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and the Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Nan Liu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Jiaren Yang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Shicheng Liu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Jinglei Yuan
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Dongzhi Gou
- School of Public Health, Chongqing Medical University, Chongqing, China
| | - Shi Li
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Xueying Dong
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Yufei Xie
- School of food science and bioengineering, Changsha University of Science & Technology, Changsha, China
| | - Junling He
- Department of Nephrology, Daping hospital, Army Medical Center, Army Medical University, Chongqing, China
| | - Hua Bai
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Mengyu Hu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Zhiquan Liao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Yuan Huang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Hao Lyu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Shuai Xiao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Dong Guo
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | | | - Marek Michalak
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Cong Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and the Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Xing-Zhen Chen
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China.
| | - Cefan Zhou
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China.
| |
Collapse
|
4
|
Yang YP, Lai WY, Lin TW, Lin YY, Chien Y, Tsai YC, Tai HY, Wang CL, Liu YY, Huang PI, Chen YW, Lo WL, Wang CY. Autophagy reprogramming stem cell pluripotency and multiple-lineage differentiation. J Chin Med Assoc 2022; 85:667-671. [PMID: 35385421 DOI: 10.1097/jcma.0000000000000728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The cellular process responsible for the degradation of cytosolic proteins and subcellular organelles in lysosomes was termed "autophagy." This process occurs at a basal level in most tissues as part of tissue homeostasis that redounds to the regular turnover of components inside cytoplasm. The breakthrough in the autophagy field is the identification of key players in the autophagy pathway, compounded under the name "autophagy-related genes" (ATG) encoding for autophagy effector proteins. Generally, the function of autophagy can be classified into two divisions: intracellular clearance of defective macromolecules and organelles and generation of degradation products. Therapeutic strategies using stem cell-based approach come as a promising therapy and develop rapidly recently as stem cells have high self-renewability and differentiation capability as known as mesenchymal stem cells (MSCs). They are defined as adherent fibroblast-like population with the abilities to self-renew and multi-lineage differentiate into osteogenic, adipogenic, and chondrogenic lineage cells. To date, they are the most extensively applied adult stem cells in clinical trials. The properties of MSCs, such as immunomodulation, neuroprotection, and tissue repair pertaining to cell differentiation, processes to replace lost, or damaged cells, for aiding cell repair and revival. Autophagy has been viewed as a remarkable mechanism for maintaining homeostasis, ensuring the adequate function and survival of long-lived stem cells. In addition, authophagy also plays a remarkable role in protecting stem cells against cellular stress when the stem cell regenerative capacity is harmed in aging and cellular degeneration. Understanding the under-explored mechanisms of MSC actions and expanding the spectrum of their clinical applications may improve the utility of the MSC-based therapeutic approach in the future.
Collapse
Affiliation(s)
- Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Wei-Yi Lai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Tzu-Wei Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yi-Ying Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yueh Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yi-Ching Tsai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Hsiao-Yun Tai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Chia-Lin Wang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yung-Yang Liu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Pin-I Huang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Cancer Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yi-Wei Chen
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Cancer Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Wen-Liang Lo
- Institute of Oral Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Division of Oral and Maxillofacial Surgery, Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Dentistry, School of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Chien-Ying Wang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Division of Trauma, Department of Emergency Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Physical Education and Health, University of Taipei, Taipei, Taiwan, ROC
| |
Collapse
|
5
|
Zheng LY, Zou X, Wang YL, Zou M, Ma F, Wang N, Li JW, Wang MS, Hung HY, Wang Q. Betulinic acid-nucleoside hybrid prevents acute alcohol -induced liver damage by promoting anti-oxidative stress and autophagy. Eur J Pharmacol 2022; 914:174686. [PMID: 34883073 DOI: 10.1016/j.ejphar.2021.174686] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/03/2021] [Accepted: 12/03/2021] [Indexed: 12/18/2022]
Abstract
Alcoholic abuse is one of the most serious causes of liver diseases worldwide. Although detailed molecular pathogenesis of alcohol-induced liver damages remains elusive with intensive debates, it has been widely recognized that hepatic damage caused by free radicals generated from alcohol metabolism is one of the most critical factors for alcohol-induced liver diseases. Betulinic acid is a potent antioxidant with additional known pharmacological safety characteristics and minimal toxicity. However, poor solubility limited its usage. In this study, we assessed the efficacy of BAN, a betulinic acid and nucleoside hybrid with good water solubility, in reversing acute liver damages using an established alcohol overdose animal model. The results indicated that BAN is an extremely promising therapeutic agent against acute alcohol-induced liver damage. BAN effectively protects liver from alcohol damage by reducing serum ALT level by up to 47%, as well as liver oxidative stress indicated by significantly increased SOD, CAT, and GSH-Px levels. Moreover, hepatic FXR activation and a corresponding downstream anti-oxidative stress transcriptional cascade including Nrf2, HO-1, and NOQ1 induce the protective role of BAN. On the other hand, BAN administration also leads to increase cellular autophagy response, as indicated by the key ATG protein activation. We concluded that BAN, comparing with Betulinic acid, prevents acute alcohol-induced liver damages more effectively, with the dual mechanisms of neutralizing oxidative stress and promoting autophagy.
Collapse
Affiliation(s)
- Li-Yun Zheng
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Pharmacology of Liver Diseases, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou, China
| | - Xi Zou
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Pharmacology of Liver Diseases, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou, China
| | - Yan-Li Wang
- National Health Commission Key Laboratory of Birth Defect Prevention, Henan Institute of Reproductive Health Science and Technology, Zhengzhou, China
| | - Min Zou
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Pharmacology of Liver Diseases, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou, China
| | - Fang Ma
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Pharmacology of Liver Diseases, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou, China
| | - Ning Wang
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Pharmacology of Liver Diseases, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou, China
| | - Jia-Wen Li
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Pharmacology of Liver Diseases, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou, China
| | - Ming-Sheng Wang
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China; Henan Key Laboratory for Pharmacology of Liver Diseases, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou, China
| | - Hsin-Yi Hung
- School of Pharmacy, College of Medicine, National Cheng Kung University, 701, Tainan, Taiwan.
| | - Qiang Wang
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China; National Health Commission Key Laboratory of Birth Defect Prevention, Henan Institute of Reproductive Health Science and Technology, Zhengzhou, China; High& New Technology Research Center, Henan Academy of Sciences, Zhengzhou, China.
| |
Collapse
|
6
|
Yang T, Guo R, Ofengeim D, Hwang JY, Zukin RS, Chen J, Zhang F. Molecular and Cellular Mechanisms of Ischemia-Induced Neuronal Death. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00005-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
7
|
Barati M, Darvishi B, Javidi MA, Mohammadian A, Shariatpanahi SP, Eisavand MR, Madjid Ansari A. Cellular stress response to extremely low-frequency electromagnetic fields (ELF-EMF): An explanation for controversial effects of ELF-EMF on apoptosis. Cell Prolif 2021; 54:e13154. [PMID: 34741480 PMCID: PMC8666288 DOI: 10.1111/cpr.13154] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/21/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Impaired apoptosis is one of the hallmarks of cancer, and almost all of the non‐surgical approaches of eradicating tumour cells somehow promote induction of apoptosis. Indeed, numerous studies have stated that non‐ionizing non‐thermal extremely low‐frequency magnetic fields (ELF‐MF) can modulate the induction of apoptosis in exposed cells; however, much controversy exists in observations. When cells are exposed to ELF‐EMF alone, very low or no statistically significant changes in apoptosis are observed. Contrarily, exposure to ELF‐EMF in the presence of a co‐stressor, including a chemotherapeutic agent or ionizing radiation, can either potentiate or inhibit apoptotic effects of the co‐stressor. In our idea, the main point neglected in interpreting these discrepancies is “the cellular stress responses” of cells following ELF‐EMF exposure and its interplay with apoptosis. The main purpose of the current review was to outline the triangle of ELF‐EMF, the cellular stress response of cells and apoptosis and to interpret and unify discrepancies in results based on it. Therefore, initially, we will describe studies performed on identifying the effect of ELF‐EMF on induction/inhibition of apoptosis and enumerate proposed pathways through which ELF‐EMF exposure may affect apoptosis; then, we will explain cellular stress response and cues for its induction in response to ELF‐EMF exposure; and finally, we will explain why such controversies have been observed by different investigators.
Collapse
Affiliation(s)
- Mojdeh Barati
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Behrad Darvishi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mohammad Amin Javidi
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Ali Mohammadian
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Mohammad Reza Eisavand
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Alireza Madjid Ansari
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| |
Collapse
|
8
|
Fang C, Woo JAA, Liu T, Zhao X, Cazzaro S, Yan Y, Matlack J, Kee T, LePochat P, Kang DE. SSH1 impedes SQSTM1/p62 flux and MAPT/Tau clearance independent of CFL (cofilin) activation. Autophagy 2021; 17:2144-2165. [PMID: 33044112 PMCID: PMC8496729 DOI: 10.1080/15548627.2020.1816663] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 01/08/2023] Open
Abstract
Accumulation of toxic protein assemblies and damaged mitochondria are key features of neurodegenerative diseases, which arise in large part from clearance defects in the Macroautophagy/autophagy-lysosome system. The autophagy cargo receptor SQSTM1/p62 plays a major role in the clearance of ubiquitinated cargo through Ser403 phosphorylation by multiple kinases. However, no phosphatase is known to physiologically dephosphorylate SQSTM1 on this activating residue. RNAi-mediated knockdown and overexpression experiments using genetically encoded fluorescent reporters and defined mutant constructs in cell lines, primary neurons, and brains show that SSH1, the canonical CFL (cofilin) phosphatase, mediates the dephosphorylation of phospho-Ser403-SQSTM1, thereby impairing SQSTM1 flux and phospho-MAPT/tau clearance. The inhibitory action of SSH1 on SQSTM1 is fully dependent on SQSTM1 Ser403 phosphorylation status and is separable from SSH1-mediated CFL activation. These findings reveal a unique action of SSH1 on SQSTM1 independent of CFL and implicate an inhibitory role of SSH1 in SQSTM1-mediated clearance of autophagic cargo, including phospho-MAPT/tau. Abbreviations: AAV: adeno-associated virus; Aβ42O: amyloid β1-42 oligomers; AD: Alzheimer disease; CA3: cornu Ammonis 3; CSNK2/CK2: casein kinase 2; FCCP: 2-[2-[4-(trifluoromethoxy)phenyl]hydrazinylidene]-propanedinitrile; FTLD: frontotemporal lobar degeneration; GFP: green fluorescent protein; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; SQSTM1/p62: sequestosome-1; PLA: proximity ligation assay; RFP: red fluorescent protein; RIPA: radioimmunoprecipitation assay; shRNA: short hairpin RNA; siRNA: small interfering RNA; Ser403: Serine403; SSH1: slingshot protein phosphatase 1; TBK1: TANK-binding kinase 1; ULK: unc-51 like kinase 1.
