1
|
Justice JL, Greco TM, Hutton JE, Reed TJ, Mair ML, Botas J, Cristea IM. Multi-epitope immunocapture of huntingtin reveals striatum-selective molecular signatures. Mol Syst Biol 2025; 21:492-522. [PMID: 40169779 PMCID: PMC12048488 DOI: 10.1038/s44320-025-00096-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 03/03/2025] [Accepted: 03/13/2025] [Indexed: 04/03/2025] Open
Abstract
Huntington's disease (HD) is a debilitating neurodegenerative disorder affecting an individual's cognitive and motor abilities. HD is caused by a mutation in the huntingtin gene producing a toxic polyglutamine-expanded protein (mHTT) and leading to degeneration in the striatum and cortex. Yet, the molecular signatures that underlie tissue-specific vulnerabilities remain unclear. Here, we investigate this aspect by leveraging multi-epitope protein interaction assays, subcellular fractionation, thermal proteome profiling, and genetic modifier assays. The use of human cell, mouse, and fly models afforded capture of distinct subcellular pools of epitope-enriched and tissue-dependent interactions linked to dysregulated cellular pathways and disease relevance. We established an HTT association with nearly all subunits of the transcriptional regulatory Mediator complex (20/26), with preferential enrichment of MED15 in the tail domain. Using HD and KO models, we find HTT modulates the subcellular localization and assembly of the Mediator. We demonstrated striatal enriched and functional interactions with regulators of calcium homeostasis and chromatin remodeling, whose disease relevance was supported by HD fly genetic modifiers assays. Altogether, we offer insights into tissue- and localization-dependent (m)HTT functions and pathobiology.
Collapse
Affiliation(s)
- Joshua L Justice
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ, 08544, USA
| | - Todd M Greco
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ, 08544, USA
| | - Josiah E Hutton
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ, 08544, USA
| | - Tavis J Reed
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ, 08544, USA
| | - Megan L Mair
- Jan and Dan Duncan Neurological Research Institute, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Juan Botas
- Jan and Dan Duncan Neurological Research Institute, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ, 08544, USA.
| |
Collapse
|
2
|
Li Y, Li X, Zhu L, Liu T, Huang L. Chitosan-based biomaterials for bone tissue engineering. Int J Biol Macromol 2025; 304:140923. [PMID: 39947561 DOI: 10.1016/j.ijbiomac.2025.140923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/30/2025] [Accepted: 02/09/2025] [Indexed: 02/17/2025]
Abstract
Common critical size bone defects encountered in clinical practice often result in inadequate bone regeneration,primarily due to the extent of damage surpassing the inherent capacity of the body for self-healing. Bone tissue engineering scaffolds possess the desirable characteristics of biomimetic bone structure, simulated extracellular matrix, optimal mechanical strength, and biological functionality, rendering them the preferred option for the treatment of bone defects. Chitosan demonstrates favorable biocompatibility, plasticity, and a range of biological activities, rendering it a highly appealing material. Chitosan and its derivatives have been found to exert an impact on bone repair through their ability to modulate macrophage polarization, angiogenesis, and the delicate equilibrium of bone remodeling. However, the efficacy of pure chitosan is constrained, necessitating its combination with other bioactive substances to achieve an optimal biomimetic scaffold that is compatible with the specific bone defect site. Chitosan is commonly utilized in the field of bone repair in four different application forms: rigid scaffold, hydrogel, membranes, and microspheres. In order to enhance comprehension of the benefits and constraints associated with chitosan, this review provides a comprehensive overview of the structure and biological properties of chitosan, the molecular mechanisms by which chitosan promotes osteogenic differentiation, the diverse methods of chitosan preparation for various applications, and the impacts of chitosan when loaded with bioactive substances.
Collapse
Affiliation(s)
- Youbin Li
- The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Xudong Li
- The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Liwei Zhu
- The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Tengyue Liu
- The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Lanfeng Huang
- The Second Hospital of Jilin University, Changchun 130041, PR China.
| |
Collapse
|
3
|
Chen JK, Ramesh S, Islam MN, Shibu MA, Kuo CH, Hsieh DJY, Lin SZ, Kuo WW, Huang CY, Ho TJ. Artemisia argyi mitigates doxorubicin-induced cardiotoxicity by inhibiting mitochondrial dysfunction through the IGF-IIR/Drp1/GATA4 signaling pathway. Biotechnol Appl Biochem 2025; 72:388-401. [PMID: 39375847 DOI: 10.1002/bab.2671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 10/09/2024]
Abstract
Doxorubicin (DOX) is mostly utilized as a wide range of antitumor anthracycline to treat different cancers. The severe antagonistic impacts of DOX on cardiotoxicity constrain its clinical application. Many mechanisms are involved in cardiac toxicity induced by DOX in the human body. Mitochondria is a central part of fatty acid and glucose metabolism. Thus, impaired mitochondrial metabolism can increase heart failure risk, which can play a vital role in cardiomyocyte mitochondrial dysfunction. This study aimed to assess the possible cardioprotective effect of water-extracted Artemisia argyi (AA) against the side effect of DOX in H9c2 cells and whether these protective effects are mediated through IGF-IIR/Drp1/GATA4 signaling pathways. Although several studies proved that AA extract has benefits for various diseases, its cardiac effects have not yet been identified. The H9c2 cells were exposed to 1 μM to establish a model of cardiac toxicity. The results revealed that water-extracted AA could block the expression of IGF-IIR/calcineurin signaling pathways induced by DOX. Notably, our results also showed that AA treatment markedly attenuated Akt phosphorylation and cleaved caspase 3, and the nuclear translocation markers NFATC3 and p-GATA4. Using actin staining for hypertrophy, we determined that AA can reduce the effect of mitochondrial reactive oxygen species and cell size. These findings suggest that water-extracted AA could be a suitable candidate for preventing DOX-induced cardiac damage.
Collapse
Affiliation(s)
- Jhong-Kuei Chen
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Samiraj Ramesh
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Research and Innovation, Institute of Biotechnology, Saveetha School of Engineering (SSE), Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| | - Md Nazmul Islam
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | | | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Dennis Jine-Yuan Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shinn-Zong Lin
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
- Ph.D. Program for Biotechnology Industry, China Medical University, Taichung, Taiwan
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien, Taiwan
| | - Tsung-Jung Ho
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
- School of Post-Baccalaureate Chinese Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
4
|
Liu X, Li T, Tu X, Xu M, Wang J. Mitochondrial fission and fusion in neurodegenerative diseases:Ca 2+ signalling. Mol Cell Neurosci 2025; 132:103992. [PMID: 39863029 DOI: 10.1016/j.mcn.2025.103992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Neurodegenerative diseases (NDs) are a group of disorders characterized by the progressive loss of neuronal structure and function. The pathogenesis is intricate and involves a network of interactions among multiple causes and systems. Mitochondria and Ca2+ signaling have long been considered to play important roles in the development of various NDs. Mitochondrial fission and fusion dynamics are important processes of mitochondrial quality control, ensuring the stability of mitochondrial structure and function. Mitochondrial fission and fusion imbalance and Ca2+ signaling disorders can aggravate the disease progression of NDs. In this review, we explore the relationship between mitochondrial dynamics and Ca2+ signaling in AD, PD, ALS, and HD, focusing on the roles of key regulatory proteins (Drp1, Fis1, Mfn1/2, and Opa1) and the association structures between mitochondria and the endoplasmic reticulum (MERCs/MAMs). We provide a detailed analysis of their involvement in the pathogenesis of these four NDs. By integrating these mechanisms, we aim to clarify their contributions to disease progression and offer insights into the development of therapeutic strategies that target mitochondrial dynamics and Ca2+ signaling. We also examine the progress in drug research targeting these pathways, highlighting their potential as therapeutic targets in the treatment of NDs.
Collapse
Affiliation(s)
- Xuan Liu
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| | - Tianjiao Li
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| | - Xinya Tu
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| | - Mengying Xu
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| | - Jianwu Wang
- Xiangya School of Public Health, Central South University, Changsha, Hunan Province, PR China.
| |
Collapse
|
5
|
Park W, Lee H, Lim W, Song G, Park S. Tri-o-tolyl phosphate impedes implantation: Malfunction of mitochondria and disruption of calcium homeostasis through MAPK and AKT signaling cascades. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 958:177920. [PMID: 39662411 DOI: 10.1016/j.scitotenv.2024.177920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/21/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024]
Abstract
Tri-o-tolyl phosphate (TOTP), a flame retardant containing aryl compounds, is widely used in human living environments owing to its several applications. However, Due to the overuse of TOTP, its residue has been identified in various environments and non-targeted organisms, including humans. Although extensive research is being conducted to address the toxicity of this substance, its potential reproductive toxicity in females has not been sufficiently studied. In this study, human HTR-8/SVneo and JEG-3 trophoblasts were used to investigate the effects of TOTP on implantation. Results showed that TOTP decreased cell viability and inhibited cell proliferation by triggering cell cycle arrest. It also induced apoptosis and mitochondrial dysfunction, disrupted calcium homeostasis, increased the influx of calcium ions into the mitochondria, and disturbed cell aggregation and migration. Moreover, the MAPK and AKT cell signaling pathways were altered, and crosstalk between these pathways were distinguished. Thus, inhibitors of the MAPK and AKT pathways exhibited potential for managing the toxicity of TOTP. Overall, this study demonstrated the reproductive toxicity of TOTP in human females and elucidated the underlying mechanisms. Our results highlighted the potential risks associated with TOTP.
Collapse
Affiliation(s)
- Wonhyoung Park
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Hojun Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Whasun Lim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| | - Sunwoo Park
- Department of GreenBio Science, Gyeongsang National University, Jinju 52725, Republic of Korea.
| |
Collapse
|
6
|
Yoon G, Kam MK, Koh YH, Jo C. Palmitoyl-L-carnitine induces tau phosphorylation and mitochondrial dysfunction in neuronal cells. PLoS One 2024; 19:e0313507. [PMID: 39536002 PMCID: PMC11560007 DOI: 10.1371/journal.pone.0313507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by cognitive decline and memory loss, involving mechanisms such as tau hyperphosphorylation and mitochondrial dysfunction. Increasing evidence suggests that age-related alterations in metabolite levels are crucial for the pathogenesis of AD. Here, we analyzed serum metabolites from mice of various ages (2, 4, 14, and 21 months old) using mass spectrometry. We identified palmitoyl-L-carnitine as a key metabolite with significantly increased levels in aged mice. In vitro experiments with SH-SY5Y neuronal cells demonstrated that palmitoyl-L-carnitine treatment enhanced tau phosphorylation, increased mitochondrial fission, and elevated intracellular calcium levels. Furthermore, the increased levels of tau phosphorylation were significantly reduced by the inhibition of GSK-3β, CDK5, and calpain, indicating that tau kinases activated by calcium overload are directly involved in the increase of tau phosphorylation. Considering that mitochondrial fission is related to mitochondrial dysfunction, we propose that the elevated level of serum palmitoyl-L-carnitine during aging contributes to AD pathology through these pathways. These findings highlight the significant role of lipid metabolism in neurodegeneration and offer potential therapeutic targets for age-related diseases, including AD.
Collapse
Affiliation(s)
- Gwangho Yoon
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Min Kyoung Kam
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Young Ho Koh
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Chulman Jo
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| |
Collapse
|
7
|
Kim KH, Lee CB. Socialized mitochondria: mitonuclear crosstalk in stress. Exp Mol Med 2024; 56:1033-1042. [PMID: 38689084 PMCID: PMC11148012 DOI: 10.1038/s12276-024-01211-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/27/2024] [Accepted: 02/07/2024] [Indexed: 05/02/2024] Open
Abstract
Traditionally, mitochondria are considered sites of energy production. However, recent studies have suggested that mitochondria are signaling organelles that are involved in intracellular interactions with other organelles. Remarkably, stressed mitochondria appear to induce a beneficial response that restores mitochondrial function and cellular homeostasis. These mitochondrial stress-centered signaling pathways have been rapidly elucidated in multiple organisms. In this review, we examine current perspectives on how mitochondria communicate with the rest of the cell, highlighting mitochondria-to-nucleus (mitonuclear) communication under various stresses. Our understanding of mitochondria as signaling organelles may provide new insights into disease susceptibility and lifespan extension.
