151
|
Liu L, Xie P, Li W, Wu Y, An W. Augmenter of Liver Regeneration Protects against Ethanol-Induced Acute Liver Injury by Promoting Autophagy. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 189:552-567. [PMID: 30553838 DOI: 10.1016/j.ajpath.2018.11.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 11/02/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023]
Abstract
Alcoholic liver disease is associated with high morbidity and mortality, and treatment options are limited to date. Augmenter of liver regeneration (ALR) may protect against hepatic injury from chemical poisons, including ethanol. Autophagy appears to positively influence survival in cases of liver dysfunction, although the mechanisms are poorly understood. Herein, we investigated effects of ALR-induced autophagy in vitro and in vivo in an ethanol-induced model of acute liver injury. Decreased serum levels of alanine aminotransferase and aspartate aminotransferase and reduced histologic lesions revealed that mice overexpressing ALR experienced less liver damage than wild-type. ALR-knockdown mice experienced more severe liver damage than wild-type. ALR-transfected HepG2 cells showed increased survival rates, improved maintenance of mitochondrial membrane potential, and increased ATP levels after ethanol treatment. The observed protection was associated with up-regulation of autophagy-markers, including light chain 3II, beclin-1, and autophagy-related gene 5, and down-regulation of p62 by ALR. Autophagy was inhibited in ALR-knockdown mice and HepG2 cells, and autophagy inhibitor bafilomycin A1 attenuated the protective effects of ALR. Results showed phosphorylated mammalian target of rapamycin (mTOR) was down-regulated when ALR was overexpressed and up-regulated when ALR was knocked down. These data show that ALR is protective against ethanol-induced acute liver injury by promoting autophagy, probably via repressing the mTOR pathway. These results have potential implications for the clinical treatment of alcoholic liver disease patients.
Collapse
Affiliation(s)
- Limin Liu
- Department of Cell Biology, Capital Medical University and the Municipal Key Laboratory for Liver Protection and Regulation of Regeneration, Beijing, People's Republic of China
| | - Ping Xie
- Department of Cell Biology, Capital Medical University and the Municipal Key Laboratory for Liver Protection and Regulation of Regeneration, Beijing, People's Republic of China
| | - Wen Li
- Department of Cell Biology, Capital Medical University and the Municipal Key Laboratory for Liver Protection and Regulation of Regeneration, Beijing, People's Republic of China
| | - Yuan Wu
- Department of Cell Biology, Capital Medical University and the Municipal Key Laboratory for Liver Protection and Regulation of Regeneration, Beijing, People's Republic of China
| | - Wei An
- Department of Cell Biology, Capital Medical University and the Municipal Key Laboratory for Liver Protection and Regulation of Regeneration, Beijing, People's Republic of China.
| |
Collapse
|
152
|
Hibshman JD, Leuthner TC, Shoben C, Mello DF, Sherwood DR, Meyer JN, Baugh LR. Nonselective autophagy reduces mitochondrial content during starvation in Caenorhabditis elegans. Am J Physiol Cell Physiol 2018; 315:C781-C792. [PMID: 30133321 PMCID: PMC6336938 DOI: 10.1152/ajpcell.00109.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Starvation significantly alters cellular physiology, and signs of aging have been reported to occur during starvation. Mitochondria are essential to the regulation of cellular energetics and aging. We sought to determine whether mitochondria exhibit signs of aging during starvation and whether quality control mechanisms regulate mitochondrial physiology during starvation. We describe effects of starvation on mitochondria in the first and third larval stages of the nematode Caenorhabditis elegans. When starved, C. elegans larvae enter developmental arrest. We observed fragmentation of the mitochondrial network, a reduction in mitochondrial DNA (mtDNA) copy number, and accumulation of DNA damage during starvation-induced developmental arrest. Mitochondrial function was also compromised by starvation. Starved worms had lower basal, maximal, and ATP-linked respiration. These observations are consistent with reduced mitochondrial quality, similar to mitochondrial phenotypes during aging. Using pharmacological and genetic approaches, we found that worms deficient for autophagy were short-lived during starvation and recovered poorly from extended starvation, indicating sensitivity to nutrient stress. Autophagy mutants unc-51/Atg1 and atg-18/Atg18 maintained greater mtDNA content than wild-type worms during starvation, suggesting that autophagy promotes mitochondrial degradation during starvation. unc-51 mutants also had a proportionally smaller reduction in oxygen consumption rate during starvation, suggesting that autophagy also contributes to reduced mitochondrial function. Surprisingly, mutations in genes involved in mitochondrial fission and fusion as well as selective mitophagy of damaged mitochondria did not affect mitochondrial content during starvation. Our results demonstrate the profound influence of starvation on mitochondrial physiology with organismal consequences, and they show that these physiological effects are influenced by autophagy.
Collapse
Affiliation(s)
- Jonathan D. Hibshman
- 1Department of Biology, Duke University, Durham, North Carolina,2University Program in Genetics and Genomics, Duke University, Durham, North Carolina,3Department of Biology, University of North Carolina, Chapel Hill, North Carolina
| | - Tess C. Leuthner
- 4Nicholas School of the Environment, Duke University, Durham, North Carolina
| | - Chelsea Shoben
- 1Department of Biology, Duke University, Durham, North Carolina
| | - Danielle F. Mello
- 4Nicholas School of the Environment, Duke University, Durham, North Carolina
| | - David R. Sherwood
- 1Department of Biology, Duke University, Durham, North Carolina,2University Program in Genetics and Genomics, Duke University, Durham, North Carolina
| | - Joel N. Meyer
- 4Nicholas School of the Environment, Duke University, Durham, North Carolina
| | - L. Ryan Baugh
- 1Department of Biology, Duke University, Durham, North Carolina,2University Program in Genetics and Genomics, Duke University, Durham, North Carolina
| |
Collapse
|
153
|
Zhong Z, Lemasters JJ. A Unifying Hypothesis Linking Hepatic Adaptations for Ethanol Metabolism to the Proinflammatory and Profibrotic Events of Alcoholic Liver Disease. Alcohol Clin Exp Res 2018; 42:2072-2089. [PMID: 30132924 PMCID: PMC6214771 DOI: 10.1111/acer.13877] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/13/2018] [Indexed: 02/06/2023]
Abstract
The pathogenesis of alcoholic liver disease (ALD) remains poorly understood but is likely a multihit pathophysiological process. Here, we propose a hypothesis of how early mitochondrial adaptations for alcohol metabolism lead to ALD pathogenesis. Acutely, ethanol (EtOH) feeding causes a near doubling of hepatic EtOH metabolism and oxygen consumption within 2 to 3 hours. This swift increase in alcohol metabolism (SIAM) is an adaptive response to hasten metabolic elimination of both EtOH and its more toxic metabolite, acetaldehyde (AcAld). In association with SIAM, EtOH causes widespread hepatic mitochondrial depolarization (mtDepo), which stimulates oxygen consumption. In parallel, voltage-dependent anion channels (VDAC) in the mitochondrial outer membrane close. Together, VDAC closure and respiratory stimulation promote selective and more rapid oxidation of EtOH first to AcAld in the cytosol and then to nontoxic acetate in mitochondria, since membrane-permeant AcAld does not require VDAC to enter mitochondria. VDAC closure also inhibits mitochondrial fatty acid oxidation and ATP release, promoting steatosis and a decrease in cytosolic ATP. After acute EtOH, these changes revert as EtOH is eliminated with little hepatocellular cytolethality. mtDepo also stimulates mitochondrial autophagy (mitophagy). After chronic high EtOH exposure, the capacity to process depolarized mitochondria by mitophagy becomes compromised, leading to intra- and extracellular release of damaged mitochondria, mitophagosomes, and/or autolysosomes containing mitochondrial damage-associated molecular pattern (mtDAMP) molecules. mtDAMPs cause inflammasome activation and promote inflammatory and profibrogenic responses, causing hepatitis and fibrosis. We propose that persistence of mitochondrial responses to EtOH metabolism becomes a tipping point, which links initial adaptive EtOH metabolism to maladaptive changes initiating onset and progression of ALD.
Collapse
Affiliation(s)
- Zhi Zhong
- Department of Drug Discovery & Biomedical Sciences and
| | - John J. Lemasters
- Department of Drug Discovery & Biomedical Sciences and
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|
154
|
Kornelius E, Li HH, Peng CH, Yang YS, Chen WJ, Chang YZ, Bai YC, Liu S, Huang CN, Lin CL. Liraglutide protects against glucolipotoxicity-induced RIN-m5F β-cell apoptosis through restoration of PDX1 expression. J Cell Mol Med 2018; 23:619-629. [PMID: 30353648 PMCID: PMC6307795 DOI: 10.1111/jcmm.13967] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/24/2018] [Accepted: 09/27/2018] [Indexed: 12/29/2022] Open
Abstract
Prolonged exposure to high levels of glucose and fatty acid (FFA) can induce tissue damage commonly referred to as glucolipotoxicity and is particularly harmful to pancreatic β-cells. Glucolipotoxicity-mediated β-cell failure is a critical causal factor in the late stages of diabetes, which suggests that mechanisms that prevent or reverse β-cell death may play a critical role in the treatment of the disease. Transcription factor PDX1 was recently reported to play a key role in maintaining β-cell function and survival, and glucolipotoxicity can activate mammalian sterile 20-like kinase 1 (Mst1), which, in turn, stimulates PDX1 degradation and causes dysfunction and apoptosis of β-cells. Interestingly, previous research has demonstrated that increased glucagon-like peptide-1 (GLP-1) signalling effectively protects β cells from glucolipotoxicity-induced apoptosis. Unfortunately, few studies have examined the related mechanism in detail, especially the role in Mst1 and PDX1 regulation. In the present study, we investigate the toxic effect of high glucose and FFA levels on rat pancreatic RINm5F β-cells and demonstrate that the GLP-1 analogue liraglutide restores the expression of PDX1 by inactivating Mst1, thus ameliorating β-cell impairments. In addition, liraglutide also upregulates mitophagy, which may help restore mitochondrial function and protect β-cells from oxidative stress damage. Our study suggests that liraglutide may serve as a potential agent for developing new therapies to reduce glucolipotoxicity.
Collapse
Affiliation(s)
- Edy Kornelius
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Hsin-Hua Li
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chiung-Huei Peng
- Division of Basic Medical Science, Hungkuang University, Taichung, Taiwan
| | - Yi-Sun Yang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Wei-Jen Chen
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Yan-Zin Chang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Chiao Bai
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Stanley Liu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chien-Ning Huang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chih-Li Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
155
|
Brayboy LM, Clark H, Knapik LO, Schnirman RE, Wessel GM. Nitrogen mustard exposure perturbs oocyte mitochondrial physiology and alters reproductive outcomes. Reprod Toxicol 2018; 82:80-87. [PMID: 30308227 DOI: 10.1016/j.reprotox.2018.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/28/2018] [Accepted: 10/04/2018] [Indexed: 12/18/2022]
Abstract
Nitrogen mustard (NM) is an alkylating chemical warfare agent, and its derivatives are used in chemotherapy. Alkylating agents can cause mitochondrial damage, so exposed females may transmit damaged genomes to their children, since mitochondria are maternally inherited and oocytes are not thought to undergo mitophagy (Boudoures et al. [1]). The objective of this study is to investigate NM's effects on oocyte mitochondria to understand risks facing female soldiers, cancer patients, and their children. Mice were injected intraperitoneally with NM, monitored for reproductive outcomes, and ovaries and oocytes were isolated for analysis. Escalating doses of NM increased oxidative stress in parental and F1 generation oocytes, suggesting that mitochondrial damage by NM is enhanced by mitochondrial superoxide. NM-treated ovaries in vitro exhibited smaller mitochondrial volume, more electron-dense and multivesicular structures, and lower birth weight litters. These results demonstrate that females must be protected from alkylating agents for their health, and the health of their offspring.