Collapse
Affiliation(s)
- Cenxiao Fang
- USF Health Byrd Institute & Alzheimer Center, Tampa, FL, USA
- Department of Molecular of Medicine, USF Health Morsani College of Medicine, Tampa, FL, USA
| | - Jung-A A. Woo
- USF Health Byrd Institute & Alzheimer Center, Tampa, FL, USA
- Department of Molecular Pharmacology and Physiology, USF Health Morsani College of Medicine, Tampa, FL, USA
| | - Tian Liu
- USF Health Byrd Institute & Alzheimer Center, Tampa, FL, USA
- Department of Molecular of Medicine, USF Health Morsani College of Medicine, Tampa, FL, USA
| | - Xingyu Zhao
- USF Health Byrd Institute & Alzheimer Center, Tampa, FL, USA
- Department of Molecular of Medicine, USF Health Morsani College of Medicine, Tampa, FL, USA
| | - Sara Cazzaro
- USF Health Byrd Institute & Alzheimer Center, Tampa, FL, USA
- Department of Molecular of Medicine, USF Health Morsani College of Medicine, Tampa, FL, USA
| | - Yan Yan
- USF Health Byrd Institute & Alzheimer Center, Tampa, FL, USA
- Department of Molecular of Medicine, USF Health Morsani College of Medicine, Tampa, FL, USA
| | - Jenet Matlack
- USF Health Byrd Institute & Alzheimer Center, Tampa, FL, USA
- Department of Molecular of Medicine, USF Health Morsani College of Medicine, Tampa, FL, USA
| | - Teresa Kee
- USF Health Byrd Institute & Alzheimer Center, Tampa, FL, USA
- Department of Molecular of Medicine, USF Health Morsani College of Medicine, Tampa, FL, USA
| | - Patrick LePochat
- USF Health Byrd Institute & Alzheimer Center, Tampa, FL, USA
- Department of Molecular of Medicine, USF Health Morsani College of Medicine, Tampa, FL, USA
| | - David E. Kang
- USF Health Byrd Institute & Alzheimer Center, Tampa, FL, USA
- Department of Molecular of Medicine, USF Health Morsani College of Medicine, Tampa, FL, USA
- Department of Research Service, James A. Haley Veterans Administration Hospital, Tampa, FL, USA
| |
Collapse
|
9
|
Sun X, Alford J, Qiu H. Structural and Functional Remodeling of Mitochondria in Cardiac Diseases. Int J Mol Sci 2021; 22:ijms22084167. [PMID: 33920673 PMCID: PMC8072869 DOI: 10.3390/ijms22084167] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 12/31/2022] Open
Abstract
Mitochondria undergo structural and functional remodeling to meet the cell demand in response to the intracellular and extracellular stimulations, playing an essential role in maintaining normal cellular function. Merging evidence demonstrated that dysregulation of mitochondrial remodeling is a fundamental driving force of complex human diseases, highlighting its crucial pathophysiological roles and therapeutic potential. In this review, we outlined the progress of the molecular basis of mitochondrial structural and functional remodeling and their regulatory network. In particular, we summarized the latest evidence of the fundamental association of impaired mitochondrial remodeling in developing diverse cardiac diseases and the underlying mechanisms. We also explored the therapeutic potential related to mitochondrial remodeling and future research direction. This updated information would improve our knowledge of mitochondrial biology and cardiac diseases’ pathogenesis, which would inspire new potential strategies for treating these diseases by targeting mitochondria remodeling.
Collapse
Affiliation(s)
| | | | - Hongyu Qiu
- Correspondence: ; Tel.: +404-413-3371; Fax: +404-413-9566
| |
Collapse
|
10
|
Geng X, Wang F, Tian D, Huang L, Streator E, Zhu J, Kurihara H, He R, Yao X, Zhang Y, Tang J. Cardiac glycosides inhibit cancer through Na/K-ATPase-dependent cell death induction. Biochem Pharmacol 2020; 182:114226. [PMID: 32976831 DOI: 10.1016/j.bcp.2020.114226] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023]
Abstract
Successful drug repurposing relies on the understanding of molecular mechanisms of the target compound. Cardiac glycosides have demonstrated potent anticancer activities; however, the pharmacological mechanisms underlying their anticancer effects remained elusive, which has restricted their further development in cancer treatment. A bottleneck is the lack of comprehensive understanding about genes and signaling pathways that are altered at the early stage of drug treatment, which is key to understand how they inhibit cancer. To address this issue, we first investigated the anticancer effects of a panel of 68 naturally isolated cardiac glycosides. Our results illustrate critical structure activity relationship of these compounds on cancer cell survival. We confirmed the anticancer effect of cardiac glycoside in mouse tumor xenografts. Through RNA sequencing, quantitative PCR and immunoblotting, we show that cardiac glycoside first activated autophagy and then induced apoptosis. Further activating autophagy by rapamycin or inhibiting apoptosis by caspase inhibitor mitigated cardiac glycoside-induced cell death, whereas inhibiting autophagy by RNA interference-mediated depletion of critical autophagy genes enhanced cell death. While depletion of Na/K-ATPase, the protein target of cardiac glycosides, by RNA interference inhibited both autophagy activation and apoptosis induction by cardiac glycoside, expression of human, but not rodent Na/K-ATPase, increased cell sensitivity to cardiac glycoside. In conclusion, our analyses reveal sequential activation of autophagy and apoptosis during early stages of cardiac glycoside treatment and indicate the importance of Na/K-ATPase in their anticancer effects.
Collapse
Affiliation(s)
- Xinran Geng
- Department of Pharmacology, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, People's Republic of China
| | - Fangfang Wang
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, People's Republic of China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drug Research, Jinan University, Guangzhou 510632, People's Republic of China
| | - Danmei Tian
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, People's Republic of China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drug Research, Jinan University, Guangzhou 510632, People's Republic of China
| | - Lihua Huang
- International Academic Support & Delivery Unit, BGI Genomics, Co., Ltd., Shenzhen 518083, People's Republic of China
| | - Evan Streator
- Department of Pharmacology, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Jingjing Zhu
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, People's Republic of China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drug Research, Jinan University, Guangzhou 510632, People's Republic of China
| | - Hiroshi Kurihara
- Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou, Guangdong 510632, People's Republic of China
| | - Rongrong He
- Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou, Guangdong 510632, People's Republic of China
| | - Xinsheng Yao
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, People's Republic of China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drug Research, Jinan University, Guangzhou 510632, People's Republic of China
| | - Youwei Zhang
- Department of Pharmacology, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Jinshan Tang
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, People's Republic of China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drug Research, Jinan University, Guangzhou 510632, People's Republic of China.
| |
Collapse
|
11
|
Li XY, Tan LC, Dong LW, Zhang WQ, Shen XX, Lu X, Zheng H, Lu YG. Susceptibility and Resistance Mechanisms During Photodynamic Therapy of Melanoma. Front Oncol 2020; 10:597. [PMID: 32528867 PMCID: PMC7247862 DOI: 10.3389/fonc.2020.00597] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 04/01/2020] [Indexed: 12/19/2022] Open
Abstract
Melanoma is the most aggressive malignant skin tumor and arises from melanocytes. The resistance of melanoma cells to various treatments results in rapid tumor growth and high mortality. As a local therapeutic modality, photodynamic therapy has been successfully applied for clinical treatment of skin diseases. Photodynamic therapy is a relatively new treatment method for various types of malignant tumors in humans and, compared to conventional treatment methods, has fewer side effects, and is more accurate and non-invasive. Although several in vivo and in vitro studies have shown encouraging results regarding the potential benefits of photodynamic therapy as an adjuvant treatment for melanoma, its clinical application remains limited owing to its relative inefficiency. This review article discusses the use of photodynamic therapy in melanoma treatment as well as the latest progress made in deciphering the mechanism of tolerance. Lastly, potential targets are identified that may improve photodynamic therapy against melanoma cells.
Collapse
Affiliation(s)
- Xin-Ying Li
- Department of Plastic Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Liu-Chang Tan
- Department of Plastic Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Li-Wen Dong
- Department of Plastic Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Wan-Qi Zhang
- Department of Plastic Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiao-Xiao Shen
- Department of Plastic Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiao Lu
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Hong Zheng
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yuan-Gang Lu
- Department of Plastic Surgery, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
12
|
Molecular mechanisms and systemic targeting of NRF2 dysregulation in cancer. Biochem Pharmacol 2020; 177:114002. [PMID: 32360363 DOI: 10.1016/j.bcp.2020.114002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 04/24/2020] [Indexed: 12/15/2022]
Abstract
NF-E2-related factor 2 (NRF2) is a master regulator of redox homeostasis and provides cellular protection against oxidants and electrophiles by inducing the expression of a wide array of phase II cytoprotective genes. Until now, a number of NRF2 activators have been developed for treatment of chronic diseases and some are under evaluation in the clinical studies. On the other hand, accumulating evidence indicates that NRF2 confers chemoresistance and radioresistance, and its expression is correlated with poor prognosis in cancer patients. Studies in the last decade demonstrate that diverse mechanisms such as somatic mutations, accumulation of KEAP1 binding proteins, transcriptional dysregulation, oncogene activation, and accumulation of reactive metabolites contribute to NRF2 activation in cancer. In the present review, we illustrate the molecular mechanisms governing the function of NRF2 and explain how they are hijacked in cancer. We also provide some examples of NRF2 inhibitors together with a brief explanation of their mechanisms of action.
Collapse
|
13
|
Alpha 1-Antitrypsin Deficiency: A Disorder of Proteostasis-Mediated Protein Folding and Trafficking Pathways. Int J Mol Sci 2020; 21:ijms21041493. [PMID: 32098273 PMCID: PMC7073043 DOI: 10.3390/ijms21041493] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 12/30/2022] Open
Abstract
Human cells express large amounts of different proteins continuously that must fold into well-defined structures that need to remain correctly folded and assemble in order to ensure their cellular and biological functions. The integrity of this protein balance/homeostasis, also named proteostasis, is maintained by the proteostasis network (PN). This integrated biological system, which comprises about 2000 proteins (chaperones, folding enzymes, degradation components), control and coordinate protein synthesis folding and localization, conformational maintenance, and degradation. This network is particularly challenged by mutations such as those found in genetic diseases, because of the inability of an altered peptide sequence to properly engage PN components that trigger misfolding and loss of function. Thus, deletions found in the ΔF508 variant of the Cystic Fibrosis (CF) transmembrane regulator (CFTR) triggering CF or missense mutations found in the Z variant of Alpha 1-Antitrypsin deficiency (AATD), leading to lung and liver diseases, can accelerate misfolding and/or generate aggregates. Conversely to CF variants, for which three correctors are already approved (ivacaftor, lumacaftor/ivacaftor, and most recently tezacaftor/ivacaftor), there are limited therapeutic options for AATD. Therefore, a more detailed understanding of the PN components governing AAT variant biogenesis and their manipulation by pharmacological intervention could delay, or even better, avoid the onset of AATD-related pathologies.
Collapse
|
14
|
Liu D, Li Q, Zhang X, Wang H, Cao W, Li D, Xing W, Song M, Wang W, Meng Q, Wang Y. Systematic Review: Immunoglobulin G N-Glycans as Next-Generation Diagnostic Biomarkers for Common Chronic Diseases. ACTA ACUST UNITED AC 2019; 23:607-614. [DOI: 10.1089/omi.2019.0032] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Di Liu
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China
| | - Qihuan Li
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China
| | - Xiaoyu Zhang
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China
| | - Hao Wang
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | - Weijie Cao
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China
| | - Dong Li
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Weijia Xing
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Manshu Song
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China
| | - Wei Wang
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Qun Meng
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China
| | - Youxin Wang
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Di Domenico F, Zuliani I, Tramutola A. Shining a light on defective autophagy by proteomics approaches: implications for neurodegenerative illnesses. Expert Rev Proteomics 2019; 16:951-964. [PMID: 31709850 DOI: 10.1080/14789450.2019.1691919] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Autophagy is one of the most conserved clearance systems through which eukaryotes manage to handle dysfunctional and excess organelles and macromolecules. This catabolic process has not only a role in the maintenance of basal turnover of cellular components, but it is also essential in cells adaptation to stress conditions. In the last decades, defects in autophagic machinery have been identified as a feature in neurodegenerative diseases. In this context, mass spectrometry-based proteomics has become an important tool in the comprehensive analysis of proteins involved in the autophagic flux.Area covered: In this review, we discuss recent contributions of proteomic techniques in the study of defective autophagy related to neurodegenerative illness. Particular emphasis is given to the identification of i) shared autophagic markers between different disorders, which support common pathological mechanisms; ii) unique autophagic signature, which could aid to discriminate among diseases.Expert opinion: Proteomic approaches are valuable in the identification of alterations of components to the autophagic process at different steps of the process. The investigation of autophagic defects associated with neurological disorders is crucial in order to unravel all the potential mechanism leading to neurodegeneration and propose effective therapeutic strategies targeting autophagy.
Collapse
Affiliation(s)
- Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Ilaria Zuliani
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
16
|
Colorectal cancer cells respond differentially to autophagy inhibition in vivo. Sci Rep 2019; 9:11316. [PMID: 31383875 PMCID: PMC6683171 DOI: 10.1038/s41598-019-47659-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/19/2019] [Indexed: 12/19/2022] Open
Abstract
Autophagy has both tumor-promoting and -suppressing effects in cancer, including colorectal cancer (CRC), with transformed cells often exhibiting high autophagic flux. In established tumors, autophagy inhibition can lead to opposite responses resulting in either tumor cell death or hyperproliferation. The functional mechanisms underlying these differences are poorly understood. The present study aimed to investigate the relationship between the autophagic capacities of CRC cells and their sensitivities to autophagy inhibition. All studied CRC cell lines showed high basal autophagic flux. However, only HCT116 and Caco-2/15 cells displayed regulated autophagic flux upon starvation. Knockdown of ATG5 (which disrupts autophagosome elongation) or RAB21 (which decreases autophagosome/lysosome fusion) had little effect on CRC cell proliferation in vitro. Nonetheless, inhibition of autophagy in vivo had a substantial cell line-dependent impact on tumor growth, with some cells displaying decreased (HCT116 and Caco-2/15) or increased (SW480 and LoVo) proliferation. RNA sequencing and Western blot analyses in hyperproliferative SW480 tumors revealed that the mTORC2 and AKT pathways were hyperactivated following autophagy impairment. Inhibition of either mTOR or AKT activities rescued the observed hyperproliferation in autophagy-inhibited SW480 and reduced tumor growth. These results highlight that autophagy inhibition can lead, in specific cellular contexts, to compensatory mechanisms promoting tumor growth.
