1
|
Yokoya M, Takata F, Iwao T, Matsumoto J, Tanaka Y, Aridome H, Yasunaga M, Mizoguchi J, Sano K, Dohgu S. α-Synuclein Degradation in Brain Pericytes Is Mediated via Akt, ERK, and p38 MAPK Signaling Pathways. Int J Mol Sci 2025; 26:1615. [PMID: 40004079 PMCID: PMC11855147 DOI: 10.3390/ijms26041615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/07/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Parkinson's disease (PD) is characterized by widespread distribution of Lewy bodies, which are composed of phosphorylated and aggregated forms of α-Synuclein (α-Syn), in the brain. Although the accumulation and propagation of α-Syn contribute to the development of PD, the involvement of the blood-brain barrier (BBB) in these processes remains unknown. Pericytes, one of the cell types that constitute the BBB, degrade various forms of α-Syn. However, the detailed mechanisms involved in α-Syn degradation by pericytes remain poorly understood. Therefore, in this study, we aimed to determine the ability of the BBB-constituting cells, particularly primary cultures of rat pericytes, brain endothelial cells, and astrocytes, to degrade α-Syn. After α-Syn uptake by the cells, intracellular α-Syn decreased only in pericytes. This pericyte-specific α-Syn decrease was inhibited by an autophagy inhibitor, bafilomycin A1, and a proteasome inhibitor, MG132. siRNA-mediated knockdown of degradation enzymes or familial PD-associated genes, including cathepsin D, DJ-1, and LRRK2, did not affect α-Syn clearance in pericytes. However, pharmacological inhibitors of Akt, ERK, and p38 MAPK inhibited α-Syn degradation by pericytes. In conclusion, our results suggest that α-Syn degradation by pericytes is mediated by an autophagy-lysosome system and a ubiquitin-proteasome system via α-Syn-activated Akt, ERK, and p38 MAPK signaling pathways.
Collapse
Affiliation(s)
- Miki Yokoya
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (M.Y.); (F.T.); (T.I.); (J.M.); (Y.T.); (H.A.); (M.Y.); (J.M.)
| | - Fuyuko Takata
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (M.Y.); (F.T.); (T.I.); (J.M.); (Y.T.); (H.A.); (M.Y.); (J.M.)
| | - Takuro Iwao
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (M.Y.); (F.T.); (T.I.); (J.M.); (Y.T.); (H.A.); (M.Y.); (J.M.)
| | - Junichi Matsumoto
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (M.Y.); (F.T.); (T.I.); (J.M.); (Y.T.); (H.A.); (M.Y.); (J.M.)
| | - Yasuyoshi Tanaka
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (M.Y.); (F.T.); (T.I.); (J.M.); (Y.T.); (H.A.); (M.Y.); (J.M.)
| | - Hisataka Aridome
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (M.Y.); (F.T.); (T.I.); (J.M.); (Y.T.); (H.A.); (M.Y.); (J.M.)
| | - Miho Yasunaga
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (M.Y.); (F.T.); (T.I.); (J.M.); (Y.T.); (H.A.); (M.Y.); (J.M.)
| | - Junko Mizoguchi
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (M.Y.); (F.T.); (T.I.); (J.M.); (Y.T.); (H.A.); (M.Y.); (J.M.)
| | - Kazunori Sano
- Department of Physiology and Pharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan;
| | - Shinya Dohgu
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (M.Y.); (F.T.); (T.I.); (J.M.); (Y.T.); (H.A.); (M.Y.); (J.M.)
| |
Collapse
|
2
|
Wu LY, Zhang JL, Zeeshan M, Zhou Y, Zhang YT, He WT, Jin N, Dai Y, Chi W, Ou Z, Dong GH, Lin LZ. Caspase-8 promotes NLRP3 inflammasome activation mediates eye development defects in zebrafish larvae exposed to perfulorooctane sulfonate (PFOS). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 356:124252. [PMID: 38815886 DOI: 10.1016/j.envpol.2024.124252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/18/2024] [Accepted: 05/26/2024] [Indexed: 06/01/2024]
Abstract
Epidemiological evidence showed that serum high perfluorooctane sulfonate (PFOS) levels are associated with multiple eye related diseases, but the potential underlying molecular mechanisms remain poorly understood. Zebrafish and photoreceptor cell (661w) models were used to investigate the molecular mechanism of PFOS induced eye development defects. Our results showed a novel molecular mechanism of PFOS-induced inflammation response-mediated photoreceptor cell death associated with eye development defects. Inhibition of Caspase-8 activation significantly decreased photoreceptor cell death in PFOS exposure. Mechanistically, Toll-like receptor 4 (TLR4) mediates activation of Caspase-8 promote activation of NLR family pyrin domain-containing 3 (NLRP3) inflammasome to elicit maturation of interleukin-1 beta (IL-1β) via Caspase-1 activation, facilitating photoreceptor cell inflammation damage in PFOS exposure. In addition, we also made a novel finding that Caspase-3 activation was increased via Caspase-8 activation and directly intensified cell death. Our results show the important role of Caspase-8 activation in PFOS induced eye development defects and highlight Caspase-8 mediated activation of the NLRP3 inflammation triggers activation of Caspase-1 and promote the maturation of IL-1β in retinal inflammatory injury.
Collapse
Affiliation(s)
- Lu-Yin Wu
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jing-Lin Zhang
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Mohammed Zeeshan
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Yang Zhou
- State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Environmental Protection, Guangzhou, 510655, China
| | - Yun-Ting Zhang
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Wan-Ting He
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Nanxiang Jin
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70210, Kuopio, Finland
| | - Ye Dai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, Guangdong, China
| | - Wei Chi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, Guangdong, China
| | - Zejin Ou
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Guang-Hui Dong
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Li-Zi Lin
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
3
|
Li L, Shi C, Dong F, Xu G, Lei M, Zhang F. Targeting pyroptosis to treat ischemic stroke: From molecular pathways to treatment strategy. Int Immunopharmacol 2024; 133:112168. [PMID: 38688133 DOI: 10.1016/j.intimp.2024.112168] [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: 01/17/2024] [Revised: 04/19/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
Ischemic stroke is the primary reason for human disability and death, but the available treatment options are limited. Hence, it is imperative to explore novel and efficient therapies. In recent years, pyroptosis (a pro-inflammatory cell death characterized by inflammation) has emerged as an important pathological mechanism in ischemic stroke that can cause cell death through plasma membrane rupture and release of inflammatory cytokines. Pyroptosis is closely associated with inflammation, which exacerbates the inflammatory response in ischemic stroke. The level of inflammasomes, GSDMD, Caspases, and inflammatory factors is increased after ischemic stroke, exacerbating brain injury by mediating pyroptosis. Hence, inhibition of pyroptosis can be a therapeutic strategy for ischemic stroke. In this review, we have summarized the relationship between pyroptosis and ischemic stroke, as well as a series of treatments to attenuate pyroptosis, intending to provide insights for new therapeutic targets on ischemic stroke.
Collapse
Affiliation(s)
- Lina Li
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Chonglin Shi
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Guangyu Xu
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Mingcheng Lei
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China.
| |
Collapse
|
4
|
Ye H, Lu M, Tu C, Min L. Necroptosis in the sarcoma immune microenvironment: From biology to therapy. Int Immunopharmacol 2023; 122:110603. [PMID: 37467689 DOI: 10.1016/j.intimp.2023.110603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/23/2023] [Accepted: 07/02/2023] [Indexed: 07/21/2023]
Abstract
Apoptosis resistance remains a major obstacle to treatment failure in sarcoma. Necroptosis is a caspase-independent programmed cell death, investigated as a novel strategy to eradicate anti-apoptotic tumor cells. The process is mediated by the receptor-interacting proteins kinase family and mixed lineage kinase domain-like proteins, which is morphologically similar to necrosis. Recent studies suggest that necroptosis in the tumor microenvironment has pro- or anti-tumor effects on immune response and cancer development. Necroptosis-related molecules display a remarkable value in prognosis prediction and therapeutic response evaluation of sarcoma. Furthermore, the induction of tumor necroptosis has been explored as a feasible therapeutic strategy against sarcoma and to synergize with immunotherapy. This review discusses the dual roles of necroptosis in the immune microenvironment and tumor progression, and explores the potential of necroptosis as a new target for sarcoma treatment.
Collapse
Affiliation(s)
- Huali Ye
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Minxun Lu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Chongqi Tu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Li Min
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
5
|
Zhang L, Cui T, Wang X. The Interplay Between Autophagy and Regulated Necrosis. Antioxid Redox Signal 2023; 38:550-580. [PMID: 36053716 PMCID: PMC10025850 DOI: 10.1089/ars.2022.0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022]
Abstract
Significance: Autophagy is critical to cellular homeostasis. Emergence of the concept of regulated necrosis, such as necroptosis, ferroptosis, pyroptosis, and mitochondrial membrane-permeability transition (MPT)-derived necrosis, has revolutionized the research into necrosis. Both altered autophagy and regulated necrosis contribute to major human diseases. Recent studies reveal an intricate interplay between autophagy and regulated necrosis. Understanding the interplay at the molecular level will provide new insights into the pathophysiology of related diseases. Recent Advances: Among the three forms of autophagy, macroautophagy is better studied for its crosstalk with regulated necrosis. Macroautophagy seemingly can either antagonize or promote regulated necrosis, depending upon the form of regulated necrosis, the type of cells or stimuli, and other cellular contexts. This review will critically analyze recent advances in the molecular mechanisms governing the intricate dialogues between macroautophagy and main forms of regulated necrosis. Critical Issues: The dual roles of autophagy, either pro-survival or pro-death characteristics, intricate the mechanistic relationship between autophagy and regulated necrosis at molecular level in various pathological conditions. Meanwhile, key components of regulated necrosis are also involved in the regulation of autophagy, which further complicates the interrelationship. Future Directions: Resolving the controversies over causation between altered autophagy and a specific form of regulated necrosis requires approaches that are more definitive, where rigorous evaluation of autophagic flux and the development of more reliable and specific methods to quantify each form of necrosis will be essential. The relationship between chaperone-mediated autophagy or microautophagy and regulated necrosis remains largely unstudied. Antioxid. Redox Signal. 38, 550-580.
Collapse
Affiliation(s)
- Lei Zhang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
| | - Taixing Cui
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Xuejun Wang
- Division of Basic Biomedical Sciences, The University of South Dakota Sanford School of Medicine, Vermillion, South Dakota, USA
| |
Collapse
|
6
|
Zhao W, Chen C, Zhou J, Chen X, Cai K, Shen M, Chen X, Jiang L, Wang G. Inhibition of Autophagy Promotes the Anti-Tumor Effect of Metformin in Oral Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14174185. [PMID: 36077722 PMCID: PMC9454503 DOI: 10.3390/cancers14174185] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Oral Squamous Cell Carcinoma (OSCC) is the most common malignant tumor in the head and neck. Due to its high malignancy and easy recurrence, the five-year survival rate is only 50–60%. Currently, commonly used chemotherapy drugs for OSCC include cisplatin, paclitaxel, and fluorouracil, which are highly cytotoxic and cause drug resistance in patients. Therefore, a safe and effective treatment strategy for OSCC is urgent. To address this issue, our study investigated the anti-tumor activity of metformin (the first-line diabetes drug) in OSCC. We found that metformin could inhibit OSCC cell proliferation by promoting apoptosis and blocking the cell cycle in G1 phase. Additionally, we also found that metformin could induce protective autophagy of OSCC cells. After inhibiting autophagy with hydroxychloroquine (HCQ), the metformin-induced apoptosis was enhanced. In vitro, metformin inhibited the growth of subcutaneous xenograft tumor in nude mice and HCQ enhanced this effect of metformin. Therefore, metformin combined with HCQ may become a safe and effective treatment strategy for OSCC.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Chen Chen
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Jianjun Zhou
- Department of Stomatology, 900 Hospital of the Joint Logistics Team, Fuzhou 350025, China
| | - Xiaoqing Chen
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Kuan Cai
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Miaomiao Shen
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Xuan Chen
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Lei Jiang
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Guodong Wang
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
- Correspondence:
| |
Collapse
|
7
|
Chen YS, Chuang WC, Kung HN, Cheng CY, Huang DY, Sekar P, Lin WW. Pan-Caspase Inhibitor zVAD Induces Necroptotic and Autophagic Cell Death in TLR3/4-Stimulated Macrophages. Mol Cells 2022; 45:257-272. [PMID: 34949739 PMCID: PMC9001149 DOI: 10.14348/molcells.2021.0193] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/24/2021] [Accepted: 10/15/2021] [Indexed: 11/27/2022] Open
Abstract
In addition to inducing apoptosis, caspase inhibition contributes to necroptosis and/or autophagy depending on the cell type and cellular context. In macrophages, necroptosis can be induced by co-treatment with Toll-like receptor (TLR) ligands (lipopolysaccharide [LPS] for TLR4 and polyinosinic-polycytidylic acid [poly I:C] for TLR3) and a cell-permeable pan-caspase inhibitor zVAD. Here, we elucidated the signaling pathways and molecular mechanisms of cell death. We showed that LPS/zVAD- and poly I:C/zVAD-induced cell death in bone marrow-derived macrophages (BMDMs) was inhibited by receptor-interacting protein kinase 1 (RIP1) inhibitor necrostatin-1 and autophagy inhibitor 3-methyladenine. Electron microscopic images displayed autophagosome/autolysosomes, and immunoblotting data revealed increased LC3II expression. Although zVAD did not affect LPS- or poly I:C-induced activation of IKK, JNK, and p38, it enhanced IRF3 and STAT1 activation as well as type I interferon (IFN) expression. In addition, zVAD inhibited ERK and Akt phosphorylation induced by LPS and poly I:C. Of note, zVAD-induced enhancement of the IRF3/IFN/STAT1 axis was abolished by necrostatin-1, while zVAD-induced inhibition of ERK and Akt was not. Our data further support the involvement of autocrine IFNs action in reactive oxygen species (ROS)-dependent necroptosis, LPS/zVAD-elicited ROS production was inhibited by necrostatin-1, neutralizing antibody of IFN receptor (IFNR) and JAK inhibitor AZD1480. Accordingly, both cell death and ROS production induced by TLR ligands plus zVAD were abrogated in STAT1 knockout macrophages. We conclude that enhanced TRIF-RIP1-dependent autocrine action of IFNβ, rather than inhibition of ERK or Akt, is involved in TLRs/zVAD-induced autophagic and necroptotic cell death via the JAK/STAT1/ROS pathway.