Collapse
Affiliation(s)
- Lynae M Brayboy
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, 101 Dudley Street, Providence, RI, 02905, USA; Alpert Medical School of Brown University, Providence, RI, 02903, USA; Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA.
| | - Haley Clark
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Laura O Knapik
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, 101 Dudley Street, Providence, RI, 02905, USA
| | - Ruby E Schnirman
- University of Chicago, 5801 South Ellis Avenue, Chicago, IL 60637, USA
| | - Gary M Wessel
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
156
|
Yamada T, Murata D, Adachi Y, Itoh K, Kameoka S, Igarashi A, Kato T, Araki Y, Huganir RL, Dawson TM, Yanagawa T, Okamoto K, Iijima M, Sesaki H. Mitochondrial Stasis Reveals p62-Mediated Ubiquitination in Parkin-Independent Mitophagy and Mitigates Nonalcoholic Fatty Liver Disease. Cell Metab 2018; 28:588-604.e5. [PMID: 30017357 PMCID: PMC6170673 DOI: 10.1016/j.cmet.2018.06.014] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 03/07/2018] [Accepted: 06/15/2018] [Indexed: 12/15/2022]
Abstract
It is unknown what occurs if both mitochondrial division and fusion are completely blocked. Here, we introduced mitochondrial stasis by deleting two dynamin-related GTPases for division (Drp1) and fusion (Opa1) in livers. Mitochondrial stasis rescues liver damage and hypotrophy caused by the single knockout (KO). At the cellular level, mitochondrial stasis re-establishes mitochondrial size and rescues mitophagy defects caused by division deficiency. Using Drp1KO livers, we found that the autophagy adaptor protein p62/sequestosome-1-which is thought to function downstream of ubiquitination-promotes mitochondrial ubiquitination. p62 recruits two subunits of a cullin-RING ubiquitin E3 ligase complex, Keap1 and Rbx1, to mitochondria. Resembling Drp1KO, diet-induced nonalcoholic fatty livers enlarge mitochondria and accumulate mitophagy intermediates. Resembling Drp1Opa1KO, Opa1KO rescues liver damage in this disease model. Our data provide a new concept that mitochondrial stasis leads the spatial dimension of mitochondria to a stationary equilibrium and a new mechanism for mitochondrial ubiquitination in mitophagy.
Collapse
Affiliation(s)
- Tatsuya Yamada
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daisuke Murata
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yoshihiro Adachi
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kie Itoh
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shoichiro Kameoka
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan
| | - Atsushi Igarashi
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Takashi Kato
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yoichi Araki
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard L Huganir
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ted M Dawson
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130, USA
| | - Toru Yanagawa
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Koji Okamoto
- Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan
| | - Miho Iijima
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
157
|
De Paoli SH, Tegegn TZ, Elhelu OK, Strader MB, Patel M, Diduch LL, Tarandovskiy ID, Wu Y, Zheng J, Ovanesov MV, Alayash A, Simak J. Dissecting the biochemical architecture and morphological release pathways of the human platelet extracellular vesiculome. Cell Mol Life Sci 2018; 75:3781-3801. [PMID: 29427073 PMCID: PMC11105464 DOI: 10.1007/s00018-018-2771-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 01/11/2018] [Accepted: 02/01/2018] [Indexed: 01/08/2023]
Abstract
Platelet extracellular vesicles (PEVs) have emerged as potential mediators in intercellular communication. PEVs exhibit several activities with pathophysiological importance and may serve as diagnostic biomarkers. Here, imaging and analytical techniques were employed to unveil morphological pathways of the release, structure, composition, and surface properties of PEVs derived from human platelets (PLTs) activated with the thrombin receptor activating peptide (TRAP). Based on extensive electron microscopy analysis, we propose four morphological pathways for PEVs release from TRAP-activated PLTs: (1) plasma membrane budding, (2) extrusion of multivesicular α-granules and cytoplasmic vacuoles, (3) plasma membrane blistering and (4) "pearling" of PLT pseudopodia. The PLT extracellular vesiculome encompasses ectosomes, exosomes, free mitochondria, mitochondria-containing vesicles, "podiasomes" and PLT "ghosts". Interestingly, a flow cytometry showed a population of TOM20+LC3+ PEVs, likely products of platelet mitophagy. We found that lipidomic and proteomic profiles were different between the small PEV (S-PEVs; mean diameter 103 nm) and the large vesicle (L-PEVs; mean diameter 350 nm) fractions separated by differential centrifugation. In addition, the majority of PEVs released by activated PLTs was composed of S-PEVs which have markedly higher thrombin generation activity per unit of PEV surface area compared to L-PEVs, and contribute approximately 60% of the PLT vesiculome procoagulant potency.
Collapse
Affiliation(s)
- Silvia H De Paoli
- Laboratory of Cellular Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, WO Bldg. 52/72, Room 4210, Silver Spring, MD, USA
| | - Tseday Z Tegegn
- Laboratory of Cellular Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, WO Bldg. 52/72, Room 4210, Silver Spring, MD, USA
| | - Oumsalama K Elhelu
- Laboratory of Cellular Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, WO Bldg. 52/72, Room 4210, Silver Spring, MD, USA
| | - Michael B Strader
- Laboratory of Biochemistry and Vascular Biology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, WO Bldg. 52/72, Silver Spring, MD, 20993-0002, USA
| | - Mehulkumar Patel
- Laboratory of Cellular Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, WO Bldg. 52/72, Room 4210, Silver Spring, MD, USA
| | - Lukas L Diduch
- Dakota Consulting, Inc., 1110 Bonifant St., Silver Spring, MD, USA
| | - Ivan D Tarandovskiy
- Laboratory of Cellular Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, WO Bldg. 52/72, Room 4210, Silver Spring, MD, USA
| | - Yong Wu
- Division of Biology, Chemistry and Materials Science, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Jiwen Zheng
- Division of Biology, Chemistry and Materials Science, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Mikhail V Ovanesov
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, USA
| | - Abdu Alayash
- Laboratory of Biochemistry and Vascular Biology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, WO Bldg. 52/72, Silver Spring, MD, 20993-0002, USA
| | - Jan Simak
- Laboratory of Cellular Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, WO Bldg. 52/72, Room 4210, Silver Spring, MD, USA.
| |
Collapse
|
158
|
Zhang Y, Xu B, Yao M, Dong T, Mao Z, Hang B, Han X, Lin Z, Bian Q, Li M, Xia Y. Titanium dioxide nanoparticles induce proteostasis disruption and autophagy in human trophoblast cells. Chem Biol Interact 2018; 296:124-133. [PMID: 30273564 DOI: 10.1016/j.cbi.2018.09.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/20/2018] [Accepted: 09/24/2018] [Indexed: 12/13/2022]
Abstract
Titanium dioxide nanoparticles (TiO2 NPs) exist in many nano-products and concerns have been raised about their potential toxicity on human beings. One such issue is their potential effects on placental function, and the studies on this topic are limited and the mechanism remains unclear. Here we employed human trophoblast HTR-8/SVneo cells to investigate the effects of TiO2 NPs on trophoblast. Results showed that TiO2 NPs could enter cells and were mostly distributed in lysosomes, with some in the cytoplasm. TiO2 NPs and protein aggregation were found in both fetal bovine serum (FBS) in culture medium and cytoplasm of HTR-8/SVneo cells. In consistence with that, proteostasis of HTR-8/SVneo cells was significantly disrupted and endoplasmic reticulum (ER) stress related markers including PERK, IRE1-α were increased. After high speed centrifugation, the proteins PERK and IRE1-α were dramatically decreased in the highest TiO2 NPs treatment group, which indicated interactions between TiO2 NPs and these two proteins. Meanwhile, the protein expressions of LC3-II/LC3-I and P62, the autophagy biomarkers, were increased and the autophagy flux was not blocked. Cellular ROS stress increased and mitophagy related genes including PINK and Parkin increased along with the increased co-localization of LC3 and mitochondria. Taken together, these results indicated that TiO2 NPs interacted with intracellular proteins and activated ER stress and mitophagy in HTR-8/SVneo cells, which might do damage to placental function.
Collapse
Affiliation(s)
- Yuqing Zhang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Bo Xu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Mengmeng Yao
- Healthcare Management, International Business Center of Nanjing University, Nanjing, 211166, China
| | - Tianyu Dong
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Zhilei Mao
- The Affiliated Changzhou Maternity and Child Health Care Hospital, Nanjing Medical University, Changzhou, 213003, China
| | - Bo Hang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Xiumei Han
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Zhongning Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Qian Bian
- Department of Toxicology and Function Assessment, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China
| | - Min Li
- Department of Anatomy, Nanjing Medical University, Nanjing, 211166, China.
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing, 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
159
|
Diot A, Agnew T, Sanderson J, Liao C, Carver J, Neves RPD, Gupta R, Guo Y, Waters C, Seto S, Daniels MJ, Dombi E, Lodge T, Morten K, Williams SA, Enver T, Iborra FJ, Votruba M, Poulton J. Validating the RedMIT/GFP-LC3 Mouse Model by Studying Mitophagy in Autosomal Dominant Optic Atrophy Due to the OPA1Q285STOP Mutation. Front Cell Dev Biol 2018; 6:103. [PMID: 30283778 PMCID: PMC6156146 DOI: 10.3389/fcell.2018.00103] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 08/13/2018] [Indexed: 01/02/2023] Open
Abstract
Background: Autosomal dominant optic atrophy (ADOA) is usually caused by mutations in the essential gene, OPA1. This encodes a ubiquitous protein involved in mitochondrial dynamics, hence tissue specificity is not understood. Dysregulated mitophagy (mitochondria recycling) is implicated in ADOA, being increased in OPA1 patient fibroblasts. Furthermore, autophagy may be increased in retinal ganglion cells (RGCs) of the OPA1Q285STOP mouse model. Aims: We developed a mouse model for studying mitochondrial dynamics in order to investigate mitophagy in ADOA. Methods: We crossed the OPA1Q285STOP mouse with our RedMIT/GFP-LC3 mouse, harboring red fluorescent mitochondria and green fluorescent autophagosomes. Colocalization between mitochondria and autophagosomes, the hallmark of mitophagy, was quantified in fluorescently labeled organelles in primary cell cultures, using two high throughput imaging methods Imagestream (Amnis) and IN Cell Analyzer 1000 (GE Healthcare Life Sciences). We studied colocalization between mitochondria and autophagosomes in fixed sections using confocal microscopy. Results: We validated our imaging methods for RedMIT/GFP-LC3 mouse cells, showing that colocalization of red fluorescent mitochondria and green fluorescent autophagosomes is a useful indicator of mitophagy. We showed that colocalization increases when lysosomal processing is impaired. Further, colocalization of mitochondrial fragments and autophagosomes is increased in cultures from the OPA1Q285STOP/RedMIT/GFP-LC3 mice compared to RedMIT/GFP-LC3 control mouse cells that were wild type for OPA1. This was apparent in both mouse embryonic fibroblasts (MEFs) using IN Cell 1000 and in splenocytes using ImageStream imaging flow cytometer (Amnis). We confirmed that this represents increased mitophagic flux using lysosomal inhibitors. We also used microscopy to investigate the level of mitophagy in the retina from the OPA1Q285STOP/RedMIT/GFP-LC3 mice and the RedMIT/GFP-LC3 control mice. However, the expression levels of fluorescent proteins and the image signal-to-background ratios precluded the detection of colocalization so we were unable to show any difference in colocalization between these mice. Conclusions: We show that colocalization of fluorescent mitochondria and autophagosomes in cell cultures, but not fixed tissues from the RedMIT/GFP-LC3, can be used to detect mitophagy. We used this model to confirm that mitophagy is increased in a mouse model of ADOA. It will be useful for cell based studies of diseases caused by impaired mitochondrial dynamics.
Collapse
Affiliation(s)
- Alan Diot
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Thomas Agnew
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Jeremy Sanderson
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Chunyan Liao
- Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
| | - Janet Carver
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | | | - Rajeev Gupta
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Yanping Guo
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Caroline Waters
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, United Kingdom
| | - Sharon Seto
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, United Kingdom
| | - Matthew J. Daniels
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Headington, United Kingdom
| | - Eszter Dombi
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Tiffany Lodge
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Karl Morten
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Suzannah A. Williams
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Tariq Enver
- UCL Cancer Institute, University College London, London, United Kingdom
| | | | - Marcela Votruba
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, United Kingdom
| | - Joanna Poulton
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
160
|
Zhao N, Guo FF, Xie KQ, Zeng T. Targeting Nrf-2 is a promising intervention approach for the prevention of ethanol-induced liver disease. Cell Mol Life Sci 2018; 75:3143-3157. [PMID: 29947925 PMCID: PMC11105722 DOI: 10.1007/s00018-018-2852-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/18/2018] [Accepted: 06/06/2018] [Indexed: 02/07/2023]
Abstract
Alcoholic liver disease (ALD) remains to be a worldwide health problem. It is generally accepted that oxidative stress plays critical roles in the pathogenesis of ALD, and antioxidant therapy represents a logical strategy for the prevention and treatment of ALD. Nuclear factor erythroid-derived 2-like 2 (NFE2L2 or Nrf-2) is essential for the antioxidant responsive element (ARE)-mediated induction of endogenous antioxidant enzymes such as heme oxygenase 1 (HO-1) and glutamate-cysteine ligase [GCL, the rate-limiting enzyme in the synthesis of glutathione (GSH)]. Activation of Nrf-2 pathway by genetic manipulation or pharmacological agents has been demonstrated to provide protection against ALD, which suggests that targeting Nrf-2 may be a promising approach for the prevention and treatment of ALD. Herein, we review the relevant literature about the potential hepatoprotective roles of Nrf-2 activation against ALD.