Collapse
Affiliation(s)
- Kyung Hwa Kim
- Department of Health Sciences, The Graduate School of Dong-A University, 840 Hadan-dong, Saha-gu, Busan, 49315, Korea.
| | - Cho Bi Lee
- Department of Health Sciences, The Graduate School of Dong-A University, 840 Hadan-dong, Saha-gu, Busan, 49315, Korea
| |
Collapse
|
8
|
Martin N, Zhu K, Czarnecka-Herok J, Vernier M, Bernard D. Regulation and role of calcium in cellular senescence. Cell Calcium 2023; 110:102701. [PMID: 36736165 DOI: 10.1016/j.ceca.2023.102701] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/28/2023]
Abstract
Cellular senescence is a state of stable cell proliferation arrest accompanied by a distinct secretory program impacting the senescent cell microenvironment. This phenotype can be induced by many stresses, including telomere shortening, oncogene activation, oxidative or genotoxic stress. Cellular senescence plays a key role in the organism throughout life, with beneficial effects at a young age for instance in embryonic development and wound healing, and deleterious effects during aging and in aging-related diseases. In the last decade calcium and calcium signaling have been established as critical factors in the implementation and regulation of cellular senescence. In this review we will present and discuss the main discoveries in this field, from the observation of an increased intracellular calcium concentration in senescent cells to the identification of calcium-binding proteins, calcium channels (TRP, ITPR, …) and MERCs (mitochondria-endoplasmic reticulum contact sites) as key players in this context.
Collapse
Affiliation(s)
- Nadine Martin
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Equipe Labellisée la Ligue Contre le Cancer, Université de Lyon, Lyon, France.
| | - Kexin Zhu
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Equipe Labellisée la Ligue Contre le Cancer, Université de Lyon, Lyon, France
| | - Joanna Czarnecka-Herok
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Equipe Labellisée la Ligue Contre le Cancer, Université de Lyon, Lyon, France
| | - Mathieu Vernier
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Equipe Labellisée la Ligue Contre le Cancer, Université de Lyon, Lyon, France
| | - David Bernard
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR 5286, Centre Léon Bérard, Equipe Labellisée la Ligue Contre le Cancer, Université de Lyon, Lyon, France.
| |
Collapse
|
9
|
Zhai R, Fang B, Lai Y, Peng B, Bai H, Liu X, Li L, Huang W. Small-molecule fluorogenic probes for mitochondrial nanoscale imaging. Chem Soc Rev 2023; 52:942-972. [PMID: 36514947 DOI: 10.1039/d2cs00562j] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mitochondria are inextricably linked to the development of diseases and cell metabolism disorders. Super-resolution imaging (SRI) is crucial in enhancing our understanding of mitochondrial ultrafine structures and functions. In addition to high-precision instruments, super-resolution microscopy relies heavily on fluorescent materials with unique photophysical properties. Small-molecule fluorogenic probes (SMFPs) have excellent properties that make them ideal for mitochondrial SRI. This paper summarizes recent advances in the field of SMFPs, with a focus on the chemical and spectroscopic properties required for mitochondrial SRI. Finally, we discuss future challenges in this field, including the design principles of SMFPs and nanoscopic techniques.
Collapse
Affiliation(s)
- Rongxiu Zhai
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, Xi'an 710072, China.
| | - Bin Fang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, Xi'an 710072, China. .,School of Materials Science and Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China
| | - Yaqi Lai
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, Xi'an 710072, China.
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, Xi'an 710072, China.
| | - Hua Bai
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, Xi'an 710072, China.
| | - Xiaowang Liu
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, Xi'an 710072, China.
| | - Lin Li
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, Xi'an 710072, China. .,The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, Fujian, China
| | - Wei Huang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, Xi'an 710072, China. .,The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, Fujian, China
| |
Collapse
|
10
|
Bassot A, Chen J, Takahashi-Yamashiro K, Yap MC, Gibhardt CS, Le GNT, Hario S, Nasu Y, Moore J, Gutiérrez T, Mina L, Mast H, Moses A, Bhat R, Ballanyi K, Lemieux H, Sitia R, Zito E, Bogeski I, Campbell RE, Simmen T. The endoplasmic reticulum kinase PERK interacts with the oxidoreductase ERO1 to metabolically adapt mitochondria. Cell Rep 2023; 42:111899. [PMID: 36586409 DOI: 10.1016/j.celrep.2022.111899] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 10/04/2022] [Accepted: 12/08/2022] [Indexed: 12/31/2022] Open
Abstract
Endoplasmic reticulum (ER) homeostasis requires molecular regulators that tailor mitochondrial bioenergetics to the needs of protein folding. For instance, calnexin maintains mitochondria metabolism and mitochondria-ER contacts (MERCs) through reactive oxygen species (ROS) from NADPH oxidase 4 (NOX4). However, induction of ER stress requires a quick molecular rewiring of mitochondria to adapt to new energy needs. This machinery is not characterized. We now show that the oxidoreductase ERO1⍺ covalently interacts with protein kinase RNA-like ER kinase (PERK) upon treatment with tunicamycin. The PERK-ERO1⍺ interaction requires the C-terminal active site of ERO1⍺ and cysteine 216 of PERK. Moreover, we show that the PERK-ERO1⍺ complex promotes oxidization of MERC proteins and controls mitochondrial dynamics. Using proteinaceous probes, we determined that these functions improve ER-mitochondria Ca2+ flux to maintain bioenergetics in both organelles, while limiting oxidative stress. Therefore, the PERK-ERO1⍺ complex is a key molecular machinery that allows quick metabolic adaptation to ER stress.
Collapse
Affiliation(s)
- Arthur Bassot
- Department of Cell Biology, Faculty of Medicine and Dentistry, Edmonton, AB T6G 2G2, Canada
| | - Junsheng Chen
- Department of Cell Biology, Faculty of Medicine and Dentistry, Edmonton, AB T6G 2G2, Canada
| | | | - Megan C Yap
- Department of Cell Biology, Faculty of Medicine and Dentistry, Edmonton, AB T6G 2G2, Canada
| | - Christine Silvia Gibhardt
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Giang N T Le
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Saaya Hario
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yusuke Nasu
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Jack Moore
- Alberta Proteomics and Mass Spectrometry Facility, University of Alberta, 4096 Katz Research Building, Edmonton AB T6G2E1, Canada
| | - Tomas Gutiérrez
- Department of Cell Biology, Faculty of Medicine and Dentistry, Edmonton, AB T6G 2G2, Canada
| | - Lucas Mina
- Department of Cell Biology, Faculty of Medicine and Dentistry, Edmonton, AB T6G 2G2, Canada
| | - Heather Mast
- Faculty Saint-Jean, Department of Medicine, Faculty of Medicine and Dentistry, Edmonton, AB T6G2H7, Canada
| | - Audric Moses
- Department of Pediatrics, Edmonton, AB T6G2H7, Canada
| | - Rakesh Bhat
- Precision Biolaboratories, St. Albert, AB T8N 5A7, Canada
| | - Klaus Ballanyi
- Department of Physiology, University of Alberta, Edmonton, AB T6G2H7, Canada
| | - Hélène Lemieux
- Faculty Saint-Jean, Department of Medicine, Faculty of Medicine and Dentistry, Edmonton, AB T6G2H7, Canada
| | - Roberto Sitia
- Division of Genetics and Cell Biology, Università Vita-Salute IRCCS Ospedale San Raffaele, 20132 Milano, Italy
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri, 20156 Milano, Italy; Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino PU, Italy
| | - Ivan Bogeski
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Robert E Campbell
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada; Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Thomas Simmen
- Department of Cell Biology, Faculty of Medicine and Dentistry, Edmonton, AB T6G 2G2, Canada.
| |
Collapse
|
11
|
Bar-Peled L, Kory N. Principles and functions of metabolic compartmentalization. Nat Metab 2022; 4:1232-1244. [PMID: 36266543 PMCID: PMC10155461 DOI: 10.1038/s42255-022-00645-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/24/2022] [Indexed: 01/20/2023]
Abstract
Metabolism has historically been studied at the levels of whole cells, whole tissues and whole organisms. As a result, our understanding of how compartmentalization-the spatial and temporal separation of pathways and components-shapes organismal metabolism remains limited. At its essence, metabolic compartmentalization fulfils three important functions or 'pillars': establishing unique chemical environments, providing protection from reactive metabolites and enabling the regulation of metabolic pathways. However, how these pillars are established, regulated and maintained at both the cellular and systemic levels remains unclear. Here we discuss how the three pillars are established, maintained and regulated within the cell and discuss the consequences of dysregulation of metabolic compartmentalization in human disease. Organelles are increasingly emerging as 'command-and-control centres' and the increased understanding of metabolic compartmentalization is revealing new aspects of metabolic homeostasis, with this knowledge being translated into therapies for the treatment of cancer and certain neurodegenerative diseases.
Collapse
Affiliation(s)
- Liron Bar-Peled
- Center for Cancer Research, Massachusetts General Hospital and Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Nora Kory
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
12
|
Dang CP, Issara-Amphorn J, Charoensappakit A, Udompornpitak K, Bhunyakarnjanarat T, Saisorn W, Sae-Khow K, Leelahavanichkul A. BAM15, a Mitochondrial Uncoupling Agent, Attenuates Inflammation in the LPS Injection Mouse Model: An Adjunctive Anti-Inflammation on Macrophages and Hepatocytes. J Innate Immun 2021; 13:359-375. [PMID: 34062536 PMCID: PMC8613553 DOI: 10.1159/000516348] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/30/2021] [Indexed: 11/19/2022] Open
Abstract
Controlof immune responses through the immunometabolism interference is interesting for sepsis treatment. Then, expression of immunometabolism-associated genes and BAM15, a mitochondrial uncoupling agent, was explored in a proinflammatory model using lipopolysaccharide (LPS) injection. Accordingly, the decreased expression of mitochondrial uncoupling proteins was demonstrated by transcriptomic analysis on metabolism-associated genes in macrophages (RAW246.7) and by polymerase chain reaction in LPS-stimulated RAW246.7 and hepatocytes (Hepa 1-6). Pretreatment with BAM15 at 24 h prior to LPS in macrophages attenuated supernatant inflammatory cytokines (IL-6, TNF-α, and IL-10), downregulated genes of proinflammatory M1 polarization (iNOS and IL-1β), upregulated anti-inflammatory M2 polarization (Arg1 and FIZZ), and decreased cell energy status (extracellular flux analysis and ATP production). Likewise, BAM15 decreased expression of proinflammatory genes (IL-6, TNF-α, IL-10, and iNOS) and reduced cell energy in hepatocytes. In LPS-administered mice, BAM15 attenuated serum cytokines, organ injury (liver enzymes and serum creatinine), and tissue cytokines (livers and kidneys), in part, through the enhanced phosphorylated αAMPK, a sensor of ATP depletion with anti-inflammatory property, in the liver, and reduced inflammatory monocytes/macrophages (Ly6C +ve, CD11b +ve) in the liver as detected by Western blot and flow cytometry, respectively. In conclusion, a proof of concept for inflammation attenuation of BAM15 through metabolic interference-induced anti-inflammation on macrophages and hepatocytes was demonstrated as a new strategy of anti-inflammation in sepsis.