Collapse
|
17
|
Park HS, Jo E, Han JH, Jung SH, Lee DH, Park I, Heo KS, Na M, Myung CS. Hepatoprotective effects of an Acer tegmentosum Maxim extract through antioxidant activity and the regulation of autophagy. JOURNAL OF ETHNOPHARMACOLOGY 2019; 239:111912. [PMID: 31029758 DOI: 10.1016/j.jep.2019.111912] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 04/23/2019] [Accepted: 04/23/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acer tegmentosum Maxim (AT), the East Asian stripe maple, is an herb used to treat liver disease and is approved as a functional food in Korea. AT protects against hepatic disorders, atopic dermatitis, and diabetes mellitus. AIM OF THE STUDY We explored the mechanism of the hepatoprotective effects of AT extract in in vitro and in vivo levels. MATERIALS AND METHODS AT extract from Acer tegmentosum Maxim was extracted by hot water. Hepatoprotective effects of AT extract were confirmed using carbon tetrachloride (CCl4)- or alcohol-induced mouse model, and H2O2- or alcohol-induced HepG2 (liver hepatocellular carcinoma cell line) cells by measuring GOT, GPT, TG, and MDA levels. Hematoxylin and eosin (H&E) staining was used to observe the pathological analysis. Cytotoxicity or protective effect of AT extract was confirmed using MTT assay in HepG2 cells. Antioxidant effect of AT extract was measured using DPPH or H2DCFDA assay. Mechanism study of antioxidant and autophagy was carried out using western blotting and immunofluorescence analysis. RESULTS AT extract increased the viability of HepG2 cells treated with H2O2 and ethanol, and protected the liver against damage induced by CCl4 and alcohol. The AT extract increased the levels of nuclear respiratory factor 2 (Nrf2) and heme oxygenase-1 (HO-1). The level of microtubule-associated protein light chain 3 (LC3)-Ⅱ, beclin-1, autophagy-related genes (Atg) such as Atg3 and Atg12-5 as markers of autophagy activation was also increased. Moreover, the AT extract increased activation of mitogen-activated protein kinase (MAPK), which regulated autophagy and HO-1. CONCLUSION Therefore, these results indicate that the AT extract has a hepatoprotective effect by increasing antioxidant activity and inducing autophagy.
Collapse
Affiliation(s)
- Hyun-Soo Park
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, 34134, Republic of Korea.
| | - Eunji Jo
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, 34134, Republic of Korea.
| | - Joo-Hui Han
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, 34134, Republic of Korea.
| | - Sang-Hyuk Jung
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, 34134, Republic of Korea.
| | - Do-Hyung Lee
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, 34134, Republic of Korea.
| | - InWha Park
- Pharmacognosy, Chungnam National University College of Pharmacy, Daejeon, 34134, Republic of Korea.
| | - Kyung-Sun Heo
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, 34134, Republic of Korea.
| | - MinKyun Na
- Pharmacognosy, Chungnam National University College of Pharmacy, Daejeon, 34134, Republic of Korea; Institute of Drug Research & Development, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | - Chang-Seon Myung
- Department of Pharmacology, Chungnam National University College of Pharmacy, Daejeon, 34134, Republic of Korea; Institute of Drug Research & Development, Chungnam National University, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
18
|
Wang X, Li W, Zhang N, Zheng X, Jing Z. Opportunities and challenges of co-targeting epidermal growth factor receptor and autophagy signaling in non-small cell lung cancer. Oncol Lett 2019; 18:499-506. [PMID: 31289521 DOI: 10.3892/ol.2019.10372] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 04/11/2019] [Indexed: 12/17/2022] Open
Abstract
Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) are a standard therapy for patients with non-small cell lung cancer (NSCLC) with sensitive mutations. However, acquired resistance emerges following a median of 6-12 months. Several studies demonstrated that EGFR-TKI-induced tumor microenvironment stresses and autophagy are important causes of resistance. The current review summarizes the molecular mechanisms involved in EGFR-mediated regulation of autophagy. The role of autophagy in EGFR-TKI treatment, which may serve a role in protection or cell death, was discussed. Furthermore, co-inhibiting EGFR and autophagy signaling as a rational therapeutic strategy in the treatment of patients with NSCLC was explored.
Collapse
Affiliation(s)
- Xiaoju Wang
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, P.R. China
| | - Wenxin Li
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, P.R. China
| | - Ni Zhang
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, P.R. China
| | - Xiaoli Zheng
- Cancer Research Institute, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, P.R. China
| | - Zhao Jing
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, P.R. China
| |
Collapse
|
19
|
Li Q, Jia S, Xu L, Li B, Chen N. Metformin-induced autophagy and irisin improves INS-1 cell function and survival in high-glucose environment via AMPK/SIRT1/PGC-1α signal pathway. Food Sci Nutr 2019; 7:1695-1703. [PMID: 31139382 PMCID: PMC6526663 DOI: 10.1002/fsn3.1006] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 03/04/2019] [Indexed: 12/15/2022] Open
Abstract
In order to explore the protective function of metformin on pancreatic β cells to alleviate insulin resistance and underlying mechanisms, INS-1 cells were cultured into normal control (N), high glucose (H), high glucose and metformin (H + Met), high glucose and chloroquine (H + CQ), and high glucose and Ex527 (H + Ex527) groups, respectively. Upon 24-hr cultivation, the proliferation and glucose-stimulated insulin secretion (GSIS) of INS-1 cells were determined, and the expression of irisin and other proteins associated with AMPK/SIRT1/PGC-1α signal pathway, autophagy, and apoptosis was evaluated. Compared with the N group, the cells from the H group revealed lower proliferation, GSIS, and expression of irisin and proteins associated with AMPK/SIRT1/PGC-1α signal pathway and autophagy, but higher expression of proteins associated with apoptosis; in contrast, metformin could significantly rescue lower cell proliferation, GSIS, and expression of proteins associated with AMPK/SIRT1/PGC-1α signal pathway and autophagy, as well as irisin, and suppress apoptosis in high-glucose environment. Meanwhile, autophagy inhibitor CQ and SIRT1 inhibitor Ex527 can block above functions of metformin. Therefore, metformin can promote INS-1 cell proliferation, enhance GSIS, and suppress apoptosis by activating AMPK/SIRT1/PGC-1α signal pathway, up-regulating irisin expression, and inducing autophagy in INS-1 cells in high-glucose environment.
Collapse
Affiliation(s)
- Qingxue Li
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Sport Training and Monitoring, College of Health ScienceWuhan Sports UniversityWuhanChina
- School of Sports and HealthLinyi UniversityLinyiChina
| | - Shaohui Jia
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Sport Training and Monitoring, College of Health ScienceWuhan Sports UniversityWuhanChina
| | - Lei Xu
- Graduate SchoolWuhan Sports UniversityWuhanChina
| | - Biao Li
- Graduate SchoolWuhan Sports UniversityWuhanChina
| | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Sport Training and Monitoring, College of Health ScienceWuhan Sports UniversityWuhanChina
| |
Collapse
|
20
|
Qi N, Liao S, Abuzeid AMI, Li J, Wu C, Lv M, Lin X, Hu J, Yu L, Xiao W, Sun M, Li G. The effect of autophagy on the survival and invasive activity of Eimeria tenella sporozoites. Sci Rep 2019; 9:5835. [PMID: 30967577 PMCID: PMC6456608 DOI: 10.1038/s41598-019-41947-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 03/19/2019] [Indexed: 12/11/2022] Open
Abstract
Autophagy is a cellular process that is vital for the maintenance of homeostasis in eukaryotic cells. Currently, autophagy-related genes (atgs) in the Eimeria tenella genome database have been reported, but very little is known about the effects of autophagy on the survival and invasive activity of this protozoan. In this study, we investigated the autophagy in E. tenella sporozoites under starvation and autophagy-modulators treatments and evaluated the autophagy influence on cellular adenosine triphosphate (ATP) levels, the survival rate and the invasive activity of the sporozoites. The results showed that the autophagy could be induced in the sporozoites by starvation or inducer rapamycin (RP), but it could be inhibited by 3-methyladenine (3-MA) treatment. The sporozoites after starvation and RP-treatment displayed punctate signals of EtATG8 and formed autophagosomes. The survival rate of the sporozoites under starvation was significantly lower than that in the control group, whereas the ATP levels in sporozoite were far greater than those in the control. The quantitative real-time reverse transcriptase polymerase chain reaction (qRT-PCR) showed that the invasive activity of the sporozoites was up- and down-regulated by RP and 3-MA induction, respectively. Our results indicate that autophagy has effects on the survival and invasive activity of E. tenella sporozoites, which may provide new insights into anti-coccidial drugs.
Collapse
Affiliation(s)
- Nanshan Qi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture, Guangzhou, P. R. China.,Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, Guangdong, P. R. China
| | - Shenquan Liao
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture, Guangzhou, P. R. China.,Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, Guangdong, P. R. China
| | - Asmaa M I Abuzeid
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China
| | - Juan Li
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture, Guangzhou, P. R. China.,Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, Guangdong, P. R. China
| | - Caiyan Wu
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture, Guangzhou, P. R. China.,Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, Guangdong, P. R. China
| | - Minna Lv
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture, Guangzhou, P. R. China.,Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, Guangdong, P. R. China
| | - Xuhui Lin
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture, Guangzhou, P. R. China.,Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, Guangdong, P. R. China
| | - Junjing Hu
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture, Guangzhou, P. R. China.,Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, Guangdong, P. R. China
| | - Linzeng Yu
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture, Guangzhou, P. R. China.,Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, Guangdong, P. R. China
| | - Wenwan Xiao
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture, Guangzhou, P. R. China.,Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, Guangdong, P. R. China
| | - Mingfei Sun
- Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture, Guangzhou, P. R. China. .,Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, Guangdong, P. R. China.
| | - Guoqing Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.
| |
Collapse
|
21
|
Yoo YM, Jung EM, Jeung EB. Rapamycin-induced autophagy decreases Myf5 and MyoD proteins in C2C12 myoblast cells. Toxicol In Vitro 2019; 58:132-141. [PMID: 30905858 DOI: 10.1016/j.tiv.2019.03.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 03/20/2019] [Accepted: 03/20/2019] [Indexed: 12/19/2022]
Abstract
Rapamycin is an immunosuppressant that inhibits the mammalian or mechanistic target of rapamycin (mTOR) protein kinase and extends lifespan in organisms including mice. Myf5 and MyoD act as muscle-specific transcriptional factors for skeletal muscle differentiation. In this study, we determined whether rapamycin-induced autophagy causes the decrease of Myf5 and MyoD protein in C2C12 myoblast cells. Rapamycin induced a significant increase in the expression of the microtubule-associated protein 1 light chain 3 (LC3) II protein in a dose-dependent manner for 12 h. Rapamycin treatment also significantly increased p-ERK, p-Akt, and catalase expressions, and decreased Mn-SOD expression in a dose-dependent manner. Bax expression was significantly high compared to Bcl-2 expression in a dose-dependent manner of rapamycin for 12 h. For further study of rapamycin-induced autophagy in C2C12 myoblast cells, we investigated rapamycin treatment for 24, 36, and 48 h. Cell viability did not change with rapamycin treatment for 24, 36, and 48 h. Rapamycin-induced LC3-II, Beclin-1, Bax, and Bcl-2 proteins were significantly increased compared to without rapamycin. p-ERK expression increased with rapamycin treatment for 24 and 36 h compared to that without rapamycin, but decreased for 48 h. p-Akt expression decreased with rapamycin treatment for 36 and 48 h compared to that without rapamycin. In the same conditions, rapamycin-induced autophagy significantly reduced the Myf5 and MyoD proteins. Together, these results suggest that rapamycin-induced autophagy results in the decrease of Myf5 and MyoD proteins in C2C12 myoblast cells.