Collapse
Affiliation(s)
- Ning Zhao
- Institute of Toxicology, School of Public Health, Shandong University, 44 Wenhua West Road, Jinan, 250012, Shandong, China
| | - Fang-Fang Guo
- Department of Pharmacy, Qilu Hospital of Shandong University, 107 Wenhua West Road, Jinan, 250012, Shandong, China
| | - Ke-Qin Xie
- Institute of Toxicology, School of Public Health, Shandong University, 44 Wenhua West Road, Jinan, 250012, Shandong, China
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Shandong University, 44 Wenhua West Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
161
|
|
162
|
Lemasters JJ, Zhong Z. Mitophagy in hepatocytes: Types, initiators and role in adaptive ethanol metabolism☆. LIVER RESEARCH 2018; 2:125-132. [PMID: 31157120 PMCID: PMC6541449 DOI: 10.1016/j.livres.2018.09.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mitophagy (mitochondrial autophagy) in hepatocytes is an essential quality control mechanism that removes for lysosomal digestion damaged, effete and superfluous mitochondria. Mitophagy has distinct variants. In type 1 mitophagy, typical of nutrient deprivation, cup-shaped sequestration membranes (phagophores) grow, surround and sequester individual mitochondria into mitophagosomes, often in coordination with mitochondrial fission. After sequestration, the outer compartment of the mitophagosome acidifies and the entrapped mitochondrion depolarizes, followed by fusion with lysosomes. By contrast, mitochondrial depolarization stimulates type 2 mitophagy, which is characterized by coalescence of autophagic microtubule-associated protein 1A/1B-light chain 3 (LC3)-containing structures on mitochondrial surfaces without the formation of a phagophore or mitochondrial fission. Oppositely to type 1 mitophagy, the inhibition of phosphoinositide-3-kinase (PI3K) does not block type 2 mitophagy. In type 3 mitophagy, or micromitophagy, mitochondria-derived vesicles (MDVs) enriched in oxidized proteins bud off from mitochondrial inner and outer membranes and incorporate into multivesicular bodies by vesicle scission into the lumen. In response to ethanol feeding, widespread ethanol-induced hepatocellular mitochondrial depolarization occurs to facilitate hepatic ethanol metabolism. As a consequence, type 2 mitophagy develops in response to the mitochondrial depolarization. After chronic high ethanol feeding, processing of depolarized mitochondria by mitophagy becomes compromised, leading to release of mitochondrial damage-associated molecular patterns (mtDAMPs) that promote inflammatory and profibrogenic responses. We propose that the persistence of mitochondrial responses for acute ethanol metabolism links initial adaptive ethanol metabolism to mitophagy and then to chronic maladaptive changes initiating onset and the progression of alcoholic liver disease (ALD).
Collapse
Affiliation(s)
- John J. Lemasters
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Zhi Zhong
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
163
|
Hyttinen JMT, Viiri J, Kaarniranta K, Błasiak J. Mitochondrial quality control in AMD: does mitophagy play a pivotal role? Cell Mol Life Sci 2018; 75:2991-3008. [PMID: 29777261 PMCID: PMC11105454 DOI: 10.1007/s00018-018-2843-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/25/2018] [Accepted: 05/16/2018] [Indexed: 01/05/2023]
Abstract
Age-related macular degeneration (AMD) is the predominant cause of visual loss in old people in the developed world, whose incidence is increasing. This disease is caused by the decrease in macular function, due to the degeneration of retinal pigment epithelium (RPE) cells. The aged retina is characterised by increased levels of reactive oxygen species (ROS), impaired autophagy, and DNA damage that are linked to AMD pathogenesis. Mitophagy, a mitochondria-specific type of autophagy, is an essential part of mitochondrial quality control, the collective mechanism responsible for this organelle's homeostasis. The abundance of ROS, DNA damage, and the excessive energy consumption in the ageing retina all contribute to the degeneration of RPE cells and their mitochondria. We discuss the role of mitophagy in the cell and argue that its impairment may play a role in AMD pathogenesis. Thus, mitophagy as a potential therapeutic target in AMD and other degenerative diseases is as well explored.
Collapse
Affiliation(s)
- Juha M T Hyttinen
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| | - Johanna Viiri
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland
- Department of Ophthalmology, Kuopio University Hospital, P.O. Box 100, 70029 KYS, Kuopio, Finland
| | - Janusz Błasiak
- Department of Molecular Genetics, University of Łódź, Pomorska 141/143, 90-236, Łódź, Poland
| |
Collapse
|
164
|
Beyond autophagy: a novel role for autism-linked Wdfy3 in brain mitophagy. Sci Rep 2018; 8:11348. [PMID: 30054502 PMCID: PMC6063930 DOI: 10.1038/s41598-018-29421-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 07/05/2018] [Indexed: 01/12/2023] Open
Abstract
WD repeat and FYVE domain-containing 3 (WDFY3; also known as Autophagy-Linked FYVE or Alfy) is an identified intellectual disability, developmental delay and autism risk gene. This gene encodes for a scaffolding protein that is expressed in both the developing and adult central nervous system and required for autophagy and aggrephagy with yet unexplored roles in mitophagy. Given that mitochondrial trafficking, dynamics and remodeling have key roles in synaptic plasticity, we tested the role of Wdfy3 on brain bioenergetics by using Wdfy3+/lacZ mice, the only known Wdfy3 mutant animal model with overt neurodevelopmental anomalies that survive to adulthood. We found that Wdfy3 is required for sustaining brain bioenergetics and morphology via mitophagy. Decreased mitochondrial quality control by conventional mitophagy was partly compensated for by the increased formation of mitochondria-derived vesicles (MDV) targeted to lysosomal degradation (micromitophagy). These observations, extended through proteomic analysis of mitochondria-enriched cortical fractions, showed significant enrichment for pathways associated with mitophagy, mitochondrial transport and axon guidance via semaphorin, Robo, L1cam and Eph-ephrin signaling. Collectively, our findings support a critical role for Wdfy3 in mitochondrial homeostasis with implications for neuron differentiation, neurodevelopment and age-dependent neurodegeneration.
Collapse
|
165
|
Siasos G, Tsigkou V, Kosmopoulos M, Theodosiadis D, Simantiris S, Tagkou NM, Tsimpiktsioglou A, Stampouloglou PK, Oikonomou E, Mourouzis K, Philippou A, Vavuranakis M, Stefanadis C, Tousoulis D, Papavassiliou AG. Mitochondria and cardiovascular diseases-from pathophysiology to treatment. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:256. [PMID: 30069458 DOI: 10.21037/atm.2018.06.21] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mitochondria are the source of cellular energy production and are present in different types of cells. However, their function is especially important for the heart due to the high demands in energy which is achieved through oxidative phosphorylation. Mitochondria form large networks which regulate metabolism and the optimal function is achieved through the balance between mitochondrial fusion and mitochondrial fission. Moreover, mitochondrial function is upon quality control via the process of mitophagy which removes the damaged organelles. Mitochondrial dysfunction is associated with the development of numerous cardiac diseases such as atherosclerosis, ischemia-reperfusion (I/R) injury, hypertension, diabetes, cardiac hypertrophy and heart failure (HF), due to the uncontrolled production of reactive oxygen species (ROS). Therefore, early control of mitochondrial dysfunction is a crucial step in the therapy of cardiac diseases. A number of anti-oxidant molecules and medications have been used but the results are inconsistent among the studies. Eventually, the aim of future research is to design molecules which selectively target mitochondrial dysfunction and restore the capacity of cellular anti-oxidant enzymes.
Collapse
Affiliation(s)
- Gerasimos Siasos
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.,Division of Cardiovascular, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vasiliki Tsigkou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Marinos Kosmopoulos
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Dimosthenis Theodosiadis
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Spyridon Simantiris
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Nikoletta Maria Tagkou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Athina Tsimpiktsioglou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Panagiota K Stampouloglou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Evangelos Oikonomou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Konstantinos Mourouzis
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Anastasios Philippou
- Department of Experimental Physiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Manolis Vavuranakis
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | | | - Dimitris Tousoulis
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
166
|
Hu Z, Cai HY, Luo YY, Xiao JM, Li L, Guo T. Effect of varying hypoxia reoxygenation times on autophagy of cardiomyocytes. Acta Cir Bras 2018; 33:223-230. [PMID: 29668775 DOI: 10.1590/s0102-865020180030000004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/06/2018] [Indexed: 12/11/2022] Open
Abstract
PURPOSE To investigate the impact of different hypoxia reoxygenation (HR) times on autophagy of rat cardiomyocytes (H9C2). METHODS Rat cardiomyocytes were randomly divided into normal control group (group A), hypoxia group (group B), 2 h HR group (group C), 12 h HR group (group D), and 24 h HR group (group E). LC3 II/LC3 I was determined via western blotting, and cell viabilities of cardiomyocytes were measured using methyl thiazolyl tetrazolium (MTT) assay. RESULTS Cell viabilities in HR model groups were significantly lower than those of group A (P<0.05). LC3 II/LC3 I levels in groups B to D were significantly higher than those of group A (P<0.05), and group D showed the highest LC3 II/LC3 I levels. Cell viabilities in groups B to D were significantly lower than those of group A (P<0.05), with group D showing the lowest cell viabilities (P<0.05). CONCLUSIONS Hypoxia can induce autophagy in rat cardiomyocytes, which can be further activated by reoxygenation; most notable after 12 h. Hypoxia-induced cell injury can be aggravated by reoxygenation. The lowest cell viability was observed at 12 h after reoxygenation; however, cell viability can be recovered after 24 h.
Collapse
Affiliation(s)
- Zhao Hu
- Department of Cardiology, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Hong-Yan Cai
- Department of Cardiology, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yun-Yan Luo
- Department of Cardiology, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Jian-Ming Xiao
- Department of Cardiology, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Lin Li
- Department of Cardiology, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Tao Guo
- Department of Cardiology, First Affiliated Hospital, Kunming Medical University, Kunming, China
| |
Collapse
|
167
|
Qi QM, Xue YC, Lv J, Sun D, Du JX, Cai SQ, Li YH, Gu TC, Wang MB. Ginkgolic acids induce HepG2 cell death via a combination of apoptosis, autophagy and the mitochondrial pathway. Oncol Lett 2018; 15:6400-6408. [PMID: 29725398 PMCID: PMC5920365 DOI: 10.3892/ol.2018.8177] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/12/2018] [Indexed: 01/07/2023] Open
Abstract
Ginkgolic acids may induce malignant cell death via the B-cell lymphoma 2 (Bcl-2)-associated X protein (Bax)/Bcl-2 apoptosis pathway. Concurrently, apoptosis, autophagy and mitochondrial dysfunction may also be involved in bringing about this endpoint. The anticancer effect of Ginkgolic acids (GAs) was investigated using the HepG2 cell line. The median lethal dose of the GAs of the HepG2 was measured via an MTT assay, the dose-response curves were evaluated and changes in cell morphology were monitored by microscopy. Autophagy in HepG2 cells was down regulated using 3-methyladenine (3-MA) or Beclin-1-specific small interfering RNA (siRNA) and the expression of apoptosis associated proteins caspase-3, Bax/Bcl-2, and the autophagy-associated protein 5 and microtubule-associated protein 1A/1B-light chain 3 in the GA-treated HepG2 cells were all measured by western blot analysis. The level of apoptosis in the GA-treated cells was also assessed using terminal deoxynucleotidyl-transferase-mediated dUTP nick-end labeling (TUNEL) assay, and the mitochondrial membrane potential (Δψm) was detected by immunofluorescence. The results of the MTT and TUNEL assays indicated that the proliferation of HepG2 cells treated with GAs was significantly reduced compared with the control group, and the rate of the inhibition was dose-dependent. Western blot analysis indicated that treatment with the Gas induced apoptosis and autophagy in the HepG2 cells. The Δψm of the GA-treated HepG2 cells was decreased compared with the control, as monitored by immunofluorescence. However, upon the administration of 3-MA or Beclin-1-specific siRNAs (inhibitors of the autophagy), the expression levels of the apoptosis- and autophagy-associated proteins were decreased. In conclusion, the results of the present study indicated that GAs are potent anticancer agents that function through a combination of the apoptosis, autophagy and mitochondrial pathways.