Collapse
Affiliation(s)
- Cong Phi Dang
- Medical Microbiology, Interdisciplinary and International Program, Graduate School, Chulalongkorn University, Bangkok, Thailand,
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand,
| | | | - Awirut Charoensappakit
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Kanyarat Udompornpitak
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Wilasinee Saisorn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Kritsanawan Sae-Khow
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Translational Research in Inflammation and Immunology Research Unit (TRIRU), Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
13
|
Klier PEZ, Martin JG, Miller EW. Imaging Reversible Mitochondrial Membrane Potential Dynamics with a Masked Rhodamine Voltage Reporter. J Am Chem Soc 2021; 143:4095-4099. [PMID: 33710896 DOI: 10.1021/jacs.0c13110] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mitochondria are the site of aerobic respiration, producing ATP via oxidative phosphorylation as protons flow down their electrochemical gradient through ATP synthase. This negative membrane potential across the inner mitochondrial membrane (ΔΨm) represents a fundamental biophysical parameter central to cellular life. Traditional, electrode-based methods for recording membrane potential are impossible to implement on mitochondria within intact cells. Fluorescent ΔΨm indicators based on cationic, lipophilic dyes are a common alternative, but these indicators are complicated by concentration-dependent artifacts and the requirement to maintain dye in the extracellular solution to visualize reversible ΔΨm dynamics. Here, we report the first example of a fluorescent ΔΨm reporter that does not rely on ΔΨm-dependent accumulation. We redirected the localization of a photoinduced electron transfer (PeT)-based indicator, Rhodamine Voltage Reporter (RhoVR), to mitochondria by masking the carboxylate of RhoVR 1 as an acetoxymethyl (AM) ester. Once within mitochondria, esterases remove the AM ester, trapping RhoVR inside of the mitochondrial matrix, where it can incorporate within the inner membrane and reversibly report on changes in ΔΨm. We show that this Small molecule, Permeable, Internally Redistributing for Inner membrane Targeting Rhodamine Voltage Reporter, or SPIRIT RhoVR, localizes to mitochondria across a number of different cell lines and responds reversibly to changes in ΔΨm induced by exceptionally low concentrations of the uncoupler FCCP without the need for exogenous pools of dye (unlike traditional, accumulation-based rhodamine esters). SPIRIT RhoVR is compatible with multi-color imaging, enabling simultaneous, real-time observation of cytosolic Ca2+, plasma membrane potential, and reversible ΔΨm dynamics.
Collapse
|
14
|
Zhang X, Griepentrog JE, Zou B, Xu L, Cyr AR, Chambers LM, Zuckerbraun BS, Shiva S, Rosengart MR. CaMKIV regulates mitochondrial dynamics during sepsis. Cell Calcium 2020; 92:102286. [PMID: 32932146 DOI: 10.1016/j.ceca.2020.102286] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022]
Abstract
Sepsis and shock states impose mitochondrial stress, and in response, adaptive mechanisms such as fission, fusion and mitophagy are induced to eliminate damaged portions of or entire dysfunctional mitochondria. The mechanisms underlying these events are being elucidated; yet a direct link between loss of mitochondrial membrane potential ΔΨm and the initiation of fission, fusion and mitophagy remains to be well characterized. The direct association between the magnitude of the ΔΨm and the capacity for mitochondria to buffer Ca2+ renders Ca2+ uniquely suited as the signal engaging these mechanisms in circumstances of mitochondrial stress that lower the ΔΨm. Herein, we show that the calcium/calmodulin-dependent protein kinase (CaMK) IV mediates an adaptive slowing in oxidative respiration that minimizes oxidative stress in the kidneys of mice subjected to either cecal ligation and puncture (CLP) sepsis or endotoxemia. CaMKIV shifts the balance towards mitochondrial fission and away from fusion by 1) directly phosphorylating an activating Serine616 on the fission protein DRP1 and 2) reducing the expression of the fusion proteins Mfn1/2 and OPA-1. CaMKIV, through its function as a direct PINK1 kinase and regulator of Parkin expression, also enables mitophagy. These data support that CaMKIV serves as a keystone linking mitochondrial stress with the adaptive mechanisms of mitochondrial fission, fusion and mitophagy that mitigate oxidative stress in the kidneys of mice responding to sepsis.
Collapse
Affiliation(s)
- Xianghong Zhang
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - John E Griepentrog
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Baobo Zou
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Li Xu
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States; Department of Emergency, Union Hospital, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan, China
| | - Anthony R Cyr
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lauran M Chambers
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Brian S Zuckerbraun
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Sruti Shiva
- Department of Pharmacology & Chemical Biology, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Matthew R Rosengart
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
15
|
Karakaidos P, Rampias T. Mitonuclear Interactions in the Maintenance of Mitochondrial Integrity. Life (Basel) 2020; 10:life10090173. [PMID: 32878185 PMCID: PMC7555762 DOI: 10.3390/life10090173] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/28/2020] [Indexed: 12/27/2022] Open
Abstract
In eukaryotic cells, mitochondria originated in an α-proteobacterial endosymbiont. Although these organelles harbor their own genome, the large majority of genes, originally encoded in the endosymbiont, were either lost or transferred to the nucleus. As a consequence, mitochondria have become semi-autonomous and most of their processes require the import of nuclear-encoded components to be functional. Therefore, the mitochondrial-specific translation has evolved to be coordinated by mitonuclear interactions to respond to the energetic demands of the cell, acquiring unique and mosaic features. However, mitochondrial-DNA-encoded genes are essential for the assembly of the respiratory chain complexes. Impaired mitochondrial function due to oxidative damage and mutations has been associated with numerous human pathologies, the aging process, and cancer. In this review, we highlight the unique features of mitochondrial protein synthesis and provide a comprehensive insight into the mitonuclear crosstalk and its co-evolution, as well as the vulnerabilities of the animal mitochondrial genome.
Collapse
|
16
|
English K, Shepherd A, Uzor NE, Trinh R, Kavelaars A, Heijnen CJ. Astrocytes rescue neuronal health after cisplatin treatment through mitochondrial transfer. Acta Neuropathol Commun 2020; 8:36. [PMID: 32197663 PMCID: PMC7082981 DOI: 10.1186/s40478-020-00897-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 02/13/2020] [Indexed: 12/28/2022] Open
Abstract
Neurodegenerative disorders, including chemotherapy-induced cognitive impairment, are associated with neuronal mitochondrial dysfunction. Cisplatin, a commonly used chemotherapeutic, induces neuronal mitochondrial dysfunction in vivo and in vitro. Astrocytes are key players in supporting neuronal development, synaptogenesis, axonal growth, metabolism and, potentially mitochondrial health. We tested the hypothesis that astrocytes transfer healthy mitochondria to neurons after cisplatin treatment to restore neuronal health.We used an in vitro system in which astrocytes containing mito-mCherry-labeled mitochondria were co-cultured with primary cortical neurons damaged by cisplatin. Culture of primary cortical neurons with cisplatin reduced neuronal survival and depolarized neuronal mitochondrial membrane potential. Cisplatin induced abnormalities in neuronal calcium dynamics that were characterized by increased resting calcium levels, reduced calcium responses to stimulation with KCl, and slower calcium clearance. The same dose of cisplatin that caused neuronal damage did not affect astrocyte survival or astrocytic mitochondrial respiration. Co-culture of cisplatin-treated neurons with astrocytes increased neuronal survival, restored neuronal mitochondrial membrane potential, and normalized neuronal calcium dynamics especially in neurons that had received mitochondria from astrocytes which underlines the importance of mitochondrial transfer. These beneficial effects of astrocytes were associated with transfer of mitochondria from astrocytes to cisplatin-treated neurons. We show that siRNA-mediated knockdown of the Rho-GTPase Miro-1 in astrocytes reduced mitochondrial transfer from astrocytes to neurons and prevented the normalization of neuronal calcium dynamics.In conclusion, we showed that transfer of mitochondria from astrocytes to neurons rescues neurons from the damage induced by cisplatin treatment. Astrocytes are far more resistant to cisplatin than cortical neurons. We propose that transfer of functional mitochondria from astrocytes to neurons is an important repair mechanism to protect the vulnerable cortical neurons against the toxic effects of cisplatin.
Collapse
Affiliation(s)
- Krystal English
- Division of Internal Medicine, Department of Symptom Research, Laboratories of Neuroimmunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Andrew Shepherd
- Division of Internal Medicine, Department of Symptom Research, Laboratories of Neuroimmunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Ndidi-Ese Uzor
- Department of Neurobiology & Anatomy, The University of Texas McGovern Medical School, Houston, TX 77030 USA
- Department of Neurology, The University of Texas McGovern Medical School, Houston, TX 77030 USA
| | - Ronnie Trinh
- Division of Internal Medicine, Department of Symptom Research, Laboratories of Neuroimmunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Annemieke Kavelaars
- Division of Internal Medicine, Department of Symptom Research, Laboratories of Neuroimmunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Cobi J. Heijnen
- Division of Internal Medicine, Department of Symptom Research, Laboratories of Neuroimmunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| |
Collapse
|
17
|
Pulli I, Löf C, Blom T, Asghar M, Lassila T, Bäck N, Lin KL, Nyström J, Kemppainen K, Toivola D, Dufour E, Sanz A, Cooper H, Parys J, Törnquist K. Sphingosine kinase 1 overexpression induces MFN2 fragmentation and alters mitochondrial matrix Ca2+ handling in HeLa cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1475-1486. [DOI: 10.1016/j.bbamcr.2019.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 06/02/2019] [Accepted: 06/13/2019] [Indexed: 01/08/2023]
|
18
|
Lambert M, Capuano V, Boet A, Tesson L, Bertero T, Nakhleh MK, Remy S, Anegon I, Pechoux C, Hautefort A, Rucker-Martin C, Manoury B, Domergue V, Mercier O, Girerd B, Montani D, Perros F, Humbert M, Antigny F. Characterization of Kcnk3-Mutated Rat, a Novel Model of Pulmonary Hypertension. Circ Res 2019; 125:678-695. [PMID: 31347976 DOI: 10.1161/circresaha.119.314793] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
RATIONALE Pulmonary arterial hypertension is a severe lethal cardiopulmonary disease. Loss of function mutations in KCNK3 (potassium channel subfamily K member 3) gene, which encodes an outward rectifier K+ channel, have been identified in pulmonary arterial hypertension patients. OBJECTIVE We have demonstrated that KCNK3 dysfunction is common to heritable and nonheritable pulmonary arterial hypertension and to experimental pulmonary hypertension (PH). Finally, KCNK3 is not functional in mouse pulmonary vasculature. METHODS AND RESULTS Using CRISPR/Cas9 technology, we generated a 94 bp out of frame deletion in exon 1 of Kcnk3 gene and characterized these rats at the electrophysiological, echocardiographic, hemodynamic, morphological, cellular, and molecular levels to decipher the cellular mechanisms associated with loss of KCNK3. Using patch-clamp technique, we validated our transgenic strategy by demonstrating the absence of KCNK3 current in freshly isolated pulmonary arterial smooth muscle cells from Kcnk3-mutated rats. At 4 months of age, echocardiographic parameters revealed shortening of the pulmonary artery acceleration time associated with elevation of the right ventricular systolic pressure. Kcnk3-mutated rats developed more severe PH than wild-type rats after monocrotaline exposure or chronic hypoxia exposure. Kcnk3-mutation induced a lung distal neomuscularization and perivascular extracellular matrix activation. Lungs of Kcnk3-mutated rats were characterized by overactivation of ERK1/2 (extracellular signal-regulated kinase1-/2), AKT (protein kinase B), SRC, and overexpression of HIF1-α (hypoxia-inducible factor-1 α), survivin, and VWF (Von Willebrand factor). Linked with plasma membrane depolarization, reduced endothelial-NOS expression and desensitization of endothelial-derived hyperpolarizing factor, Kcnk3-mutated rats presented predisposition to vasoconstriction of pulmonary arteries and a severe loss of sildenafil-induced pulmonary arteries relaxation. Moreover, we showed strong alteration of right ventricular cardiomyocyte excitability. Finally, Kcnk3-mutated rats developed age-dependent PH associated with low serum-albumin concentration. CONCLUSIONS We established the first Kcnk3-mutated rat model of PH. Our results confirm that KCNK3 loss of function is a key event in pulmonary arterial hypertension pathogenesis. This model presents new opportunities for understanding the initiating mechanisms of PH and testing biologically relevant therapeutic molecules in the context of PH.