Collapse
Affiliation(s)
- Yeong-Min Yoo
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Eui-Man Jung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Eui-Bae Jeung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea.
| |
Collapse
|
22
|
Zhi X, Li B, Li Z, Zhang J, Yu J, Zhang L, Xu Z. Adrenergic modulation of AMPK‑dependent autophagy by chronic stress enhances cell proliferation and survival in gastric cancer. Int J Oncol 2019; 54:1625-1638. [PMID: 30896863 PMCID: PMC6438426 DOI: 10.3892/ijo.2019.4753] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 02/22/2019] [Indexed: 12/30/2022] Open
Abstract
Epidemiological data show that chronic stress has adverse effects on the incidence and progression of cancer. As a critical target organ for stress hormones, the stomach is frequently subjected to stress-related injury. However, few reports regarding the association between stress and gastric cancer (GC) have been published. The present study aimed to investigate the effect of chronic stress on the growth and survival of GC, and the role of the autophagy process. A restraint-stress procedure over 21 days was used to establish a chronic stress mouse model. Subcutaneous xenografts and gastric orthotopic xenografts were established in BALB/c nude mice. Alzet osmotic minipumps containing either PBS or propranolol hydrochloride was inserted on the nape of the neck 7 days prior to the initiation of restraint stress. The presence of autophagosomes and autolysosomes were examined by electron microscopy. The stress hormone norepinephrine significantly enhanced the proliferation of GC cells. By inhibiting adrenoreceptor expression, it was demonstrated that β2-adrenergic receptor (ADRB2) was the specific β-adrenergic receptor subtype responsible for catecholamine release. In addition, it was demonstrated that the induction of autophagy was a novel consequence of β2-adrenergic activation in GC cells. This was demonstrated by the appearance of double-membrane vesicles, punctuate GFP-RFP-microtubule-associated protein 1 light chain 3 distribution in the cytoplasm and a corresponding increase in autophagic flux. Notably, norepinephrine-induced autophagy was shown to have a tumor-promoting role under conditions of chronic stress in vitro and in vivo. It was further demonstrated that, upon activation of cAMP-response element binding protein, chronic stress promoted autophagic flux through the adenosine 5′-monophosphate-activated protein kinase-unc-51 like autophagy activating kinase 1 (AMPK-ULK1) pathway. Tissue microarray analysis revealed a negative correlation between the expression of ADRB2 and autophagic marker p62/sequestosome-1 in GC tumor samples. Additionally, high protein levels of ADRB2 correlated positively with tumor, node, metastasis stage and poor prognosis in patients with GC. These results establish a novel pathway that chronic stress activates tumor-promoting autophagy to accelerate the progression of GC. The present study is the first, to the best of our knowledge, providing preclinical evidence that chronic stress serves a role in the progression of GC.
Collapse
Affiliation(s)
- Xiaofei Zhi
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Bowen Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zheng Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jiaxuan Zhang
- Department of Emergency Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Junbo Yu
- Department of Emergency Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Lu Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
23
|
Maiuri MC, Kroemer G. Therapeutic modulation of autophagy: which disease comes first? Cell Death Differ 2019; 26:680-689. [PMID: 30728461 PMCID: PMC6460393 DOI: 10.1038/s41418-019-0290-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 01/11/2019] [Indexed: 02/06/2023] Open
Abstract
The relentless efforts of thousands of researchers have allowed deciphering the molecular machinery that regulates and executes autophagy, thus identifying multiple molecular targets to enhance or block the process, rendering autophagy "druggable". Autophagy inhibition may be useful for preserving the life of cells that otherwise would succumb to excessive self-digestion. Moreover, autophagy blockade may reduce the fitness of cancer cells or interrupt metabolic circuitries required for their growth. Autophagy stimulation is probably useful for the prevention or treatment of aging, cancer (when stimulation of immunosurveillance is the therapeutic goal), cardiovascular disease, cystic fibrosis, infection by intracellular pathogens, obesity, and intoxication by heavy metals, just to mention a few examples. Epidemiological evidence suggests broad health-improving effects for lifestyles, micronutrients, and drugs that favor autophagy. In this review, we discuss the role of autophagy in disease pathogenesis while focusing on the question, which disease will become the first clinically approved indication for therapeutic autophagy modulation.
Collapse
Affiliation(s)
- Maria Chiara Maiuri
- Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, 75006, Paris, France.
- Cell Biology and Metabolomics Platforms, Gustave Roussy Cancer Campus, 94805, Villejuif, France.
- INSERM U1138, 75006, Paris, France.
- Université Paris Descartes, Sorbonne Paris Cité, 75006, Paris, France.
- Sorbonne Université, 75006, Paris, France.
| | - Guido Kroemer
- Cell Biology and Metabolomics Platforms, Gustave Roussy Cancer Campus, 94805, Villejuif, France.
- INSERM U1138, 75006, Paris, France.
- Université Paris Descartes, Sorbonne Paris Cité, 75006, Paris, France.
- Sorbonne Université, 75006, Paris, France.
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, 75015, Paris, France.
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, 17176, Stockholm, Sweden.
| |
Collapse
|
24
|
Palmisano NJ, Meléndez A. Autophagy in C. elegans development. Dev Biol 2019; 447:103-125. [PMID: 29709599 PMCID: PMC6204124 DOI: 10.1016/j.ydbio.2018.04.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 03/19/2018] [Accepted: 04/12/2018] [Indexed: 12/11/2022]
Abstract
Autophagy involves the sequestration of cytoplasmic contents in a double-membrane structure referred to as the autophagosome and the degradation of its contents upon delivery to lysosomes. Autophagy activity has a role in multiple biological processes during the development of the nematode Caenorhabditis elegans. Basal levels of autophagy are required to remove aggregate prone proteins, paternal mitochondria, and spermatid-specific membranous organelles. During larval development, autophagy is required for the remodeling that occurs during dauer development, and autophagy can selectively degrade components of the miRNA-induced silencing complex, and modulate miRNA-mediated silencing. Basal levels of autophagy are important in synapse formation and in the germ line, to promote the proliferation of proliferating stem cells. Autophagy activity is also required for the efficient removal of apoptotic cell corpses by promoting phagosome maturation. Finally, autophagy is also involved in lipid homeostasis and in the aging process. In this review, we first describe the molecular complexes involved in the process of autophagy, its regulation, and mechanisms for cargo recognition. In the second section, we discuss the developmental contexts where autophagy has been shown to be important. Studies in C. elegans provide valuable insights into the physiological relevance of this process during metazoan development.
Collapse
Affiliation(s)
- Nicholas J Palmisano
- Biology Department, Queens College, CUNY, Flushing, NY, USA; Biology Ph.D. Program, The Graduate Center of the City University of New York, NK, USA
| | - Alicia Meléndez
- Biology Department, Queens College, CUNY, Flushing, NY, USA; Biology Ph.D. Program, The Graduate Center of the City University of New York, NK, USA; Biochemistry Ph.D. Program, The Graduate Center of the City University of New York, NY, USA.
| |
Collapse
|
25
|
Guo T, Nan Z, Miao C, Jin X, Yang W, Wang Z, Tu Y, Bao H, Lyu J, Zheng H, Deng Q, Guo P, Xi Y, Yang X, Ge W. The autophagy-related gene Atg101 in Drosophila regulates both neuron and midgut homeostasis. J Biol Chem 2019; 294:5666-5676. [PMID: 30760524 DOI: 10.1074/jbc.ra118.006069] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 02/10/2019] [Indexed: 12/19/2022] Open
Abstract
Atg101 is an autophagy-related gene identified in worms, flies, mice, and mammals, which encodes a protein that functions in autophagosome formation by associating with the ULK1-Atg13-Fip200 complex. In the last few years, the critical role of Atg101 in autophagy has been well-established through biochemical studies and the determination of its protein structure. However, Atg101's physiological role, both during development and in adulthood, remains less understood. Here, we describe the generation and characterization of an Atg101 loss-of-function mutant in Drosophila and report on the roles of Atg101 in maintaining tissue homeostasis in both adult brains and midguts. We observed that homozygous or hemizygous Atg101 mutants were semi-lethal, with only some of them surviving into adulthood. Both developmental and starvation-induced autophagy processes were defective in the Atg101 mutant animals, and Atg101 mutant adult flies had a significantly shorter lifespan and displayed a mobility defect. Moreover, we observed the accumulation of ubiquitin-positive aggregates in Atg101 mutant brains, indicating a neuronal defect. Interestingly, Atg101 mutant adult midguts were shorter and thicker and exhibited abnormal morphology with enlarged enterocytes. Detailed analysis also revealed that the differentiation from intestinal stem cells to enterocytes was impaired in these midguts. Cell type-specific rescue experiments disclosed that Atg101 had a function in enterocytes and limited their growth. In summary, the results of our study indicate that Drosophila Atg101 is essential for tissue homeostasis in both adult brains and midguts. We propose that Atg101 may have a role in age-related processes.
Collapse
Affiliation(s)
- Ting Guo
- From the Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China 310058, and
| | - Zi Nan
- From the Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China 310058, and
| | - Chen Miao
- From the Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Department of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058
| | - Xiaoye Jin
- From the Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Department of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058
| | - Weiwei Yang
- From the Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Department of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058
| | - Zehua Wang
- From the Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China 310058, and
| | - Yinqi Tu
- From the Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China 310058, and
| | - Hongcun Bao
- From the Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China 310058, and
| | - Jialan Lyu
- From the Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Department of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058
| | - Huimei Zheng
- From the Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Department of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058
| | - Qiannan Deng
- From the Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China 310058, and
| | - Pengfei Guo
- From the Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China 310058, and
| | - Yongmei Xi
- From the Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Department of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058
| | - Xiaohang Yang
- From the Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Department of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058
| | - Wanzhong Ge
- From the Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058, .,the Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058.,the Department of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China 310058
| |
Collapse
|
26
|
Fan P, Wang N, Wang L, Xie X-Q. Autophagy and Apoptosis Specific Knowledgebases-guided Systems Pharmacology Drug Research. Curr Cancer Drug Targets 2019; 19:716-728. [PMID: 30727895 DOI: 10.2174/1568009619666190206122149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 11/20/2018] [Accepted: 01/30/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND Autophagy and apoptosis are the basic physiological processes in cells that clean up aged and mutant cellular components or even the entire cells. Both autophagy and apoptosis are disrupted in most major diseases such as cancer and neurological disorders. Recently, increasing attention has been paid to understand the crosstalk between autophagy and apoptosis due to their tightly synergetic or opposite functions in several pathological processes. OBJECTIVE This study aims to assist autophagy and apoptosis-related drug research, clarify the intense and complicated connections between two processes, and provide a guide for novel drug development. METHODS We established two chemical-genomic databases which are specifically designed for autophagy and apoptosis, including autophagy- and apoptosis-related proteins, pathways and compounds. We then performed network analysis on the apoptosis- and autophagy-related proteins and investigated the full protein-protein interaction (PPI) network of these two closely connected processes for the first time. RESULTS The overlapping targets we discovered show a more intense connection with each other than other targets in the full network, indicating a better efficacy potential for drug modulation. We also found that Death-associated protein kinase 1 (DAPK1) is a critical point linking autophagy- and apoptosis-related pathways beyond the overlapping part, and this finding may reveal some delicate signaling mechanism of the process. Finally, we demonstrated how to utilize our integrated computational chemogenomics tools on in silico target identification for small molecules capable of modulating autophagy- and apoptosis-related pathways. CONCLUSION The knowledge-bases for apoptosis and autophagy and the integrated tools will accelerate our work in autophagy and apoptosis-related research and can be useful sources for information searching, target prediction, and new chemical discovery.
Collapse
Affiliation(s)
- Peihao Fan
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, University of Pittsburgh, 3501 Terrace Street, PA, United States
| | - Nanyi Wang
- School of Pharmacy, University of Pittsburgh, 335 Sutherland Drive, 206 Salk Pavilion, PA, United States
| | - Lirong Wang
- School of Pharmacy, University of Pittsburgh, 335 Sutherland Drive, 206 Salk Pavilion, PA, United States
| | - Xie X-Q
- School of Pharmacy, University of Pittsburgh, 335 Sutherland Drive, 206 Salk Pavilion, PA, United States
| |
Collapse
|
27
|
The role of autophagy in morphogenesis and stem cell maintenance. Histochem Cell Biol 2018; 150:721-732. [PMID: 30382373 DOI: 10.1007/s00418-018-1751-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2018] [Indexed: 12/21/2022]
Abstract
During embryonic development, cells need to undergo a number of morphological changes that are decisive for the shaping of the embryo's body, initiating organogenesis and differentiation into functional tissues. These remodeling processes are accompanied by profound changes in the cell membrane, the cytoskeleton, organelles, and extracellular matrix composition. While considerably detailed insight into the role of autophagy in stem cells biology has been gained in the recent years, information regarding the participation of autophagy in morphogenetic processes is only sparse. This review, therefore, focuses on the role of autophagy in cell morphogenesis through its regulatory activity in TGFβ signaling, expression of adhesion molecules and cell cycle modification. It also discusses the role of autophagy in stem cell maintenance which is very fundamental for cell renewal and replacement during development, pathogenesis of certain diseases and development of therapies. We are thus addressing here perspectives for further potentially rewarding research topics.