Collapse
Affiliation(s)
- Yuan-Shen Chen
- Department of Neurosurgery, National Taiwan University Hospital Yunlin Branch, Douliu 64041, Taiwan
| | - Wei-Chu Chuang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
| | - Hsiu-Ni Kung
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei 10617, Taiwan
| | - Ching-Yuan Cheng
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
| | - Duen-Yi Huang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
| | - Ponarulselvam Sekar
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 11031, Taiwan
| | - Wan-Wan Lin
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 11031, Taiwan
- Department of Pharmacology, National Defense Medical Center, Taipei 11490, Taiwan
| |
Collapse
|
8
|
Needs SH, Bootman MD, Grotzke JE, Kramer HB, Allman SA. Off‐target inhibition of NGLY1 by the polycaspase inhibitor Z‐VAD‐fmk induces cellular autophagy. FEBS J 2022; 289:3115-3131. [PMID: 34995415 PMCID: PMC9304259 DOI: 10.1111/febs.16345] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 09/14/2021] [Accepted: 01/05/2022] [Indexed: 12/28/2022]
Affiliation(s)
- Sarah H. Needs
- School of Life, Health and Chemical Sciences The Open University Milton Keynes UK
- Reading School of Pharmacy University of Reading UK
| | - Martin D. Bootman
- School of Life, Health and Chemical Sciences The Open University Milton Keynes UK
| | | | - Holger B. Kramer
- Department of Physiology, Anatomy and Genetics University of Oxford UK
- MRC London Institute of Medical Sciences UK
| | - Sarah A. Allman
- School of Life, Health and Chemical Sciences The Open University Milton Keynes UK
- Reading School of Pharmacy University of Reading UK
- Leicester School of Pharmacy De Montfort University Leicester UK
| |
Collapse
|
9
|
Chen Y, Li P, Peng Y, Xie X, Zhang Y, Jiang Y, Li T, Qin X, Li S, Yang H, Wu C, Zheng C, Zhu J, You F, Liu Y. Protective autophagy attenuates soft substrate-induced apoptosis through ROS/JNK signaling pathway in breast cancer cells. Free Radic Biol Med 2021; 172:590-603. [PMID: 34242793 DOI: 10.1016/j.freeradbiomed.2021.07.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 07/03/2021] [Indexed: 02/06/2023]
Abstract
Tumor microenvironments are characterized not only in terms of chemical composition, but also by physical properties such as stiffness, which influences morphology, proliferation, and fate of tumor cells. However, the underlying mechanisms between matrix stiffness and the apoptosis-autophagy balance remain largely unexplored. In this study, we cultured human breast cancer MDA-MB-231 cells on rigid (57 kPa), stiff (38 kPa) or soft (10 kPa) substrates and demonstrated that increasing autophagy levels and autophagic flux in the cells cultured on soft substrates partly attenuated soft substrate-induced apoptosis. Mechanistically, this protective autophagy is regulated by intracellular reactive oxygen species (ROS) accumulation, which triggers the downstream signals of JNK, Bcl-2 and Beclin-1. More importantly, soft substrate-induced activation of ROS/JNK signaling promotes cell apoptosis through the mitochondrial pathway, whereas it increases protective autophagy by suppressing the interaction of Bcl-2 and Beclin-1. Taken together, our data suggest that JNK is the mediator of soft substrate-induced breast cancer cell apoptosis and autophagy which is likely to be the mechanism that partly attenuates mitochondrial apoptosis. This study provides new insights into the molecular mechanism by which autophagy plays a protective role against soft substrate-induced apoptosis in human breast cancer cells.
Collapse
Affiliation(s)
- Yu Chen
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Ping Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Yueting Peng
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Xiaoxue Xie
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Yixi Zhang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Ying Jiang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Tingting Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Xiang Qin
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Shun Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Hong Yang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Chunhui Wu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China
| | - Chuan Zheng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, 610072, Sichuan, PR China
| | - Jie Zhu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, 610072, Sichuan, PR China
| | - Fengming You
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, 610072, Sichuan, PR China
| | - Yiyao Liu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, PR China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, 610072, Sichuan, PR China.
| |
Collapse
|
10
|
Xu C, Chen S, Xu M, Chen X, Wang X, Zhang H, Dong X, Zhang R, Chen X, Gao W, Huang S, Chen L. Cadmium Impairs Autophagy Leading to Apoptosis by Ca 2+-Dependent Activation of JNK Signaling Pathway in Neuronal Cells. Neurochem Res 2021; 46:2033-2045. [PMID: 34021889 DOI: 10.1007/s11064-021-03341-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 01/04/2023]
Abstract
Autophagy, a process for self-degradation of intracellular components and dysfunctional organelles, is closely related with neurodegenerative diseases. It has been shown that cadmium (Cd) induces neurotoxicity partly by impairing autophagy. However, the underlying mechanism is not fully elucidated. In this study, we show that Cd induced expansion of autophagosomes with a concomitant abnormal expression of autophagy-related (Atg) proteins in PC12 cells and primary murine neurons. 3-MA, a classical inhibitor of autophagy, attenuated Cd-induced expansion of autophagosomes and apoptosis in the cells. Further investigation demonstrated that Cd activated JNK pathway contributing to autophagosome expansion-dependent neuronal apoptosis. This is supported by the findings that pharmacological inhibition of JNK with SP600125 or expression of dominant negative c-Jun markedly attenuated Cd-induced expansion of autophagosomes and abnormal expression of Atg proteins, as well as apoptosis in PC12 cells and/or primary neurons. Furthermore, we noticed that chelating intracellular free Ca2+ ([Ca2+]i) with BAPTA/AM profoundly blocked Cd-elicited activation of JNK pathway and consequential expansion of autophagosomes, abnormal expression of Atg proteins, and apoptosis in the neuronal cells. Similar events were also seen following prevention of [Ca2+]i elevation with EGTA or 2-APB, implying a Ca2+-dependent mechanism involved. Taken together, the results indicate that Cd impairs autophagy leading to apoptosis by Ca2+-dependent activation of JNK signaling pathway in neuronal cells. Our findings highlight that manipulation of intracellular Ca2+ level and/or JNK activity to ameliorate autophagy may be a promising intervention against Cd-induced neurotoxicity and neurodegeneration.
Collapse
Affiliation(s)
- Chong Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Sujuan Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, 453003, People's Republic of China
| | - Ming Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
| | - Xiaoling Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
| | - Xiaoxue Wang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
| | - Hai Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
| | - Xiaoqing Dong
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
| | - Ruijie Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
| | - Xin Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
| | - Wei Gao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71130-3932, USA.
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, 71130-3932, USA.
| | - Long Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
11
|
Pan X, He G, Hai B, Liu Y, Bian L, Yong L, Zhang H, Yang C, Du C, Mao T, Ma Y, Jia F, Dou X, Zhai S, Liu X. VPS34 regulates dynamin to determine the endocytosis of mitochondria-targeted zinc oxide nanoparticles in human osteosarcoma cells. J Mater Chem B 2021; 9:2641-2655. [PMID: 33683276 DOI: 10.1039/d1tb00226k] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In our previous study, zinc oxide nanoparticles (ZnO NPs) presented satisfying therapeutic effects with cancer cell selectivity in osteosarcoma cells and, thus, have been considered as a potential nanomedicine for human osteosarcoma treatment. However, the poorly investigated internalization process, including their endocytic pathway into tumor cells and intracellular fate, limits the clinical application. Here, we further clarified these aspects. First, ZnO NPs were rapidly internalized by osteosarcoma cells and accumulated in mitochondria, before being entrapped into lysosomes. Second, dynasore (a dynamin inhibitor) was demonstrated to be the most effective in blocking ZnO NP uptake and rescuing ZnO NP-induced osteosarcoma cell autophagic death and apoptosis. Third, we confirmed the key role of dynamin 2 in ZnO NP endocytosis and subsequent autophagic cell death in vitro and in vivo. Furthermore, we proved that VPS34 transferred from cell cytoplasm to cell membrane to interact with dynamin under ZnO NP treatment. Altogether, combined with our previous study, the current research further revealed that ZnO NPs entered human osteosarcoma cells through the VPS34/dynamin 2-dependent endocytic pathway, directly targeting and damaging the mitochondria before being entrapped into the lysosomes, thereby initiating mitophagy-Zn2+-reactive oxygen species-mitophagy axis mediated cell apoptosis.
Collapse
Affiliation(s)
- Xiaoyu Pan
- Department of Orthopedics, Beijing International Cooperation Base for Science and Technology on Biomimetic Titanium Orthopedic Implants, Peking University Third Hospital, Beijing 100191, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Wang T, Wu X, Al Rudaisat M, Song Y, Cheng H. Curcumin induces G2/M arrest and triggers autophagy, ROS generation and cell senescence in cervical cancer cells. J Cancer 2020; 11:6704-6715. [PMID: 33046993 PMCID: PMC7545669 DOI: 10.7150/jca.45176] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 09/11/2020] [Indexed: 12/13/2022] Open
Abstract
Our study explored the tumor-suppressive effect of curcumin on cervical cancer cells. Cervical cancer is one of the most common cancers among women worldwide. Acquired resistance to chemotherapeutics and toxicity of such drugs has undermined the effectiveness of cervical cancer treatments. Therefore, the identification of novel chemotherapeutics is key to improving the survival of patients with cervical cancer. Curcumin has been shown to have various bioactivities, including antioxidant and antitumor effects; however, its effect on cervical cancer remains elusive. Here, we used the SiHa human cervical cancer cell line to test various concentrations of curcumin on the proliferation and apoptosis of cervical cancer cells. The involvement of autophagy and reactive oxygen species (ROS) in these effects were also tested by using specific autophagy inhibitors and ROS scavengers. Our results showed that curcumin induced ROS accumulation, apoptosis, autophagy, cell cycle arrest, and cellular senescence accompanied by upregulation of p53 and p21 proteins in SiHa cells.
Collapse
Affiliation(s)
- Tuan Wang
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 3 Qingchun Road, Zhejiang, 310016, P.R. China
| | - Xia Wu
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 3 Qingchun Road, Zhejiang, 310016, P.R. China
| | - Mus'ab Al Rudaisat
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 3 Qingchun Road, Zhejiang, 310016, P.R. China
| | - Yinjing Song
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 3 Qingchun Road, Zhejiang, 310016, P.R. China
| | - Hao Cheng
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 3 Qingchun Road, Zhejiang, 310016, P.R. China
| |
Collapse
|
13
|
Hassanpour M, Rezaie J, Darabi M, Hiradfar A, Rahbarghazi R, Nouri M. Autophagy modulation altered differentiation capacity of CD146 + cells toward endothelial cells, pericytes, and cardiomyocytes. Stem Cell Res Ther 2020; 11:139. [PMID: 32216836 PMCID: PMC7099797 DOI: 10.1186/s13287-020-01656-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/11/2020] [Accepted: 03/18/2020] [Indexed: 12/15/2022] Open
Abstract
Background To date, many attempts are employed to increase the regenerative potential of stem cells. In this study, we evaluated the hypothesis of whether an autophagy modulation could alter differentiation potency of CD146+ cells into mature pericyte, endothelial, and cardiomyocyte lineage. Methods In this study, CD146+cells were enriched from the human bone marrow aspirates and trans-differentiated into mature endothelial cells, pericytes, and cardiomyocytes after exposure to autophagy stimulator (50-μM Met)/inhibitor (15-μM HCQ). The protein levels of autophagy proteins were monitored by western blotting. NO content was measured using the Griess assay. Using real-time PCR assay and western blotting, we monitored the lineage protein and gene levels. Pro-inflammatory cytokine and angiocrine factors were measured by ELISA. The fatty acid change was determined by gas chromatography. We also measured exosome secretion capacity by measuring AChE activity and real-time PCR assay. Result Data revealed the modulation of autophagy factors, Beclin-1, P62, and LC3 II/I ratio in differentiating CD146+ cells after exposure to Met and HCQ (p < 0.05). The inhibition of autophagy increased NO content compared to the Met-treated cells (p < 0.05). Real-time PCR analysis showed that the treatment of CD146+ cells with autophagy modulators altered the expression of VE-cadherin, cTnI, and α-SMA (p < 0.05). Met increased the expression of VE-cadherin, α-SMA, and cTnI compared to the HCQ-treated cells (p < 0.05) while western blotting revealed the protein synthesis of all lineage-specific proteins under the stimulation and inhibition of autophagy. None statistically significant differences were found in the levels of Tie-1, Tie-2, VEGFR-1, and VEGFR-2 after autophagy modulation. Fatty acid profile analysis revealed the increase of unsaturated fatty acids after exposure to HCQ (p < 0.05). The treatment of cells with HCQ increased the levels of TNF-α and IL-6 compared to the Met-treated cells. Data revealed the increase of exosome biogenesis and secretion to the supernatant in cells treated with HCQ compared to the Met groups (p < 0.05). Conclusions In summary, autophagy modulation could alter differentiation potency of CD146+cells which is important in cardiac regeneration.
Collapse
Affiliation(s)
- Mehdi Hassanpour
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, 5166614756, Iran.,Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Masoud Darabi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirataollah Hiradfar
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, 5166614756, Iran. .,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Nouri
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. .,Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, 5166614756, Iran.
| |
Collapse
|
14
|
All-trans-retinal induces autophagic cell death via oxidative stress and the endoplasmic reticulum stress pathway in human retinal pigment epithelial cells. Toxicol Lett 2020; 322:77-86. [PMID: 31931077 DOI: 10.1016/j.toxlet.2020.01.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/03/2020] [Accepted: 01/06/2020] [Indexed: 11/20/2022]
Abstract
Failure of all-trans-retinal (atRAL) clearance contributes to retina degeneration. However, whether autophagy can be activated by excess atRAL accumulation in retinal pigment epithelial (RPE) cells is not known. This study showed that atRAL provoked mitochondria-associated reactive oxygen species (ROS) production, activated the nuclear factor (erythroid-derived 2)-like 2 and apoptosis in a human RPE cell line, ARPE-19 cells. Moreover, we found that autophagic flux was functionally activated after atRAL treatment. The antioxidant N-acetylcysteine attenuated the expression of autophagy markers, suggesting that ROS triggered atRAL-activated autophagy. In addition, autophagic cell death was observed in atRAL-treated RPE cells, while inhibition of autophagy with 3-methyladenine or LC3, Beclin1, p62 silencing ameliorated atRAL-induced cytotoxicity. Suppression of autophagy quenched mitochondrial ROS and inhibited HO-1 and γ-GCSh expression, indicating that atRAL-activated autophagy enhances intracellular oxidative stress, thereby promoting RPE cell apoptosis. Furthermore, we found that inhibiting endoplasmic reticulum (ER) stress suppressed atRAL-induced mitochondrial ROS generation, subsequently attenuated autophagy and apoptosis in RPE cells. Taken together, these results suggest that atRAL-induced oxidative stress and ER stress modulate autophagy, which may contribute to RPE degeneration. There may be positive feedback regulatory mechanisms between atRAL-induced oxidative stress and autophagy or ER stress.