Collapse
Affiliation(s)
- Mélanie Lambert
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M. H., F.A.).,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.).,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.)
| | - Véronique Capuano
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M. H., F.A.).,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.).,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.)
| | - Angèle Boet
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M. H., F.A.).,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.).,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.)
| | - Laurent Tesson
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, France (L.T., S.R., I.A.).,PTransgenic Rat ImmunoPhenomic (TRIP) facility Nantes, Nantes, France (L.T., S.R., I.A.)
| | - Thomas Bertero
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France (T.B.)
| | - Morad K Nakhleh
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M. H., F.A.).,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.).,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.)
| | - Séverine Remy
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, France (L.T., S.R., I.A.).,PTransgenic Rat ImmunoPhenomic (TRIP) facility Nantes, Nantes, France (L.T., S.R., I.A.)
| | - Ignacio Anegon
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, France (L.T., S.R., I.A.).,PTransgenic Rat ImmunoPhenomic (TRIP) facility Nantes, Nantes, France (L.T., S.R., I.A.)
| | - Christine Pechoux
- GABI, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France (C.P.)
| | - Aurélie Hautefort
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M. H., F.A.).,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.).,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.)
| | - Catherine Rucker-Martin
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M. H., F.A.).,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.).,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.)
| | - Boris Manoury
- Signalisation et Physiopathologie Cardiovasculaire - UMR_S 1180, Univ. Paris-Sud, INSERM, Université Paris-Saclay, Châtenay-Malabry, France (B.M.)
| | - Valérie Domergue
- Animal Facility, Institut Paris Saclay d'Innovation Thérapeutique (UMS IPSIT), Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France (V.D.)
| | - Olaf Mercier
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M. H., F.A.).,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.).,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.)
| | - Barbara Girerd
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M. H., F.A.).,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.).,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.)
| | - David Montani
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M. H., F.A.).,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.).,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.)
| | - Frédéric Perros
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M. H., F.A.).,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.).,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.).,Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Canada (F.P.)
| | - Marc Humbert
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M. H., F.A.).,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.).,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.)
| | - Fabrice Antigny
- From the University Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M. H., F.A.).,Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.).,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France (M.L., V.C., A.B., M.K.N., A.H., C.R.-M., O.M., B.G., D.M., F.P., M.H., F.A.)
| |
Collapse
|
19
|
A novel mitochondrial micropeptide MPM enhances mitochondrial respiratory activity and promotes myogenic differentiation. Cell Death Dis 2019; 10:528. [PMID: 31296841 PMCID: PMC6624212 DOI: 10.1038/s41419-019-1767-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/25/2019] [Accepted: 06/26/2019] [Indexed: 12/14/2022]
Abstract
Micropeptides belong to a class of newly identified small molecules with <100 amino acids in length, and their functions remain largely unknown. Here, we identified a novel muscle-enriched micropeptide that was localized to mitochondria (named MPM, micropeptide in mitochondria) and upregulated during in vitro differentiation of C2C12 myoblasts and in vivo early postnatal skeletal muscle development, and muscle regeneration after cardiotoxin (CTX) damage. Downregulation of MPM was observed in the muscular tissues of tibial muscular dystrophy and Duchenne muscular dystrophy patients. Furthermore, MPM silencing inhibited the differentiation of C2C12 myoblasts into myotubes, whereas MPM overexpression stimulated it. MPM−/− mice exhibited smaller skeletal muscle fibers and worse muscle performance, such as decrease in the maximum grip force of limbs, the latency to fall off rotarod, and the exhausting swimming time. Muscle regeneration was also impaired in MPM−/− mice, as evidenced by lower expression of Pax7, MyoD, and MyoG after CTX injection and smaller regenerated myofibers, compared with wild-type mice. Mechanistical investigations based on both gain- and loss-of function studies revealed that MPM increased oxygen consumption and ATP production of mitochondria. Moreover, ectopic expression of PGC-1α, which can enhance mitochondrial respiration, attenuated the inhibitory effect of siMPM on myogenic differentiation. These results imply that MPM may promote myogenic differentiation and muscle fiber growth by enhancing mitochondrial respiratory activity, which highlights the importance of micropeptides in the elaborate regulatory network of both myogenesis and mitochondrial activity and implicates MPM as a potential target for muscular dystrophy therapy.
Collapse
|
20
|
Soledad RB, Charles S, Samarjit D. The secret messages between mitochondria and nucleus in muscle cell biology. Arch Biochem Biophys 2019; 666:52-62. [PMID: 30935885 PMCID: PMC6538274 DOI: 10.1016/j.abb.2019.03.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 03/01/2019] [Accepted: 03/28/2019] [Indexed: 02/06/2023]
Abstract
Over two thousand proteins are found in the mitochondrial compartment but the mitochondrial genome codes for only 13 proteins. The majority of mitochondrial proteins are products of nuclear genes and are synthesized in the cytosol, then translocated into the mitochondria. Most of the subunits of the five respiratory chain complexes in the inner mitochondrial membrane, which generate a proton gradient across the membrane and produce ATP, are encoded by nuclear genes. Therefore, it is quite clear that import of nuclear-encoded proteins into the mitochondria is essential for mitochondrial function. Nuclear to mitochondrial communication is well studied. However, there is another arm to this communication, mitochondria to nucleus retrograde signaling. This plays an important role in the maintenance of cellular homeostasis, and is less well studied. Several transcription factors, including Sp1, SIRT3 and GSP2, are activated by altered mitochondrial function. These activated transcription factors then translocate to the nucleus. Based on the mitochondrially generated molecular signal, nuclear genes are targeted, which alters transcription of nuclear genes that code for mitochondrial proteins. This review article will mainly focus on this interactive and bi-directional communication between mitochondria and nucleus, and how this communication plays a significant role in muscle cell biology.
Collapse
Affiliation(s)
| | - Steenbergen Charles
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Das Samarjit
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
21
|
Abstract
Together, the nuclear and mitochondrial genomes encode the oxidative phosphorylation (OXPHOS) complexes that reside in the mitochondrial inner membrane and enable aerobic life. Mitochondria maintain their own genome that is expressed and regulated by factors distinct from their nuclear counterparts. For optimal function, the cell must ensure proper stoichiometric production of OXPHOS subunits by coordinating two physically separated and evolutionarily distinct gene expression systems. Here, we review our current understanding of mitonuclear coregulation primarily at the levels of transcription and translation. Additionally, we discuss other levels of coregulation that may exist but remain largely unexplored, including mRNA modification and stability and posttranslational protein degradation.
Collapse
Affiliation(s)
- R Stefan Isaac
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA; , ,
| | - Erik McShane
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA; , ,
| | - L Stirling Churchman
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA; , ,
| |
Collapse
|
22
|
Duncan OF, Granat L, Ranganathan R, Singh VK, Mazaud D, Fanto M, Chambers D, Ballard CG, Bateman JM. Ras-ERK-ETS inhibition alleviates neuronal mitochondrial dysfunction by reprogramming mitochondrial retrograde signaling. PLoS Genet 2018; 14:e1007567. [PMID: 30059502 PMCID: PMC6085068 DOI: 10.1371/journal.pgen.1007567] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 08/09/2018] [Accepted: 07/16/2018] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial dysfunction activates the mitochondrial retrograde signaling pathway, resulting in large scale changes in gene expression. Mitochondrial retrograde signaling in neurons is poorly understood and whether retrograde signaling contributes to cellular dysfunction or is protective is unknown. We show that inhibition of Ras-ERK-ETS signaling partially reverses the retrograde transcriptional response to alleviate neuronal mitochondrial dysfunction. We have developed a novel genetic screen to identify genes that modify mitochondrial dysfunction in Drosophila. Knock-down of one of the genes identified in this screen, the Ras-ERK-ETS pathway transcription factor Aop, alleviates the damaging effects of mitochondrial dysfunction in the nervous system. Inhibition of Ras-ERK-ETS signaling also restores function in Drosophila models of human diseases associated with mitochondrial dysfunction. Importantly, Ras-ERK-ETS pathway inhibition partially reverses the mitochondrial retrograde transcriptional response. Therefore, mitochondrial retrograde signaling likely contributes to neuronal dysfunction through mis-regulation of gene expression. Loss of mitochondrial function activates the mitochondrial retrograde signaling pathway resulting in large scale changes in nuclear gene transcription. Very little is known about retrograde signaling in the nervous system and how the transcriptional changes affect neuronal function. Here we identify Ras-ERK-ETS signaling as a novel mitochondrial retrograde signaling pathway in the Drosophila nervous system. Inhibition of Ras-ERK-ETS signaling improves neuronal function in Drosophila models of mitochondrial disease. Targeting Ras-ERK-ETS signaling may therefore have therapeutic potential in mitochondrial disease patients. Using a transcriptomic approach, we find that inhibition of Ras-ERK-ETS signaling partially reverses the mitochondrial retrograde transcriptional response. Surprisingly therefore, the mitochondrial retrograde transcriptional response contributes to neuronal dysfunction.
Collapse
Affiliation(s)
- Olivia F. Duncan
- Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
| | - Lucy Granat
- Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
| | - Ramya Ranganathan
- Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
| | - Vandana K. Singh
- Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
| | - David Mazaud
- Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
| | - Manolis Fanto
- Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
| | - David Chambers
- Wolfson Centre for Age-Related Diseases, King’s College London, London, United Kingdom
| | - Clive G. Ballard
- Medical School Building, St Luke's Campus, University of Exeter, Exeter, United Kingdom
| | - Joseph M. Bateman
- Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
23
|
Bi S, Wang H, Kuang W. Stem cell rejuvenation and the role of autophagy in age retardation by caloric restriction: An update. Mech Ageing Dev 2018; 175:46-54. [PMID: 30031008 DOI: 10.1016/j.mad.2018.07.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 07/17/2018] [Accepted: 07/17/2018] [Indexed: 12/14/2022]
Abstract
Stem cells being pluripotent in nature can differentiate into a wide array of specific cells and asymmetrically divide to produce new ones but may undergo aging by themselves. Aging has both quantitative and qualitative effects on stem cells, and could eventually restrain them from replenishing into progenitor cells. Reactive oxygen species (ROS) accumulated in the aging cells could not only block the cell cycle but also affect autophagy by damaging the mitochondria. Autophagy could eliminate redundant production of ROS in aging stem cells and helps to maintain the proliferation capacity by restraining the expression of p16INK4a. Current studies showed that improving autophagy could restore the regenerative ability of aging stem cells. Therefore, it is important for an organism to maintain the appropriate autophagy. Caloric restriction (CR) was shown to retard the stem cell aging by a certain basic level of autophagy, suggesting that CR was an effective way to extend longevity in mammals. However, little is known about the underlying mechanisms. In this review, we tried to explore the molecular mechanisms on how CR induces appropriate autophagy to restore aging stem cell regenerative ability.
Collapse
Affiliation(s)
- Shanrong Bi
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hanyu Wang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weihong Kuang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
24
|
Park D, Chang S. Soluble Aβ 1-42 increases the heterogeneity in synaptic vesicle pool size among synapses by suppressing intersynaptic vesicle sharing. Mol Brain 2018; 11:10. [PMID: 29463281 PMCID: PMC5819658 DOI: 10.1186/s13041-018-0353-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 02/13/2018] [Indexed: 11/10/2022] Open
Abstract
Growing evidence has indicated that prefibrillar form of soluble amyloid beta (sAβ1-42) is the major causative factor in the synaptic dysfunction associated with AD. The molecular changes leading to presynaptic dysfunction caused by sAβ1-42, however, still remains elusive. Recently, we found that sAβ1-42 inhibits chemically induced long-term potentiation-induced synaptogenesis by suppressing the intersynaptic vesicle trafficking through calcium (Ca2+) dependent hyperphosphorylation of synapsin and CaMKIV. However, it is still unclear how sAβ1-42 increases intracellular Ca2+ that induces hyperphosphorylation of CaMKIV and synapsin, and what is the functional consequences of sAβ1-42-induced defects in intersynaptic vesicle trafficking in physiological conditions. In this study, we showed that sAβ1-42elevated intracellular Ca2+ through not only extracellular Ca2+ influx but also Ca2+ release from mitochondria. Surprisingly, without Ca2+ release from mitochondria, sAβ1-42 failed to increase intracellular Ca2+ even in the presence of normal extracellular Ca2+. We further found that sAβ1-42-induced mitochondria Ca2+ release alone sufficiently increased Serine 9 phosphorylation of synapsin. By blocking synaptic vesicle reallocation, sAβ1-42 significantly increased heterogeneity of total synaptic vesicle pool size among synapses. Together, our results suggested that by disrupting the axonal vesicle trafficking, sAβ1-42 disabled neurons to adjust synaptic pool sizes among synapses, which might prevent homeostatic rescaling in synaptic strength of individual neurons.