Collapse
Affiliation(s)
- Qian-Ming Qi
- Department of Medical Laboratory, The 359th Hospital of The People's Liberation Army, Zhenjiang, Jiangsu 212000, P.R. China
| | - Yin-Cun Xue
- Department of General Surgery, Jingjiang People's Hospital, Taizhou, Jiangsu 214500, P.R. China
| | - Jian Lv
- Department of General Surgery, Jingjiang People's Hospital, Taizhou, Jiangsu 214500, P.R. China
| | - Di Sun
- Department of General Surgery, Jingjiang People's Hospital, Taizhou, Jiangsu 214500, P.R. China
| | - Jian-Xin Du
- Department of General Surgery, Jingjiang People's Hospital, Taizhou, Jiangsu 214500, P.R. China
| | - Sheng-Qiang Cai
- Department of General Surgery, Jingjiang People's Hospital, Taizhou, Jiangsu 214500, P.R. China
| | - Yun-He Li
- Department of General Surgery, Jingjiang People's Hospital, Taizhou, Jiangsu 214500, P.R. China
| | - Tian-Cun Gu
- Department of General Surgery, Jingjiang People's Hospital, Taizhou, Jiangsu 214500, P.R. China
| | - Mu-Bing Wang
- Department of General Surgery, Jingjiang People's Hospital, Taizhou, Jiangsu 214500, P.R. China,Correspondence to: Dr Mu-Bing Wang, Department of General Surgery, Jingjiang People's Hospital, 28 Zhongzhou Road, Taizhou, Jiangsu 214500, P.R. China, E-mail:
| |
Collapse
|
168
|
Tekirdag K, Cuervo AM. Chaperone-mediated autophagy and endosomal microautophagy: Joint by a chaperone. J Biol Chem 2018; 293:5414-5424. [PMID: 29247007 PMCID: PMC5900761 DOI: 10.1074/jbc.r117.818237] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
A variety of mechanisms deliver cytosolic materials to the lysosomal compartment for degradation through autophagy. Here, we focus on two autophagic pathways, the chaperone-mediated autophagy and the endosomal microautophagy that rely on the cytosolic chaperone hsc70 for substrate targeting. Although hsc70 participates in the triage of proteins for degradation by different proteolytic systems, the common characteristic shared by these two forms of autophagy is that hsc70 binds directly to a specific five-amino acid motif in the cargo protein for its autophagic targeting. We summarize the current understanding of the molecular machineries behind each of these types of autophagy.
Collapse
Affiliation(s)
- Kumsal Tekirdag
- From the Department of Developmental and Molecular Biology, Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Ana Maria Cuervo
- From the Department of Developmental and Molecular Biology, Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
169
|
Thurairajah K, Briggs GD, Balogh ZJ. The source of cell-free mitochondrial DNA in trauma and potential therapeutic strategies. Eur J Trauma Emerg Surg 2018; 44:325-334. [PMID: 29633007 PMCID: PMC6002458 DOI: 10.1007/s00068-018-0954-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 04/03/2018] [Indexed: 01/04/2023]
Abstract
Mitochondria play a key role in the pathophysiology of post-injury inflammation. Cell-free mitochondrial DNA (cf-mtDNA) is now understood to catalyse sterile inflammation after trauma. Observations in trauma cohorts have identified high cf-mtDNA in patients with systemic inflammatory response syndrome and multiple organ failure as well as following major surgery. The source of cf-mtDNA can be various cells affected by mechanical and hypoxic injury (passive mechanism) or induced by inflammatory mechanisms (active mechanism). Multiple forms of cf-mtDNA exist; mtDNA fragments, mtDNA in microparticles/vesicles and cell-free mitochondria. Trauma to cells that are rich in mitochondria are believed to release more cf-mtDNA. This review describes the current understanding of the mechanisms of cf-mtDNA release, its systemic effects and the potential therapeutic implications related to its modification. Although current understanding is insufficient to change trauma management, focussed research goals have been identified to pave the way for monitoring and manipulation of cf-mtDNA release and effects in trauma.
Collapse
Affiliation(s)
- Kabilan Thurairajah
- Department of Traumatology, John Hunter Hospital and University of Newcastle, Newcastle, NSW, Australia
| | - Gabrielle Daisy Briggs
- Department of Traumatology, John Hunter Hospital and University of Newcastle, Newcastle, NSW, Australia
| | - Zsolt Janos Balogh
- Department of Traumatology, John Hunter Hospital and University of Newcastle, Newcastle, NSW, Australia.
| |
Collapse
|
170
|
Chu CT. Multiple pathways for mitophagy: A neurodegenerative conundrum for Parkinson's disease. Neurosci Lett 2018; 697:66-71. [PMID: 29626647 DOI: 10.1016/j.neulet.2018.04.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/15/2018] [Accepted: 04/02/2018] [Indexed: 01/05/2023]
Abstract
It has been nearly a decade since the first landmark studies implicating familial recessive Parkinson's disease genes in the regulation of selective mitochondrial autophagy. The PTEN-induced kinase 1 (PINK1) and the E3 ubiquitin ligase Parkin (encoded by the PARK2 gene) act together to mark depolarized mitochondria for degradation. There is now an extensive body of literature detailing key mediators and steps in this pathway, based mostly on work in transformed cell lines. However, the degree to which PINK1-triggered mitophagy contributes to mitochondrial quality control in the mammalian brain, and the extent to which its disruption contributes to Parkinson's disease pathogenesis remain uncertain. In recent years, it has become clear that there are multiple, potentially redundant, pathways of cargo specification for mitophagy. Important mitophagy-independent functions of PINK1 and Parkin are also emerging. This review summarizes key features of three major mitophagy cargo recognition systems: receptor-mediated, ubiquitin-mediated and cardiolipin-mediated. New animal models that may be useful for tracking the delivery of mitochondria into lysosomes in different neuronal populations will be highlighted. Combining these research tools with methods to selectively disrupt specific mitophagy pathways may lead to a better understanding of the potential role of mitophagy in modulating neuronal vulnerability in Parkinson's spectrum (PD/PDD/DLB) and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Charleen T Chu
- Departments of Pathology and Ophthalmology, Pittsburgh Institute for Neurodegenerative Diseases, McGowan Institute for Regenerative Medicine and Center for Neuroscience at the University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
171
|
Pellacani C, Costa LG. Role of autophagy in environmental neurotoxicity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 235:791-805. [PMID: 29353798 DOI: 10.1016/j.envpol.2017.12.102] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 12/08/2017] [Accepted: 12/25/2017] [Indexed: 06/07/2023]
Abstract
Human exposure to neurotoxic pollutants (e.g. metals, pesticides and other chemicals) is recognized as a key risk factor in the pathogenesis of neurodegenerative disorders. Emerging evidence indicates that an alteration in autophagic pathways may be correlated with the onset of the neurotoxicity resulting from chronic exposure to these pollutants. In fact, autophagy is a natural process that permits to preserving cell homeostasis, through the seizure and degradation of the cytosolic damaged elements. However, when an excessive level of intracellular damage is reached, the autophagic process may also induce cell death. A correct modulation of specific stages of autophagy is important to maintain the correct balance in the organism. In this review, we highlight the critical role that autophagy plays in neurotoxicity induced by the most common classes of environmental contaminants. The understanding of this mechanism may be helpful to discover a potential therapeutic strategy to reduce side effects induced by these compounds.
Collapse
Affiliation(s)
- C Pellacani
- Dept. of Medicine and Surgery, University of Parma, Parma, Italy.
| | - L G Costa
- Dept. of Medicine and Surgery, University of Parma, Parma, Italy; Dept. of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| |
Collapse
|
172
|
Gerards M, Cannino G, González de Cózar JM, Jacobs HT. Intracellular vesicle trafficking plays an essential role in mitochondrial quality control. Mol Biol Cell 2018; 29:809-819. [PMID: 29343549 PMCID: PMC5905294 DOI: 10.1091/mbc.e17-10-0619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The Drosophila gene products Bet1, Slh, and CG10144, predicted to function in intracellular vesicle trafficking, were previously found to be essential for mitochondrial nucleoid maintenance. Here we show that Slh and Bet1 cooperate to maintain mitochondrial functions. In their absence, mitochondrial content, membrane potential, and respiration became abnormal, accompanied by mitochondrial proteotoxic stress, but without direct effects on mtDNA. Immunocytochemistry showed that both Slh and Bet1 are localized at the Golgi, together with a proportion of Rab5-positive vesicles. Some Bet1, as well as a tiny amount of Slh, cofractionated with highly purified mitochondria, while live-cell imaging showed coincidence of fluorescently tagged Bet1 with most Lysotracker-positive and a small proportion of Mitotracker-positive structures. This three-way association was disrupted in cells knocked down for Slh, although colocalized lysosomal and mitochondrial signals were still seen. Neither Slh nor Bet1 was required for global mitophagy or endocytosis, but prolonged Slh knockdown resulted in G2 growth arrest, with increased cell diameter. These effects were shared with knockdown of betaCOP but not of CG1044, Snap24, or Syntaxin6. Our findings implicate vesicle sorting at the cis-Golgi in mitochondrial quality control.
Collapse
Affiliation(s)
- Mike Gerards
- Faculty of Medicine and Life Sciences and Tampere University Hospital, FI-33014 University of Tampere, Finland.,Maastricht Center for Systems Biology (MaCSBio), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Giuseppe Cannino
- Faculty of Medicine and Life Sciences and Tampere University Hospital, FI-33014 University of Tampere, Finland
| | - Jose M González de Cózar
- Faculty of Medicine and Life Sciences and Tampere University Hospital, FI-33014 University of Tampere, Finland
| | - Howard T Jacobs
- Faculty of Medicine and Life Sciences and Tampere University Hospital, FI-33014 University of Tampere, Finland.,Institute of Biotechnology, FI-00014 University of Helsinki, Finland
| |
Collapse
|
173
|
Um JH, Yun J. Emerging role of mitophagy in human diseases and physiology. BMB Rep 2018; 50:299-307. [PMID: 28366191 PMCID: PMC5498140 DOI: 10.5483/bmbrep.2017.50.6.056] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Indexed: 12/25/2022] Open
Abstract
Mitophagy is a process of selective removal of damaged or unnecessary mitochondria using autophagic machinery. Mitophagy plays an essential role in maintaining mitochondrial quality control and homeostasis. Mitochondrial dysfunctions and defective mitophagy in neurodegenerative diseases, cancer, and metabolic diseases indicate a close link between human disease and mitophagy. Furthermore, recent studies showing the involvement of mitophagy in differentiation and development, suggest that mitophagy may play a more active role in controlling cellular functions. A better understanding of mitophagy will provide insights about human disease and offer novel chance for treatment. This review mainly focuses on the recent implications for mitophagy in human diseases and normal physiology.
Collapse
Affiliation(s)
- Jee-Hyun Um
- Department of Biochemistry, and Peripheral Neuropathy Research Center, College of Medicine, Dong-A University, Busan 49201, Korea
| | - Jeanho Yun
- Department of Biochemistry, and Peripheral Neuropathy Research Center, College of Medicine, Dong-A University, Busan 49201, Korea
| |
Collapse
|
174
|
Punzi E, Milani L, Ghiselli F, Passamonti M. Lose it or keep it: (how bivalves can provide) insights into mitochondrial inheritance mechanisms. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2018; 330:41-51. [PMID: 29393570 DOI: 10.1002/jez.b.22788] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 12/02/2017] [Accepted: 01/09/2018] [Indexed: 01/22/2023]
Abstract
The strictly maternal inheritance (SMI) is a pattern of mitochondrial inheritance observed across the whole animal kingdom. However, some interesting exceptions are known for the class Bivalvia, in which several species show an unusual pattern called doubly uniparental inheritance (DUI) whose outcome is a heteroplasmic pool of mtDNA in males. Even if DUI has been studied for long, its molecular basis has not been established yet. The aim of this work is to select classes of proteins known to be involved in the maintenance of SMI and to compare their features in two clam species differing for their mitochondrial inheritance mechanism, that is, the SMI species Ruditapes decussatus and the DUI species Ruditapes philippinarum. Data have been obtained from the transcriptomes of male and female ripe gonads of both species. Our analysis focused on nucleases and polymerases, ubiquitination and ubiquitin-like modifier pathways, and proteins involved in autophagy and mitophagy. For each protein group of interest, transcription bias (male or female), annotation, and mitochondrial targeting (when appropriate) were assessed. We did not find evidence supporting a role of nucleases/polymerases or autophagic machinery in the enforcement of SMI in R. decussatus. On the other hand, ubiquitinating enzymes with the expected features have been retrieved, providing us with two alternative testable models for mitochondrial inheritance mechanisms at the molecular level.