Collapse
|
15
|
Zhu H, Tang L, Zhang C, Wei B, Yang P, He D, Zheng L, Zhang Y. Synthesis of Chalcone Derivatives: Inducing Apoptosis of HepG2 Cells via Regulating Reactive Oxygen Species and Mitochondrial Pathway. Front Pharmacol 2019; 10:1341. [PMID: 31803052 PMCID: PMC6874057 DOI: 10.3389/fphar.2019.01341] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/22/2019] [Indexed: 12/27/2022] Open
Abstract
Chalcone derivatives, as a hot research field, exhibit a variety of physiological bioactivities and target multiple biological receptors. Based on the skeleton of (E)-1,3-diphenyl-2-propene-1-one, 14 chalcone derivatives were designed and synthesized, and evaluated as the antitumor candidates agents against four human cancer cell lines (A549, Hela, HepG2, and HL-60) as well as one normal cell line (WI-38). Among the title compounds, compound a14 showed better inhibitory activity against HepG2 cells (IC50 = 38.33 µM) and had relatively weak cytotoxicity towards normal cells WI-38 (IC50 = 121.29 µM). In this study, apoptosis, cycle arrest, assessment of reactive oxygen species (ROS) level, and measurement of mitochondrial membrane potential were adopted to explore the inhibitory mechanism of a14 towards HepG2. Compound a14 could effectively block the division of HepG2 cell lines in the G2/M phase and robustly induced generation of ROS, demonstrating that the generation of ROS induced by a14 was the main reason for resulting in the apoptosis of HepG2 cells. Moreover, the mitochondrial membrane potential (MMP) of HepG2 cells treated with a14 was significantly decreased, which was closely related to the enhanced ROS level. Furthermore, based on Western blot experiment, cell apoptosis induced by a14 also involved the expression of B-cell lymphoma-2 (Bcl-2) family and Caspase 3 protein. In summary, compound a14 could contribute to the apoptosis of HepG2 cells through regulating ROS-mitochondrial pathway, which provides valuable hints for the discovery of novel anti-tumor drug candidates.
Collapse
Affiliation(s)
- Hongtian Zhu
- Materia Medica Development Group, Institute of Medicinal Chemistry, Lanzhou University School of Pharmacy, Lanzhou, China.,Innovative Drug Research Department, Lanzhou Weihuan Biological Science and Technology Development Co, Ltd., Lanzhou, China
| | - Lei Tang
- Materia Medica Development Group, Institute of Medicinal Chemistry, Lanzhou University School of Pharmacy, Lanzhou, China.,Innovative Drug Research Department, Lanzhou Weihuan Biological Science and Technology Development Co, Ltd., Lanzhou, China
| | - Chenghong Zhang
- Materia Medica Development Group, Institute of Medicinal Chemistry, Lanzhou University School of Pharmacy, Lanzhou, China.,Innovative Drug Research Department, Lanzhou Weihuan Biological Science and Technology Development Co, Ltd., Lanzhou, China
| | - Baochu Wei
- Materia Medica Development Group, Institute of Medicinal Chemistry, Lanzhou University School of Pharmacy, Lanzhou, China.,Pharmacy Department, Lanzhou Pulmonary Hospital Pharmacy, Lanzhou, China
| | - Pingrong Yang
- Materia Medica Development Group, Institute of Medicinal Chemistry, Lanzhou University School of Pharmacy, Lanzhou, China.,NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Chinese Materia Medica and Prepared Slices), Gansu Institute for Drug Control, Lanzhou, China
| | - Dian He
- Materia Medica Development Group, Institute of Medicinal Chemistry, Lanzhou University School of Pharmacy, Lanzhou, China.,NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Chinese Materia Medica and Prepared Slices), Gansu Institute for Drug Control, Lanzhou, China
| | - Lifang Zheng
- Materia Medica Development Group, Institute of Medicinal Chemistry, Lanzhou University School of Pharmacy, Lanzhou, China
| | - Yang Zhang
- Innovative Drug Research Department, Lanzhou Weihuan Biological Science and Technology Development Co, Ltd., Lanzhou, China.,School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
16
|
Zhang L, Song Y, Chen L, Li D, Feng H, Lu Z, Fan T, Chen Z, Livingston MJ, Geng Q. MiR-20a-containing exosomes from umbilical cord mesenchymal stem cells alleviates liver ischemia/reperfusion injury. J Cell Physiol 2019; 235:3698-3710. [PMID: 31566731 DOI: 10.1002/jcp.29264] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSCs) have been proved to exert considerable therapeutic effects on ischemia-reperfusion (I/R)-induced injury, but the underlying mechanism remains unknown. In this study, we aimed to explore the potential molecular mechanism underlying the therapeutic effect of MSCs-derived exosome reinforced with miR-20a in reversing liver I/R injury. Quantitative real-time polymerase chain reaction, Western blot, and IHC were carried out to compare the differential expressions of miR-20a, Beclin-I, FAS, Caspase-3, mTOR and P62 in IR rats and normal rats. TUNEL was performed to assess IR-induced apoptosis in IR rats, and luciferase assay was used to confirm the inhibitory effect of miR-20a on Beclin-I and FAS expression. Among the 12 candidate microRNAs (miRNAs), miR-486, miR-25, miR-24, miR-20a,miR-466 and miR-433-3p were significantly downregulated in I/R. In particular, miR-20a, a miRNA highly expressed in umbilical cord-derived mesenchymal stem cells, was proved to bind to the 3' UTR of Beclin-I and FAS to exert an inhibitory effect on their expressions. Since Beclin-I and FAS were aberrantly upregulated in IR, exosomes separated from UC-MSCs showed therapeutic efficacy in reversing I/R induced apoptosis. In addition, exosomes reinforced with miR-20a and separated from UC-MSCs almost fully alleviated I/R injury. Furthermore, our results showed that miR-20a could alleviate the abnormal expression of genes related to apoptosis and autophagy, such as active Caspase-3, mTOR, P62, and LC3II. This study presented detailed evidence to clarify the mechanism underlying the therapeutic efficacy of UC-MSCs in the treatment of I/R injury.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yaolin Song
- Department of Thoracic Surgery, Ezhou Central Hospital, Ezhou, Hubei, China
| | - Lei Chen
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Donghang Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Haohao Feng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zilong Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Tao Fan
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zubin Chen
- Department of Hepatological Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Man J Livingston
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
17
|
Nicolini A, Ferrari P, Rossi G, Carpi A. Tumour growth and immune evasion as targets for a new strategy in advanced cancer. Endocr Relat Cancer 2018; 25:R577–R604. [PMID: 30306784 DOI: 10.1530/erc-18-0142] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
It has become clearer that advanced cancer, especially advanced breast cancer, is an entirely displayed pathological system that is much more complex than previously considered. However, the direct relationship between tumour growth and immune evasion can represent a general rule governing the pathological cancer system from the initial cancer cells to when the system is entirely displayed. Accordingly, a refined pathobiological model and a novel therapeutic strategy are proposed. The novel therapeutic strategy is based on therapeutically induced conditions (undetectable tumour burden and/or a prolonged tumour ‘resting state’), which enable an efficacious immune response in advanced breast and other types of solid cancers.
Collapse
Affiliation(s)
- Andrea Nicolini
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Paola Ferrari
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Giuseppe Rossi
- Unit of Epidemiology and Biostatistics, Institute of Clinical Physiology, National Council of Research, Pisa, Italy
| | - Angelo Carpi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
18
|
Ryan F, Khodagholi F, Dargahi L, Minai-Tehrani D, Ahmadiani A. Temporal Pattern and Crosstalk of Necroptosis Markers with Autophagy and Apoptosis Associated Proteins in Ischemic Hippocampus. Neurotox Res 2018; 34:79-92. [PMID: 29313217 DOI: 10.1007/s12640-017-9861-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 12/12/2022]
Abstract
Necroptosis, a novel type of programmed cell death, has been recently implicated as a possible mechanism for cerebral ischemia-reperfusion (I/R) injury. We herein studied time-dependent changes of necroptosis markers along with apoptosis- and autophagy-associated proteins in rat hippocampus at 1, 3, 6, 12, 24, and 48 h after global cerebral I/R injury. Furthermore, to determine the cross talk between autophagy and necroptosis, we examined the effects of pretreatment with bafilomycin-A1 (Baf-A1), as a late-stage autophagy inhibitor, on necroptosis. Highest levels of receptor-interacting protein 1 and 3 (RIP1 and RIP3), as key mediators of necroptosis, were observed at 24 h after reperfusion. Alongside, activity of glutamate dehydrogenase (GLUD1), downstream enzyme of RIP3, was increased. Peak time of necroptosis was subsequent to caspase-3-dependent cell death that peaked at 12 h of reperfusion but concurrent with autophagy. Administration of Baf-A1 could attenuate necroptosis, verified by decrease in RIP1 and RIP3 protein levels, as well as GLUD1 activity. However, there was no significant change in caspase-3-dependent cell death. Taken together, our results highlight that global cerebral I/R activates necroptosis that could be triggered by autophagy and interacts reversely with caspase-3-dependent apoptosis.
Collapse
Affiliation(s)
- Fari Ryan
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Dariush Minai-Tehrani
- Bioresearch Lab, Faculty of Biological Sciences, Shahid Beheshti University G.C, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Zhou J, Fan Y, Zhong J, Huang Z, Huang T, Lin S, Chen H. TAK1 mediates excessive autophagy via p38 and ERK in cisplatin-induced acute kidney injury. J Cell Mol Med 2018; 22:2908-2921. [PMID: 29504713 PMCID: PMC5908118 DOI: 10.1111/jcmm.13585] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 01/24/2018] [Indexed: 12/31/2022] Open
Abstract
The ability of cisplatin (cis-diamminedichloroplatinum II) toxicity to induce acute kidney injury (AKI) has attracted people's attention and concern for a long time, but its molecular mechanisms are still widely unknown. We found that the expression of transforming growth factor-β (TGF-β)-activated kinase 1 (TAK1) could be increased in kidneys of mice administrated with cisplatin. Autophagy is an evolutionarily conserved catabolic pathway and is involved in various acute and chronic injuries. Moreover, p38 MAPK (mitogen-activated protein kinase) and ERK regulate autophagy in response to various stimuli. Therefore, our hypothesis is that cisplatin activates TAK1, which phosphorylates p38 and ERK, leading to excessive autophagy of tubular epithelial cells and thus exacerbating kidney damage. Here, BALB/c mice were intraperitoneally injected with a TAK1 inhibitor and were then administrated with sham or cisplatin at 20 mg/kg by intraperitoneal injection. Compared with mice in the vehicle cisplatin group, mice intraperitoneally injected with a TAK1 inhibitor were found to have lower serum creatinine and less tubular damage following cisplatin-induced AKI. Furthermore, inhibition of TAK1 reduced p38 and Erk phosphorylation, decreased expression of LC3II and reversed the down-regulation of P62 expression induced by cisplatin. The hypothesis was verified with tubular epithelial cells administrated with cisplatin in vitro. Finally, p38 inhibitor or ERK inhibitor abated autophagy activation and cell viability reduction in tubular epithelial cells treated with cisplatin plus TAK1 overexpression vector. Taken together, our results show that cisplatin activates TAK1, which phosphorylates p38 and ERK, leading to excessive autophagy of tubular epithelial cells that exacerbates kidney damage.
Collapse
Affiliation(s)
- Jun Zhou
- Department of AnesthesiologyThe First People's Hospital of FoshanFoshanChina
| | - Youling Fan
- Department of AnesthesiologyPanyu Central HospitalGuangzhouChina
| | - Jiying Zhong
- Department of AnesthesiologyThe First People's Hospital of FoshanFoshanChina
| | - Zhenxing Huang
- Department of AnesthesiologyThe First People's Hospital of FoshanFoshanChina
| | - Teng Huang
- Department of AnesthesiologyThe First People's Hospital of FoshanFoshanChina
| | - Sen Lin
- Department of AnesthesiologyThe First People's Hospital of FoshanFoshanChina
| | - Hongtao Chen
- Department of AnesthesiologyEighth People's Hospital of GuangzhouGuangzhouChina
| |
Collapse
|
20
|
Hasanpourghadi M, Majid NA, Mustafa MR. Activation of autophagy by stress-activated signals as a cellular self-defense mechanism against the cytotoxic effects of MBIC in human breast cancer cells in vitro. Biochem Pharmacol 2018; 152:174-186. [PMID: 29608909 DOI: 10.1016/j.bcp.2018.03.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/28/2018] [Indexed: 12/16/2022]
Abstract
We recently reported that methyl 2-(-5-fluoro-2-hydroxyphenyl)-1H-benzo[d]imidazole-5-carboxylate (MBIC) is a microtubule targeting agent (MTA) with multiple mechanisms of action including apoptosis in two human breast cancer cell-lines MCF-7 and MDA-MB-231. In the present study, investigation of early molecular events following MBIC treatment demonstrated the induction of autophagy. This early (<24 h) response to MBIC was characterized by accumulation of autophagy markers; LC3-II, Beclin1, autophagic proteins (ATGs) and collection of autophagosomes but with different variations in the two cell-lines. MBIC-induced autophagy was associated with generation of reactive oxygen species (ROS). In parallel, an increased activation of SAPK/JNK pathway was detected, as an intersection of ROS production and induction of autophagy. The cytotoxic effect of MBIC was enhanced by inhibition of autophagy through blockage of SAPK/JNK signaling, suggesting that MBIC-induced autophagy, is a possible cellular self-defense mechanism against toxicity of this agent in both breast cancer cell-lines. The present findings suggest that inhibition of autophagy eliminates the cytoprotective activity of MDA-MB-231 and MCF-7 cells, and sensitizes both the aggressive and non-aggressive human breast cancer cell-lines to the cytotoxic effects of MBIC.