Collapse
Affiliation(s)
- Daehun Park
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Sunghoe Chang
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea. .,Neuroscience Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, 03080, South Korea.
| |
Collapse
|
25
|
Pomatto LCD, Davies KJA. The role of declining adaptive homeostasis in ageing. J Physiol 2017; 595:7275-7309. [PMID: 29028112 PMCID: PMC5730851 DOI: 10.1113/jp275072] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 09/01/2017] [Indexed: 12/12/2022] Open
Abstract
Adaptive homeostasis is "the transient expansion or contraction of the homeostatic range for any given physiological parameter in response to exposure to sub-toxic, non-damaging, signalling molecules or events, or the removal or cessation of such molecules or events" (Davies, 2016). Adaptive homeostasis enables biological systems to make continuous short-term adjustments for optimal functioning despite ever-changing internal and external environments. Initiation of adaptation in response to an appropriate signal allows organisms to successfully cope with much greater, normally toxic, stresses. These short-term responses are initiated following effective signals, including hypoxia, cold shock, heat shock, oxidative stress, exercise-induced adaptation, caloric restriction, osmotic stress, mechanical stress, immune response, and even emotional stress. There is now substantial literature detailing a decline in adaptive homeostasis that, unfortunately, appears to manifest with ageing, especially in the last third of the lifespan. In this review, we present the hypothesis that one hallmark of the ageing process is a significant decline in adaptive homeostasis capacity. We discuss the mechanistic importance of diminished capacity for short-term (reversible) adaptive responses (both biochemical and signal transduction/gene expression-based) to changing internal and external conditions, for short-term survival and for lifespan and healthspan. Studies of cultured mammalian cells, worms, flies, rodents, simians, apes, and even humans, all indicate declining adaptive homeostasis as a potential contributor to age-dependent senescence, increased risk of disease, and even mortality. Emerging work points to Nrf2-Keap1 signal transduction pathway inhibitors, including Bach1 and c-Myc, both of whose tissue concentrations increase with age, as possible major causes for age-dependent loss of adaptive homeostasis.
Collapse
Affiliation(s)
- Laura C. D. Pomatto
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology CenterUniversity of Southern CaliforniaLos AngelesCA 90089USA
| | - Kelvin J. A. Davies
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology CenterUniversity of Southern CaliforniaLos AngelesCA 90089USA
- Molecular and Computational Biology Program, Department of Biological Sciences of the Dornsife College of LettersArts & Sciences: the University of Southern CaliforniaLos AngelesCA 90089‐0191USA
| |
Collapse
|
26
|
Brinker AE, Vivian CJ, Koestler DC, Tsue TT, Jensen RA, Welch DR. Mitochondrial Haplotype Alters Mammary Cancer Tumorigenicity and Metastasis in an Oncogenic Driver-Dependent Manner. Cancer Res 2017; 77:6941-6949. [PMID: 29070615 DOI: 10.1158/0008-5472.can-17-2194] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/20/2017] [Accepted: 10/19/2017] [Indexed: 12/13/2022]
Abstract
Using a novel mouse model, a mitochondrial-nuclear exchange model termed MNX, we tested the hypothesis that inherited mitochondrial haplotypes alter primary tumor latency and metastatic efficiency. Male FVB/N-Tg(MMTVneu)202Mul/J (Her2) transgenic mice were bred to female MNX mice having FVB/NJ nuclear DNA with either FVB/NJ, C57BL/6J, or BALB/cJ mtDNA. Pups receiving the C57BL/6J or BALB/cJ mitochondrial genome (i.e., females crossed with Her2 males) showed significantly (P < 0.001) longer tumor latency (262 vs. 293 vs. 225 days), fewer pulmonary metastases (5 vs. 7 vs. 15), and differences in size of lung metastases (1.2 vs. 1.4 vs. 1.0 mm diameter) compared with FVB/NJ mtDNA. Although polyoma virus middle T-driven tumors showed altered primary and metastatic profiles in previous studies, depending upon nuclear and mtDNA haplotype, the magnitude and direction of changes were not the same in the HER2-driven mammary carcinomas. Collectively, these results establish mitochondrial polymorphisms as quantitative trait loci in mammary carcinogenesis, and they implicate distinct interactions between tumor drivers and mitochondria as critical modifiers of tumorigenicity and metastasis. Cancer Res; 77(24); 6941-9. ©2017 AACR.
Collapse
Affiliation(s)
- Amanda E Brinker
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, Kansas.,Department of Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, Kansas.,Heartland Center for Mitochondrial Medicine, The University of Kansas Medical Center, Kansas City, Kansas
| | - Carolyn J Vivian
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, Kansas.,Heartland Center for Mitochondrial Medicine, The University of Kansas Medical Center, Kansas City, Kansas
| | - Devin C Koestler
- Department of Biostatistics, The University of Kansas Medical Center, Kansas City, Kansas.,The University Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, Kansas
| | - Trevor T Tsue
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Roy A Jensen
- The University Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, Kansas.,Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, Kansas
| | - Danny R Welch
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, Kansas. .,Department of Molecular and Integrative Physiology, The University of Kansas Medical Center, Kansas City, Kansas.,Heartland Center for Mitochondrial Medicine, The University of Kansas Medical Center, Kansas City, Kansas.,The University Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
27
|
Zhang X, Yuan D, Sun Q, Xu L, Lee E, Lewis AJ, Zuckerbraun BS, Rosengart MR. Calcium/calmodulin-dependent protein kinase regulates the PINK1/Parkin and DJ-1 pathways of mitophagy during sepsis. FASEB J 2017; 31:4382-4395. [PMID: 28615325 DOI: 10.1096/fj.201601096rrr] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 06/06/2017] [Indexed: 11/11/2022]
Abstract
During sepsis and shock states, mitochondrial dysfunction occurs. Consequently, adaptive mechanisms, such as fission, fusion, and mitophagy, are induced to eliminate damaged portions or entire dysfunctional mitochondria. The regulatory PINK1/Parkin and DJ-1 pathways are strongly induced by mitochondrial depolarization, although a direct link between loss of mitochondrial membrane potential (ΔΨ) and mitophagy has not been identified. Mitochondria also buffer Ca2+, and their buffering capacity is dependent on ΔΨ Here, we characterize a role for calcium/calmodulin-dependent protein kinase (CaMK) I in the regulation of these mechanisms. Loss of ΔΨ with either pharmacologic depolarization or LPS leads to Ca2+-dependent mitochondrial recruitment and activation of CaMKI that precedes the colocalization of PINK1/Parkin and DJ-1. CaMKI is required and serves as both a PINK1 and Parkin kinase. The mechanisms operate in both immune and nonimmune cells and are induced in in vivo models of endotoxemia, sepsis, and hemorrhagic shock. These data support the idea that CaMKI links mitochondrial stress with the PINK1/Parkin and DJ-1 mechanisms of mitophagy.-Zhang, X., Yuan, D., Sun, Q., Xu, L., Lee, E., Lewis, A. J., Zuckerbraun, B. S., Rosengart, M. R. Calcium/calmodulin-dependent protein kinase regulates the PINK1/Parkin and DJ-1 pathways of mitophagy during sepsis.
Collapse
Affiliation(s)
- Xianghong Zhang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Du Yuan
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Qian Sun
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Li Xu
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Emma Lee
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anthony J Lewis
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Brian S Zuckerbraun
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Matthew R Rosengart
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA;
| |
Collapse
|
28
|
Upadhyay M, Agarwal S, Bhadauriya P, Ganesh S. Loss of laforin or malin results in increased Drp1 level and concomitant mitochondrial fragmentation in Lafora disease mouse models. Neurobiol Dis 2017; 100:39-51. [DOI: 10.1016/j.nbd.2017.01.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 12/24/2016] [Accepted: 01/03/2017] [Indexed: 02/02/2023] Open
|
29
|
Srinivasan S, Guha M, Kashina A, Avadhani NG. Mitochondrial dysfunction and mitochondrial dynamics-The cancer connection. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2017; 1858:602-614. [PMID: 28104365 DOI: 10.1016/j.bbabio.2017.01.004] [Citation(s) in RCA: 286] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 01/03/2017] [Accepted: 01/05/2017] [Indexed: 02/07/2023]
Abstract
Mitochondrial dysfunction is a hallmark of many diseases. The retrograde signaling initiated by dysfunctional mitochondria can bring about global changes in gene expression that alters cell morphology and function. Typically, this is attributed to disruption of important mitochondrial functions, such as ATP production, integration of metabolism, calcium homeostasis and regulation of apoptosis. Recent studies showed that in addition to these factors, mitochondrial dynamics might play an important role in stress signaling. Normal mitochondria are highly dynamic organelles whose size, shape and network are controlled by cell physiology. Defective mitochondrial dynamics play important roles in human diseases. Mitochondrial DNA defects and defective mitochondrial function have been reported in many cancers. Recent studies show that increased mitochondrial fission is a pro-tumorigenic phenotype. In this paper, we have explored the current understanding of the role of mitochondrial dynamics in pathologies. We present new data on mitochondrial dynamics and dysfunction to illustrate a causal link between mitochondrial DNA defects, excessive fission, mitochondrial retrograde signaling and cancer progression. This article is part of a Special Issue entitled Mitochondria in Cancer, edited by Giuseppe Gasparre, Rodrigue Rossignol and Pierre Sonveaux.
Collapse
Affiliation(s)
- Satish Srinivasan
- The Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, #189E, Philadelphia, PA 19104, United States
| | - Manti Guha
- The Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, #189E, Philadelphia, PA 19104, United States
| | - Anna Kashina
- The Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, #189E, Philadelphia, PA 19104, United States
| | - Narayan G Avadhani
- The Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, #189E, Philadelphia, PA 19104, United States.
| |
Collapse
|
30
|
Abstract
Hormesis is a process whereby exposure to a low dose of a potentially harmful stressor promotes adaptive changes to the cell that enables it to better tolerate subsequent stress. In recent years this concept has been applied specifically to the mitochondria (mitohormesis), suggesting that in response to a perturbation the mitochondria can initiate and transduce a signal to the nucleus that coordinates a transcriptional response resulting in both mitochondrial and non-mitochondrial adaptations that return and maintain cellular homeostasis. In this review we summarize the evidence that mitohormesis is a significant adaptive-response signaling pathway, and suggest that it plays a role in mediating exercise-induced adaptations. We discuss potential mitochondrial emitters of retrograde signals that may activate known exercise-sensitive transcription factors to modulate transcription responses to exercise, and draw on evidence from mitochondrial dysfunction animal models to support a role for mitohormesis in mitochondrial biogenesis. Studies directly linking mitohormesis to the exercise training response are lacking, however mounting evidence suggests numerous signals are emitted from the mitochondria during exercise and have the potential to induce a nuclear transcription response, with reactive oxygen species (ROS) being the primary candidate.