Collapse
Affiliation(s)
- Elisabetta Punzi
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Liliana Milani
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Fabrizio Ghiselli
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Marco Passamonti
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
175
|
Yoo SM, Jung YK. A Molecular Approach to Mitophagy and Mitochondrial Dynamics. Mol Cells 2018; 41:18-26. [PMID: 29370689 PMCID: PMC5792708 DOI: 10.14348/molcells.2018.2277] [Citation(s) in RCA: 190] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 11/22/2017] [Accepted: 11/23/2017] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial quality control systems are essential for the maintenance of functional mitochondria. At the organelle level, they include mitochondrial biogenesis, fusion and fission, to compensate for mitochondrial function, and mitophagy, for degrading damaged mitochondria. Specifically, in mitophagy, the target mitochondria are recognized by the autophagosomes and delivered to the lysosome for degradation. In this review, we describe the mechanisms of mitophagy and the factors that play an important role in this process. In particular, we focus on the roles of mitophagy adapters and receptors in the recognition of damaged mitochondria by autophagosomes. In addition, we also address a functional association of mitophagy with mitochondrial dynamics through the interaction of mitophagy adaptor and receptor proteins with mitochondrial fusion and fission proteins.
Collapse
Affiliation(s)
- Seung-Min Yoo
- Global Research Laboratory, School of Biological Sciences, Seoul National University, Seoul 08826,
Korea
| | - Yong-Keun Jung
- Global Research Laboratory, School of Biological Sciences, Seoul National University, Seoul 08826,
Korea
| |
Collapse
|
176
|
Lee JH, Mand MR, Kao CH, Zhou Y, Ryu SW, Richards AL, Coon JJ, Paull TT. ATM directs DNA damage responses and proteostasis via genetically separable pathways. Sci Signal 2018; 11:eaan5598. [PMID: 29317520 PMCID: PMC5898228 DOI: 10.1126/scisignal.aan5598] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The protein kinase ATM is a master regulator of the DNA damage response but also responds directly to oxidative stress. Loss of ATM causes ataxia telangiectasia, a neurodegenerative disorder with pleiotropic symptoms that include cerebellar dysfunction, cancer, diabetes, and premature aging. We genetically separated the activation of ATM by DNA damage from that by oxidative stress using separation-of-function mutations. We found that deficient activation of ATM by the Mre11-Rad50-Nbs1 complex and DNA double-strand breaks resulted in loss of cell viability, checkpoint activation, and DNA end resection in response to DNA damage. In contrast, loss of oxidative activation of ATM had minimal effects on DNA damage-related outcomes but blocked ATM-mediated initiation of checkpoint responses after oxidative stress and resulted in deficiencies in mitochondrial function and autophagy. In addition, expression of a variant ATM incapable of activation by oxidative stress resulted in widespread protein aggregation. These results indicate a direct relationship between the mechanism of ATM activation and its effects on cellular metabolism and DNA damage responses in human cells and implicate ATM in the control of protein homeostasis.
Collapse
Affiliation(s)
- Ji-Hoon Lee
- Howard Hughes Medical Institute, Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Michael R Mand
- Howard Hughes Medical Institute, Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Chung-Hsuan Kao
- Howard Hughes Medical Institute, Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Yi Zhou
- Howard Hughes Medical Institute, Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Seung W Ryu
- Howard Hughes Medical Institute, Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Alicia L Richards
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Joshua J Coon
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Tanya T Paull
- Howard Hughes Medical Institute, Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
177
|
Bian X, Teng T, Zhao H, Qin J, Qiao Z, Sun Y, Liun Z, Xu Z. Zinc prevents mitochondrial superoxide generation by inducing mitophagy in the setting of hypoxia/reoxygenation in cardiac cells. Free Radic Res 2017; 52:80-91. [PMID: 29216769 DOI: 10.1080/10715762.2017.1414949] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Zinc plays a role in autophagy and protects cardiac cells from ischemia/reperfusion injury. This study aimed to test if zinc can induce mitophagy leading to attenuation of mitochondrial superoxide generation in the setting of hypoxia/reoxygenation (H/R) in cardiac cells. H9c2 cells were subjected to 4 h hypoxia followed by 2 h reoxygenation. Under normoxic conditions, treatments of cells with ZnCl2 increased both the LC3-II/LC3-I ratio and GFP-LC3 puncta, implying that zinc induces autophagy. Further experiments showed that endogenous zinc is required for the autophagy induced by starvation and rapamycin. Zinc down-regulated TOM20, TIM23, and COX4 both in normoxic cells and the cells subjected to H/R, indicating that zinc can trigger mitophagy. Zinc increased ERK activity and Beclin1 expression, and zinc-induced mitophagy was inhibited by PD98059 and Beclin1 siRNA during reoxygenation. Zinc-induced Beclin1 expression was reversed by PD98059, implying that zinc promotes Beclin1 expression via ERK. In addition, zinc failed to induce mitophagy in cells transfected with PINK1 siRNA and stabilized PINK1 in mitochondria. Moreover, zinc-induced PINK1 stabilization was inhibited by PD98059. Finally, zinc prevented mitochondrial superoxide generation and dissipation of mitochondrial membrane potential (ΔΨm) at reoxygenation, which was blocked by both the Beclin1 and PINK1 siRNAs, suggesting that zinc prevents mitochondrial oxidative stress through mitophagy. In summary, zinc induces mitophagy through PINK1 and Beclin1 via ERK leading to the prevention of mitochondrial superoxide generation in the setting of H/R. Clearance of damaged mitochondria may account for the cardioprotective effect of zinc on H/R injury.
Collapse
Affiliation(s)
- Xiyun Bian
- a Department of Physiology and Pathophysiology , Tianjin Medical University , Tianjin , China
| | - Tianming Teng
- b Department of Cardiology , General Hospital, Tianjin Medical University , Tianjin , China
| | - Huanhuan Zhao
- a Department of Physiology and Pathophysiology , Tianjin Medical University , Tianjin , China
| | - Jiangyu Qin
- a Department of Physiology and Pathophysiology , Tianjin Medical University , Tianjin , China
| | - Zhen Qiao
- a Department of Physiology and Pathophysiology , Tianjin Medical University , Tianjin , China
| | - Yuemin Sun
- b Department of Cardiology , General Hospital, Tianjin Medical University , Tianjin , China
| | - Zhiqiang Liun
- a Department of Physiology and Pathophysiology , Tianjin Medical University , Tianjin , China
| | - Zhelong Xu
- a Department of Physiology and Pathophysiology , Tianjin Medical University , Tianjin , China
| |
Collapse
|
178
|
Biel TG, Rao VA. Mitochondrial dysfunction activates lysosomal-dependent mitophagy selectively in cancer cells. Oncotarget 2017; 9:995-1011. [PMID: 29416672 PMCID: PMC5787530 DOI: 10.18632/oncotarget.23171] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 11/26/2017] [Indexed: 01/13/2023] Open
Abstract
Molecules designed to target and accumulate in the mitochondria are an emerging therapeutic approach for cancer and other indications. Mitochondria-targeted redox agents (MTAs) induce mitochondrial damage and autophagy in cancer cells. However, the mechanisms for these molecules to induce mitophagy, the clearance of damaged mitochondria, are largely unknown. Using breast derived cell lines and a series of targeted molecules, mitochondrial dysfunction and autophagy was established to be selective for MDA-MB-231 cancer cells as compared to the non-cancerous MCF-12A cells. Kinetic analyses revealed that mitochondrial dysfunction precedes the activation of autophagy in these cancer cells. To determine the onset of mitophagy, stably expressing mitochondrial mKeima, a mitochondrial pH sensor, cell lines were generated and revealed that these drugs activate lysosomal dependent mitochondrial degradation in MDA-MB-231 cells. Mitophagy was confirmed by identifying the accumulation of a PINK1, mitochondria located in autophagosomes, and the formation of an autophagosome-mitochondria protein (MFN2-LC3-II) complex. These results are the first to demonstrate that mitochondrial redox agents selectively induce mitophagy in a breast cancer cell line and their potential application both as tools for investigating mitochondrial biomechanics and as therapeutic strategies that target mitochondrial metabolism.
Collapse
Affiliation(s)
- Thomas G Biel
- Laboratory of Applied Biochemistry, Division of Biotechnology Review and Research III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Silver Spring, MD 20993, USA
| | - V Ashutosh Rao
- Laboratory of Applied Biochemistry, Division of Biotechnology Review and Research III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Silver Spring, MD 20993, USA
| |
Collapse
|
179
|
Amanullah A, Upadhyay A, Joshi V, Mishra R, Jana NR, Mishra A. Progressing neurobiological strategies against proteostasis failure: Challenges in neurodegeneration. Prog Neurobiol 2017; 159:1-38. [DOI: 10.1016/j.pneurobio.2017.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 06/01/2017] [Accepted: 08/25/2017] [Indexed: 02/07/2023]
|
180
|
Meyer JN, Leuthner TC, Luz AL. Mitochondrial fusion, fission, and mitochondrial toxicity. Toxicology 2017; 391:42-53. [PMID: 28789970 PMCID: PMC5681418 DOI: 10.1016/j.tox.2017.07.019] [Citation(s) in RCA: 329] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/10/2017] [Accepted: 07/31/2017] [Indexed: 12/17/2022]
Abstract
Mitochondrial dynamics are regulated by two sets of opposed processes: mitochondrial fusion and fission, and mitochondrial biogenesis and degradation (including mitophagy), as well as processes such as intracellular transport. These processes maintain mitochondrial homeostasis, regulate mitochondrial form, volume and function, and are increasingly understood to be critical components of the cellular stress response. Mitochondrial dynamics vary based on developmental stage and age, cell type, environmental factors, and genetic background. Indeed, many mitochondrial homeostasis genes are human disease genes. Emerging evidence indicates that deficiencies in these genes often sensitize to environmental exposures, yet can also be protective under certain circumstances. Inhibition of mitochondrial dynamics also affects elimination of irreparable mitochondrial DNA (mtDNA) damage and transmission of mtDNA mutations. We briefly review the basic biology of mitodynamic processes with a focus on mitochondrial fusion and fission, discuss what is known and unknown regarding how these processes respond to chemical and other stressors, and review the literature on interactions between mitochondrial toxicity and genetic variation in mitochondrial fusion and fission genes. Finally, we suggest areas for future research, including elucidating the full range of mitodynamic responses from low to high-level exposures, and from acute to chronic exposures; detailed examination of the physiological consequences of mitodynamic alterations in different cell types; mechanism-based testing of mitotoxicant interactions with interindividual variability in mitodynamics processes; and incorporating other environmental variables that affect mitochondria, such as diet and exercise.
Collapse
Affiliation(s)
- Joel N Meyer
- Nicholas School of the Environment and Integrated Toxicology and Environmental Health Program, Duke University, Durham, NC 27708-0328, United States.
| | - Tess C Leuthner
- Nicholas School of the Environment and Integrated Toxicology and Environmental Health Program, Duke University, Durham, NC 27708-0328, United States.
| | - Anthony L Luz
- Nicholas School of the Environment and Integrated Toxicology and Environmental Health Program, Duke University, Durham, NC 27708-0328, United States.
| |
Collapse
|
181
|
Bochimoto H, Matsuno N, Ishihara Y, Shonaka T, Koga D, Hira Y, Nishikawa Y, Furukawa H, Watanabe T. The ultrastructural characteristics of porcine hepatocytes donated after cardiac death and preserved with warm machine perfusion preservation. PLoS One 2017; 12:e0186352. [PMID: 29023512 PMCID: PMC5638504 DOI: 10.1371/journal.pone.0186352] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 10/01/2017] [Indexed: 12/15/2022] Open
Abstract
The effects of warm machine perfusion preservation of liver grafts donated after cardiac death on the intracellular three-dimensional ultrastructure of the organelles in hepatocytes remain unclear. Here we analyzed comparatively the ultrastructure of the endomembrane systems in porcine hepatocytes under warm ischemia and successive hypothermic and midthermic machine perfusion preservation, a type of the warm machine perfusion. Porcine liver grafts which had a warm ischemia time of 60 minutes were perfused for 4 hours with modified University of Wisconsin gluconate solution. Group A grafts were preserved with hypothermic machine perfusion preservation at 8°C constantly for 4 hours. Group B grafts were preserved with rewarming up to 22°C by warm machine perfusion preservation for 4 hours. An analysis of hepatocytes after 60 minutes of warm ischemia by scanning electron microscope revealed the appearance of abnormal vacuoles and invagination of mitochondria. In the hepatocytes preserved by subsequent hypothermic machine perfusion preservation, strongly swollen mitochondria were observed. In contrast, the warm machine perfusion preservation could preserve the functional appearance of mitochondria in hepatocytes. Furthermore, abundant vacuoles and membranous structures sequestrating cellular organelles like autophagic vacuoles were frequently observed in hepatocytes after warm machine perfusion preservation. In conclusion, the ultrastructure of the endomembrane systems in the hepatocytes of liver grafts changed in accordance with the temperature conditions of machine perfusion preservation. In addition, temperature condition of the machine perfusion preservation may also affect the condition of the hepatic graft attributed to autophagy systems, and consequently alleviate the damage of the hepatocytes.