Collapse
|
28
|
Chronic heart failure is characterized by altered mitochondrial function and structure in circulating leucocytes. Oncotarget 2018; 9:35028-35040. [PMID: 30416677 PMCID: PMC6205552 DOI: 10.18632/oncotarget.26164] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 09/13/2018] [Indexed: 12/21/2022] Open
Abstract
Oxidative stress is currently viewed as a key factor in the genesis and progression of Heart Failure (HF). The aim of this study was to characterize the mitochondrial changes linked to oxidative stress generation in circulating peripheral blood mononuclear cells isolated from chronic HF patients (HF_PBMCs) in order to highlight the involvement of mitochondrial dysfunction in the pathophysiology of HF. To assess the production of reactive oxygen species (ROS), mitochondrial function and ultrastructure and the mitophagic flux in circulating PBMCs we enrolled 15 patients with HF and a control group of ten healthy subjects. The HF_PBMCs showed a mitochondrial population consisting of damaged and less functional organelles responsible of higher superoxide anion production both at baseline and under in vitro stress conditions, with evidence of cellular apoptosis. Although the mitophagic flux at baseline was enhanced in HF_PBMCs at level similar to those that could be achieved in control PBMCs only under inflammatory stress conditions, the activation of mitophagy was unable to preserve a proper mitochondrial dynamics upon stress stimuli in HF. In summary, circulating HF_PBMCs show structural and functional derangements of mitochondria with overproduction of reactive oxidant species. This mitochondrial failure sustains a leucocyte dysfunctional status in the blood that may contribute to development and persistence of stress conditions within the cardiovascular system in HF.
Collapse
|
29
|
Eberhardt W, Nasrullah U, Pfeilschifter J. Activation of renal profibrotic TGFβ controlled signaling cascades by calcineurin and mTOR inhibitors. Cell Signal 2018; 52:1-11. [PMID: 30145216 DOI: 10.1016/j.cellsig.2018.08.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 12/11/2022]
Abstract
The calcineurin inhibitors (CNI) cyclosporine A (CsA) and tacrolimus represent potent immunosuppressive agents frequently used for solid organ transplantation and treatment of autoimmune disorders. Despite of their immense therapeutic benefits, residual fibrosis mainly in the kidney represents a common side effect of long-term therapy with CNI. Regardless of the immunosuppressive action, an increasing body of evidence implicates that a drug-induced increase in TGFβ and subsequent activation of TGFβ-initiated signaling pathways is closely associated with the development and progression of CNI-induced nephropathy. Mechanistically, an increase in reactive oxygen species (ROS) generation due to drug-induced changes in the intracellular redox homeostasis functions as an important trigger of the profibrotic signaling cascades activated under therapy with CNI. Although, inhibitors of the mechanistic target of rapamycin (mTOR) kinase have firmly been established as alternative compounds with a lower nephrotoxic potential, an activation of fibrogenic signaling cascades has been reported for these drugs as well. This review will comprehensively summarize recent advances in the understanding of profibrotic signaling events modulated by these widely used compounds with a specific focus put on mechanisms occurring independent of their respective immunosuppressive action. Herein, the impact of redox modulation, the activation of canonical TGFβ and non-Smad pathways and modulation of autophagy by both classes of immunosuppressive drugs will be highlighted and discussed in a broader perspective. The comprehensive knowledge of profibrotic signaling events specifically accompanying the immunomodulatory activity of these widely used drugs is needed for a reliable benefit-risk assessment under therapeutic regimens.
Collapse
Affiliation(s)
- Wolfgang Eberhardt
- Pharmazentrum frankfurt/ZAFES, Universitätsklinikum und Goethe-Universität, Frankfurt am Main, Germany.
| | - Usman Nasrullah
- Pharmazentrum frankfurt/ZAFES, Universitätsklinikum und Goethe-Universität, Frankfurt am Main, Germany
| | - Josef Pfeilschifter
- Pharmazentrum frankfurt/ZAFES, Universitätsklinikum und Goethe-Universität, Frankfurt am Main, Germany
| |
Collapse
|
30
|
Long-Zhi Decoction Medicated Serum Promotes Angiogenesis in Human Umbilical Vein Endothelial Cells Based on Autophagy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:6857398. [PMID: 29853968 PMCID: PMC5964498 DOI: 10.1155/2018/6857398] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 04/11/2018] [Indexed: 12/19/2022]
Abstract
Ischemic stroke (IS) is a fatal subtype of stroke that lacks effective treatments. Angiogenesis following IS is an effective response that mediates brain recovery and repair. Our previous study demonstrated that long-zhi decoction (LZD), a Chinese herbal formula, promoted angiogenesis in rats of IS model. To further investigate the association between the proangiogenic mechanism of an LZD-medicated serum and cellular autophagy, we evaluated its promotional effect on angiogenesis in human umbilical vein endothelial cells (HUVECs) in vitro. We used HUVECs subjected to H2O2 to induce injury and observed the effects of the LZD-medicated serum treatment. Cell-based assays included proliferation, migration, and tube formation. To assess the extent of autophagy, transmission electron microscopy was used to measure the number of autophagosomes. Immunofluorescence and Western blotting were performed to evaluate the autophagy-related protein of LC3-II and Beclin-1. The LZD-medicated serum promoted proliferation, migration, and tube formation in HUVECs. The LZD-medicated serum also increased the autophagosomes and the autophagic protein expressions of LC3-II and Beclin-1. The proangiogenic and autophagic activity of LZD provides new cogitations to its clinical application and may lead to potential drug development for treating various vascular diseases, especially in the elderly, in the future.
Collapse
|
31
|
Zhou GZ, Li AF, Sun YH, Sun GC. A novel synthetic curcumin derivative MHMM-41 induces ROS-mediated apoptosis and migration blocking of human lung cancer cells A549. Biomed Pharmacother 2018; 103:391-398. [PMID: 29674274 DOI: 10.1016/j.biopha.2018.04.086] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 04/10/2018] [Accepted: 04/10/2018] [Indexed: 11/17/2022] Open
Abstract
Many curcumin derivatives were produced and characterized to improve the physiochemical instability and low solubility of curcumin. Here, MHMM-41 (a novel curcumin derivative) was used to treat non-small lung cancer cells of human (known as A549) and to identify its anti-proliferative activities. Our results suggested that MHMM-41 display no significant cytotoxicity toward normal human lung fibroblast 2BS cells and mouse embryonal fibroblast 3T3 cells. It also had better anti-proliferative activity than curcumin in A549 cells. Further study showed a significant increase of apoptotic A549 cells in time and dose dependent manners. The activation of caspase-3, 8, 9, 12, Bax and PARP proteins were detected. Consequently, MHMM-41 treatment led to the reduction of mitochondrial membrane potential by JC-1 staining and characteristic nuclei fragmentation by Hoechst 33,342 staining, respectively, which showed that A549 apoptosis could be triggered by the extrinsic and intrinsic mitochondrial pathways. The release of ROS was also measured by flow cytometry. Further, wound healing assay and transwell experiments confirmed the anti-migration ability of MHMM-41 in A549 cells. Our current study suggested the potentials of MHMM-41 to inhibit the A549 cell proliferation. However, the intensive mechanical research on the anti-proliferation of A549 cells needs to be performed in the future.
Collapse
Affiliation(s)
- Guang-Zhou Zhou
- Department of Biotechnology, College of Bioengineering, Henan University of Technology, Zhengzhou, Henan, 450001, China.
| | - A-Fang Li
- Department of Biotechnology, College of Bioengineering, Henan University of Technology, Zhengzhou, Henan, 450001, China
| | - Yan-He Sun
- Department of Biotechnology, College of Bioengineering, Henan University of Technology, Zhengzhou, Henan, 450001, China
| | - Gang-Chun Sun
- College of Chemistry and Chemical Engineering, Henan University of Technology, Zhengzhou, 450001, China.
| |
Collapse
|
32
|
Ma J, Weng L, Wang Z, Jia Y, Liu B, Wu S, Cao Y, Sun X, Yin X, Shang M, Mao A. MiR-124 induces autophagy-related cell death in cholangiocarcinoma cells through direct targeting of the EZH2-STAT3 signaling axis. Exp Cell Res 2018. [PMID: 29530475 DOI: 10.1016/j.yexcr.2018.02.037] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cholangiocarcinoma (CCA) is a lethal cancer associated with chronic inflammation that has increased in prevalence in recent decades. The dysregulated expression of microRNAs (miRNAs) has been detected in various types of malignancies, and depending on the target genes this can result in miRNAs functioning as tumor suppressors or oncogenes. In this study, we investigated the role of miR-124 in cholangiocarcinoma (CCA) and found that its expression was significantly downregulated in the tumor tissue of patients and in CCA cell lines. Our results provided evidence that miR-124 induces apoptotic cell death and triggers the autophagic flux in CCA cells. EZH2 and STAT3 were identified as direct targets of miR-124. The effect of miR-124 on EZH2 expression in CCA cells was evaluated using cell transfection, xenotransplantation into nude mice and a luciferase reporter assay. Silencing of EZH2 restored the effects of miR-124, whereas overexpression of EZH2 abrogated the effects of miR-124. Silencing of Beclin1 or ATG5 abrogated the effects of miR-124 or siEZH2. In vivo, overexpression of miR-124 dramatically induced autophagy-related cell death and suppressed tumorigenicity. Taken together, our findings indicated that downregulation of miR-124 expression was associated with disease progression in human CCA and we revealed that miR-124 exerts a tumor suppressive function in CCA by inducing autophagy-related cell death via direct targeting of the EZH2-STAT3 signaling axis.
Collapse
Affiliation(s)
- Jun Ma
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xian Xia Road, Shanghai 200336, China
| | - Li Weng
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xian Xia Road, Shanghai 200336, China.
| | - Zhongmin Wang
- Affiliated Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China.
| | - Yiping Jia
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xian Xia Road, Shanghai 200336, China
| | - Bingyan Liu
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xian Xia Road, Shanghai 200336, China
| | - Shaoqiu Wu
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xian Xia Road, Shanghai 200336, China
| | - Yan Cao
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xian Xia Road, Shanghai 200336, China
| | - Xianjun Sun
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xian Xia Road, Shanghai 200336, China
| | - Xiang Yin
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xian Xia Road, Shanghai 200336, China
| | - Mingyi Shang
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xian Xia Road, Shanghai 200336, China.
| | - Aiwu Mao
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xian Xia Road, Shanghai 200336, China.
| |
Collapse
|
33
|
Mitochondria and Sex-Specific Cardiac Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1065:241-256. [PMID: 30051389 DOI: 10.1007/978-3-319-77932-4_16] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The focus of this chapter is the gender differences in mitochondria in cardiovascular disease. There is broad evidence suggesting that some of the gender differences in cardiovascular outcome may be partially related to differences in mitochondrial biology (Ventura-Clapier R, Moulin M, Piquereau J, Lemaire C, Mericskay M, Veksler V, Garnier A, Clin Sci (Lond) 131(9):803-822, 2017)). Mitochondrial disorders are causally affected by mutations in either nuclear or mitochondrial genes involved in the synthesis of respiratory chain subunits or in their posttranslational control. This can be due to mutations of the mtDNA which are transmitted by the mother or mutations in the nuclear DNA. Because natural selection on mitochondria operates only in females, mutations may have had more deleterious effects in males than in females (Ventura-Clapier R, Moulin M, Piquereau J, Lemaire C, Mericskay M, Veksler V, Garnier A, Clin Sci (Lond) 131(9):803-822, 2017; Camara AK, Lesnefsky EJ, Stowe DF. Antioxid Redox Signal 13(3):279-347, 2010). As mitochondrial mutations can affect all tissues, they are responsible for a large panel of pathologies including neuromuscular disorders, encephalopathies, metabolic disorders, cardiomyopathies, neuropathies, renal dysfunction, etc. Many of these pathologies present sex/gender specificity. Thus, alleviating or preventing mitochondrial dysfunction will contribute to mitigating the severity or progression of the development of diseases. Here, we present evidence for the involvement of mitochondria in the sex specificity of cardiovascular disorders.