Collapse
Affiliation(s)
- Mohadeseh Hasanpourghadi
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Nazia Abdul Majid
- Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Mohd Rais Mustafa
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia.
| |
Collapse
|
21
|
Bhat J, Sosna J, Fritsch J, Quabius ES, Schütze S, Zeissig S, Ammerpohl O, Adam D, Kabelitz D. Expression of non-secreted IL-4 is associated with HDAC inhibitor-induced cell death, histone acetylation and c-Jun regulation in human gamma/delta T-cells. Oncotarget 2018; 7:64743-64756. [PMID: 27556516 PMCID: PMC5323112 DOI: 10.18632/oncotarget.11462] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/11/2016] [Indexed: 01/24/2023] Open
Abstract
Previously, the expression of a non-secreted IL-4 variant (IL-4δ13) has been described in association with apoptosis and age-dependent Th2 T-cell polarization. Signaling pathways involved in this process have so far not been studied. Here we report the induction of IL-4δ13 expression in human γδ T-cells upon treatment with a sublethal dose of histone deacetylase (HDACi) inhibitor valproic acid (VPA). Induction of IL-4δ13 was associated with increased cytoplasmic IL-4Rα and decreased IL-4 expression, while mRNA for mature IL-4 was concomitantly down-regulated. Importantly, only the simultaneous combination of apoptosis and necroptosis inhibitors prevented IL-4δ13 expression and completely abrogated VPA-induced global histone H3K9 acetylation mark. Further, our work reveals a novel involvement of transcription factor c-Jun in the signaling network of IL-4, HDAC1, caspase-3 and mixed lineage kinase domain-like protein (MLKL). This study provides novel insights into the effects of epigenetic modulator VPA on human γδ T-cell differentiation.
Collapse
Affiliation(s)
- Jaydeep Bhat
- Institute of Immunology, Christian-Albrechts-University, Kiel, Germany
| | - Justyna Sosna
- Institute of Immunology, Christian-Albrechts-University, Kiel, Germany.,Current address: Department of Molecular Biology and Biochemistry, University of California-Irvine, Irvine, CA, USA
| | - Jürgen Fritsch
- Institute of Immunology, Christian-Albrechts-University, Kiel, Germany
| | - Elgar Susanne Quabius
- Institute of Immunology, Christian-Albrechts-University, Kiel, Germany.,Department of Otorhinolaryngology, Head and Neck Surgery, Christian-Albrechts-University, Kiel, Germany
| | - Stefan Schütze
- Institute of Immunology, Christian-Albrechts-University, Kiel, Germany
| | - Sebastian Zeissig
- Department of Internal Medicine I, Christian-Albrechts-University, Kiel, Germany.,Current address: Department of Medicine I, University Medical Center Dresden, Technical University Dresden, Dresden, Germany.,Current address: Center for Regenerative Therapies Dresden (CRTD), Technical University Dresden, Dresden, Germany
| | - Ole Ammerpohl
- Institute of Human Genetics, University Medical Center Schleswig-Holstein Kiel, Christian-Albrechts-University, Kiel, Germany
| | - Dieter Adam
- Institute of Immunology, Christian-Albrechts-University, Kiel, Germany
| | - Dieter Kabelitz
- Institute of Immunology, Christian-Albrechts-University, Kiel, Germany
| |
Collapse
|
22
|
Vegliante R, Ciriolo MR. Autophagy and Autophagic Cell Death: Uncovering New Mechanisms Whereby Dehydroepiandrosterone Promotes Beneficial Effects on Human Health. VITAMINS AND HORMONES 2018; 108:273-307. [PMID: 30029730 DOI: 10.1016/bs.vh.2018.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dehydroepiandrosterone (DHEA) is the most abundant steroid hormone in human serum and a precursor of sexual hormones. Its levels, which are maximum between the age of 20 and 30, dramatically decline with aging thus raising the question that many pathological conditions typical of the elderly might be associated with the decrement of circulating DHEA. Moreover, since its very early discovery, DHEA and its metabolites have been shown to be active in many pathophysiological contexts, including cardiovascular disease, brain disorders, and cancer. Indeed, treatment with DHEA has beneficial effects for the cure of these and many other pathologies in vitro, in vivo, and in patient studies. However, the molecular mechanisms underlying DHEA effects have been only partially elucidated. Autophagy is a self-digestive process, by which cell homeostasis is maintained, damaged organelles removed, and cell survival assured upon stress stimuli. However, high rate of autophagy is detrimental and leads to a form of programmed cell death known as autophagic cell death (ACD). In this chapter, we describe the process of autophagy and the morphological and biochemical features of ACD. Moreover, we analyze the beneficial effects of DHEA in several pathologies and the molecular mechanisms with particular emphasis on its regulation of cell death processes. Finally, we review data indicating DHEA and structurally related steroid hormones as modulators of both autophagy and ACD, a research field that opens new avenues in the therapeutic use of these compounds.
Collapse
Affiliation(s)
- Rolando Vegliante
- MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Hopital Civil-Institut d'Hématologie et Immunologie, Strasbourg, France
| | - Maria R Ciriolo
- University of Rome 'Tor Vergata', Rome, Italy; IRCCS San Raffaele 'La Pisana', Rome, Italy.
| |
Collapse
|
23
|
Kesarwani P, Chakraborty P, Gudi R, Chatterjee S, Scurti G, Toth K, Simms P, Husain M, Armeson K, Husain S, Garrett-Mayer E, Vasu C, Nishimura MI, Mehrotra S. Blocking TCR restimulation induced necroptosis in adoptively transferred T cells improves tumor control. Oncotarget 2018; 7:69371-69383. [PMID: 27750220 PMCID: PMC5342484 DOI: 10.18632/oncotarget.12674] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 10/02/2016] [Indexed: 02/04/2023] Open
Abstract
Advancements in adoptive cell transfer therapy (ACT) has led to the use of T cells engineered with tumor specific T cell receptors, which after rapid expansion can be obtained in sufficient numbers for treating patients. However, due to massive proliferation these cells are close to replicative senescence, exhibit exhausted phenotype, and also display increased susceptibility to activation induced cell death. We have previously shown that tumor reactive T cells undergo caspase-independent cell death upon TCR restimulation with cognate antigen, which involves reactive oxygen species and c-jun N-terminal kinase. Herein, we show that a large fraction of the human melanoma epitope tyrosinase reactive TCR transduced T cells that exhibit effector memory (TEM) phenotype and undergo programmed necrosis, or necroptosis, upon TCR restimulation. As compared to the T central memory (TCM) subsets, the TEM subset displayed an increased expression of genes involved in necroptotic cell death, and a necrotic phenotype upon TCR restimulation as confirmed by electron microscopy. Higher expression of receptor-interacting kinases (RIPK) that mediate necroptosis was also observed in the TEM fraction. Further, the TEM cells were rescued from undergoing necroptosis when pretreated with necroptotic inhibitor NecroX2 before TCR restimulation. Importantly, NecroX2 pretreated tumor reactive T cells also exhibited better tumor control and increased in vivo persistence when adoptively-transferred to treat subcutaneously established murine melanoma B16-F10. Thus, it is likely that the outcome of ACT could be vastly improved by interfering with the necroptotic cell death pathway in activated tumor reactive T cells used in immunotherapy.
Collapse
Affiliation(s)
- Pravin Kesarwani
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Paramita Chakraborty
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Radhika Gudi
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Shilpak Chatterjee
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Gina Scurti
- Department of Surgery, Loyola University, Maywood, IL, USA
| | - Kyle Toth
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Patt Simms
- Department of Surgery, Loyola University, Maywood, IL, USA
| | - Mahvash Husain
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Kent Armeson
- Department of Public Health, Medical University of South Carolina, Charleston, SC, USA
| | - Shahid Husain
- Department of Opthamology, Medical University of South Carolina, Charleston, SC, USA
| | | | - Chethamarakshan Vasu
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | | | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
24
|
Pushkarev VM, Guda BB, Pushkarev VV, Tronko ND. Oncogene toxicity in thyroid carcinomas and other types of tumors. CYTOL GENET+ 2018. [DOI: 10.3103/s0095452718010103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
25
|
Ogura K, Terasaki Y, Miyoshi-Akiyama T, Terasaki M, Moss J, Noda M, Yahiro K. Vibrio cholerae Cholix Toxin-Induced HepG2 Cell Death is Enhanced by Tumor Necrosis Factor-Alpha Through ROS and Intracellular Signal-Regulated Kinases. Toxicol Sci 2018; 156:455-468. [PMID: 28087840 DOI: 10.1093/toxsci/kfx009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Cholix toxin (Cholix) from Vibrio cholerae is a potent virulence factor exhibiting ADP-ribosyltransferase activity on eukaryotic elongation factor 2 (eEF2) of host cells, resulting in the inhibition of protein synthesis. Administration of Cholix or its homologue Pseudomonas exotoxin A (PEA) to mice causes lethal hepatocyte damage. In this study, we demonstrate cytotoxicity of Cholix on human hepatocytes in the presence of tumor necrosis factor α (TNF-α), which has been reported to play a fatal role in PEA administered to mice. Compared with incubating HepG2 cells with Cholix alone, co-treatment with TNF-α and Cholix (TNF-α/Cholix) significantly enhanced the activation of caspases, cytochrome c release from mitochondria into cytoplasm, and poly-ADP-ribose polymerase (PARP) cleavage, while incubation with TNF-α alone or co-treatment with TNF-α/catalytically inactive Cholix did not. In the early stage of cell death, Cholix increased phosphorylation of mitogen-activated protein kinases (e.g., p38, ERK, JNK) and Akt, which was not affected by TNF-α alone. MAPK inhibitors (SP600125, SB20852, and U0126) suppressed PARP cleavage induced by TNF-α/Cholix. Protein kinase inhibitor Go6976 suppressed JNK phosphorylation and PARP cleavage by TNF-α/Cholix. In contrast, PKC activator PMA in the absence of TNF-α promoted Cholix-induced PARP cleavage. Reactive oxygen species (ROS) inhibitor, N-acetyl cysteine (NAC), suppressed TNF-α/Cholix-induced JNK and ERK phosphorylation, resulting in inhibition of PARP cleavage. These data suggest that ROS and JNK pathways are important mediators of TNF-α/Cholix-induced HepG2 cell death.
Collapse
Affiliation(s)
- Kohei Ogura
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yasuhiro Terasaki
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Tohru Miyoshi-Akiyama
- Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Mika Terasaki
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Joel Moss
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892-1590
| | - Masatoshi Noda
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kinnosuke Yahiro
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
26
|
Matsuzawa-Ishimoto Y, Shono Y, Gomez LE, Hubbard-Lucey VM, Cammer M, Neil J, Dewan MZ, Lieberman SR, Lazrak A, Marinis JM, Beal A, Harris PA, Bertin J, Liu C, Ding Y, van den Brink MRM, Cadwell K. Autophagy protein ATG16L1 prevents necroptosis in the intestinal epithelium. J Exp Med 2017; 214:3687-3705. [PMID: 29089374 PMCID: PMC5716041 DOI: 10.1084/jem.20170558] [Citation(s) in RCA: 218] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/20/2017] [Accepted: 09/01/2017] [Indexed: 12/22/2022] Open
Abstract
Matsuzawa-Ishimoto et al. show that autophagy gene ATG16L1, which is associated with inflammatory diseases of the gastrointestinal tract, is essential for preventing necroptotic cell death and loss of Paneth cells in the intestinal epithelium. A variant of the autophagy gene ATG16L1 is associated with Crohn’s disease, an inflammatory bowel disease (IBD), and poor survival in allogeneic hematopoietic stem cell transplant recipients. We demonstrate that ATG16L1 in the intestinal epithelium is essential for preventing loss of Paneth cells and exaggerated cell death in animal models of virally triggered IBD and allogeneic hematopoietic stem cell transplantation. Intestinal organoids lacking ATG16L1 reproduced this loss in Paneth cells and displayed TNFα-mediated necroptosis, a form of programmed necrosis. This cytoprotective function of ATG16L1 was associated with the role of autophagy in promoting mitochondrial homeostasis. Finally, therapeutic blockade of necroptosis through TNFα or RIPK1 inhibition ameliorated disease in the virally triggered IBD model. These findings indicate that, in contrast to tumor cells in which autophagy promotes caspase-independent cell death, ATG16L1 maintains the intestinal barrier by inhibiting necroptosis in the epithelium.
Collapse
Affiliation(s)
- Yu Matsuzawa-Ishimoto
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY.,Department of Microbiology, New York University School of Medicine, New York, NY
| | - Yusuke Shono
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Luis E Gomez
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY
| | - Vanessa M Hubbard-Lucey
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY
| | - Michael Cammer
- Microscopy Core, Office of Collaborative Science, New York University School of Medicine, New York, NY
| | - Jessica Neil
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY.,Department of Microbiology, New York University School of Medicine, New York, NY
| | - M Zahidunnabi Dewan
- Histopathology Core, Office of Collaborative Science, New York University School of Medicine, New York, NY
| | - Sophia R Lieberman
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Amina Lazrak
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jill M Marinis
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA
| | - Allison Beal
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA
| | - Philip A Harris
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA
| | - John Bertin
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA
| | - Chen Liu
- Departments of Pathology and Laboratory Medicine, New Jersey Medical School and Robert Wood Johnson Medical School, Rutgers University, Newark, NJ
| | - Yi Ding
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY
| | - Marcel R M van den Brink
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY .,Adult BMT Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Weil Medical College of Cornell University, New York, NY
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY .,Department of Microbiology, New York University School of Medicine, New York, NY
| |
Collapse
|
27
|
A novel chalcone derivative S17 induces apoptosis through ROS dependent DR5 up-regulation in gastric cancer cells. Sci Rep 2017; 7:9873. [PMID: 28852176 PMCID: PMC5575266 DOI: 10.1038/s41598-017-10400-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/08/2017] [Indexed: 01/09/2023] Open
Abstract
A new series of etherification chalcone derivatives were designed and synthesized through Willimison etherification and Claisen-Schmidt condensation. Among them, compound 2-c which was given chemical name of S17, has been successfully screened out as the most potent one on gastric cancer cell line(MGC803) through the investigation for their effects against the growth of five cancer cell lines (EC109, HepG2, MCF7, MGC803, SKNSH). S17 exhibited strong anti-proliferative activity on other two gastric cancer cells (HGC27 and SGC7901), but less cytotoxicity to non-malignant gastric epithelial cells GES1. S17 potently killed gastric cancer cells with causing modulation of Bcl-2 family proteins and activation of caspase 9/3 cascade. S17 also up-regulated DR5 expression and DR5 knockdown partially reversed S17-induced apoptosis, caspase activation and MMP decrease. S17 robustly induced generation of ROS with Keap/Nrf2 pathway activated and the application of ROS scavenger N-acetyl cysteine (NAC) completely blocked these effects by S17 in MGC803 cells. Intraperitoneal administration of S17 significantly inhibited the growth of MGC803 cells in vivo in a xenograft mouse model without observed toxicity. These results indicated that S17 is a leadbrominated chalcone derivate and deserves further investigation for prevention and treatment of gastric cancer.