Collapse
Affiliation(s)
- Hiroki Bochimoto
- Health Care Administration Center, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Naoto Matsuno
- Department of Surgery, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
- * E-mail:
| | - Yo Ishihara
- Department of Microscopic Anatomy and Cell Biology, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Tatsuya Shonaka
- Department of Surgery, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Daisuke Koga
- Department of Microscopic Anatomy and Cell Biology, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Yoshiki Hira
- Area of Functional Anatomy, Department of Nursing, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Yuji Nishikawa
- Department of Pathology, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Hiroyuki Furukawa
- Department of Surgery, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Tsuyoshi Watanabe
- Department of Microscopic Anatomy and Cell Biology, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| |
Collapse
|
182
|
Transient Oxygen/Glucose Deprivation Causes a Delayed Loss of Mitochondria and Increases Spontaneous Calcium Signaling in Astrocytic Processes. J Neurosci 2017; 36:7109-27. [PMID: 27383588 DOI: 10.1523/jneurosci.4518-15.2016] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 05/24/2016] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Recently, mitochondria have been localized to astrocytic processes where they shape Ca(2+) signaling; this relationship has not been examined in models of ischemia/reperfusion. We biolistically transfected astrocytes in rat hippocampal slice cultures to facilitate fluorescent confocal microscopy, and subjected these slices to transient oxygen/glucose deprivation (OGD) that causes delayed excitotoxic death of CA1 pyramidal neurons. This insult caused a delayed loss of mitochondria from astrocytic processes and increased colocalization of mitochondria with the autophagosome marker LC3B. The losses of neurons in area CA1 and mitochondria in astrocytic processes were blocked by ionotropic glutamate receptor (iGluR) antagonists, tetrodotoxin, ziconotide (Ca(2+) channel blocker), two inhibitors of reversed Na(+)/Ca(2+) exchange (KB-R7943, YM-244769), or two inhibitors of calcineurin (cyclosporin-A, FK506). The effects of OGD were mimicked by NMDA. The glutamate uptake inhibitor (3S)-3-[[3-[[4-(trifluoromethyl)benzoyl]amino]phenyl]methoxy]-l-aspartate increased neuronal loss after OGD or NMDA, and blocked the loss of astrocytic mitochondria. Exogenous glutamate in the presence of iGluR antagonists caused a loss of mitochondria without a decrease in neurons in area CA1. Using the genetic Ca(2+) indicator Lck-GCaMP-6S, we observed two types of Ca(2+) signals: (1) in the cytoplasm surrounding mitochondria (mitochondrially centered) and (2) traversing the space between mitochondria (extramitochondrial). The spatial spread, kinetics, and frequency of these events were different. The amplitude of both types was doubled and the spread of both types changed by ∼2-fold 24 h after OGD. Together, these data suggest that pathologic activation of glutamate transport and increased astrocytic Ca(2+) through reversed Na(+)/Ca(2+) exchange triggers mitochondrial loss and dramatic increases in Ca(2+) signaling in astrocytic processes. SIGNIFICANCE STATEMENT Astrocytes, the most abundant cell type in the brain, are vital integrators of signaling and metabolism. Each astrocyte consists of many long, thin branches, called processes, which ensheathe vasculature and thousands of synapses. Mitochondria occupy the majority of each process. This occupancy is decreased by ∼50% 24 h after an in vitro model of ischemia/reperfusion injury, due to delayed fragmentation and mitophagy. The mechanism appears to be independent of neuropathology, instead involving an extended period of high glutamate uptake into astrocytes. Our data suggest that mitochondria serve as spatial buffers, and possibly even as a source of calcium signals in astrocytic processes. Loss of mitochondria resulted in drastically altered calcium signaling that could disrupt neurovascular coupling and gliotransmission.
Collapse
|
183
|
Modulating mitochondrial quality in disease transmission: towards enabling mitochondrial DNA disease carriers to have healthy children. Biochem Soc Trans 2017; 44:1091-100. [PMID: 27528757 PMCID: PMC4984448 DOI: 10.1042/bst20160095] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Indexed: 12/19/2022]
Abstract
One in 400 people has a maternally inherited mutation in mtDNA potentially causing incurable disease. In so-called heteroplasmic disease, mutant and normal mtDNA co-exist in the cells of carrier women. Disease severity depends on the proportion of inherited abnormal mtDNA molecules. Families who have had a child die of severe, maternally inherited mtDNA disease need reliable information on the risk of recurrence in future pregnancies. However, prenatal diagnosis and even estimates of risk are fraught with uncertainty because of the complex and stochastic dynamics of heteroplasmy. These complications include an mtDNA bottleneck, whereby hard-to-predict fluctuations in the proportions of mutant and normal mtDNA may arise between generations. In ‘mitochondrial replacement therapy’ (MRT), damaged mitochondria are replaced with healthy ones in early human development, using nuclear transfer. We are developing non-invasive alternatives, notably activating autophagy, a cellular quality control mechanism, in which damaged cellular components are engulfed by autophagosomes. This approach could be used in combination with MRT or with the regular management, pre-implantation genetic diagnosis (PGD). Mathematical theory, supported by recent experiments, suggests that this strategy may be fruitful in controlling heteroplasmy. Using mice that are transgenic for fluorescent LC3 (the hallmark of autophagy) we quantified autophagosomes in cleavage stage embryos. We confirmed that the autophagosome count peaks in four-cell embryos and this correlates with a drop in the mtDNA content of the whole embryo. This suggests removal by mitophagy (mitochondria-specific autophagy). We suggest that modulating heteroplasmy by activating mitophagy may be a useful complement to mitochondrial replacement therapy.
Collapse
|
184
|
Demine S, Tejerina S, Bihin B, Thiry M, Reddy N, Renard P, Raes M, Jadot M, Arnould T. Mild mitochondrial uncoupling induces HSL/ATGL-independent lipolysis relying on a form of autophagy in 3T3-L1 adipocytes. J Cell Physiol 2017; 233:1247-1265. [PMID: 28488768 DOI: 10.1002/jcp.25994] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 05/08/2017] [Indexed: 12/15/2022]
Abstract
Obesity is characterized by an excessive triacylglycerol accumulation in white adipocytes. Various mechanisms allowing the tight regulation of triacylglycerol storage and mobilization by lipid droplet-associated proteins as well as lipolytic enzymes have been identified. Increasing energy expenditure by inducing a mild uncoupling of mitochondria in adipocytes might represent a putative interesting anti-obesity strategy as it reduces the adipose tissue triacylglycerol content (limiting alterations caused by cell hypertrophy) by stimulating lipolysis through yet unknown mechanisms, limiting the adverse effects of adipocyte hypertrophy. Herein, the molecular mechanisms involved in lipolysis induced by a mild uncoupling of mitochondria in white 3T3-L1 adipocytes were characterized. Mitochondrial uncoupling-induced lipolysis was found to be independent from canonical pathways that involve lipolytic enzymes such as HSL and ATGL. Finally, enhanced lipolysis in response to mitochondrial uncoupling relies on a form of autophagy as lipid droplets are captured by endolysosomal vesicles. This new mechanism of triacylglycerol breakdown in adipocytes exposed to mild uncoupling provides new insights on the biology of adipocytes dealing with mitochondria forced to dissipate energy.
Collapse
Affiliation(s)
- Stéphane Demine
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), Namur, Belgium
| | - Silvia Tejerina
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), Namur, Belgium
| | - Benoît Bihin
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), Namur, Belgium
| | - Marc Thiry
- Laboratory of Cell Biology, GIGA-R, University of Liège, Liège, Belgium
| | - Nagabushana Reddy
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), Namur, Belgium
| | - Patricia Renard
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), Namur, Belgium
| | - Martine Raes
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), Namur, Belgium
| | - Michel Jadot
- Laboratory of Molecular Physiology (URPhyM), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), Namur, Belgium
| | - Thierry Arnould
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), Namur, Belgium
| |
Collapse
|
185
|
Li MX, Mu DZ. [Mitophagy and nervous system disease]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:724-729. [PMID: 28606244 PMCID: PMC7390300 DOI: 10.7499/j.issn.1008-8830.2017.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 02/15/2017] [Indexed: 06/07/2023]
Abstract
Mitophagy is a process during which the cell selectively removes the mitochondria via the mechanism of autophagy. It is crucial to the functional completeness of the whole mitochondrial network and determines cell survival and death. On the one hand, the damaged mitochondria releases pro-apoptotic factors which induce cell apoptosis; on the other hand, the damaged mitochondria eliminates itself via autophagy, which helps to maintain cell viability. Mitophagy is of vital importance for the development and function of the nervous system. Neural cells rely on autophagy to control protein quality and eliminate the damaged mitochondria, and under normal circumstances, mitophagy can protect the neural cells. Mutations in genes related to mitophagy may cause the development and progression of neurodegenerative diseases. An understanding of the role of mitophagy in nervous system diseases may provide new theoretical bases for clinical treatment. This article reviews the research advances in the relationship between mitophagy and different types of nervous system diseases.
Collapse
Affiliation(s)
- Ming-Xi Li
- Department of Pediatrics, West China Second Hospital, Sichuan University/Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education/Key Laboratory of Development and Related Diseases of Women and Children, Chengdu 610041, China.
| | | |
Collapse
|
186
|
Mitophagy Transcriptome: Mechanistic Insights into Polyphenol-Mediated Mitophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017. [PMID: 28626500 PMCID: PMC5463118 DOI: 10.1155/2017/9028435] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mitochondria are important bioenergetic and signalling hubs critical for myriad cellular functions and homeostasis. Dysfunction in mitochondria is a central theme in aging and diseases. Mitophagy, a process whereby damaged mitochondria are selectively removed by autophagy, plays a key homeostatic role in mitochondrial quality control. Upregulation of mitophagy has shown to mitigate superfluous mitochondrial accumulation and toxicity to safeguard mitochondrial fitness. Hence, mitophagy is a viable target to promote longevity and prevent age-related pathologies. Current challenge in modulating mitophagy for cellular protection involves identification of physiological ways to activate the pathway. Till date, mitochondrial stress and toxins remain the most potent inducers of mitophagy. Polyphenols have recently been demonstrated to protect mitochondrial health by facilitating mitophagy, thus suggesting the exciting prospect of augmenting mitophagy through dietary intake. In this review, we will first discuss the different surveillance mechanisms responsible for the removal of damaged mitochondrial components, followed by highlighting the transcriptional regulatory mechanisms of mitophagy. Finally, we will review the functional connection between polyphenols and mitophagy and provide insight into the underlying mechanisms that potentially govern polyphenol-induced mitophagy.
Collapse
|
187
|
Fedorovich SV, Waseem TV, Puchkova LV. Biogenetic and morphofunctional heterogeneity of mitochondria: the case of synaptic mitochondria. Rev Neurosci 2017; 28:363-373. [DOI: 10.1515/revneuro-2016-0077] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 12/29/2016] [Indexed: 12/26/2022]
Abstract
AbstractThe mitochondria of different cells are different in their morphological and biochemical properties. These organelles generate free radicals during activity, leading inevitably to mitochondrial DNA damage. It is not clear how this problem is addressed in long-lived cells, such as neurons. We propose the hypothesis that mitochondria within the same cell also differ in lifespan and ability to divide. According to our suggestion, cells have a pool of ‘stem’ mitochondria with low metabolic activity and a pool of ‘differentiated’ mitochondria with significantly shorter lifespans and high metabolic activity. We consider synaptic mitochondria as a possible example of ‘differentiated’ mitochondria. They are significantly smaller than mitochondria from the cell body, and they are different in key enzyme activity levels, proteome, and lipidome. Synaptic mitochondria are more sensitive to different damaging factors. It has been established that neurons have a sorting mechanism that sends mitochondria with high membrane potential to presynaptic endings. This review describes the properties of synaptic mitochondria and their role in the regulation of synaptic transmission.
Collapse
Affiliation(s)
- Sergei V. Fedorovich
- Institute of Biophysics and Cell Engineering, Akademicheskaya St., 27, Minsk 220072, Belarus
| | - Tatyana V. Waseem
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Ludmila V. Puchkova
- Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya str., 29, St. Petersburg 195251, Russia
- ITMO University, Kronverksky av., 49, St.Petersburg 197101, Russia
- Institute of Experimental Medicine, Pavlova str., 12, St.Petersburg 197376, Russia
| |
Collapse
|
188
|
Boulinguiez A, Staels B, Duez H, Lancel S. Mitochondria and endoplasmic reticulum: Targets for a better insulin sensitivity in skeletal muscle? Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:901-916. [PMID: 28529179 DOI: 10.1016/j.bbalip.2017.05.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 05/15/2017] [Accepted: 05/17/2017] [Indexed: 12/16/2022]
Abstract
Obesity and its associated metabolic disorders represent a major health burden, with economic and social consequences. Although adapted lifestyle and bariatric surgery are effective in reducing body weight, obesity prevalence is still rising. Obese individuals often become insulin-resistant. Obesity impacts on insulin responsive organs, such as the liver, adipose tissue and skeletal muscle, and increases the risk of cardiovascular diseases, type 2 diabetes and cancer. In this review, we discuss the effects of obesity and insulin resistance on skeletal muscle, an important organ for the control of postprandial glucose. The roles of mitochondria and the endoplasmic reticulum in insulin signaling are highlighted and potential innovative research and treatment perspectives are proposed.