Collapse
Affiliation(s)
- Troy L Merry
- Energy Metabolism Laboratory, Swiss Federal Institute of Technology (ETH), 8603 Zurich, Switzerland.
| | - Michael Ristow
- Energy Metabolism Laboratory, Swiss Federal Institute of Technology (ETH), 8603 Zurich, Switzerland
| |
Collapse
|
31
|
Quirós PM, Mottis A, Auwerx J. Mitonuclear communication in homeostasis and stress. Nat Rev Mol Cell Biol 2016; 17:213-26. [PMID: 26956194 DOI: 10.1038/nrm.2016.23] [Citation(s) in RCA: 526] [Impact Index Per Article: 58.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mitochondria participate in crucial cellular processes such as energy harvesting and intermediate metabolism. Although mitochondria possess their own genome--a vestige of their bacterial origins and endosymbiotic evolution--most mitochondrial proteins are encoded in the nucleus. The expression of the mitochondrial proteome hence requires tight coordination between the two genomes to adapt mitochondrial function to the ever-changing cellular milieu. In this Review, we focus on the pathways that coordinate the communication between mitochondria and the nucleus during homeostasis and mitochondrial stress. These pathways include nucleus-to-mitochondria (anterograde) and mitochondria-to-nucleus (retrograde) communication, mitonuclear feedback signalling and proteostasis regulation, the integrated stress response and non-cell-autonomous communication. We discuss how mitonuclear communication safeguards cellular and organismal fitness and regulates lifespan.
Collapse
Affiliation(s)
- Pedro M Quirós
- Laboratory for Integrative and Systems Physiology, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Adrienne Mottis
- Laboratory for Integrative and Systems Physiology, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory for Integrative and Systems Physiology, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| |
Collapse
|
32
|
Foufelle F, Fromenty B. Role of endoplasmic reticulum stress in drug-induced toxicity. Pharmacol Res Perspect 2016; 4:e00211. [PMID: 26977301 PMCID: PMC4777263 DOI: 10.1002/prp2.211] [Citation(s) in RCA: 179] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 12/14/2015] [Indexed: 12/13/2022] Open
Abstract
Drug‐induced toxicity is a key issue for public health because some side effects can be severe and life‐threatening. These adverse effects can also be a major concern for the pharmaceutical companies since significant toxicity can lead to the interruption of clinical trials, or the withdrawal of the incriminated drugs from the market. Recent studies suggested that endoplasmic reticulum (ER) stress could be an important event involved in drug liability, in addition to other key mechanisms such as mitochondrial dysfunction and oxidative stress. Indeed, drug‐induced ER stress could lead to several deleterious effects within cells and tissues including accumulation of lipids, cell death, cytolysis, and inflammation. After recalling important information regarding drug‐induced adverse reactions and ER stress in diverse pathophysiological situations, this review summarizes the main data pertaining to drug‐induced ER stress and its potential involvement in different adverse effects. Drugs presented in this review are for instance acetaminophen (APAP), arsenic trioxide and other anticancer drugs, diclofenac, and different antiretroviral compounds. We also included data on tunicamycin (an antibiotic not used in human medicine because of its toxicity) and thapsigargin (a toxic compound of the Mediterranean plant Thapsia garganica) since both molecules are commonly used as prototypical toxins to induce ER stress in cellular and animal models.
Collapse
|
33
|
Mitochondrial Retrograde Signaling: Triggers, Pathways, and Outcomes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:482582. [PMID: 26583058 PMCID: PMC4637108 DOI: 10.1155/2015/482582] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/08/2015] [Accepted: 05/13/2015] [Indexed: 12/22/2022]
Abstract
Mitochondria are essential organelles for eukaryotic homeostasis. Although these organelles possess their own DNA, the vast majority (>99%) of mitochondrial proteins are encoded in the nucleus. This situation makes systems that allow the communication between mitochondria and the nucleus a requirement not only to coordinate mitochondrial protein synthesis during biogenesis but also to communicate eventual mitochondrial malfunctions, triggering compensatory responses in the nucleus. Mitochondria-to-nucleus retrograde signaling has been described in various organisms, albeit with differences in effector pathways, molecules, and outcomes, as discussed in this review.
Collapse
|
34
|
MacVicar TDB, Mannack LVJC, Lees RM, Lane JD. Targeted siRNA Screens Identify ER-to-Mitochondrial Calcium Exchange in Autophagy and Mitophagy Responses in RPE1 Cells. Int J Mol Sci 2015; 16:13356-80. [PMID: 26110381 PMCID: PMC4490499 DOI: 10.3390/ijms160613356] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 06/04/2015] [Accepted: 06/05/2015] [Indexed: 12/25/2022] Open
Abstract
Autophagy is an important stress response pathway responsible for the removal and recycling of damaged or redundant cytosolic constituents. Mitochondrial damage triggers selective mitochondrial autophagy (mitophagy), mediated by a variety of response factors including the Pink1/Parkin system. Using human retinal pigment epithelial cells stably expressing autophagy and mitophagy reporters, we have conducted parallel screens of regulators of endoplasmic reticulum (ER) and mitochondrial morphology and function contributing to starvation-induced autophagy and damage-induced mitophagy. These screens identified the ER chaperone and Ca2+ flux modulator, sigma non-opioid intracellular receptor 1 (SIGMAR1), as a regulator of autophagosome expansion during starvation. Screens also identified phosphatidyl ethanolamine methyl transferase (PEMT) and the IP3-receptors (IP3Rs) as mediators of Parkin-induced mitophagy. Further experiments suggested that IP3R-mediated transfer of Ca2+ from the ER lumen to the mitochondrial matrix via the mitochondrial Ca2+ uniporter (MCU) primes mitochondria for mitophagy. Importantly, recruitment of Parkin to damaged mitochondria did not require IP3R-mediated ER-to-mitochondrial Ca2+ transfer, but mitochondrial clustering downstream of Parkin recruitment was impaired, suggesting involvement of regulators of mitochondrial dynamics and/or transport. Our data suggest that Ca2+ flux between ER and mitochondria at presumed ER/mitochondrial contact sites is needed both for starvation-induced autophagy and for Parkin-mediated mitophagy, further highlighting the importance of inter-organellar communication for effective cellular homeostasis.
Collapse
Affiliation(s)
- Thomas D B MacVicar
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol BS81TD, UK.
| | - Lilith V J C Mannack
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol BS81TD, UK.
| | - Robert M Lees
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol BS81TD, UK.
| | - Jon D Lane
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol BS81TD, UK.
| |
Collapse
|
35
|
Identification of tetrahydrocarbazoles as novel multifactorial drug candidates for treatment of Alzheimer's disease. Transl Psychiatry 2014; 4:e489. [PMID: 25514752 PMCID: PMC4270312 DOI: 10.1038/tp.2014.132] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 10/12/2014] [Accepted: 11/17/2014] [Indexed: 01/08/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative brain disorder and the most frequent cause of dementia. To date, there are only a few approved drugs for AD, which show little or no effect on disease progression. Impaired intracellular calcium homeostasis is believed to occur early in the cascade of events leading to AD. Here, we examined the possibility of normalizing the disrupted calcium homeostasis in the endoplasmic reticulum (ER) store as an innovative approach for AD drug discovery. High-throughput screening of a small-molecule compound library led to the identification of tetrahydrocarbazoles, a novel multifactorial class of compounds that can normalize the impaired ER calcium homeostasis. We found that the tetrahydrocarbazole lead structure, first, dampens the enhanced calcium release from ER in HEK293 cells expressing familial Alzheimer's disease (FAD)-linked presenilin 1 mutations. Second, the lead structure also improves mitochondrial function, measured by increased mitochondrial membrane potential. Third, the same lead structure also attenuates the production of amyloid-beta (Aβ) peptides by decreasing the cleavage of amyloid precursor protein (APP) by β-secretase, without notably affecting α- and γ-secretase cleavage activities. Considering the beneficial effects of tetrahydrocarbazoles addressing three key pathological aspects of AD, these compounds hold promise for the development of potentially effective AD drug candidates.
Collapse
|
36
|
Wei Y, Zhang YJ, Cai Y, Xu MH. The role of mitochondria in mTOR-regulated longevity. Biol Rev Camb Philos Soc 2014; 90:167-81. [PMID: 24673778 DOI: 10.1111/brv.12103] [Citation(s) in RCA: 272] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Revised: 02/07/2014] [Accepted: 02/27/2014] [Indexed: 11/27/2022]
Abstract
Several unbiased genome-wide RNA interference (RNAi) screens have pointed to mitochondrial metabolism as the major factor for lifespan regulation. However, conflicting data remain to be clarified concerning the mitochondrial free radical theory of aging (MFRTA). Recently, mTOR (mechanistic target of rapamycin) has been proposed to be the central regulator of aging although how mTOR modulates lifespan is poorly understood. Interestingly, mTOR has been shown to regulate many aspects of mitochondrial function, such as mitochondrial biogenesis, apoptosis, mitophagy and mitochondrial hormesis (mitohormesis) including the retrograde response and mitochondrial unfolded protein response (mito-UPR). Here we discuss the data linking mitochondrial metabolism to mTOR regulation of lifespan, suggesting that hormetic effects may be key to explaining some controversial results regarding the MFRTA. We also discuss the possibility that dysfunction of mitochondrial adaptive responses rather than free radicals per se contributes to the aging process.
Collapse
Affiliation(s)
- Yuehua Wei
- No.3 People's Hospital, School of Medicine, Shanghai Jiao Tong University, 280 Mohe Road, Shanghai, 201900, China
| | | | | | | |
Collapse
|
37
|
Guha M, Avadhani NG. Mitochondrial retrograde signaling at the crossroads of tumor bioenergetics, genetics and epigenetics. Mitochondrion 2013; 13:577-91. [PMID: 24004957 DOI: 10.1016/j.mito.2013.08.007] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 08/20/2013] [Accepted: 08/27/2013] [Indexed: 12/25/2022]
Abstract
Mitochondria play a central role not only in energy production but also in the integration of metabolic pathways as well as signals for apoptosis and autophagy. It is becoming increasingly apparent that mitochondria in mammalian cells play critical roles in the initiation and propagation of various signaling cascades. In particular, mitochondrial metabolic and respiratory states and status on mitochondrial genetic instability are communicated to the nucleus as an adaptive response through retrograde signaling. Each mammalian cell contains multiple copies of the mitochondrial genome (mtDNA). A reduction in mtDNA copy number has been reported in various human pathological conditions such as diabetes, obesity, neurodegenerative disorders, aging and cancer. Reduction in mtDNA copy number disrupts mitochondrial membrane potential (Δψm) resulting in dysfunctional mitochondria. Dysfunctional mitochondria trigger retrograde signaling and communicate their changing metabolic and functional state to the nucleus as an adaptive response resulting in an altered nuclear gene expression profile and altered cell physiology and morphology. In this review, we provide an overview of the various modes of mitochondrial retrograde signaling focusing particularly on the Ca(2+)/Calcineurin mediated retrograde signaling. We discuss the contribution of the key factors of the pathway such as Calcineurin, IGF1 receptor, Akt kinase and HnRNPA2 in the propagation of signaling and their role in modulating genetic and epigenetic changes favoring cellular reprogramming towards tumorigenesis.
Collapse
Affiliation(s)
- Manti Guha
- Department of Animal Biology and the Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| | | |
Collapse
|
38
|
Hsu CC, Wang CH, Wu LC, Hsia CY, Chi CW, Yin PH, Chang CJ, Sung MT, Wei YH, Lu SH, Lee HC. Mitochondrial dysfunction represses HIF-1α protein synthesis through AMPK activation in human hepatoma HepG2 cells. Biochim Biophys Acta Gen Subj 2013; 1830:4743-51. [PMID: 23791554 DOI: 10.1016/j.bbagen.2013.06.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 05/09/2013] [Accepted: 06/04/2013] [Indexed: 01/11/2023]
Abstract
BACKGROUND Hypoxia-inducible factor-1α (HIF-1α) is an important transcription factor that modulates cellular responses to hypoxia and also plays critical roles in cancer progression. Recently, somatic mutations and decreased copy number of mitochondrial DNA (mtDNA) were detected in hepatocellular carcinoma (HCC). These mutations were shown to have the potential to cause mitochondrial dysfunction. However, the effects and mechanisms of mitochondrial dysfunction on HIF-1α function are not fully understood. This study aims to explore the underlying mechanism by which mitochondrial dysfunction regulates HIF-1α expression. METHODS Human hepatoma HepG2 cells were treated with various mitochondrial respiration inhibitors and an uncoupler, respectively, and the mRNA and protein expressions as well as transactivation activity of HIF-1α were determined. The role of AMP-activated protein kinase (AMPK) was further analyzed by compound C and AMPK knock-down. RESULTS Treatments of mitochondrial inhibitors and an uncoupler respectively reduced both the protein level and transactivation activity of HIF-1α in HepG2 cells under normoxia or hypoxia. The mitochondrial dysfunction-repressed HIF-1α protein synthesis was associated with decreased phosphorylations of p70(S6K) and 4E-BP-1. Moreover, mitochondrial dysfunction decreased intracellular ATP content and elevated the phosphorylation of AMPK. Treatments with compound C, an AMPK inhibitor, and knock-down of AMPK partially rescued the mitochondrial dysfunction-repressed HIF-1α expression. CONCLUSIONS Mitochondrial dysfunctions resulted in reduced HIF-1α protein synthesis through AMPK-dependent manner in HepG2 cells. GENERAL SIGNIFICANCE Our results provided a mechanism for communication from mitochondria to the nucleus through AMPK-HIF-1α. Mitochondrial function is important for HIF-1α expression in cancer progression.