Collapse
|
28
|
Yang M, Hao Y, Gao J, Zhang Y, Xu W, Tao L. Spinosad induces autophagy of Spodoptera frugiperda Sf9 cells and the activation of AMPK/mTOR signaling pathway. Comp Biochem Physiol C Toxicol Pharmacol 2017; 195:52-59. [PMID: 28223193 DOI: 10.1016/j.cbpc.2017.02.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/16/2017] [Accepted: 02/16/2017] [Indexed: 10/20/2022]
Abstract
Spinosad, a high-selectivity neural toxin, has been widely used in agricultural production. However, the mode of action of spinosad on insect non-neural cells is not yet clear and hence requires further investigation. Therefore, to reveal the cytotoxic mechanisms of spinosad, we investigated whether and how it can induce autophagic cell death. After treating Sf9 cells with spinosad, the resulting autophagosome was observed by transmission electron microscopy and monodansylcadaverine staining. Interestingly, spinosad induced the accumulation of Beclin-1, degradation of p62, and intensification of LC3-B formation and translocation and thus autophagy, whereas, 3-MA treatment reverted the phenotype. Under ATP depletion conditions, spinosad induced autophagy of Sf9 cells and activation of the AMPK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Mingjun Yang
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Youwu Hao
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Jufang Gao
- College of Life and Environmental Sciences, Shanghai Normal University, 100 Guilin Road, Shanghai 200234, China
| | - Yang Zhang
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Wenping Xu
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Liming Tao
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| |
Collapse
|
29
|
Zhao Y, Fan D, Zheng ZP, Li ETS, Chen F, Cheng KW, Wang M. 8-C-(E-phenylethenyl)quercetin from onion/beef soup induces autophagic cell death in colon cancer cells through ERK activation. Mol Nutr Food Res 2016; 61. [PMID: 27670274 DOI: 10.1002/mnfr.201600437] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/19/2016] [Accepted: 09/20/2016] [Indexed: 11/10/2022]
Abstract
SCOPE Quercetin, a flavonoid, widely distributed in edible fruits and vegetables, was reported to effectively inhibit 2-amino-1-methyl-6-phenylimidazo[4, 5-b]pyridine (PhIP) formation in a food model (roast beef patties) with itself being converted into a novel compound 8-C-(E-phenylethenyl)quercetin (8-CEPQ). Here we investigated whether 8-CEPQ could be formed in a real food system, and tested its anticancer activity in human colon cancer cell lines. METHODS AND RESULTS LC-MS was applied for the determination of 8-CEPQ formation in onion/beef soup. Anticancer activity of 8-CEPQ was evaluated by using cell viability assay and flow cytometry. Results showed that 8-CEPQ suppressed proliferation and caused G2 phase arrest in colon cancer cells. Based on immunofluorescent staining assay, western blot assay, and RNA knockdown data, we found that 8-CEPQ did not cause apoptotic cell death. Instead, it induced autophagic cell death. Moreover, treatment with 8-CEPQ induced phosphorylation of extracellular signal-regulated kinase (ERK). Inhibition of ERK phosphorylation by the mitogen-activated protein kinase kinase (MEK)/ERK inhibitor U0126 attenuated 8-CEPQ-induced autophagy and reversed 8-CEPQ-mediated cell growth inhibition. CONCLUSION Our results demonstrate that 8-CEPQ, a novel quercetin derivative, could be formed in onion/beef soup. 8-CEPQ inhibited colon cancer cell growth by inducing autophagic cell death through ERK activation.
Collapse
Affiliation(s)
- Yueliang Zhao
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Daming Fan
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Zong-Ping Zheng
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Edmund T S Li
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Feng Chen
- College of Engineering, Institute for Food & Bioresource Engineering, Peking University, Beijing, China
| | - Ka-Wing Cheng
- College of Engineering, Institute for Food & Bioresource Engineering, Peking University, Beijing, China
| | - Mingfu Wang
- School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
30
|
Cang X, Wang X, Liu P, Wu X, Yan J, Chen J, Wu G, Jin Y, Xu F, Su J, Wan C, Wang X. PINK1 alleviates palmitate induced insulin resistance in HepG2 cells by suppressing ROS mediated MAPK pathways. Biochem Biophys Res Commun 2016; 478:431-438. [DOI: 10.1016/j.bbrc.2016.07.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 07/01/2016] [Indexed: 11/17/2022]
|
31
|
Vibrio vulnificus VvhA induces autophagy-related cell death through the lipid raft-dependent c-Src/NOX signaling pathway. Sci Rep 2016; 6:27080. [PMID: 27250250 PMCID: PMC4890043 DOI: 10.1038/srep27080] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 05/15/2016] [Indexed: 12/22/2022] Open
Abstract
VvhA, a virulent factor of Vibrio (V.) vulnificus, induces acute cell death in a destructive manner. Autophagy plays an important role in cell death, but the functional role of VvhA in autophagy-related cell death has not been elucidated yet. We found that rVvhA significantly increased LC3 puncta formation and autophagic flux in promoting the cell death of human intestinal epithelial Caco-2 cells. The cell death induced by rVvhA was independent of lysosomal permeabilizaton and caspase activation. rVvhA induced rapid phosphorylation of c-Src in the membrane lipid raft, which resulted in an increased interaction between lipid raft molecule caveolin-1 and NADPH oxidase (NOX) complex Rac1 for ROS production. NOX-mediated ROS signaling induced by rVvhA increased the phosphorylation of extracellular signal-regulated kinase (ERK) and eukaryotic translation initiation factor 2α (eIF2α) which are required for mRNA expression of Atg5 and Atg16L1 involved in autophagosome formation. In an in vivo model, VvhA increased autophagy activation and paracellular permeabilization in intestinal epithelium. Collectively, the results here show that VvhA plays a pivotal role in the pathogenesis and dissemination of V. vulnificus by autophagy upregulation, through the lipid raft-mediated c-Src/NOX signaling pathway and ERK/eIF2α activation.
Collapse
|
32
|
Janda E, Lascala A, Carresi C, Parafati M, Aprigliano S, Russo V, Savoia C, Ziviani E, Musolino V, Morani F, Isidoro C, Mollace V. Parkinsonian toxin-induced oxidative stress inhibits basal autophagy in astrocytes via NQO2/quinone oxidoreductase 2: Implications for neuroprotection. Autophagy 2016; 11:1063-80. [PMID: 26046590 PMCID: PMC4590600 DOI: 10.1080/15548627.2015.1058683] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Oxidative stress (OS) stimulates autophagy in different cellular systems, but it remains controversial if this rule can be generalized. We have analyzed the effect of chronic OS induced by the parkinsonian toxin paraquat (PQ) on autophagy in astrocytoma cells and primary astrocytes, which represent the first cellular target of neurotoxins in the brain. PQ decreased the basal levels of LC3-II and LC3-positive vesicles, and its colocalization with lysosomal markers, both in the absence and presence of chloroquine. This was paralleled by increased number and size of SQSTM1/p62 aggregates. Downregulation of autophagy was also observed in cells chronically exposed to hydrogen peroxide or nonlethal concentrations of PQ, and it was associated with a reduced astrocyte capability to protect dopaminergic cells from OS in co-cultures. Surprisingly, PQ treatment led to inhibition of MTOR, activation of MAPK8/JNK1 and MAPK1/ERK2-MAPK3/ERK1 and upregulation of BECN1/Beclin 1 expression, all signals typically correlating with induction of autophagy. Reduction of OS by NMDPEF, a specific NQO2 inhibitor, but not by N-acetylcysteine, abrogated the inhibitory effect of PQ and restored autophagic flux. Activation of NQO2 by PQ or menadione and genetic manipulation of its expression confirmed the role of this enzyme in the inhibitory action of PQ on autophagy. PQ did not induce NFE2L2/NRF2, but when it was co-administered with NMDPEF NFE2L2 activity was enhanced in a SQSTM1-independent fashion. Thus, a prolonged OS in astrocytes inhibits LC3 lipidation and impairs autophagosome formation and autophagic flux, in spite of concomitant activation of several pro-autophagic signals. These findings outline an unanticipated neuroprotective role of astrocyte autophagy and identify in NQO2 a novel pharmacological target for its positive modulation.
Collapse
Key Words
- AVs, autophagic vacuoles
- Ab, antibody
- BNAH, benzyldihydronicotinamide riboside
- CA-DCF-DA, 5(6)-carboxy-2′,7′ dichlorofluorescein diacetate
- CQ, chloroquine
- DMEM, Dulbecco's modified Eagle's medium
- DMSO, dimethyl sulfoxide
- FACS, flow cytometry
- GFAP, glial fibrillary acidic protein
- GFP, green fluorescent protein
- K3, menadione
- MAPK, mitogen-activated protein kinase
- MFI, mean fluorescence intensity
- MPTP, 1-methyl 4-phenyl 1,2,3,6-tetraidro-piridine
- MitoSOX, 3,8-phenanthridinediamine, 5-(6′-triphenylphosphoniumhexyl)-5,6 dihydro-6-phenyl
- NFE2L2, nuclear factor, erythroid 2-like 2
- NMDPEF, N-[2-(2-methoxy-6H-dipyrido[2,3-a:3,2-e]pyrrolizin-11-yl)ethyl]-2-furamide]
- NQO2
- OS, oxidative stress
- PBS, phosphate-buffered saline
- PQ, paraquat
- ROS
- ROS, reactive oxygen species
- RT, room temperature
- SN, substantia nigra
- TTBS, Tween-Tris buffered saline
- WB, western blotting
- astrocytes
- macroautophagy
- p-, phosphorylated
- paraquat
- parkinson disease
- shRNA, short harpin ribonucleic acid
- siRNA, small interfering ribonucleic acid
Collapse
Affiliation(s)
- Elzbieta Janda
- a Department of Health Sciences; University "Magna Graecia"; Campus Germaneto ; Catanzaro , Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Vegliante R, Desideri E, Di Leo L, Ciriolo MR. Dehydroepiandrosterone triggers autophagic cell death in human hepatoma cell line HepG2 via JNK-mediated p62/SQSTM1 expression. Carcinogenesis 2016; 37:233-44. [PMID: 26762228 DOI: 10.1093/carcin/bgw003] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 01/03/2016] [Indexed: 01/17/2023] Open
Abstract
Autophagy is a catabolic process that cancer cells usually exploit during stress conditions to provide energy by recycling organelles and proteins. Beyond its prosurvival role, it is well accepted that occurrence of autophagy is often associated with a particular type of programmed cell death known as autophagic cell death (ACD). Dehydroepiandrosterone (DHEA) is an endogenous hormone showing anticancer properties even if the underlying mechanisms are not fully clear yet. Here, we provide evidence that DHEA induces ACD in human hepatoma cell line, HepG2. Indeed, autophagy inhibitors (i.e. 3-methyladenine or Atg5 siRNA) significantly reduced the percentage of dead cells. DHEA induces p62-dependent autophagy, which turns detrimental and brings about death. DHEA stimulates reactive oxygen species-independent jun N-terminal kinase (JNK) phosphoactivation and the treatment with JNK inhibitor reduces p62 mRNA levels, as well as DHEA-induced ACD. The transcription factor nuclear factor (erythroid-derived-2)-like-2 (Nrf2) constitutes the link between JNK and p62 since its migration to the nucleus is suppressed by JNK inhibitor and its inhibition through a dominant negative Nrf2 plasmid transfection decreases p62 protein levels. Overall, our data indicate that DHEA induces ACD in HepG2 via a JNK-Nrf2-p62 axis. Thus, DHEA could represent a new appealing drug for eliminating tumor cells through autophagy particularly in apoptosis-resistant cases.
Collapse
Affiliation(s)
- Rolando Vegliante
- Department of Biology, University of Rome 'Tor Vergata', Rome 00133, Italy and
| | - Enrico Desideri
- Department of Biology, University of Rome 'Tor Vergata', Rome 00133, Italy and Present address: Department of Microbiology, Immunobiology and Genetics, University of Vienna, Vienna 1030, Austria
| | - Luca Di Leo
- Department of Biology, University of Rome 'Tor Vergata', Rome 00133, Italy and
| | - Maria Rosa Ciriolo
- Department of Biology, University of Rome 'Tor Vergata', Rome 00133, Italy and IRCCS San Raffaele, Rome 00166, Italy
| |
Collapse
|
34
|
CNOT3 suppression promotes necroptosis by stabilizing mRNAs for cell death-inducing proteins. Sci Rep 2015. [DOI: 10.1038/srep14779 205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
35
|
CNOT3 suppression promotes necroptosis by stabilizing mRNAs for cell death-inducing proteins. Sci Rep 2015; 5:14779. [PMID: 26437789 PMCID: PMC4594005 DOI: 10.1038/srep14779] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/08/2015] [Indexed: 12/02/2022] Open
Abstract
The CCR4-NOT complex is conserved in eukaryotes and is involved in mRNA metabolism, though its molecular physiological roles remain to be established. We show here that CNOT3-depleted mouse embryonic fibroblasts (MEFs) undergo cell death. Levels of other complex subunits are decreased in CNOT3-depleted MEFs. The death phenotype is rescued by introduction of wild-type (WT), but not mutated CNOT3, and is not suppressed by the pan-caspase inhibitor, zVAD-fluoromethylketone. Gene expression profiling reveals that mRNAs encoding cell death-related proteins, including receptor-interacting protein kinase 1 (RIPK1) and RIPK3, are stabilized in CNOT3-depleted MEFs. Some of these mRNAs bind to CNOT3, and in the absence of CNOT3 their poly(A) tails are elongated. Inhibition of RIPK1-RIPK3 signaling by a short-hairpin RNA or a necroptosis inhibitor, necrostatin-1, confers viability upon CNOT3-depleted MEFs. Therefore, we conclude that CNOT3 targets specific mRNAs to prevent cells from being disposed to necroptotic death.