Collapse
Affiliation(s)
- Alexis Boulinguiez
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000, Lille, France.
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000, Lille, France.
| | - Hélène Duez
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000, Lille, France.
| | - Steve Lancel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000, Lille, France.
| |
Collapse
|
189
|
Boudoures AL, Saben J, Drury A, Scheaffer S, Modi Z, Zhang W, Moley KH. Obesity-exposed oocytes accumulate and transmit damaged mitochondria due to an inability to activate mitophagy. Dev Biol 2017; 426:126-138. [PMID: 28438607 DOI: 10.1016/j.ydbio.2017.04.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/22/2017] [Accepted: 04/17/2017] [Indexed: 11/26/2022]
Abstract
Mitochondria are the most prominent organelle in the oocyte. Somatic cells maintain a healthy population of mitochondria by degrading damaged mitochondria via mitophagy, a specialized autophagy pathway. However, evidence from previous work investigating the more general macroautophagy pathway in oocytes suggests that mitophagy may not be active in the oocyte. This would leave the vast numbers of mitochondria - poised to be inherited by the offspring - vulnerable to damage. Here we test the hypothesis that inactive mitophagy in the oocyte underlies maternal transmission of dysfunctional mitochondria. To determine whether oocytes can complete mitophagy, we used either CCCP or AntimycinA to depolarize mitochondria and trigger mitophagy. After depolarization, we did not detect co-localization of mitochondria with autophagosomes and mitochondrial DNA copy number remained unchanged, indicating the non-functional mitochondrial population was not removed. To investigate the impact of an absence of mitophagy in oocytes with damaged mitochondria on offspring mitochondrial function, we utilized in vitro fertilization of high fat high sugar (HF/HS)-exposed oocytes, which have lower mitochondrial membrane potential and damaged mitochondria. Here, we demonstrate that blastocysts generated from HF/HS oocytes have decreased mitochondrial membrane potential, lower metabolites involved in ATP generation, and accumulation of PINK1, a mitophagy marker protein. This mitochondrial phenotype in the blastocyst mirrors the phenotype we show in HF/HS exposed oocytes. Taken together, these data suggest that the mechanisms governing oocyte mitophagy are fundamentally distinct from those governing somatic cell mitophagy and that the absence of mitophagy in the setting of HF/HS exposure contributes to the oocyte-to-blastocyst transmission of dysfunctional mitochondria.
Collapse
Affiliation(s)
- Anna L Boudoures
- Center for Reproductive and Health Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Jessica Saben
- Center for Reproductive and Health Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Andrea Drury
- Center for Reproductive and Health Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Suzanne Scheaffer
- Center for Reproductive and Health Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Zeel Modi
- Center for Reproductive and Health Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Wendy Zhang
- Center for Reproductive and Health Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Kelle H Moley
- Center for Reproductive and Health Sciences, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
190
|
Krech J, Tong G, Wowro S, Walker C, Rosenthal LM, Berger F, Schmitt KRL. Moderate therapeutic hypothermia induces multimodal protective effects in oxygen-glucose deprivation/reperfusion injured cardiomyocytes. Mitochondrion 2017; 35:1-10. [PMID: 28396253 DOI: 10.1016/j.mito.2017.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/01/2017] [Accepted: 04/04/2017] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Therapeutic hypothermia has been shown to attenuate myocardial cell death due to ischemia/reperfusion injury. However, cellular mechanisms of cooling remain to be elucidated. Especially during reperfusion, mitochondrial dysfunction contributes to cell death by releasing apoptosis inductors. The aim of the present study was to investigate the effects of moderate therapeutic hypothermia (33.5°C) on mitochondrial mediated apoptosis in ischemia/reperfusion-injured cardiomyocytes. METHODS Ischemic injury was simulated by oxygen-glucose deprivation for 6h in glucose/serum-free medium at 0.2% O2 in mouse atrial HL-1 cardiomyocytes. Simulation of reperfusion was achieved by restoration of nutrients in complete supplemented medium and incubation at 21% O2. Early application of therapeutic hypothermia, cooling during the oxygen-glucose deprivation phase, was initiated after 3h of oxygen-glucose deprivation and maintained for 24h. Mitochondrial membrane integrity was assessed by cytochrome c and AIF protein releases. Furthermore, mitochondria were stained with MitoTracker Red and intra-cellular cytochrome c localization was visualized by immunofluorescence staining. Moreover, anti-apoptotic Bcl-2 and Hsp70 as well as phagophore promoting LC3-II protein expressions were analyzed by Western-blot analysis. RESULTS Therapeutic hypothermia initiated during oxygen-glucose deprivation significantly reduced mitochondrial release of cytochrome c and AIF in cardiomyocytes during reperfusion. Secondly, anti-apoptotic Bcl-2/Bax ratio and Hsp70 protein expressions were significantly upregulated due to hypothermia, indicating an inhibition of both caspase-dependent and -independent apoptosis. Furthermore, cardiomyocytes treated with therapeutic hypothermia showed increased LC3-II protein levels associated with the mitochondria during the first 3h of reperfusion, indicating the initiation of phagophores formation and sequestration of presumably damaged mitochondrion. CONCLUSION Early application of therapeutic hypothermia effectively inhibited cardiomyocyte cell death due to oxygen-glucose deprivation/reperfusion-induced injury via multiple pathways. As hypothermia preserved mitochondrial membrane integrity, which resulted in reduced cytochrome c and AIF releases, induction of both caspase-dependent and -independent apoptosis was minimized. Secondly, cooling attenuated intrinsic apoptosis via Hsp70 upregulation and increasing anti-apoptotic Bcl-2/Bax ratio. Moreover, therapeutic hypothermia promoted mitochondrial associated LC3-II during the early phase of reperfusion, possibly leading to the sequestration and degradation of damaged mitochondrion to attenuate the activation of cell death.
Collapse
Affiliation(s)
- Jana Krech
- Department of Congenital Heart Disease/Pediatric Cardiology, German Heart Institute Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Giang Tong
- Department of Congenital Heart Disease/Pediatric Cardiology, German Heart Institute Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Sylvia Wowro
- Department of Congenital Heart Disease/Pediatric Cardiology, German Heart Institute Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Christoph Walker
- Department of Congenital Heart Disease/Pediatric Cardiology, German Heart Institute Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Lisa-Maria Rosenthal
- Department of Congenital Heart Disease/Pediatric Cardiology, German Heart Institute Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Felix Berger
- Department of Congenital Heart Disease/Pediatric Cardiology, German Heart Institute Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Department of Pediatric Cardiology, Charité - University Medical Center, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Katharina Rose Luise Schmitt
- Department of Congenital Heart Disease/Pediatric Cardiology, German Heart Institute Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
191
|
Rovira-Llopis S, Bañuls C, Diaz-Morales N, Hernandez-Mijares A, Rocha M, Victor VM. Mitochondrial dynamics in type 2 diabetes: Pathophysiological implications. Redox Biol 2017; 11:637-645. [PMID: 28131082 PMCID: PMC5284490 DOI: 10.1016/j.redox.2017.01.013] [Citation(s) in RCA: 392] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 01/05/2017] [Accepted: 01/13/2017] [Indexed: 12/18/2022] Open
Abstract
Mitochondria play a key role in maintaining cellular metabolic homeostasis. These organelles have a high plasticity and are involved in dynamic processes such as mitochondrial fusion and fission, mitophagy and mitochondrial biogenesis. Type 2 diabetes is characterised by mitochondrial dysfunction, high production of reactive oxygen species (ROS) and low levels of ATP. Mitochondrial fusion is modulated by different proteins, including mitofusin-1 (MFN1), mitofusin-2 (MFN2) and optic atrophy (OPA-1), while fission is controlled by mitochondrial fission 1 (FIS1), dynamin-related protein 1 (DRP1) and mitochondrial fission factor (MFF). PARKIN and (PTEN)-induced putative kinase 1 (PINK1) participate in the process of mitophagy, for which mitochondrial fission is necessary. In this review, we discuss the molecular pathways of mitochondrial dynamics, their impairment under type 2 diabetes, and pharmaceutical approaches for targeting mitochondrial dynamics, such as mitochondrial division inhibitor-1 (mdivi-1), dynasore, P110 and 15-oxospiramilactone. Furthermore, we discuss the pathophysiological implications of impaired mitochondrial dynamics, especially in type 2 diabetes.
Collapse
Affiliation(s)
- Susana Rovira-Llopis
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Celia Bañuls
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Noelia Diaz-Morales
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Antonio Hernandez-Mijares
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain; Department of Medicine, University of Valencia, Valencia, Spain
| | - Milagros Rocha
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain; CIBERehd - Department of Pharmacology, University of Valencia, Valencia, Spain
| | - Victor M Victor
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain; CIBERehd - Department of Pharmacology, University of Valencia, Valencia, Spain; Department of Physiology, University of Valencia, Valencia, Spain.
| |
Collapse
|
192
|
Mitochondrial dynamics as regulators of cancer biology. Cell Mol Life Sci 2017; 74:1999-2017. [PMID: 28083595 DOI: 10.1007/s00018-016-2451-3] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/22/2016] [Accepted: 12/29/2016] [Indexed: 02/07/2023]
Abstract
Mitochondria are dynamic organelles that supply energy required to drive key cellular processes, such as survival, proliferation, and migration. Critical to all of these processes are changes in mitochondrial architecture, a mechanical mechanism encompassing both fusion and fragmentation (fission) of the mitochondrial network. Changes to mitochondrial shape, size, and localization occur in a regulated manner to maintain energy and metabolic homeostasis, while deregulation of mitochondrial dynamics is associated with the onset of metabolic dysfunction and disease. In cancers, oncogenic signals that drive excessive proliferation, increase intracellular stress, and limit nutrient supply are all able to alter the bioenergetic and biosynthetic requirements of cancer cells. Consequently, mitochondrial function and shape rapidly adapt to these hostile conditions to support cancer cell proliferation and evade activation of cell death programs. In this review, we will discuss the molecular mechanisms governing mitochondrial dynamics and integrate recent insights into how changes in mitochondrial shape affect cellular migration, differentiation, apoptosis, and opportunities for the development of novel targeted cancer therapies.
Collapse
|
193
|
Go KL, Lee S, Behrns KE, Kim JS. Mitochondrial Damage and Mitophagy in Ischemia/Reperfusion-Induced Liver Injury. MOLECULES, SYSTEMS AND SIGNALING IN LIVER INJURY 2017:183-219. [DOI: 10.1007/978-3-319-58106-4_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
194
|
Liao C, Ashley N, Diot A, Morten K, Phadwal K, Williams A, Fearnley I, Rosser L, Lowndes J, Fratter C, Ferguson DJP, Vay L, Quaghebeur G, Moroni I, Bianchi S, Lamperti C, Downes SM, Sitarz KS, Flannery PJ, Carver J, Dombi E, East D, Laura M, Reilly MM, Mortiboys H, Prevo R, Campanella M, Daniels MJ, Zeviani M, Yu-Wai-Man P, Simon AK, Votruba M, Poulton J. Dysregulated mitophagy and mitochondrial organization in optic atrophy due to OPA1 mutations. Neurology 2016; 88:131-142. [PMID: 27974645 PMCID: PMC5224718 DOI: 10.1212/wnl.0000000000003491] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/04/2016] [Indexed: 01/13/2023] Open
Abstract
Objective: To investigate mitophagy in 5 patients with severe dominantly inherited optic atrophy (DOA), caused by depletion of OPA1 (a protein that is essential for mitochondrial fusion), compared with healthy controls. Methods: Patients with severe DOA (DOA plus) had peripheral neuropathy, cognitive regression, and epilepsy in addition to loss of vision. We quantified mitophagy in dermal fibroblasts, using 2 high throughput imaging systems, by visualizing colocalization of mitochondrial fragments with engulfing autophagosomes. Results: Fibroblasts from 3 biallelic OPA1(−/−) patients with severe DOA had increased mitochondrial fragmentation and mitochondrial DNA (mtDNA)–depleted cells due to decreased levels of OPA1 protein. Similarly, in siRNA-treated control fibroblasts, profound OPA1 knockdown caused mitochondrial fragmentation, loss of mtDNA, impaired mitochondrial function, and mitochondrial mislocalization. Compared to controls, basal mitophagy (abundance of autophagosomes colocalizing with mitochondria) was increased in (1) biallelic patients, (2) monoallelic patients with DOA plus, and (3) OPA1 siRNA–treated control cultures. Mitophagic flux was also increased. Genetic knockdown of the mitophagy protein ATG7 confirmed this by eliminating differences between patient and control fibroblasts. Conclusions: We demonstrated increased mitophagy and excessive mitochondrial fragmentation in primary human cultures associated with DOA plus due to biallelic OPA1 mutations. We previously found that increased mitophagy (mitochondrial recycling) was associated with visual loss in another mitochondrial optic neuropathy, Leber hereditary optic neuropathy (LHON). Combined with our LHON findings, this implicates excessive mitochondrial fragmentation, dysregulated mitophagy, and impaired response to energetic stress in the pathogenesis of mitochondrial optic neuropathies, potentially linked with mitochondrial mislocalization and mtDNA depletion.