Collapse
Affiliation(s)
- Chia-Chi Hsu
- Department and Institute of Pharmacology, National Yang-Ming University, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Monteith A, Marszalec W, Chan P, Logan J, Yu W, Schwarz N, Wokosin D, Hockberger P. Imaging of mitochondrial and non-mitochondrial responses in cultured rat hippocampal neurons exposed to micromolar concentrations of TMRM. PLoS One 2013; 8:e58059. [PMID: 23483968 PMCID: PMC3587568 DOI: 10.1371/journal.pone.0058059] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 01/31/2013] [Indexed: 11/24/2022] Open
Abstract
Tetramethylrhodamine methyl ester (TMRM) is a fluorescent dye used to study mitochondrial function in living cells. Previously, we reported that TMRM effectively labeled mitochondria of neurons deep within mouse brain slices. Use of micromolar concentration of dye, which was required to get sufficient staining for two-photon imaging, resulted in typical fluctuations of TMRM. With prolonged exposure, we recorded additional responses in some neurons that included slow oscillations and propagating waves of fluorescence. (Note: We use the terms “fluctuation” to refer to a change in the fluorescent state of an individual mitochondrion, “oscillation” to refer to a localized change in fluorescence in the cytosol, and “wave” to refer to a change in cytosolic fluorescence that propagated within a cell. Use of these terms does not imply any underlying periodicity.) In this report we describe similar results using cultured rat hippocampal neurons. Prolonged exposure of cultures to 2.5 µM TMRM produced a spontaneous increase in fluorescence in some neurons, but not glial cells, after 45–60 minutes that was followed by slow oscillations, waves, and eventually apoptosis. Spontaneous increases in fluorescence were insensitive to high concentrations of FCCP (100 µM) and thapsigargin (10 µM) indicating that they originated, at least in part, from regions outside of mitochondria. The oscillations did not correlate with changes in intracellular Ca2+, but did correlate with differences in fluorescence lifetime of the dye. Fluorescence lifetime and one-photon ratiometric imaging of TMRM suggested that the spontaneous increase and subsequent oscillations were due to movement of dye between quenched (hydrophobic) and unquenched (hydrophilic) compartments. We propose that these movements may be correlates of intracellular events involved in early stages of apoptosis.
Collapse
Affiliation(s)
- Andrew Monteith
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - William Marszalec
- Department of Molecular Pharmacology and Biological Chemistry, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Philip Chan
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Juliette Logan
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Weiming Yu
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Nicholas Schwarz
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - David Wokosin
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Philip Hockberger
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
40
|
Duguez S, Duddy WJ, Gnocchi V, Bowe J, Dadgar S, Partridge TA. Atmospheric oxygen tension slows myoblast proliferation via mitochondrial activation. PLoS One 2012; 7:e43853. [PMID: 22937109 PMCID: PMC3427224 DOI: 10.1371/journal.pone.0043853] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 07/30/2012] [Indexed: 11/23/2022] Open
Abstract
Background Mitochondrial activity inhibits proliferation and is required for differentiation of myoblasts. Myoblast proliferation is also inhibited by the ∼20% oxygen level used in standard tissue culture. We hypothesize that mitochondrial activity would be greater at hyperoxia (20% O2) relative to more physiological oxygen (5% O2). Methodology/Principal Findings Murine primary myoblasts from isolated myofibres and conditionally immortalized H-2K myoblasts were cultured at 5% and 20% oxygen. Proliferation, assayed by cell counts, EdU labeling, and CFSE dilution, was slower at 20% oxygen. Expression of MyoD in primary myoblasts was delayed at 20% oxygen, but myogenicity, as measured by fusion index, was slightly higher. FACS-based measurement of mitochondrial activity indicators and luminometric measurement of ATP levels revealed that mitochondria exhibited greater membrane potential and higher levels of Reactive Oxygen Species (ROS) at 20% oxygen with concomitant elevation of intracellular ATP. Mitochondrial mass was unaffected. Low concentrations of CCCP, a respiratory chain uncoupler, and Oligomycin A, an ATP synthase inhibitor, each increased the rate of myoblast proliferation. ROS were investigated as a potential mechanism of mitochondrial retrograde signaling, but scavenging of ROS levels by N-acetyl-cysteine (NAC) or α-Phenyl-N-tert-butylnitrone (PBN) did not rescue the suppressed rate of cell division in hyperoxic conditions, suggesting other pathways. Primary myoblasts from older mice showed a slower proliferation than those from younger adult mice at 20% oxygen but no difference at 5% oxygen. Conclusions/Significance These results implicate mitochondrial regulation as a mechanistic explanation for myoblast response to oxygen tension. The rescue of proliferation rate in myoblasts of aged mice by 5% oxygen suggests a major artefactual component to age-related decline of satellite cell proliferation in standard tissue culture at 20% oxygen. It lends weight to the idea that these age-related changes result at least in part from environmental factors rather than characteristics intrinsic to the satellite cell.
Collapse
Affiliation(s)
- Stephanie Duguez
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, District of Columbia, United States of America
- Université Pierre et Marie Curie (UPMC UMR S 974)-Institut National de la Santé et de la Recherche Médicale (Inserm U974)-Centre National de la Recherche Scientifique (CNRS UMR 7215), Institut de Myologie, Paris, France
| | - William J. Duddy
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, District of Columbia, United States of America
- Université Pierre et Marie Curie (UPMC UMR S 974)-Institut National de la Santé et de la Recherche Médicale (Inserm U974)-Centre National de la Recherche Scientifique (CNRS UMR 7215), Institut de Myologie, Paris, France
- * E-mail:
| | - Viola Gnocchi
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, District of Columbia, United States of America
| | - James Bowe
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, District of Columbia, United States of America
| | - Sherry Dadgar
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, District of Columbia, United States of America
| | - Terence A. Partridge
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, District of Columbia, United States of America
| |
Collapse
|
41
|
Roth JA, Sridhar S, Singleton ST. Effect of glutamate and riluzole on manganese-induced apoptotic cell signaling in neuronally differentiated mouse P19 Cells. Neurochem Int 2012; 61:25-33. [PMID: 22543103 DOI: 10.1016/j.neuint.2012.04.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Revised: 04/11/2012] [Accepted: 04/13/2012] [Indexed: 12/21/2022]
Abstract
Excess exposure to Mn causes a neurological disorder known as manganism which is similar to dystonic movements associated with Parkinson's disease. Manganism is largely restricted to occupations in which high atmospheric levels are prevalent which include Mn miners, welders and those employed in the ferroalloy processing or related industrial settings. T1 weighted MRI images reveal that Mn is deposited to the greatest extent in the globus pallidus, an area of the brain that is presumed to be responsible for the major CNS associated symptoms. Neurons within the globus pallidus receive glutamatergic input from the subthalamic nuclei which has been suggested to be involved in the toxic actions of Mn. The neurotoxic actions of Mn and glutamate are similar in that they both affect calcium accumulation in the mitochondria leading to apoptotic cell death. In this paper, we demonstrate that the combination of Mn and glutamate potentiates toxicity of neuronally differentiated P19 cells over that observed with either agent alone. Apoptotic signals ROS, caspase 3 and JNK were increased in an additive fashion when the two neurotoxins were combined. The anti-glutamatergic drug, riluzole, was shown to attenuate these apoptotic signals and prevent P19 cell death. Results of this study confirm, for the first time, that Mn toxicity is potentiated in the presence of glutamate and that riluzole is an effective antioxidant which protects against both Mn and glutamate toxicity.
Collapse
Affiliation(s)
- Jerome A Roth
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14214, USA.
| | | | | |
Collapse
|
42
|
Jones AWE, Yao Z, Vicencio JM, Karkucinska-Wieckowska A, Szabadkai G. PGC-1 family coactivators and cell fate: roles in cancer, neurodegeneration, cardiovascular disease and retrograde mitochondria-nucleus signalling. Mitochondrion 2011; 12:86-99. [PMID: 21983689 DOI: 10.1016/j.mito.2011.09.009] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Revised: 09/08/2011] [Accepted: 09/16/2011] [Indexed: 12/29/2022]
Abstract
Over the past two decades, a complex nuclear transcriptional machinery controlling mitochondrial biogenesis and function has been described. Central to this network are the PGC-1 family coactivators, characterised as master regulators of mitochondrial biogenesis. Recent literature has identified a broader role for PGC-1 coactivators in both cell death and cellular adaptation under conditions of stress, here reviewed in the context of the pathology associated with cancer, neurodegeneration and cardiovascular disease. Moreover, we propose that these studies also imply a novel conceptual framework on the general role of mitochondrial dysfunction in disease. It is now well established that the complex nuclear transcriptional control of mitochondrial biogenesis allows for adaptation of mitochondrial mass and function to environmental conditions. On the other hand, it has also been suggested that mitochondria alter their function according to prevailing cellular energetic requirements and thus function as sensors that generate signals to adjust fundamental cellular processes through a retrograde mitochondria-nucleus signalling pathway. Therefore, altered mitochondrial function can affect cell fate not only directly by modifying cellular energy levels or redox state, but also indirectly, by altering nuclear transcriptional patterns. The current literature on such retrograde signalling in both yeast and mammalian cells is thus reviewed, with an outlook on its potential contribution to disease through the regulation of PGC-1 family coactivators. We propose that further investigation of these pathways will lead to the identification of novel pharmacological targets and treatment strategies to combat disease.
Collapse
Affiliation(s)
- Aleck W E Jones
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, UK
| | | | | | | | | |
Collapse
|
43
|
Baloyannis SJ. Mitochondria are related to synaptic pathology in Alzheimer's disease. Int J Alzheimers Dis 2011; 2011:305395. [PMID: 21922047 PMCID: PMC3171689 DOI: 10.4061/2011/305395] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2011] [Accepted: 07/12/2011] [Indexed: 01/08/2023] Open
Abstract
Morphological alterations of mitochondria may play an important role in the pathogenesis of Alzheimer's disease, been associated with oxidative stress and Aβ-peptide-induced toxicity. We proceeded to estimation of mitochondria on electron micrographs of autopsy specimens of Alzheimer's disease. We found substantial morphological and morphometric changes of the mitochondria in the neurons of the hippocampus, the neocortex, the cerebellar cortex, the thalamus, the globus pallidus, the red nucleus, the locus coeruleus, and the climbing fibers. The alterations consisted of considerable changes of the cristae, accumulation of osmiophilic material, and modification of the shape and size. Mitochondrial alterations were prominent in neurons, which showed a depletion of dendritic spines and loss of dendritic branches. Mitochondrial alterations are not related with the accumulation of amyloid deposits, but are prominent whenever fragmentation of the Golgi apparatus exists. Morphometric analysis showed also that mitochondria are significantly reduced in neurons, which demonstrated synaptic pathology.