Collapse
|
34
|
Liu D, Xu J, Qian G, Hamid M, Gan F, Chen X, Huang K. Selenizing astragalus polysaccharide attenuates PCV2 replication promotion caused by oxidative stress through autophagy inhibition via PI3K/AKT activation. Int J Biol Macromol 2017; 108:350-359. [PMID: 29217185 DOI: 10.1016/j.ijbiomac.2017.12.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/01/2017] [Accepted: 12/04/2017] [Indexed: 02/07/2023]
Abstract
Our previous studies have shown that oxidative stress could promote the porcine circovirus type 2 (PCV2) replication, and astragalus polysaccharide (APS)/selenium could suppress PCV2 replication. However, whether selenizing astragalus polysaccharide (sAPS) provides protection against oxidative stress-induced PCV2 replication promotion and the mechanism involved remain unclear. The present study aimed to explore the mechanism of the PCV2 replication promotion induced by oxidative stress and a novel pharmacotherapeutic approach involving the regulation of autophagy of sAPS. Our results showed that H2O2 promoted PCV2 replication via enhancing autophagy by using 3-methyladenine (3-MA) and autophagy-related gene 5 (ATG5) knockdown. Sodium selenite, APS, the mixture of sodium selenite and APS, and sAPS significantly inhibited H2O2-induced PCV2 replication promotion, respectively. Among these, sAPS exerted maximal inhibitory effect. sAPS could also significantly inhibit autophagy activated by H2O2 and increase the Akt and mTOR phosphorylation. Moreover, LY294002, the specific phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) inhibitor, significantly alleviated the effects of sAPS on autophagy and PCV2 replication. Taken together, we conclude that H2O2 promotes PCV2 replication by inducing autophagy and sAPS attenuates the PCV2 replication promotion through autophagy inhibition via PI3K/AKT activation.
Collapse
Affiliation(s)
- Dandan Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Jing Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Gang Qian
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Mohammed Hamid
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Fang Gan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Xingxiang Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China.
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China.
| |
Collapse
|
35
|
Sohn EJ, Park HT. Natural agents mediated autophagic signal networks in cancer. Cancer Cell Int 2017; 17:110. [PMID: 29209152 PMCID: PMC5704453 DOI: 10.1186/s12935-017-0486-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 11/23/2017] [Indexed: 01/01/2023] Open
Abstract
Recent studies suggested that natural compounds are important in finding targets for cancer treatments. Autophagy (“self-eating”) plays important roles in multiple diseases and acts as a tumor suppressor in cancer. Here, we examined the molecular mechanism by which natural agents regulate autophagic signals. Understanding the relationship between natural agents and cellular autophagy may provide more information for cancer diagnosis and chemoprevention.
Collapse
Affiliation(s)
- Eun Jung Sohn
- College of Korean Medicine, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul, 130-701 Republic of Korea.,Peripheral Neuropathy Research Center, Department of Physiology, College of Medicine, Dong-A University, Dongdaesin-Dong, Seo-Gu, Busan, 602-714 Republic of Korea
| | - Hwan Tae Park
- Peripheral Neuropathy Research Center, Department of Physiology, College of Medicine, Dong-A University, Dongdaesin-Dong, Seo-Gu, Busan, 602-714 Republic of Korea
| |
Collapse
|
36
|
Chen SY, Lin CH, Lin JT, Cheng YF, Chen HM, Kao SH. Adenine causes cell cycle arrest and autophagy of chronic myelogenous leukemia K562 cells via AMP-activated protein kinase signaling. Oncol Lett 2017; 14:5575-5580. [PMID: 29113185 DOI: 10.3892/ol.2017.6890] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 05/11/2017] [Indexed: 01/14/2023] Open
Abstract
AMP-activated protein kinase (AMPK) is known as a pivotal regulator of cellular metabolism. Mounting evidences have demonstrated that AMPK activation exerts tumor suppressive activity on leukemia cells. The present study reported that adenine, an AMPK activator, triggers cell cycle arrest and autophagy of human chronic myelogenous leukemia K562 cells consequently suppressing cell viability. The present findings revealed that adenine treatment (4.0-8.0 mM) significantly inhibited the viability of K562 cells to 69.3±2.5% (24 h) and 53.4±2.1% (48 h) of the control. Flow cytometric analysis revealed that there was a significant accumulation in G2/M phase, but not sub-G1 phase K562 cells following exposure to adenine. Additional investigation demonstrated that adenine treatments significantly increased the number of acidic vesicular organelles and the level of autophagosomal microtubule associated protein 1 light chain 3 α (LC3) marker. By contrast, cleavage of caspase-9, caspase-3 and poly-ADP-ribose polymerase was insignificantly affected in K562 cells following adenine treatment. In K562 cells, adenine was able to markedly promote the phosphorylation of AMPKα and suppress the phosphorylation of mammalian target of rapamycin (mTOR), a downstream target of AMPK. In addition, inhibiting AMPK phosphorylation using dorsomorphin restored mTOR phosphorylation, inhibited the accumulation of LC3 and significantly recovered the suppressed cell viability in response to adenine. Taken together, the present results demonstrated that adenine induced G2/M phase arrest and autophagic cell death, consequently suppressing the viability of K562 cells, which may attribute to the AMPK activation triggered by adenine. These findings provide evidence that adenine may be beneficial to chronic myelogenous leukemia therapy by suppressing excessive cell proliferation.
Collapse
Affiliation(s)
- San-Yuan Chen
- Department of Traditional Chinese Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 600, Taiwan R.O.C
| | - Chun-Hsiang Lin
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung 402, Taiwan R.O.C
| | - Jiun-Tsai Lin
- Institute of Applied Science and Engineering, Catholic Fu Jen University, New Taipei 242, Taiwan R.O.C
| | - Yi-Fang Cheng
- Energenesis Biomedical Co. Ltd., New Taipei 235, Taiwan R.O.C
| | - Han-Min Chen
- Institute of Applied Science and Engineering, Catholic Fu Jen University, New Taipei 242, Taiwan R.O.C
| | - Shao-Hsuan Kao
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung 402, Taiwan R.O.C.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402, Taiwan R.O.C
| |
Collapse
|
37
|
Zhou Y, Hong F, Tian Y, Zhao X, Hong J, Ze Y, Wang L. Nanoparticulate titanium dioxide-inhibited dendritic development is involved in apoptosis and autophagy of hippocampal neurons in offspring mice. Toxicol Res (Camb) 2017; 6:889-901. [PMID: 30090551 PMCID: PMC6062220 DOI: 10.1039/c7tx00153c] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/05/2017] [Indexed: 12/24/2022] Open
Abstract
Background: Numerous studies have demonstrated that, upon maternal exposure, nano-TiO2 can cross the placental barrier, accumulate in offspring animals, and cause neurotoxicity. However, the neurotoxic mechanisms are not fully understood. The aim of this study is to determine the effects of nano-TiO2 on the dendritic outgrowth of hippocampal neurons and confirm the role of apoptosis and excessive autophagy in the neurotoxicity of offspring mice caused by nano-TiO2, as well as its molecular mechanisms. Methods: Pregnant mice were intragastrically administered 1, 2, or 3 mg per kg body weight nano-TiO2 consecutively from prenatal day 7 to postpartum day 21. The ultrastructure, mitochondrial membrane potential (MMP), levels of reactive oxygen species (ROS) and peroxides, and ATP contents, along with the expression of apoptosis- and autophagy-related factors, were investigated. Results: The dendritic length of hippocampal neurons was lower in the group treated with nano-TiO2 than in the control group. Apoptosis, excessive autophagy, and nano-TiO2 aggregation in hippocampal neurons resulted from maternal exposure to nano-TiO2. Maternal exposure to nano-TiO2 also resulted in the over-production of ROS, increases in malondialdehyde and protein carbonylation, reductions in MMP and ATP contents, up-regulation of apoptosis- or autophagy-related factors including histone H2AX at serine 139 (γH2AX), cytochrome C (Cyt C), caspase 3, phosphoinositide 3-kinase (PI3K3C), Beclin 1, c-Jun, LC3I, LC3II, JNK and p-JNK expression, and an increase of LC3II/LC3I, as well as down-regulation of Bcl-2 expression in hippocampal neurons of offspring mice. Conclusions: Maternal exposure to nano-TiO2 inhibited the dendritic outgrowth of hippocampal neurons. This effect is closely associated with excessive autophagy, which is related to severe oxidative stress and alterations in the expressions of apoptosis- and autophagy-related factors in the hippocampal neurons of offspring mice, due to maternal exposure to nano-TiO2.
Collapse
Affiliation(s)
- Yingjun Zhou
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection , Huaiyin Normal University , Huaian 223300 , China .
- Laboratory for Food Safety and Nutritional Function , Huaiyin Normal University , Huaian 223300 , China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake , Huaiyin Normal University , Huaian 223300 , China
| | - Fashui Hong
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection , Huaiyin Normal University , Huaian 223300 , China .
- Laboratory for Food Safety and Nutritional Function , Huaiyin Normal University , Huaian 223300 , China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake , Huaiyin Normal University , Huaian 223300 , China
| | - Yusheng Tian
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection , Huaiyin Normal University , Huaian 223300 , China .
- Laboratory for Food Safety and Nutritional Function , Huaiyin Normal University , Huaian 223300 , China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake , Huaiyin Normal University , Huaian 223300 , China
| | - Xiangyu Zhao
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection , Huaiyin Normal University , Huaian 223300 , China .
- Laboratory for Food Safety and Nutritional Function , Huaiyin Normal University , Huaian 223300 , China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake , Huaiyin Normal University , Huaian 223300 , China
| | - Jie Hong
- Medical College of Soochow University , Suzhou 215123 , China
| | - Yuguan Ze
- Medical College of Soochow University , Suzhou 215123 , China
| | - Ling Wang
- Library of Soochow University , Suzhou 215123 , China
| |
Collapse
|
38
|
The dual role of poly(ADP-ribose) polymerase-1 in modulating parthanatos and autophagy under oxidative stress in rat cochlear marginal cells of the stria vascularis. Redox Biol 2017; 14:361-370. [PMID: 29049980 PMCID: PMC5641921 DOI: 10.1016/j.redox.2017.10.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/01/2017] [Accepted: 10/03/2017] [Indexed: 12/20/2022] Open
Abstract
Oxidative stress is reported to regulate several apoptotic and necrotic cell death pathways in auditory tissues. Poly(ADP-ribose) polymerase-1 (PARP-1) can be activated under oxidative stress, which is the hallmark of parthanatos. Autophagy, which serves either a pro-survival or pro-death function, can also be stimulated by oxidative stress, but the role of autophagy and its relationship with parthanatos underlying this activation in the inner ear remains unknown. In this study, we established an oxidative stress model in vitro by glucose oxidase/glucose (GO/G), which could continuously generate low concentrations of H2O2 to mimic continuous exposure to H2O2 in physiological conditions, for investigation of oxidative stress-induced cell death mechanisms and the regulatory role of PARP-1 in this process. We observed that GO/G induced stria marginal cells (MCs) death via upregulation of PARP-1 expression, accumulation of polyADP-ribose (PAR) polymers, decline of mitochondrial membrane potential (MMP) and nuclear translocation of apoptosis-inducing factor (AIF), which all are biochemical features of parthanatos. PARP-1 knockdown rescued GO/G-induced MCs death, as well as abrogated downstream molecular events of PARP-1 activation. In addition, we demonstrated that GO/G stimulated autophagy and PARP-1 knockdown suppressed GO/G-induced autophagy in MCs. Interestingly, autophagy suppression by 3-Methyladenine (3-MA) accelerated GO/G-induced parthanatos, indicating a pro-survival function of autophagy in GO/G-induced MCs death. Taken together, these data suggested that PARP-1 played dual roles by modulating parthanatos and autophagy in oxidative stress-induced MCs death, which may be considered as a promising therapeutic target for ameliorating oxidative stress-related hearing disorders. The model mimicked continuous exposure to H2O2 in physiological conditions. GO/G induced parthanatos and autophagy in MCs. GO/G-induced parthanatos and autophagy are mediated by PARP-1. Autophagy inhibition by 3-MA sensitizes MCs to GO/G-induced parthanatos. PARP-1 plays a dual role by modulating parthanatos and autophagy under GO/G-induced oxidative stress in MCs.