Collapse
|
36
|
Wang JS, Wu D, Huang DY, Lin WW. TAK1 inhibition-induced RIP1-dependent apoptosis in murine macrophages relies on constitutive TNF-α signaling and ROS production. J Biomed Sci 2015; 22:76. [PMID: 26381601 PMCID: PMC4574455 DOI: 10.1186/s12929-015-0182-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/05/2015] [Indexed: 12/29/2022] Open
Abstract
Background Transforming growth factor-β (TGF-β)-activated kinase 1 (TAK1) is a key regulator of signal cascades of TNF-α receptor and TLR4, and can induce NF-κB activation for preventing cell apoptosis and eliciting inflammation response. Results TAK1 inhibitor (TAKI) can decrease the cell viability of murine bone marrow-derived macrophages (BMDM), RAW264.7 and BV-2 cells, but not dermal microvascular endothelial cells, normal human epidermal keratinocytes, THP-1 monocytes, human retinal pigment epithelial cells, microglia CHME3 cells, and some cancer cell lines (CL1.0, HeLa and HCT116). In BMDM, TAKI-induced caspase activation and cell apoptosis were enhanced by lipopolysaccharide (LPS). Moreover, TAKI treatment increased the cytosolic and mitochondrial reactive oxygen species (ROS) production, and ROS scavengers NAC and BHA can inhibit cell death caused by TAKI. In addition, RIP1 inhibitor (necrostatin-1) can protect cells against TAKI-induced mitochondrial ROS production and cell apoptosis. We also observed the mitochondrial membrane potential loss after TAKI treatment and deterioration of oxygen consumption upon combination with LPS. Notably TNF-α neutralization antibody and inhibitor enbrel can decrease the cell death caused by TAKI. Conclusions TAKI-induced cytotoxicity is cell context specific, and apoptosis observed in macrophages is dependent on the constitutive autocrine action of TNF-α for RIP1 activation and ROS production.
Collapse
Affiliation(s)
- Jang-Shiun Wang
- Department of Pharmacology, College of Medicine, National Taiwan University, No 1, Sec 1, Jen-Ai Road, Taipei, Taiwan.,Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | - Dean Wu
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Duen-Yi Huang
- Department of Pharmacology, College of Medicine, National Taiwan University, No 1, Sec 1, Jen-Ai Road, Taipei, Taiwan
| | - Wan-Wan Lin
- Department of Pharmacology, College of Medicine, National Taiwan University, No 1, Sec 1, Jen-Ai Road, Taipei, Taiwan. .,Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
37
|
Wu H, Lin J, Liu P, Huang Z, Zhao P, Jin H, Wang C, Wen L, Gu N. Is the autophagy a friend or foe in the silver nanoparticles associated radiotherapy for glioma?. Biomaterials 2015; 62:47-57. [DOI: 10.1016/j.biomaterials.2015.05.033] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 05/14/2015] [Accepted: 05/18/2015] [Indexed: 12/19/2022]
|
38
|
RIP1-dependent Bid cleavage mediates TNFα-induced but Caspase-3-independent cell death in L929 fibroblastoma cells. Apoptosis 2015; 20:92-109. [PMID: 25398540 DOI: 10.1007/s10495-014-1058-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
L929 fibroblastoma cells (L929-A) and L929 fibrosarcoma cells (L929-N) are different cell lines that are commonly used to study the cytotoxicity of tumor necrosis factor alpha (TNFα). TNFα has been reported to induce necrosis in both of these cell lines. However, comparing the TNFα-induced cell death in these two cell lines, we found that, unlike the L929-N cells that show typical RIP3-dependent necrosis, TNFα-induced cell death in L929-A cells is pan-caspase inhibitor Z-VAD-FMK (Z-VAD)-sensitive, which does not depend on RIP3. We also confirmed that the cell death signal in the L929-A cells was initiated through cytosol-preassembled ripoptosome and that the knockdown of either Caspase-8 or RIP1 protein blocked cell death. Compared with the L929-N cells, the L929-A cell line had lower levels of constitutive and inducible TNFα autocrine production, and the pan-caspase inhibitors Z-VAD or Q-VD did not kill the L929-A cells as they affect the L929-N cells. Moreover, the L929-A cells expressed less RIP3 protein than the L929-N cells; therefore, TNFα failed to induce RIP3-dependent necroptosis. In addition, the ripoptosome-mediated cell death signal was transduced to the mitochondria through Caspase-8-mediated and RIP1 kinase activity-dependent Bid cleavage. The RIP1 kinase inhibitor Necrostatin-1 (Nec-1) or Caspase-8 knockdown completely blocked Bid cleavage, and the knockdown of Bid or Bax/Bak prevented TNFα-induced cell death in the L929-A cells. Although the activation of Bax/Bak decreased the mitochondrial membrane potential, the levels of mitochondrial intermembrane space proteins, including cytochrome-c (cyt-C) and Smac, declined, and western blotting and immunofluorescence staining analysis did not determine whether these proteins were redistributed to the cytosol. In addition, the mitochondrial outer membrane protein Tom20 was also reduced, indicating that the reduced mitochondria proteins may be induced by the reduced mitochondria numbers. No efficient cyt-C release was observed; therefore, the limited activation and cleavage of downstream caspases, including Caspase-9, Caspase-7, Caspase-6 and Caspase-3, was insufficient to kill the cells. The Caspase-9, Caspase-6 and Caspase-3/7 inhibitors or Caspase-9 and -3 knockdown also failed to block cell death, and the overexpression of Bcl-2 also did not abrogate cell death. Moreover, the dead cells showed necrotic-like but not apoptotic characteristics under transmission electronmicroscopy, and these features were significantly different from mitochondrial apoptosis, indicating that the effector caspases were not the executioners of cell death. These new discoveries show that TNFα-induced cell death in L929-A cells is different than typical RIP3-dependent necrosis and Caspase-8/Caspase-3-mediated apoptosis. These results highlight that caution is necessary when using different L929 cells as a model to investigate TNFα-induced cell death.
Collapse
|
39
|
Targeting mTOR/p70S6K/glycolysis signaling pathway restores glucocorticoid sensitivity to 4E-BP1 null Burkitt Lymphoma. BMC Cancer 2015; 15:529. [PMID: 26189041 PMCID: PMC4506760 DOI: 10.1186/s12885-015-1535-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 07/07/2015] [Indexed: 11/10/2022] Open
Abstract
Background Increasing evidence indicates that rapamycin could be used as a potential glucocorticoid (GC) sensitizer in lymphoblastic malignancies via genetic prevention of 4E-BP1 phosphorylation. Interestingly, we found that combined rapamycin with dexamethasone can effectively reverse GC resistance in 4E-BP1 null lymphoma cells. In this study, we investigated the potential link between mTOR/p70S6K signaling pathway, glycolysis, autophagy and GC resistance. Methods Antitumor effects of the combination of rapamycin and dexamethasone were evaluated on cell viability by MTT assay and in vivo studies, on cell cycle and apoptosis by flow cytometry, on autophagy by western blot, MDC staining and transmission electron microscopy and on cell signaling by western blot. Moreover, to test whether inhibiting glycolysis is the core mechanism in rapamycin restoring GC sensitivity, we took glycolysis inhibitor 2-deoxyglucose to replace rapamycin and then evaluated the antitumor effects in vitro. Results Raji cells are resistant to rapamycin (IC50 > 1000 nM) or dexamethasone (IC50 > 100 μM) treatment alone. The combination of rapamycin and dexamethasone synergistically inhibited the viability of Raji cells in vitro and in vivo by inducing caspase-dependent and -independent cell death and G0/G1 cell cycle arrest. These effects were achieved by the inhibition of mTOR/p70S6K signaling pathway, which led to the inhibition of glycolysis and the induction of autophagy. Pretreatment with pan-caspase inhibitor z-VAD-fmk or autophagy inhibitor 3-MA failed to protect the cells from combined treatment-induced death. Glycolysis inhibitor combined with dexamethasone produced a similar antitumor effects in vitro. Conclusions Inhibition of mTOR/p70S6K/glycolysis signaling pathway is the key point of therapy in reversing GC resistant in Burkitt lymphoma patients. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1535-z) contains supplementary material, which is available to authorized users.
Collapse
|
40
|
Dose-dependent dual role of PIT-1 (POU1F1) in somatolactotroph cell proliferation and apoptosis. PLoS One 2015; 10:e0120010. [PMID: 25822178 PMCID: PMC4379079 DOI: 10.1371/journal.pone.0120010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 02/02/2015] [Indexed: 12/11/2022] Open
Abstract
To test the role of wtPIT-1 (PITWT) or PIT-1 (R271W) (PIT271) in somatolactotroph cells, we established, using inducible lentiviral vectors, sublines of GH4C1 somatotroph cells that allow the blockade of the expression of endogenous PIT-1 and/or the expression of PITWT or PIT271, a dominant negative mutant of PIT-1 responsible for Combined Pituitary Hormone Deficiency in patients. Blocking expression of endogenous PIT-1 induced a marked decrease of cell proliferation. Overexpressing PITWT twofold led also to a dose-dependent decrease of cell proliferation that was accompanied by cell death. Expression of PIT271 induced a strong dose-dependent decrease of cell proliferation accompanied by a very pronounced cell death. These actions of PIT271 are independent of its interaction/competition with endogenous PIT-1, as they were unchanged when expression of endogenous PIT-1 was blocked. All these actions are specific for somatolactotroph cells, and could not be observed in heterologous cells. Cell death induced by PITWT or by PIT271 was accompanied by DNA fragmentation, but was not inhibited by inhibitors of caspases, autophagy or necrosis, suggesting that this cell death is a caspase-independent apoptosis. Altogether, our results indicate that under normal conditions PIT-1 is important for the maintenance of cell proliferation, while when expressed at supra-normal levels it induces cell death. Through this dual action, PIT-1 may play a role in the expansion/regression cycles of pituitary lactotroph population during and after lactation. Our results also demonstrate that the so-called “dominant-negative” action of PIT271 is independent of its competition with PIT-1 or a blockade of the actions of the latter, and are actions specific to this mutant variant of PIT-1.
Collapse
|
41
|
Ou X, Lee MR, Huang X, Messina-Graham S, Broxmeyer HE. SIRT1 positively regulates autophagy and mitochondria function in embryonic stem cells under oxidative stress. Stem Cells 2014; 32:1183-94. [PMID: 24449278 DOI: 10.1002/stem.1641] [Citation(s) in RCA: 264] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 12/03/2013] [Indexed: 12/13/2022]
Abstract
SIRT1, an NAD-dependent deacetylase, plays a role in regulation of autophagy. SIRT1 increases mitochondrial function and reduces oxidative stress, and has been linked to age-related reactive oxygen species (ROS) generation, which is highly dependent on mitochondrial metabolism. H2O2 induces oxidative stress and autophagic cell death through interference with Beclin 1 and the mTOR signaling pathways. We evaluated connections between SIRT1 activity and induction of autophagy in murine (m) and human (h) embryonic stem cells (ESCs) upon ROS challenge. Exogenous H2 O2 (1 mM) induced apoptosis and autophagy in wild-type (WT) and Sirt1-/- mESCs. High concentrations of H2O2 (1 mM) induced more apoptosis in Sirt1-/-, than in WT mESCs. However, addition of 3-methyladenine, a widely used autophagy inhibitor, in combination with H2O2 induced more cell death in WT than in Sirt1-/- mESCs. Decreased induction of autophagy in Sirt1-/- mESCs was demonstrated by decreased conversion of LC3-I to LC3-II, lowered expression of Beclin-1, and decreased LC3 punctae and LysoTracker staining. H2O2 induced autophagy with loss of mitochondrial membrane potential and disruption of mitochondrial dynamics in Sirt1-/- mESCs. Increased phosphorylation of P70/85-S6 kinase and ribosomal S6 was noted in Sirt1-/- mESCs, suggesting that SIRT1 regulates the mTOR pathway. Consistent with effects in mESCs, inhibition of SIRT1 using Lentivirus-mediated SIRT1 shRNA in hESCs demonstrated that knockdown of SIRT1 decreased H2O2-induced autophagy. This suggests a role for SIRT1 in regulating autophagy and mitochondria function in ESCs upon oxidative stress, effects mediated at least in part by the class III PI3K/Beclin 1 and mTOR pathways.
Collapse
Affiliation(s)
- Xuan Ou
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | | | | |
Collapse
|
42
|
Thapalia BA, Zhou Z, Lin X. Autophagy, a process within reperfusion injury: an update. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:8322-8341. [PMID: 25674198 PMCID: PMC4314030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 11/26/2014] [Indexed: 06/04/2023]
Abstract
Autophagy is an important constitutive intracellular catalytic process that occurs in basal conditions, as well as during stress in all tissues. It is induced during cellular growth, tissue differentiation and metabolic demands. The regulated expression is cytoprotective while its deregulation leads to varieties of diseases. It plays a vital role in ischemic heart disease, being beneficial and adaptive during ischemia while detrimental and lethal during reperfusion. Reperfusion injury is the consequence of this deregulated autophagy and the motive of its persistence during reperfusion is still obscure. A long standing debate persists as to the dual nature of autophagy and defining its clearer role in cell death as compared to the widely studied process, apoptosis. Despite the progresses in understanding of the process and identification of critical mediators, there is no therapeutic strategy to address its final outcome, the reperfusion injury. This lack of effective therapeutic strategies has even questioned the validity of the process as a single entity. We still continue to witness the devastation with standard cure of reperfusion. In this article, we review the process, highlight reperfusion injury and outline important studies being conducted for the prevention of reperfusion injury and offer cardio-protection.