Collapse
Affiliation(s)
- Chunyan Liao
- Author affiliations are provided at the end of the article
| | - Neil Ashley
- Author affiliations are provided at the end of the article
| | - Alan Diot
- Author affiliations are provided at the end of the article
| | - Karl Morten
- Author affiliations are provided at the end of the article
| | | | | | - Ian Fearnley
- Author affiliations are provided at the end of the article
| | - Lyndon Rosser
- Author affiliations are provided at the end of the article
| | - Jo Lowndes
- Author affiliations are provided at the end of the article
| | - Carl Fratter
- Author affiliations are provided at the end of the article
| | | | - Laura Vay
- Author affiliations are provided at the end of the article
| | | | | | | | | | - Susan M Downes
- Author affiliations are provided at the end of the article
| | - Kamil S Sitarz
- Author affiliations are provided at the end of the article
| | | | - Janet Carver
- Author affiliations are provided at the end of the article
| | - Eszter Dombi
- Author affiliations are provided at the end of the article
| | - Daniel East
- Author affiliations are provided at the end of the article
| | - Matilde Laura
- Author affiliations are provided at the end of the article
| | - Mary M Reilly
- Author affiliations are provided at the end of the article
| | | | - Remko Prevo
- Author affiliations are provided at the end of the article
| | | | | | | | | | | | | | - Joanna Poulton
- Author affiliations are provided at the end of the article.
| |
Collapse
|
195
|
Chen Z, Wu D, Li L, Chen L. Apelin/APJ System: A Novel Therapeutic Target for Myocardial Ischemia/Reperfusion Injury. DNA Cell Biol 2016; 35:766-775. [DOI: 10.1089/dna.2016.3391] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Zhe Chen
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drugs Study, University of South China, Hengyang, China
| | - Di Wu
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drugs Study, University of South China, Hengyang, China
| | - Lanfang Li
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drugs Study, University of South China, Hengyang, China
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drugs Study, University of South China, Hengyang, China
| |
Collapse
|
196
|
Wende AR, Young ME, Chatham J, Zhang J, Rajasekaran NS, Darley-Usmar VM. Redox biology and the interface between bioenergetics, autophagy and circadian control of metabolism. Free Radic Biol Med 2016; 100:94-107. [PMID: 27242268 PMCID: PMC5124549 DOI: 10.1016/j.freeradbiomed.2016.05.022] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 05/25/2016] [Accepted: 05/26/2016] [Indexed: 12/01/2022]
Abstract
Understanding molecular mechanisms that underlie the recent emergence of metabolic diseases such as diabetes and heart failure has revealed the need for a multi-disciplinary research integrating the key metabolic pathways which change the susceptibility to environmental or pathologic stress. At the physiological level these include the circadian control of metabolism which aligns metabolism with temporal demand. The mitochondria play an important role in integrating the redox signals and metabolic flux in response to the changing activities associated with chronobiology, exercise and diet. At the molecular level this involves dynamic post-translational modifications regulating transcription, metabolism and autophagy. In this review we will discuss different examples of mechanisms which link these processes together. An important pathway capable of linking signaling to metabolism is the post-translational modification of proteins by O-linked N-acetylglucosamine (O-GlcNAc). This is a nutrient regulated protein modification that plays an important role in impaired cellular stress responses. Circadian clocks have also emerged as critical regulators of numerous cardiometabolic processes, including glucose/lipid homeostasis, hormone secretion, redox status and cardiovascular function. Central to these pathways are the response of autophagy, bioenergetics to oxidative stress, regulated by Keap1/Nrf2 and mechanisms of metabolic control. The extension of these ideas to the emerging concept of bioenergetic health will be discussed.
Collapse
Affiliation(s)
- Adam R Wende
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Martin E Young
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - John Chatham
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Namakkal S Rajasekaran
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Victor M Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; UAB Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
197
|
Canonico B, Cesarini E, Salucci S, Luchetti F, Falcieri E, Di Sario G, Palma F, Papa S. Defective Autophagy, Mitochondrial Clearance and Lipophagy in Niemann-Pick Type B Lymphocytes. PLoS One 2016; 11:e0165780. [PMID: 27798705 PMCID: PMC5087958 DOI: 10.1371/journal.pone.0165780] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 10/18/2016] [Indexed: 12/21/2022] Open
Abstract
Niemann-Pick disease type A (NP-A) and type B (NP-B) are lysosomal storage diseases (LSDs) caused by sphingomyelin accumulation in lysosomes relying on reduced or absent acid sphingomyelinase. A considerable body of evidence suggests that lysosomal storage in many LSD impairs autophagy, resulting in the accumulation of poly-ubiquitinated proteins and dysfunctional mitochondria, ultimately leading to cell death. Here we test this hypothesis in a cellular model of Niemann-Pick disease type B, in which autophagy has never been studied. The basal autophagic pathway was first examined in order to evaluate its functionality using several autophagy-modulating substances such as rapamycin and nocodazole. We found that human NP-B B lymphocytes display considerable alteration in their autophagic vacuole accumulation and mitochondrial fragmentation, as well as mitophagy induction (for damaged mitochondria clearance). Furthermore, lipid traceability of intra and extra-cellular environments shows lipid accumulation in NP-B B lymphocytes and also reveals their peculiar trafficking/management, culminating in lipid microparticle extrusion (by lysosomal exocytosis mechanisms) or lipophagy. All of these features point to the presence of a deep autophagy/mitophagy alteration revealing autophagic stress and defective mitochondrial clearance. Hence, rapamycin might be used to regulate autophagy/mitophagy (at least in part) and to contribute to the clearance of lysosomal aberrant lipid storage.
Collapse
Affiliation(s)
- Barbara Canonico
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
- * E-mail:
| | - Erica Cesarini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Sara Salucci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Francesca Luchetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Elisabetta Falcieri
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
- IGM, CNR, Rizzoli Orthopaedic Institute, Bologna, Italy; Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Gianna Di Sario
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Fulvio Palma
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Stefano Papa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| |
Collapse
|
198
|
Podratz JL, Lee H, Knorr P, Koehler S, Forsythe S, Lambrecht K, Arias S, Schmidt K, Steinhoff G, Yudintsev G, Yang A, Trushina E, Windebank A. Cisplatin induces mitochondrial deficits in Drosophila larval segmental nerve. Neurobiol Dis 2016; 97:60-69. [PMID: 27765583 DOI: 10.1016/j.nbd.2016.10.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 10/04/2016] [Accepted: 10/16/2016] [Indexed: 12/23/2022] Open
Abstract
Cisplatin is an effective chemotherapy drug that induces peripheral neuropathy in cancer patients. In rodent dorsal root ganglion neurons, cisplatin binds nuclear and mitochondrial DNA (mtDNA) inducing DNA damage and apoptosis. Platinum-mtDNA adducts inhibit mtDNA replication and transcription leading to mitochondrial degradation. Cisplatin also induces climbing deficiencies associated with neuronal apoptosis in adult Drosophila melanogaster. Here we used Drosophila larvae that express green fluorescent protein in the mitochondria of motor neurons to observe the effects of cisplatin on mitochondrial dynamics and function. Larvae treated with 10μg/ml cisplatin had normal survival with deficiencies in righting and heat sensing behavior. Behavior was abrogated by, the pan caspase inhibitor, p35. However, active caspase 3 was not detected by immunostaining. There was a 27% decrease in mitochondrial membrane potential and a 42% increase in reactive oxygen species (ROS) in mitochondria along the axon. Examination of mitochondrial axonal trafficking showed no changes in velocity, flux or mitochondrial length. However, cisplatin treatment resulted in a greater number of stationary organelles caused by extended pausing during axonal motility. These results demonstrate that cisplatin induces behavior deficiencies in Drosophila larvae, decreased mitochondrial activity, increased ROS production and mitochondrial pausing without killing the larvae. Thus, we identified particular aspects of mitochondrial dynamics and function that are affected in cisplatin-induced peripheral neuropathy and may represent key therapeutic targets.
Collapse
Affiliation(s)
| | - Han Lee
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Patrizia Knorr
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Suzette Arias
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Kiley Schmidt
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Georgiy Yudintsev
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Amy Yang
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Eugenia Trushina
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
199
|
Hernández-Corbacho MJ, Salama MF, Canals D, Senkal CE, Obeid LM. Sphingolipids in mitochondria. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1862:56-68. [PMID: 27697478 DOI: 10.1016/j.bbalip.2016.09.019] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/01/2016] [Accepted: 09/24/2016] [Indexed: 01/16/2023]
Abstract
Sphingolipids are bioactive lipids found in cell membranes that exert a critical role in signal transduction. In recent years, it has become apparent that sphingolipids participate in growth, senescence, differentiation and apoptosis. The anabolism and catabolism of sphingolipids occur in discrete subcellular locations and consist of a strictly regulated and interconnected network, with ceramide as the central hub. Altered sphingolipid metabolism is linked to several human diseases. Hence, an advanced knowledge of how and where sphingolipids are metabolized is of paramount importance in order to understand the role of sphingolipids in cellular functions. In this review, we provide an overview of sphingolipid metabolism. We focus on the distinct pathways of ceramide synthesis, highlighting the mitochondrial ceramide generation, transport of ceramide to mitochondria and its role in the regulation of mitochondrial-mediated apoptosis, mitophagy and implications to disease. We will discuss unanswered questions and exciting future directions. This article is part of a Special Issue entitled: Lipids of Mitochondria edited by Guenther Daum.
Collapse
Affiliation(s)
- María José Hernández-Corbacho
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY 11794, USA
| | - Mohamed F Salama
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY 11794, USA; Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Daniel Canals
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY 11794, USA
| | - Can E Senkal
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY 11794, USA
| | - Lina M Obeid
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY 11794, USA; The Northport VA Medical Center, Northport, NY 11768, USA.
| |
Collapse
|
200
|
Plecitá-Hlavatá L, Ježek P. Integration of superoxide formation and cristae morphology for mitochondrial redox signaling. Int J Biochem Cell Biol 2016; 80:31-50. [PMID: 27640755 DOI: 10.1016/j.biocel.2016.09.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/09/2016] [Accepted: 09/12/2016] [Indexed: 12/16/2022]
Abstract
The mitochondrial network provides the central cell's energetic and regulatory unit, which besides ATP and metabolite production participates in cellular signaling through regulated reactive oxygen species (ROS) production and various protein/ion fluxes. The inner membrane forms extensive folds, called cristae, i.e. cavities enfolded from and situated perpendicularly to its inner boundary membrane portion, which encompasses an inner cylinder within the outer membrane tubule. Mitochondrial cristae ultramorphology reflects various metabolic, physiological or pathological states. Since the mitochondrion is typically a predominant superoxide source and generated ROS also serve for the creation of information redox signals, we review known relationships between ROS generation within the respiratory chain complexes of cristae and cristae morphology. Notably, it is emphasized that cristae shape is governed by ATP-synthase dimers, MICOS complexes, OPA1 isoforms and the umbrella of their regulation, and also dependent on local protonmotive force (electrical potential component) in cristae. Cristae are also affected by redox-sensitive kinases/phosphatases or p66SHC. ATP-synthase dimers decrease in the inflated intracristal space, diminishing pH and hypothetically having minimal superoxide formation. Matrix-released signaling superoxide/H2O2 is predominantly integrated along mitochondrial tubules, whereas the diffusion of intracristal signaling ROS species is controlled by crista junctions, the widening of which enables specific retrograde redox signaling such as during hypoxic cell adaptation. Other physiological cases of H2O2 release from the mitochondrion include the modulation of insulin release in pancreatic β-cells, enhancement of insulin signaling in peripheral tissues, signaling by T-cell receptors, retrograde signaling during the cell cycle and cell differentiation, specifically that of adipocytes.
Collapse
Affiliation(s)
- Lydie Plecitá-Hlavatá
- Department of Membrane Transport Biophysics, No.75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Petr Ježek
- Department of Membrane Transport Biophysics, No.75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| |
Collapse
|