Collapse
|
39
|
Bhattacharya S, Yin J, Winborn CS, Zhang Q, Yue J, Chaum E. Prominin-1 Is a Novel Regulator of Autophagy in the Human Retinal Pigment Epithelium. Invest Ophthalmol Vis Sci 2017; 58:2366-2387. [PMID: 28437526 PMCID: PMC5403116 DOI: 10.1167/iovs.16-21162] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Purpose Prominin-1 (Prom1) is a transmembrane glycoprotein, which is expressed in stem cell lineages, and has recently been implicated in cancer stem cell survival. Mutations in the Prom1 gene have been shown to disrupt photoreceptor disk morphogenesis and cause an autosomal dominant form of Stargardt-like macular dystrophy (STGD4). Despite the apparent structural role of Prom1 in photoreceptors, its role in other cells of the retina is unknown. The purpose of this study is to investigate the role of Prom1 in the highly metabolically active cells of the retinal pigment epithelium (RPE). Methods Lentiviral siRNA and the genome editing CRISPR/Cas9 system were used to knockout Prom1 in primary RPE and ARPE-19 cells, respectively. Western blotting, confocal microscopy, and flow sight imaging cytometry assays were used to quantify autophagy flux. Immunoprecipitation was used to detect Prom1 interacting proteins. Results Our studies demonstrate that Prom1 is primarily a cytosolic protein in the RPE. Stress signals and physiological aging robustly increase autophagy with concomitant upregulation of Prom1 expression. Knockout of Prom1 increased mTORC1 and mTORC2 signaling, decreased autophagosome trafficking to the lysosome, increased p62 accumulation, and inhibited autophagic puncta induced by activators of autophagy. Conversely, ectopic overexpression of Prom1 inhibited mTORC1 and mTORC2 activities, and potentiated autophagy flux. Through interactions with p62 and HDAC6, Prom1 regulates autophagosome maturation and trafficking, suggesting a new cytoplasmic role of Prom1 in RPE function. Conclusions Our results demonstrate that Prom1 plays a key role in the regulation of autophagy via upstream suppression of mTOR signaling and also acting as a component of a macromolecular scaffold involving p62 and HDAC6.
Collapse
Affiliation(s)
- Sujoy Bhattacharya
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Jinggang Yin
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Christina S Winborn
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Qiuhua Zhang
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Junming Yue
- Department of Pathology, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Edward Chaum
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee, United States 3Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| |
Collapse
|
40
|
Regulation of Autophagy by MiRNAs and Their Emerging Roles in Tumorigenesis and Cancer Treatment. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 334:1-26. [PMID: 28838537 DOI: 10.1016/bs.ircmb.2017.03.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Autophagy is a conserved catabolic process for the degradation and recycling of cytosolic components or organelles through a lysosome-dependent pathway. Autophagy can be induced in response to multiple stress conditions, such as nutrient deprivation, hypoxia, energy depletion, etc. As a result, autophagy can regulate many biological processes, including cell survival, metabolism, differentiation, senescence, and cell death. MicroRNAs (MiRNAs) are small noncoding molecules that regulate gene expression by silencing mRNA targets. MiRNA dysregulation exhibits great regulatory potential during organismal development, hematopoiesis, immunity, cell proliferation and death, and autophagy. Recently, increasing studies have linked MiRNAs to autophagic regulation during cancer initiation and development. Although the relationship between MiRNAs and autophagy is quite complicated and has not been well elucidated, MiRNAs may underlie key aspects of autophagy and cancer biology. Increasing evidence shows that MiRNAs play important roles as both oncogenic MiRNAs and tumor suppressive MiRNAs in cancer initiation and development. Thus, understanding the novel relationship between MiRNAs and autophagy may allow us to develop promising cancer biomarkers and therapeutic targets.
Collapse
|
41
|
Abstract
: The increased prevalence of age-related comorbidities and mortality is worrisome in ageing HIV-infected patients. Here, we aim to analyse the different ageing mechanisms with regard to HIV infection. Ageing results from the time-dependent accumulation of random cellular damage. Epigenetic modifications and mitochondrial DNA haplogroups modulate ageing. In antiretroviral treatment-controlled patients, epigenetic clock appears to be advanced, and some haplogroups are associated with HIV infection severity. Telomere shortening is enhanced in HIV-infected patients because of HIV and some nucleoside analogue reverse transcriptase inhibitors. Mitochondria-related oxidative stress and mitochondrial DNA mutations are increased during ageing and also by some nucleoside analogue reverse transcriptase inhibitors. Overall, increased inflammation or 'inflammageing' is a major driver of ageing and could result from cell senescence with secreted proinflammatory mediators, altered gut microbiota, and coinfections. In HIV-infected patients, the level of inflammation and innate immunity activation is enhanced and related to most comorbidities and to mortality. This status could result, in addition to age, from the virus itself or viral protein released from reservoirs, from HIV-enhanced gut permeability and dysbiosis, from antiretroviral treatment, from frequent cytomegalovirus and hepatitis C virus coinfections, and also from personal and environmental factors, as central fat accumulation or smoking. Adaptive immune activation and immunosenescence are associated with comorbidities and mortality in the general population but are less predictive in HIV-infected patients. Biomarkers to evaluate ageing in HIV-infected patients are required. Numerous systemic or cellular inflammatory, immune activation, oxidative stress, or senescence markers can be tested in serum or peripheral blood mononuclear cells. The novel European Study to Establish Biomarkers of Human Ageing MARK-AGE algorithm, evaluating the biological age, is currently assessed in HIV-infected patients and reveals an advanced biological age. Some enhanced inflammatory or innate immune activation markers are interesting but still not validated for the patient's follow-up. To be able to assess patients' biological age is an important objective to improve their healthspan.
Collapse
|
42
|
Abstract
Autophagy, a highly conserved mechanism for cell survival, emerges as an important pathway in many biological processes and diseases conditions. Studies of cultured renal cells, human kidney tissues and experimental animal models implicate that autophagy regulation is the critical aspects in chronic kidney diseases (CKD). Here, we summarize the current studies on the role of autophagy in CKD. Unveiling the precise regulation mechanism of autophagy in CKD is essential for developing potential prevention, diagnostic and therapeutic targets of these sticky clinical challenges.
Collapse
Affiliation(s)
- Xu Deng
- a Department of Nephrology , Children's Hospital of Nanjing Medical University , Nanjing , China.,b Jiangsu Key Laboratory of Pediatrics , Nanjing Medical University , Nanjing , China
| | - Yifan Xie
- a Department of Nephrology , Children's Hospital of Nanjing Medical University , Nanjing , China.,b Jiangsu Key Laboratory of Pediatrics , Nanjing Medical University , Nanjing , China
| | - Aihua Zhang
- a Department of Nephrology , Children's Hospital of Nanjing Medical University , Nanjing , China.,b Jiangsu Key Laboratory of Pediatrics , Nanjing Medical University , Nanjing , China
| |
Collapse
|
43
|
Sun X, Du F, Liu S. Modulation of autophagy in exJSRV-env-transfected cells through the Akt/mTOR and MAPK signaling pathway. Biochem Biophys Res Commun 2017; 485:672-678. [PMID: 28235485 DOI: 10.1016/j.bbrc.2017.02.099] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 02/18/2017] [Indexed: 12/17/2022]
Abstract
The envelope (Env) of Jaagsiekte sheep retrovirus (JSRV) is an oncoprotein of ovine pulmonary adenocarcinoma (OPA). Autophagy is involved in different cancers, but how it is carcinogenic in JSRV Env is unclear. Modulation of autophagy in exJSRV-env-NM-transfected cells through the Akt/mTOR and MAPK signaling pathway was studied, and we observed strong positive labeling of p-Akt, p-mTOR, p-MEK1/2, p-ERK1/2, p-p38 and p-JNK in tumor cells and typical type II pneumocytes in naturally infected OPA lung tissues, which was co-aligned with JSRV-Env positive cells as shown by immunohistochemical and microscopic analysis. Akt/mTOR and MAPK pathways were activated in OPA lung and JSRV-Env transfected NIH 3T3 cells. Decreased Beclin1 and LC3 II/I suggested that autophagy was inhibited in OPA lung and JSRV-Env transfected NIH 3T3 cells. Beclin1 and LC3 II/I increased in JSRV-Env transfected NIH3T3 cells treated with mTOR inhibitor (rapamycin), ERK1/2 inhibitor (PD 98059), p38 inhibitor (SB 203580) and JNK inhibitor (SP 600125), suggesting that Akt/mTOR and MAPK pathways were responsible for JSRV-Env decreased autophagy. In conclusion, JSRV Env decreased autophagy in JSRV-Env transfected NIH3T3 cells through Akt/mTOR and MAPK pathways, in particular, JNK and p38 pathways.
Collapse
Affiliation(s)
- Xiaolin Sun
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Hohhot 010018, China.
| | - Fangyuan Du
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Hohhot 010018, China
| | - Shuying Liu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Hohhot 010018, China.
| |
Collapse
|
44
|
Vaiserman AM, Lushchak OV, Koliada AK. Anti-aging pharmacology: Promises and pitfalls. Ageing Res Rev 2016; 31:9-35. [PMID: 27524412 DOI: 10.1016/j.arr.2016.08.004] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 08/06/2016] [Accepted: 08/09/2016] [Indexed: 12/12/2022]
Abstract
Life expectancy has grown dramatically in modern times. This increase, however, is not accompanied by the same increase in healthspan. Efforts to extend healthspan through pharmacological agents targeting aging-related pathological changes are now in the spotlight of geroscience, the main idea of which is that delaying of aging is far more effective than preventing the particular chronic disorders. Currently, anti-aging pharmacology is a rapidly developing discipline. It is a preventive field of health care, as opposed to conventional medicine which focuses on treating symptoms rather than root causes of illness. A number of pharmacological agents targeting basic aging pathways (i.e., calorie restriction mimetics, autophagy inducers, senolytics etc.) are now under investigation. This review summarizes the literature related to advances, perspectives and challenges in the field of anti-aging pharmacology.
Collapse
Affiliation(s)
| | - Oleh V Lushchak
- Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | | |
Collapse
|
45
|
Gong L, Di C, Xia X, Wang J, Chen G, Shi J, Chen P, Xu H, Zhang W. AKT/mTOR signaling pathway is involved in salvianolic acid B-induced autophagy and apoptosis in hepatocellular carcinoma cells. Int J Oncol 2016; 49:2538-2548. [PMID: 27779641 DOI: 10.3892/ijo.2016.3748] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 10/17/2016] [Indexed: 11/06/2022] Open
Abstract
Chinese medicines are emerging as an attractive new generation of anticancer drugs. Here, we explored the impact of salvianolic acid B (Sal B), the major water-soluble compounds of Danshen, on apoptosis and autophagy of human hepatocellular carcinoma cells (HCC). We also investigated the related molecular mechanisms. We found that Sal B exhibits potent ability to inhibit HCC cells viability in a concentration-dependent manner, and to induce apoptosis via the mitochondrial apoptosis pathway. Additionally, Sal B could also induce autophagy. Furthermore, pretreatment with the autophagy inhibitor chloroquine or 3-methyladenine showed the potential in attenuating the apoptosis rate induced by Sal B. Mechanistically, Sal B treatment inhibited the AKT/mTOR signaling cascade in vitro. Overexpression of AKT abolished the effects of Sal B on HCC cells, suggesting a critical role of the AKT/mTOR signaling pathway in Sal B-induced biological effects. Our results indicated that the mitochondrial pathway was involved in Sal B-induced apoptosis of HCC cells. Moreover, the AKT/mTOR signaling pathway was involved in Sal B-induced autophagy, which promoted apoptosis. This study may provide a promising strategy for using Sal B as a chemotherapeutic agent for patients with HCC.
Collapse
Affiliation(s)
- Ling Gong
- Department of Liver Diseases, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, P.R. China
| | - Chunhong Di
- Department of Laboratory, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, P.R. China
| | - Xiaofang Xia
- Department of Liver Diseases, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, P.R. China
| | - Jie Wang
- Department of Liver Diseases, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, P.R. China
| | - Gongying Chen
- Department of Liver Diseases, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, P.R. China
| | - Junping Shi
- Department of Liver Diseases, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang 310015, P.R. China
| | - Pengshuai Chen
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Hui Xu
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Weibing Zhang
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
46
|
Meo-Evoli N, Almacellas E, Massucci FA, Gentilella A, Ambrosio S, Kozma SC, Thomas G, Tauler A. V-ATPase: a master effector of E2F1-mediated lysosomal trafficking, mTORC1 activation and autophagy. Oncotarget 2016; 6:28057-70. [PMID: 26356814 PMCID: PMC4695044 DOI: 10.18632/oncotarget.4812] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 07/30/2015] [Indexed: 11/26/2022] Open
Abstract
In addition to being a master regulator of cell cycle progression, E2F1 regulates other associated biological processes, including growth and malignancy. Here, we uncover a regulatory network linking E2F1 to lysosomal trafficking and mTORC1 signaling that involves v-ATPase regulation. By immunofluorescence and time-lapse microscopy we found that E2F1 induces the movement of lysosomes to the cell periphery, and that this process is essential for E2F1-induced mTORC1 activation and repression of autophagy. Gain- and loss-of-function experiments reveal that E2F1 regulates v-ATPase activity and inhibition of v-ATPase activity repressed E2F1-induced lysosomal trafficking and mTORC1 activation. Immunoprecipitation experiments demonstrate that E2F1 induces the recruitment of v-ATPase to lysosomal RagB GTPase, suggesting that E2F1 regulates v-ATPase activity by enhancing the association of V0 and V1 v-ATPase complex. Analysis of v-ATPase subunit expression identified B subunit of V0 complex, ATP6V0B, as a transcriptional target of E2F1. Importantly, ATP6V0B ectopic-expression increased v-ATPase and mTORC1 activity, consistent with ATP6V0B being responsible for mediating the effects of E2F1 on both responses. Our findings on lysosomal trafficking, mTORC1 activation and autophagy suppression suggest that pharmacological intervention at the level of v-ATPase may be an efficacious avenue for the treatment of metastatic processes in tumors overexpressing E2F1.