Collapse
Affiliation(s)
- Bisharad Anil Thapalia
- Anhui Medical UniversityHefei 230032, Anhui, China
- Department of Cardiology, First Affiliated Hospital of Anhui Medical UniversityHefei 230032, Anhui, China
| | - Zhen Zhou
- Anhui Medical UniversityHefei 230032, Anhui, China
- Department of Cardiology, First Affiliated Hospital of Anhui Medical UniversityHefei 230032, Anhui, China
| | - Xianhe Lin
- Department of Cardiology, First Affiliated Hospital of Anhui Medical UniversityHefei 230032, Anhui, China
| |
Collapse
|
43
|
Autosis and autophagic cell death: the dark side of autophagy. Cell Death Differ 2014; 22:367-76. [PMID: 25257169 PMCID: PMC4326571 DOI: 10.1038/cdd.2014.143] [Citation(s) in RCA: 563] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 08/03/2014] [Accepted: 08/04/2014] [Indexed: 12/31/2022] Open
Abstract
It is controversial whether cells truly die via autophagy or whether — in dying cells — autophagy is merely an innocent bystander or a well-intentioned ‘Good Samaritan' trying to prevent inevitable cellular demise. However, there is increasing evidence that the genetic machinery of autophagy may be essential for cell death in certain settings. We recently identified a novel form of autophagy gene-dependent cell death, termed autosis, which is mediated by the Na+,K+-ATPase pump and has unique morphological features. High levels of cellular autophagy, as occurs with treatment with autophagy-inducing peptides, starvation, or in vivo during certain types of ischemia, can trigger autosis. These findings provide insights into the mechanisms and strategies for prevention of cell death during extreme stress conditions.
Collapse
|
44
|
Galluzzi L, Bravo-San Pedro JM, Vitale I, Aaronson SA, Abrams JM, Adam D, Alnemri ES, Altucci L, Andrews D, Annicchiarico-Petruzzelli M, Baehrecke EH, Bazan NG, Bertrand MJ, Bianchi K, Blagosklonny MV, Blomgren K, Borner C, Bredesen DE, Brenner C, Campanella M, Candi E, Cecconi F, Chan FK, Chandel NS, Cheng EH, Chipuk JE, Cidlowski JA, Ciechanover A, Dawson TM, Dawson VL, De Laurenzi V, De Maria R, Debatin KM, Di Daniele N, Dixit VM, Dynlacht BD, El-Deiry WS, Fimia GM, Flavell RA, Fulda S, Garrido C, Gougeon ML, Green DR, Gronemeyer H, Hajnoczky G, Hardwick JM, Hengartner MO, Ichijo H, Joseph B, Jost PJ, Kaufmann T, Kepp O, Klionsky DJ, Knight RA, Kumar S, Lemasters JJ, Levine B, Linkermann A, Lipton SA, Lockshin RA, López-Otín C, Lugli E, Madeo F, Malorni W, Marine JC, Martin SJ, Martinou JC, Medema JP, Meier P, Melino S, Mizushima N, Moll U, Muñoz-Pinedo C, Nuñez G, Oberst A, Panaretakis T, Penninger JM, Peter ME, Piacentini M, Pinton P, Prehn JH, Puthalakath H, Rabinovich GA, Ravichandran KS, Rizzuto R, Rodrigues CM, Rubinsztein DC, Rudel T, Shi Y, Simon HU, Stockwell BR, Szabadkai G, Tait SW, Tang HL, Tavernarakis N, Tsujimoto Y, Vanden Berghe T, Vandenabeele P, Villunger A, Wagner EF, et alGalluzzi L, Bravo-San Pedro JM, Vitale I, Aaronson SA, Abrams JM, Adam D, Alnemri ES, Altucci L, Andrews D, Annicchiarico-Petruzzelli M, Baehrecke EH, Bazan NG, Bertrand MJ, Bianchi K, Blagosklonny MV, Blomgren K, Borner C, Bredesen DE, Brenner C, Campanella M, Candi E, Cecconi F, Chan FK, Chandel NS, Cheng EH, Chipuk JE, Cidlowski JA, Ciechanover A, Dawson TM, Dawson VL, De Laurenzi V, De Maria R, Debatin KM, Di Daniele N, Dixit VM, Dynlacht BD, El-Deiry WS, Fimia GM, Flavell RA, Fulda S, Garrido C, Gougeon ML, Green DR, Gronemeyer H, Hajnoczky G, Hardwick JM, Hengartner MO, Ichijo H, Joseph B, Jost PJ, Kaufmann T, Kepp O, Klionsky DJ, Knight RA, Kumar S, Lemasters JJ, Levine B, Linkermann A, Lipton SA, Lockshin RA, López-Otín C, Lugli E, Madeo F, Malorni W, Marine JC, Martin SJ, Martinou JC, Medema JP, Meier P, Melino S, Mizushima N, Moll U, Muñoz-Pinedo C, Nuñez G, Oberst A, Panaretakis T, Penninger JM, Peter ME, Piacentini M, Pinton P, Prehn JH, Puthalakath H, Rabinovich GA, Ravichandran KS, Rizzuto R, Rodrigues CM, Rubinsztein DC, Rudel T, Shi Y, Simon HU, Stockwell BR, Szabadkai G, Tait SW, Tang HL, Tavernarakis N, Tsujimoto Y, Vanden Berghe T, Vandenabeele P, Villunger A, Wagner EF, Walczak H, White E, Wood WG, Yuan J, Zakeri Z, Zhivotovsky B, Melino G, Kroemer G. Essential versus accessory aspects of cell death: recommendations of the NCCD 2015. Cell Death Differ 2014; 22:58-73. [PMID: 25236395 PMCID: PMC4262782 DOI: 10.1038/cdd.2014.137] [Show More Authors] [Citation(s) in RCA: 718] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 07/30/2014] [Indexed: 02/07/2023] Open
Abstract
Cells exposed to extreme physicochemical or mechanical stimuli die in an uncontrollable manner, as a result of their immediate structural breakdown. Such an unavoidable variant of cellular demise is generally referred to as ‘accidental cell death' (ACD). In most settings, however, cell death is initiated by a genetically encoded apparatus, correlating with the fact that its course can be altered by pharmacologic or genetic interventions. ‘Regulated cell death' (RCD) can occur as part of physiologic programs or can be activated once adaptive responses to perturbations of the extracellular or intracellular microenvironment fail. The biochemical phenomena that accompany RCD may be harnessed to classify it into a few subtypes, which often (but not always) exhibit stereotyped morphologic features. Nonetheless, efficiently inhibiting the processes that are commonly thought to cause RCD, such as the activation of executioner caspases in the course of apoptosis, does not exert true cytoprotective effects in the mammalian system, but simply alters the kinetics of cellular demise as it shifts its morphologic and biochemical correlates. Conversely, bona fide cytoprotection can be achieved by inhibiting the transduction of lethal signals in the early phases of the process, when adaptive responses are still operational. Thus, the mechanisms that truly execute RCD may be less understood, less inhibitable and perhaps more homogeneous than previously thought. Here, the Nomenclature Committee on Cell Death formulates a set of recommendations to help scientists and researchers to discriminate between essential and accessory aspects of cell death.
Collapse
Affiliation(s)
- L Galluzzi
- 1] Gustave Roussy Cancer Center, Villejuif, France [2] Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France [3] Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
| | - J M Bravo-San Pedro
- 1] Gustave Roussy Cancer Center, Villejuif, France [2] Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France [3] INSERM, U1138, Gustave Roussy, Paris, France
| | - I Vitale
- Regina Elena National Cancer Institute, Rome, Italy
| | - S A Aaronson
- Department of Oncological Sciences, The Tisch Cancer Institute, Ichan School of Medicine at Mount Sinai, New York, NY, USA
| | - J M Abrams
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX, USA
| | - D Adam
- Institute of Immunology, Christian-Albrechts University, Kiel, Germany
| | - E S Alnemri
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - L Altucci
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università degli Studi di Napoli, Napoli, Italy
| | - D Andrews
- Department of Biochemistry and Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - M Annicchiarico-Petruzzelli
- Biochemistry Laboratory, Istituto Dermopatico dell'Immacolata - Istituto Ricovero Cura Carattere Scientifico (IDI-IRCCS), Rome, Italy
| | - E H Baehrecke
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - N G Bazan
- Neuroscience Center of Excellence, School of Medicine, New Orleans, LA, USA
| | - M J Bertrand
- 1] VIB Inflammation Research Center, Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - K Bianchi
- 1] Barts Cancer Institute, Cancer Research UK Centre of Excellence, London, UK [2] Queen Mary University of London, John Vane Science Centre, London, UK
| | - M V Blagosklonny
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - K Blomgren
- Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden
| | - C Borner
- Institute of Molecular Medicine and Spemann Graduate School of Biology and Medicine, Albert-Ludwigs University, Freiburg, Germany
| | - D E Bredesen
- 1] Buck Institute for Research on Aging, Novato, CA, USA [2] Department of Neurology, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - C Brenner
- 1] INSERM, UMRS769, Châtenay Malabry, France [2] LabEx LERMIT, Châtenay Malabry, France [3] Université Paris Sud/Paris XI, Orsay, France
| | - M Campanella
- Department of Comparative Biomedical Sciences and Consortium for Mitochondrial Research, University College London (UCL), London, UK
| | - E Candi
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - F Cecconi
- 1] Laboratory of Molecular Neuroembryology, IRCCS Fondazione Santa Lucia, Rome, Italy [2] Department of Biology, University of Rome Tor Vergata; Rome, Italy [3] Unit of Cell Stress and Survival, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - F K Chan
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA
| | - N S Chandel
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - E H Cheng
- Human Oncology and Pathogenesis Program and Department of Pathology, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
| | - J E Chipuk
- Department of Oncological Sciences, The Tisch Cancer Institute, Ichan School of Medicine at Mount Sinai, New York, NY, USA
| | - J A Cidlowski
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences (NIEHS), National Institute of Health (NIH), North Carolina, NC, USA
| | - A Ciechanover
- Tumor and Vascular Biology Research Center, The Rappaport Faculty of Medicine and Research Institute, Technion Israel Institute of Technology, Haifa, Israel
| | - T M Dawson
- 1] Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (ICE), Departments of Neurology, Pharmacology and Molecular Sciences, Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA [2] Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, USA
| | - V L Dawson
- 1] Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (ICE), Departments of Neurology, Pharmacology and Molecular Sciences, Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA [2] Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, USA
| | - V De Laurenzi
- Department of Experimental and Clinical Sciences, Gabriele d'Annunzio University, Chieti, Italy
| | - R De Maria
- Regina Elena National Cancer Institute, Rome, Italy
| | - K-M Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - N Di Daniele
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - V M Dixit
- Department of Physiological Chemistry, Genentech, South San Francisco, CA, USA
| | - B D Dynlacht
- Department of Pathology and Cancer Institute, Smilow Research Center, New York University School of Medicine, New York, NY, USA
| | - W S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medicine (Hematology/Oncology), Penn State Hershey Cancer Institute, Penn State College of Medicine, Hershey, PA, USA
| | - G M Fimia
- 1] Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy [2] Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases Lazzaro Spallanzani, Istituto Ricovero Cura Carattere Scientifico (IRCCS), Rome, Italy
| | - R A Flavell
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - S Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe University, Frankfurt, Germany
| | - C Garrido
- 1] INSERM, U866, Dijon, France [2] Faculty of Medicine, University of Burgundy, Dijon, France
| | - M-L Gougeon
- Antiviral Immunity, Biotherapy and Vaccine Unit, Infection and Epidemiology Department, Institut Pasteur, Paris, France
| | - D R Green
- Department of Immunology, St Jude's Children's Research Hospital, Memphis, TN, USA
| | - H Gronemeyer
- Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
| | - G Hajnoczky
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - J M Hardwick
- W Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, USA
| | - M O Hengartner
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - H Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - B Joseph
- Department of Oncology-Pathology, Cancer Centrum Karolinska (CCK), Karolinska Institute, Stockholm, Sweden
| | - P J Jost
- Medical Department for Hematology, Technical University of Munich, Munich, Germany
| | - T Kaufmann
- Institute of Pharmacology, Medical Faculty, University of Bern, Bern, Switzerland
| | - O Kepp
- 1] Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France [2] INSERM, U1138, Gustave Roussy, Paris, France [3] Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - D J Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - R A Knight
- 1] Medical Molecular Biology Unit, Institute of Child Health, University College London (UCL), London, UK [2] Medical Research Council Toxicology Unit, Leicester, UK
| | - S Kumar
- 1] Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia [2] School of Medicine and School of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia
| | - J J Lemasters
- Departments of Drug Discovery and Biomedical Sciences and Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - B Levine
- 1] Center for Autophagy Research, University of Texas, Southwestern Medical Center, Dallas, TX, USA [2] Howard Hughes Medical Institute (HHMI), Chevy Chase, MD, USA
| | - A Linkermann
- Division of Nephrology and Hypertension, Christian-Albrechts University, Kiel, Germany
| | - S A Lipton
- 1] The Scripps Research Institute, La Jolla, CA, USA [2] Sanford-Burnham Center for Neuroscience, Aging, and Stem Cell Research, La Jolla, CA, USA [3] Salk Institute for Biological Studies, La Jolla, CA, USA [4] University of California, San Diego (UCSD), San Diego, CA, USA
| | - R A Lockshin
- Department of Biological Sciences, St. John's University, Queens, NY, USA
| | - C López-Otín
- Department of Biochemistry and Molecular Biology, Faculty of Medecine, Instituto Universitario de Oncología (IUOPA), University of Oviedo, Oviedo, Spain
| | - E Lugli
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Milan, Italy
| | - F Madeo
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - W Malorni
- 1] Department of Therapeutic Research and Medicine Evaluation, Istituto Superiore di Sanita (ISS), Roma, Italy [2] San Raffaele Institute, Sulmona, Italy
| | - J-C Marine
- 1] Laboratory for Molecular Cancer Biology, Center for the Biology of Disease, Leuven, Belgium [2] Laboratory for Molecular Cancer Biology, Center of Human Genetics, Leuven, Belgium
| | - S J Martin
- Department of Genetics, The Smurfit Institute, Trinity College, Dublin, Ireland
| | - J-C Martinou
- Department of Cell Biology, University of Geneva, Geneva, Switzerland
| | - J P Medema
- Laboratory for Experiments Oncology and Radiobiology (LEXOR), Academic Medical Center (AMC), Amsterdam, The Netherlands
| | - P Meier
- Institute of Cancer Research, The Breakthrough Toby Robins Breast Cancer Research Centre, London, UK
| | - S Melino
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Rome, Italy
| | - N Mizushima
- Graduate School and Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - U Moll
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - C Muñoz-Pinedo
- Cell Death Regulation Group, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - G Nuñez
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - A Oberst
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - T Panaretakis
- Department of Oncology-Pathology, Cancer Centrum Karolinska (CCK), Karolinska Institute, Stockholm, Sweden
| | - J M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - M E Peter
- Department of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - M Piacentini
- 1] Department of Biology, University of Rome Tor Vergata; Rome, Italy [2] Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases Lazzaro Spallanzani, Istituto Ricovero Cura Carattere Scientifico (IRCCS), Rome, Italy
| | - P Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology and LTTA Center, University of Ferrara, Ferrara, Italy
| | - J H Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons, Dublin, Ireland
| | - H Puthalakath
- Department of Biochemistry, La Trobe Institute of Molecular Science, La Trobe University, Melbourne, Australia
| | - G A Rabinovich
- Laboratory of Immunopathology, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - K S Ravichandran
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - R Rizzuto
- Department Biomedical Sciences, University of Padova, Padova, Italy
| | - C M Rodrigues
- Research Institute for Medicines, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - D C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - T Rudel
- Department of Microbiology, University of Würzburg; Würzburg, Germany
| | - Y Shi
- Soochow Institute for Translational Medicine, Soochow University, Suzhou, China
| | - H-U Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - B R Stockwell
- 1] Howard Hughes Medical Institute (HHMI), Chevy Chase, MD, USA [2] Departments of Biological Sciences and Chemistry, Columbia University, New York, NY, USA
| | - G Szabadkai
- 1] Department Biomedical Sciences, University of Padova, Padova, Italy [2] Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, University College London (UCL), London, UK
| | - S W Tait
- 1] Cancer Research UK Beatson Institute, Glasgow, UK [2] Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - H L Tang
- W Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, USA
| | - N Tavernarakis
- 1] Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece [2] Department of Basic Sciences, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Y Tsujimoto
- Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - T Vanden Berghe
- 1] VIB Inflammation Research Center, Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - P Vandenabeele
- 1] VIB Inflammation Research Center, Ghent, Belgium [2] Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium [3] Methusalem Program, Ghent University, Ghent, Belgium
| | - A Villunger
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - E F Wagner
- Cancer Cell Biology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - H Walczak
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London (UCL), London, UK
| | - E White
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - W G Wood
- 1] Department of Pharmacology, University of Minnesota School of Medicine, Minneapolis, MN, USA [2] Geriatric Research, Education and Clinical Center, VA Medical Center, Minneapolis, MN, USA
| | - J Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Z Zakeri
- 1] Department of Biology, Queens College, Queens, NY, USA [2] Graduate Center, City University of New York (CUNY), Queens, NY, USA
| | - B Zhivotovsky
- 1] Division of Toxicology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden [2] Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - G Melino
- 1] Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy [2] Medical Research Council Toxicology Unit, Leicester, UK
| | - G Kroemer
- 1] Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France [2] Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France [3] INSERM, U1138, Gustave Roussy, Paris, France [4] Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France [5] Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| |
Collapse
|
45
|
Cho YS, Park SY. Harnessing of Programmed Necrosis for Fighting against Cancers. Biomol Ther (Seoul) 2014; 22:167-75. [PMID: 25009696 PMCID: PMC4060077 DOI: 10.4062/biomolther.2014.046] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 05/11/2014] [Accepted: 05/12/2014] [Indexed: 12/17/2022] Open
Abstract
Chemotherapy has long been considered as one of useful strategies for cancer treatment. It is primarily based on the apoptosis that can selectively kill cancer cells. However, cancer cells can progressively develop an acquired resistance to apoptotic cell death, rendering refractory to chemo- and radiotherapies. Although the mechanism by which cells attained resistance to drug remains to be clarified, it might be caused by either pumping out of them or interfering with apoptotic signal cascades in response to cancer drugs. In case that cancer cells are defective in some part of apoptotic machinery by repeated exposure to anticancer drugs, alternative cell death mechanistically distinct from apoptosis could be adopted to remove cancer cells refractory to apoptosis-inducing agents. This review will mainly deal with harnessing of necrotic cell death, specifically, programmed necrosis and practical uses. Here, we begin with various defects of apoptotic death machinery in cancer cells, and then provide new perspective on programmed necrosis as an alternative anticancer approach.