Collapse
Affiliation(s)
- Stavros J Baloyannis
- Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54006 Thessaloniki, Greece
| |
Collapse
|
44
|
Zampolla T, Spikings E, Rawson D, Zhang T. Cytoskeleton proteins F-actin and tubulin distribution and interaction with mitochondria in the granulosa cells surrounding stage III zebrafish (Danio rerio) oocytes. Theriogenology 2011; 76:1110-9. [PMID: 21752457 DOI: 10.1016/j.theriogenology.2011.05.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 04/20/2011] [Accepted: 05/19/2011] [Indexed: 01/30/2023]
Abstract
The distributional arrangement of mitochondria in the granulosa cells surrounding stage III zebrafish oocyte has been reported as a contiguous aggregation of mitochondria at the margin of the each granulosa cell. The aim of the present study was to further investigate the mitochondrial distribution in the granulosa cell layer in stage III ovarian follicles and the interaction between mitochondria and cytoskeleton elements actin and tubulin. To determine mitochondrial distribution/transport, immunocytochemistry analysis of tubulin and mitochondrial COX-I was carried out along with phalloidin staining of polymerised F-actin. The follicles were also exposed to a range of conditions that are known to affect mitochondria and the cytoskeleton proteins actin and tubulin. The mitochondrial inhibitor FCCP, the anti-mitotic drug nocodazole, and actin polymerisation inhibitor cytochalasin B were used. Levels of ATP, mtDNA copy number, and viability assessed by Trypan blue were also studied after exposure to inhibitors in order to determine the relationship between mitochondrial distribution/activity and ATP production. F-actin showed a hexagonal-polygonal distribution surrounding the mitochondria in granulosa cells, with the F-actin network adjacent to the plasma membrane of each granulosa cell. Tubulin structure presented a less organised distribution than F-actin, it was sparse in the cytosol. Interaction between mitochondria and tubulin was found indicating that mitochondria and tubulin are colocalised in zebrafish ovarian follicles. The exposure of ovarian follicles to inhibitors induced the loss of mitochondrial structural integrity showing that mitochondria distribution in granulosa cells of stage III zebrafish ovarian follicles is determined by the microtubules network.
Collapse
Affiliation(s)
- T Zampolla
- LIRANS, Institute of Research in the Applied Natural Sciences, University of Bedfordshire, Luton, Bedfordshire, UK
| | | | | | | |
Collapse
|
45
|
Rothenberg KG, Siedlak SL, Lee HG, Zhu X, Perry G, Smith MA. Neurodegenerative processes in Alzheimer’s disease: an overview of pathogenesis with strategic biomarker potential. FUTURE NEUROLOGY 2011. [DOI: 10.2217/fnl.10.81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Since Alzheimer’s disease (AD) is the leading cause of senile dementia in the USA, affecting 15% of people over the age of 65 years and almost 50% of those aged over 85 years, the need for an adequate and early diagnosis as well as preventative measure against disease onset and progression is increasing. Epidemiological and molecular studies suggest that AD has multiple etiologies, including genetic mutations, genetic variations affecting susceptibility and environmental factors. All these aspects can promote the formation and the accumulation of insoluble amyloid-β and hyperphosphorylated tau. Since the disease is multifactorial and clinical diagnosis is highly exclusive, the need for a sensitive, specific and reliable biomarker for the disease is crucial. While amyloid and amyloid-related compounds may be useful biomarkers in the early diagnosis of AD, the multitude of other characteristic features of AD presented in this article may be similarly appropriate. For example, genetic mutations play a role in a subset of AD patients (often with early disease onset and more severe disease progression), and genetic analysis could thus play a role in disease diagnosis. Similarly, oxidative damage to various proteins, nucleic acids and other cellular compounds, probably arising from mitochondrial abnormalities, is found early in the disease and may provide certain biochemical signatures of disease. Ultimately, specific assays for genetic, protein and oxidative profiles and mitochondrial abnormalities, as well as those for amyloid-β and its immunological response, may serve as a relevant group of biomarkers that could be informative to individuals regarding risk of disease, as well as for indicators of the progression of disease. Correspondingly, new developments in treatment options will probably be available.
Collapse
Affiliation(s)
- Kasia Gustaw Rothenberg
- Department of Psychiatry, University Hospitals Case Medical Center, Cleveland, OH, USA
- Deptartment of Neurodegenerative Diseases, Institute of Agricultural Medicine, 2 Jaczewskiego Street, 20-095, Lublin, Poland
| | - Sandra L Siedlak
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
| | - Hyoung-gon Lee
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
| | - George Perry
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
- Neurosciences Institute & Department of Biology, College of Sciences, University of Texas at San Antonio, San Antonio, TX, USA
| | - Mark A Smith
- Department of Pathology, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
| |
Collapse
|
46
|
Joodi G, Ansari N, Khodagholi F. Chitooligosaccharide-mediated neuroprotection is associated with modulation of Hsps expression and reduction of MAPK phosphorylation. Int J Biol Macromol 2011; 48:726-35. [PMID: 21356235 DOI: 10.1016/j.ijbiomac.2011.02.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2010] [Revised: 02/14/2011] [Accepted: 02/21/2011] [Indexed: 12/13/2022]
Abstract
There is mounting evidence implicating the role of oxidative stress induced by reactive oxygen species (ROS) in neurodegenerative disease, including Alzheimer's disease. In this study we aimed to investigate the possible protective effect of chitooligosaccharide (COS), an antioxidant oligosaccharide, on hydrogen peroxide induced apoptosis in NGF-differentitated rat pheochromocytoma (PC12) cells. COS treatment reversed the decrease of cell viability induced by H(2)O(2) and this was associated with diminished intracellular ROS and decreased level of cytosolic Ca(2+). Additionally, COS contributed to up-regulation of Bcl-2, down regulation of Bax protein and reduction of cleaved Caspase-3 protein. COS treatment stabilized Nrf2 in nucleus and increased the Hsp70 level within cell while down-regulated Hsp90 expression. Moreover, COS could inhibit the phosphorylation of different mitogen activated protein kinases (MAPKs), whose aberrant phosphorylation has been implicated in AD. Our findings suggest that heat shock response and MAPK cascades are both involved in cell survival, and by concomitantly regulating both pathways, COS can be a promising agent in treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Golsa Joodi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
47
|
Holmes-Hampton GP, Miao R, Garber-Morales J, Guo Y, Münck E, Lindahl PA. A nonheme high-spin ferrous pool in mitochondria isolated from fermenting Saccharomyces cerevisiae. Biochemistry 2010; 49:4227-34. [PMID: 20408527 PMCID: PMC2868115 DOI: 10.1021/bi1001823] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mössbauer spectroscopy was used to detect pools of Fe in mitochondria from fermenting yeast cells, including those consisting of nonheme high-spin (HS) Fe(II) species, Fe(III) nanoparticles, and mononuclear HS Fe(III) species. At issue was whether these species were located within mitochondria or on their exterior. None could be removed by washing mitochondria extensively with ethylene glycol tetraacetic acid or bathophenanthroline sulfonate (BPS), Fe(II) chelators that do not appear to penetrate mitochondrial membranes. However, when mitochondrial samples were sonicated, BPS coordinated the Fe(II) species, forming a low-spin Fe(II) complex. This treatment also diminished the levels of both Fe(III) species, suggesting that all of these Fe species are encapsulated by mitochondrial membranes and are protected from chelation until membranes are disrupted. 1,10-Phenanthroline is chemically similar to BPS but is membrane soluble; it coordinated nonheme HS Fe(II) in unsonicated mitochondria. Further, the HS Fe(III) species and nanoparticles were not reduced by dithionite until the detergent deoxycholate was added to disrupt membranes. There was no correlation between the percentage of nonheme HS Fe(II) species in mitochondrial samples and the level of contaminating proteins. These results collectively indicate that the observed Fe species are contained within mitochondria. Mossbauer spectra of whole cells were dominated by HS Fe(III) features; the remainder displayed spectral features typical of isolated mitochondria, suggesting that the Fe in fermenting yeast cells can be coarsely divided into two categories: mitochondrial Fe and (mostly) HS Fe(III) ions in one or more non-mitochondrial locations.
Collapse
Affiliation(s)
| | - Ren Miao
- Department of Chemistry, Texas A&M University, College Station, TX 77843-3255
| | | | - Yisong Guo
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Eckard Münck
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Paul A. Lindahl
- Department of Chemistry, Texas A&M University, College Station, TX 77843-3255
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843
| |
Collapse
|
48
|
Gustaw-Rothenberg K, Lerner A, Bonda DJ, Lee HG, Zhu X, Perry G, Smith MA. Biomarkers in Alzheimer's disease: past, present and future. Biomark Med 2010; 4:15-26. [PMID: 20387301 DOI: 10.2217/bmm.09.86] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Epidemiological and molecular studies suggest that Alzheimer's disease (AD) has multiple etiologies including genetic mutations, genetic variations affecting susceptibility and environmental factors. These aspects can promote the formation and accumulation of insoluble amyloid-beta and hyperphosphorylated tau. Since the disease is multifactorial and clinical diagnosis is highly exclusive, the need for a sensitive, specific and reliable biomarker is crucial. The concept of a biomarker implies sensitivity and specificity relative to the condition being considered. For clinical practice, AD diagnosis has been based on adherence to clinical criteria such as the NINCDS/ADRDA and DSM-IV. A more recent set of diagnostic criteria proposed incorporates imaging findings into the diagnosis of AD. In this article, we consider the most studied candidates or group of candidates for AD biomarkers, including pathological processes and proteins (amyloid-beta, tau, oxidative stress, mitochondrial/metabolic changes and cell-cycle processes), or autoantibodies thereto, as well as genetic factors.
Collapse
Affiliation(s)
- Katarzyna Gustaw-Rothenberg
- University Hospitals, Case Medical Center and University Memory and Cognitive Center, Case Western Reserve University, Cleveland, OH, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Liu HT, He JL, Li WM, Yang Z, Wang YX, Bai XF, Yu C, Du YG. Chitosan oligosaccharides protect human umbilical vein endothelial cells from hydrogen peroxide-induced apoptosis. Carbohydr Polym 2010. [DOI: 10.1016/j.carbpol.2010.01.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
50
|
Kennedy A, Martinez K, Chung S, LaPoint K, Hopkins R, Schmidt SF, Andersen K, Mandrup S, McIntosh M. Inflammation and insulin resistance induced by trans-10, cis-12 conjugated linoleic acid depend on intracellular calcium levels in primary cultures of human adipocytes. J Lipid Res 2010; 51:1906-17. [PMID: 20154361 DOI: 10.1194/jlr.m005447] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
We previously demonstrated that trans-10, cis-12 (10,12) conjugated linoleic acid (CLA) induced inflammation and insulin resistance in primary human adipocytes by activating nuclear factor kappaB (NFkappaB) and extracellular signal-related kinase (ERK) signaling. In this study, we demonstrated that the initial increase in intracellular calcium ([Ca2+]i) mediated by 10,12 CLA was attenuated by TMB-8, an inhibitor of calcium release from the endoplasmic reticulum (ER), by BAPTA, an intracellular calcium chelator, and by D609, a phospholipase C (PLC) inhibitor. Moreover, BAPTA, TMB-8, and D609 attenuated 10,12 CLA-mediated production of reactive oxygen species (ROS), activation of ERK1/2 and cJun-NH2-terminal kinase (JNK), and induction of inflammatory genes. 10,12 CLA-mediated binding of NFkappaB to the promoters of interleukin (IL)-8 and cyclooxygenase (COX)-2 and induction of calcium-calmodulin kinase II (CaMKII) beta were attenuated by TMB-8. KN-62, a CaMKII inhibitor, also suppressed 10,12 CLA-mediated ROS production and ERK1/2 and JNK activation. Additionally, KN-62 attenuated 10,12 CLA induction of inflammatory and integrated stress response genes, increase in prostaglandin F2alpha, and suppression of peroxisome proliferator activated receptor gamma protein levels and insulin-stimulated glucose uptake. These data suggest that 10,12 CLA increases inflammation and insulin resistance in human adipocytes, in part by increasing [Ca2+]i levels, particularly calcium from the ER.
Collapse
Affiliation(s)
- Arion Kennedy
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC, USA
| | | | | | | | | | | | | | | | | |
Collapse
|