Collapse
Affiliation(s)
- Nathalie Meo-Evoli
- Departament de Bioquímica i Biologia Molecular, Facultat de Farmàcia, Universitat de Barcelona, 08028 Barcelona, Catalunya, Spain.,Laboratory of Cancer Metabolism, IDIBELL, Hospital Duran i Reynals, 08908 L'Hospitalet de Llobregat, Barcelona, Catalunya, Spain
| | - Eugènia Almacellas
- Departament de Bioquímica i Biologia Molecular, Facultat de Farmàcia, Universitat de Barcelona, 08028 Barcelona, Catalunya, Spain.,Laboratory of Cancer Metabolism, IDIBELL, Hospital Duran i Reynals, 08908 L'Hospitalet de Llobregat, Barcelona, Catalunya, Spain
| | | | - Antonio Gentilella
- Laboratory of Cancer Metabolism, IDIBELL, Hospital Duran i Reynals, 08908 L'Hospitalet de Llobregat, Barcelona, Catalunya, Spain
| | - Santiago Ambrosio
- Unitat de Bioquímica, Dep. Ciències Fisiològiques II, Facultat de Medicina, Campus Universitari de Bellvitge - IDIBELL, Universitat de Barcelona, 08908 L'Hospitalet de Llobregat, Barcelona, Catalunya, Spain
| | - Sara C Kozma
- Laboratory of Cancer Metabolism, IDIBELL, Hospital Duran i Reynals, 08908 L'Hospitalet de Llobregat, Barcelona, Catalunya, Spain.,Laboratory of Cancer Metabolism, Institut Català d'Oncologia, Hospital Duran i Reynals, 08908 L'Hospitalet de Llobregat, Barcelona, Catalunya, Spain.,Division of Hematology and Oncology, Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio 45267, USA
| | - George Thomas
- Laboratory of Cancer Metabolism, IDIBELL, Hospital Duran i Reynals, 08908 L'Hospitalet de Llobregat, Barcelona, Catalunya, Spain.,Unitat de Bioquímica, Dep. Ciències Fisiològiques II, Facultat de Medicina, Campus Universitari de Bellvitge - IDIBELL, Universitat de Barcelona, 08908 L'Hospitalet de Llobregat, Barcelona, Catalunya, Spain.,Laboratory of Cancer Metabolism, Institut Català d'Oncologia, Hospital Duran i Reynals, 08908 L'Hospitalet de Llobregat, Barcelona, Catalunya, Spain.,Division of Hematology and Oncology, Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio 45267, USA
| | - Albert Tauler
- Departament de Bioquímica i Biologia Molecular, Facultat de Farmàcia, Universitat de Barcelona, 08028 Barcelona, Catalunya, Spain.,Laboratory of Cancer Metabolism, IDIBELL, Hospital Duran i Reynals, 08908 L'Hospitalet de Llobregat, Barcelona, Catalunya, Spain
| |
Collapse
|
47
|
Langer R, Streutker CJ, Swanson PE. Autophagy and its current relevance to the diagnosis and clinical management of esophageal diseases. Ann N Y Acad Sci 2016; 1381:113-121. [PMID: 27526024 DOI: 10.1111/nyas.13190] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/25/2016] [Accepted: 07/07/2016] [Indexed: 12/26/2022]
Abstract
Autophagy is an evolutionarily conserved cell survival program that degrades dysfunctional organelles and misfolded or long-lived proteins through the formation of lysosomes. Basal autophagy helps to maintain cellular homeostasis, while additional autophagy can be induced under cellular stress conditions. Autophagy has shown to be involved in a variety of diseases, such as inflammation, autoimmune diseases, degeneration, and cancer. We review the relevance of autophagy to the diagnosis and clinical management of esophageal diseases with the following questions in mind. What is autophagy and can/should we detect it in routine pathology specimens? What is the role of autophagy in gastroesophageal reflux disease/inflammatory esophageal disease? What role may autophagy play in the interaction between pro- and antiapoptotic pathways in esophageal malignancies and treatment?
Collapse
Affiliation(s)
- Rupert Langer
- Institute of Pathology, University of Bern, Bern, Switzerland.
| | - Catherine J Streutker
- Li Ka Shing Institute, St. Michael's Hospital and Department of Laboratory Medicine and Pathobiology University of Toronto, Toronto, Canada
| | - Paul E Swanson
- Cumming School of Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
48
|
Xia Q, Zheng Y, Jiang W, Huang Z, Wang M, Rodriguez R, Jin X. Valproic acid induces autophagy by suppressing the Akt/mTOR pathway in human prostate cancer cells. Oncol Lett 2016; 12:1826-1832. [PMID: 27588130 PMCID: PMC4998110 DOI: 10.3892/ol.2016.4880] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/10/2016] [Indexed: 01/28/2023] Open
Abstract
Previous studies have demonstrated that the chronic administration of valproic acid (VPA) suppresses angiogenesis in vivo; however, the mechanisms implicated in VPA-induced autophagy remain unclear. The current study aimed to assess VPA-induced autophagy in three prostate cancer cell lines (PC3, DU145 and LNCaP), in addition to analyzing the Akt/mammalian target of rapamycin (mTOR) signal pathway. Prostate cancer cell lines were cultured with various doses of VPA. Cell cycle was analyzed using flow cytometry, and autophagy markers [1A/1B-light chain 3 (LC3)-II and Beclin-1] were examined using transmission electron microscopy, fluorescent microscopy and western blotting. Activation of the Akt/mTOR signal pathway was also assessed by western blotting. The results demonstrated that VPA induced autophagosomes and suppressed the Akt/mTOR signal pathway. This was confirmed by detection of increased LC3-II and Beclin-1 in VPA-treated cells compared with untreated controls. Phosphorylated forms of Akt (PC3, P=0.048; DU145, P=0.045; LNCaP, P=0.039) and mTOR (PC3, P=0.012; DU145, P=0.41; LNCaP, P=0.35) were significantly reduced following VPA treatment. These results suggest that VPA may function as a histone deacetylase inhibitor, suppressing the growth of prostate cancer cells by modulating autophagy pathways, including inhibition of the Akt/mTOR pathway. Further experiments are required to determine the significance of all involved pathways regarding VPA-induced growth inhibition.
Collapse
Affiliation(s)
- Qinghua Xia
- Department of Minimally Invasive Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Yi Zheng
- Department of Emergency, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Wei Jiang
- Department of Minimally Invasive Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Zhongxian Huang
- Department of Urology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Muwen Wang
- Department of Minimally Invasive Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Ronald Rodriguez
- Department of Urology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Xunbo Jin
- Department of Minimally Invasive Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
49
|
Shen G, Ren H, Qiu T, Liang D, Xie B, Zhang Z, Yao Z, Yang Z, Jiang X. Implications of the Interaction Between miRNAs and Autophagy in Osteoporosis. Calcif Tissue Int 2016; 99:1-12. [PMID: 26922423 DOI: 10.1007/s00223-016-0122-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 02/15/2016] [Indexed: 01/08/2023]
Abstract
Imbalances between bone formation and resorption are the primary cause of osteoporosis. However, currently, a detailed molecular mechanism of osteoporosis is not available. Autophagy is the conserved process characterized by degrading and recycling aggregated proteins, intracellular pathogens, and damaged organelles. MicroRNAs (miRNAs) are novel regulatory factors that play important roles in numerous cellular processes, including autophagy, through the posttranscriptional regulation of gene expression. Conversely, autophagy plays a role in the regulation of miRNA homeostasis. Recent advances have revealed that both autophagy and miRNAs are involved in the maintenance of bone homoeostasis, whereas the role of the interaction of miRNAs with autophagy in osteoporosis remains unclear. In this paper, we review previous reports on autophagy, miRNAs, and their interaction in osteoporosis.
Collapse
Affiliation(s)
- Gengyang Shen
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Hui Ren
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Ting Qiu
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - De Liang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Bo Xie
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhida Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhensong Yao
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhidong Yang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xiaobing Jiang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- Laboratory Affiliated to National Key Discipline of Orthopaedic and Traumatology of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
50
|
Johansson I, Monsen VT, Pettersen K, Mildenberger J, Misund K, Kaarniranta K, Schønberg S, Bjørkøy G. The marine n-3 PUFA DHA evokes cytoprotection against oxidative stress and protein misfolding by inducing autophagy and NFE2L2 in human retinal pigment epithelial cells. Autophagy 2016; 11:1636-51. [PMID: 26237736 PMCID: PMC4590664 DOI: 10.1080/15548627.2015.1061170] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Accumulation and aggregation of misfolded proteins is a hallmark of several diseases collectively known as proteinopathies. Autophagy has a cytoprotective role in diseases associated with protein aggregates. Age-related macular degeneration (AMD) is the most common neurodegenerative eye disease that evokes blindness in elderly. AMD is characterized by degeneration of retinal pigment epithelial (RPE) cells and leads to loss of photoreceptor cells and central vision. The initial phase associates with accumulation of intracellular lipofuscin and extracellular deposits called drusen. Epidemiological studies have suggested an inverse correlation between dietary intake of marine n-3 polyunsaturated fatty acids (PUFAs) and the risk of developing neurodegenerative diseases, including AMD. However, the disease-preventive mechanism(s) mobilized by n-3 PUFAs is not completely understood. In human retinal pigment epithelial cells we find that physiologically relevant doses of the n-3 PUFA docosahexaenoic acid (DHA) induce a transient increase in cellular reactive oxygen species (ROS) levels that activates the oxidative stress response regulator NFE2L2/NRF2 (nuclear factor, erythroid derived 2, like 2). Simultaneously, there is a transient increase in intracellular protein aggregates containing SQSTM1/p62 (sequestosome 1) and an increase in autophagy. Pretreatment with DHA rescues the cells from cell cycle arrest induced by misfolded proteins or oxidative stress. Cells with a downregulated oxidative stress response, or autophagy, respond with reduced cell growth and survival after DHA supplementation. These results suggest that DHA both induces endogenous antioxidants and mobilizes selective autophagy of misfolded proteins. Both mechanisms could be relevant to reduce the risk of developing aggregate-associate diseases such as AMD.
Collapse
Affiliation(s)
- Ida Johansson
- a Department of Laboratory Medicine ; Children's and Women's Health; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway.,b Department of Technology ; University College of Sør-Trøndelag ; Trondheim , Norway.,c Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine ; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Vivi Talstad Monsen
- a Department of Laboratory Medicine ; Children's and Women's Health; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Kristine Pettersen
- a Department of Laboratory Medicine ; Children's and Women's Health; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway.,b Department of Technology ; University College of Sør-Trøndelag ; Trondheim , Norway.,c Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine ; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Jennifer Mildenberger
- b Department of Technology ; University College of Sør-Trøndelag ; Trondheim , Norway.,c Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine ; Norwegian University of Science and Technology ; Trondheim , Norway.,d Department of Cancer Research and Molecular Medicine ; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Kristine Misund
- d Department of Cancer Research and Molecular Medicine ; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway.,e KG Jebsen Center for Myeloma Research; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Kai Kaarniranta
- f Department of Ophthalmology ; Institute of Clinical Medicine; University of Eastern Finland ; Kuopio , Finland.,g Department of Ophthalmology ; Kuopio University Hospital ; Kuopio , Finland
| | - Svanhild Schønberg
- a Department of Laboratory Medicine ; Children's and Women's Health; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Geir Bjørkøy
- b Department of Technology ; University College of Sør-Trøndelag ; Trondheim , Norway.,c Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine ; Norwegian University of Science and Technology ; Trondheim , Norway
| |
Collapse
|