Collapse
Affiliation(s)
- Young Sik Cho
- College of Pharmacy, Keimyung University, Daegu 704-701, Republic of Korea
| | - Seung Yeon Park
- College of Pharmacy, Keimyung University, Daegu 704-701, Republic of Korea
| |
Collapse
|
46
|
Lin Z, Liu T, Kamp DW, Wang Y, He H, Zhou X, Li D, Yang L, Zhao B, Liu G. AKT/mTOR and c-Jun N-terminal kinase signaling pathways are required for chrysotile asbestos-induced autophagy. Free Radic Biol Med 2014; 72:296-307. [PMID: 24735948 PMCID: PMC4075764 DOI: 10.1016/j.freeradbiomed.2014.04.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 03/21/2014] [Accepted: 04/04/2014] [Indexed: 12/20/2022]
Abstract
Chrysotile asbestos is closely associated with excess mortality from pulmonary diseases such as lung cancer, mesothelioma, and asbestosis. Although multiple mechanisms in which chrysotile asbestos fibers induce pulmonary disease have been identified, the role of autophagy in human lung epithelial cells has not been examined. In this study, we evaluated whether chrysotile asbestos induces autophagy in A549 human lung epithelial cells and then analyzed the possible underlying molecular mechanism. Chrysotile asbestos induced autophagy in A549 cells based on a series of biochemical and microscopic autophagy markers. We observed that asbestos increased expression of A549 cell microtubule-associated protein 1 light chain 3 (LC3-II), an autophagy marker, in conjunction with dephosphorylation of phospho-AKT, phospho-mTOR, and phospho-p70S6K. Notably, AKT1/AKT2 double-knockout murine embryonic fibroblasts (MEFs) had negligible asbestos-induced LC3-II expression, supporting a crucial role for AKT signaling. Chrysotile asbestos also led to the phosphorylation/activation of Jun N-terminal kinase (JNK) and p38 MAPK. Pharmacologic inhibition of JNK, but not p38 MAPK, dramatically inhibited the protein expression of LC3-II. Moreover, JNK2(-/-) MEFs but not JNK1(-/-) MEFs blocked LC3-II levels induced by chrysotile asbestos. In addition, N-acetylcysteine, an antioxidant, attenuated chrysotile asbestos-induced dephosphorylation of P-AKT and completely abolished phosphorylation/activation of JNK. Finally, we demonstrated that chrysotile asbestos-induced apoptosis was not affected by the presence of the autophagy inhibitor 3-methyladenine or autophagy-related gene 5 siRNA, indicating that the chrysotile asbestos-induced autophagy may be adaptive rather than prosurvival. Our findings demonstrate that AKT/mTOR and JNK2 signaling pathways are required for chrysotile asbestos-induced autophagy. These data provide a mechanistic basis for possible future clinical applications targeting these signaling pathways in the management of asbestos-induced lung disease.
Collapse
Affiliation(s)
- Ziying Lin
- Clinical Research Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, China
| | - Tie Liu
- Department of Hematology, The Second Affiliated Hospital, Medical School of Xi׳an Jiaotong University, Xi׳an 710004, Shanxi, China
| | - David W Kamp
- Department of Medicine, Northwestern University Feinberg School of Medicine, and Jesse Brown VA Medical Center, Chicago, IL 60611, USA.
| | - Yahong Wang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, China
| | - Huijuan He
- Clinical Research Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, China
| | - Xu Zhou
- Clinical Research Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, China
| | - Donghong Li
- Clinical Research Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, China
| | - Lawei Yang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, China
| | - Bin Zhao
- Clinical Research Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, China
| | - Gang Liu
- Clinical Research Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, China.
| |
Collapse
|
47
|
DHA2, a synthesized derivative of bisbibenzyl, exerts antitumor activity against ovarian cancer through inhibition of XIAP and Akt/mTOR pathway. Food Chem Toxicol 2014; 69:163-74. [DOI: 10.1016/j.fct.2014.04.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 04/02/2014] [Accepted: 04/05/2014] [Indexed: 11/24/2022]
|
48
|
Kim J, Lim W, Kim S, Jeon S, Hui Z, ni K, Kim C, Im Y, Choi H, Kim O. Photodynamic therapy (PDT) resistance by PARP1 regulation on PDT-induced apoptosis with autophagy in head and neck cancer cells. J Oral Pathol Med 2014; 43:675-84. [DOI: 10.1111/jop.12195] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Jisun Kim
- Department of oral pathology; Dental Science Research Institute and Medical Research Center for Biomineralization Disorders; School of Dentistry; Chonnam National University; Gwangju Korea
| | - Wonbong Lim
- Department of oral pathology; Dental Science Research Institute and Medical Research Center for Biomineralization Disorders; School of Dentistry; Chonnam National University; Gwangju Korea
| | - Sangwoo Kim
- Department of oral pathology; Dental Science Research Institute and Medical Research Center for Biomineralization Disorders; School of Dentistry; Chonnam National University; Gwangju Korea
| | - Sangmi Jeon
- Department of oral pathology; Dental Science Research Institute and Medical Research Center for Biomineralization Disorders; School of Dentistry; Chonnam National University; Gwangju Korea
| | - Zheng Hui
- Department of oral pathology; Dental Science Research Institute and Medical Research Center for Biomineralization Disorders; School of Dentistry; Chonnam National University; Gwangju Korea
| | - Kou ni
- Department of oral pathology; Dental Science Research Institute and Medical Research Center for Biomineralization Disorders; School of Dentistry; Chonnam National University; Gwangju Korea
| | - Changsu Kim
- Su Dental Clinical Research Center; Yeoksam-dong; Gangnam-gu Seoul South Korea
| | - Yeonggwan Im
- Department of oral medicine; School of Dentistry; Chonnam National University; Gwangju Korea
| | - Hongran Choi
- Department of oral pathology; Dental Science Research Institute and Medical Research Center for Biomineralization Disorders; School of Dentistry; Chonnam National University; Gwangju Korea
| | - Okjoon Kim
- Department of oral pathology; Dental Science Research Institute and Medical Research Center for Biomineralization Disorders; School of Dentistry; Chonnam National University; Gwangju Korea
| |
Collapse
|
49
|
Tiwari M, Sharma LK, Vanegas D, Callaway DA, Bai Y, Lechleiter JD, Herman B. A nonapoptotic role for CASP2/caspase 2: modulation of autophagy. Autophagy 2014; 10:1054-1070. [PMID: 24879153 PMCID: PMC4091168 DOI: 10.4161/auto.28528] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Revised: 03/09/2014] [Accepted: 03/13/2014] [Indexed: 12/19/2022] Open
Abstract
CASP2/caspase 2 plays a role in aging, neurodegeneration, and cancer. The contributions of CASP2 have been attributed to its regulatory role in apoptotic and nonapoptotic processes including the cell cycle, DNA repair, lipid biosynthesis, and regulation of oxidant levels in the cells. Previously, our lab demonstrated CASP2-mediated modulation of autophagy during oxidative stress. Here we report the novel finding that CASP2 is an endogenous repressor of autophagy. Knockout or knockdown of CASP2 resulted in upregulation of autophagy in a variety of cell types and tissues. Reinsertion of Caspase-2 gene (Casp2) in mouse embryonic fibroblast (MEFs) lacking Casp2 (casp2(-/-)) suppresses autophagy, suggesting its role as a negative regulator of autophagy. Loss of CASP2-mediated autophagy involved AMP-activated protein kinase, mechanistic target of rapamycin, mitogen-activated protein kinase, and autophagy-related proteins, indicating the involvement of the canonical pathway of autophagy. The present study also demonstrates an important role for loss of CASP2-induced enhanced reactive oxygen species production as an upstream event in autophagy induction. Additionally, in response to a variety of stressors that induce CASP2-mediated apoptosis, casp2(-/-) cells demonstrate a further upregulation of autophagy compared with wild-type MEFs, and upregulated autophagy provides a survival advantage. In conclusion, we document a novel role for CASP2 as a negative regulator of autophagy, which may provide important insight into the role of CASP2 in various processes including aging, neurodegeneration, and cancer.
Collapse
Affiliation(s)
- Meenakshi Tiwari
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
- Department of Pathology and Laboratory Medicine; All India Institute of Medical Sciences; Patna, India
| | - Lokendra K Sharma
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Difernando Vanegas
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Danielle A Callaway
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Yidong Bai
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - James D Lechleiter
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Brian Herman
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| |
Collapse
|
50
|
Tiwari M, Sharma LK, Vanegas D, Callaway DA, Bai Y, Lechleiter JD, Herman B. A nonapoptotic role for CASP2/caspase 2: modulation of autophagy. Autophagy 2014. [PMID: 24879153 DOI: 10.4161/auto.28528.erratum.in:autophagy.2017;13(3):637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
CASP2/caspase 2 plays a role in aging, neurodegeneration, and cancer. The contributions of CASP2 have been attributed to its regulatory role in apoptotic and nonapoptotic processes including the cell cycle, DNA repair, lipid biosynthesis, and regulation of oxidant levels in the cells. Previously, our lab demonstrated CASP2-mediated modulation of autophagy during oxidative stress. Here we report the novel finding that CASP2 is an endogenous repressor of autophagy. Knockout or knockdown of CASP2 resulted in upregulation of autophagy in a variety of cell types and tissues. Reinsertion of Caspase-2 gene (Casp2) in mouse embryonic fibroblast (MEFs) lacking Casp2 (casp2(-/-)) suppresses autophagy, suggesting its role as a negative regulator of autophagy. Loss of CASP2-mediated autophagy involved AMP-activated protein kinase, mechanistic target of rapamycin, mitogen-activated protein kinase, and autophagy-related proteins, indicating the involvement of the canonical pathway of autophagy. The present study also demonstrates an important role for loss of CASP2-induced enhanced reactive oxygen species production as an upstream event in autophagy induction. Additionally, in response to a variety of stressors that induce CASP2-mediated apoptosis, casp2(-/-) cells demonstrate a further upregulation of autophagy compared with wild-type MEFs, and upregulated autophagy provides a survival advantage. In conclusion, we document a novel role for CASP2 as a negative regulator of autophagy, which may provide important insight into the role of CASP2 in various processes including aging, neurodegeneration, and cancer.
Collapse
Affiliation(s)
- Meenakshi Tiwari
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA; Department of Pathology and Laboratory Medicine; All India Institute of Medical Sciences; Patna, India
| | - Lokendra K Sharma
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Difernando Vanegas
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Danielle A Callaway
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Yidong Bai
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - James D Lechleiter
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| | - Brian Herman
- Department of Cellular and Structural Biology; University of Texas Health Science Center at San Antonio; South Texas Research Facility; San Antonio, TX USA
| |
Collapse
|