1
|
Alibhai FJ, Li RK. Rejuvenation of the Aging Heart: Molecular Determinants and Applications. Can J Cardiol 2024; 40:1394-1411. [PMID: 38460612 DOI: 10.1016/j.cjca.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/20/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024] Open
Abstract
In Canada and worldwide, the elderly population (ie, individuals > 65 years of age) is increasing disproportionately relative to the total population. This is expected to have a substantial impact on the health care system, as increased aged is associated with a greater incidence of chronic noncommunicable diseases. Within the elderly population, cardiovascular disease is a leading cause of death, therefore developing therapies that can prevent or slow disease progression in this group is highly desirable. Historically, aging research has focused on the development of anti-aging therapies that are implemented early in life and slow the age-dependent decline in cell and organ function. However, accumulating evidence supports that late-in-life therapies can also benefit the aged cardiovascular system by limiting age-dependent functional decline. Moreover, recent studies have demonstrated that rejuvenation (ie, reverting cellular function to that of a younger phenotype) of the already aged cardiovascular system is possible, opening new avenues to develop therapies for older individuals. In this review, we first provide an overview of the functional changes that occur in the cardiomyocyte with aging and how this contributes to the age-dependent decline in heart function. We then discuss the various anti-aging and rejuvenation strategies that have been pursued to improve the function of the aged cardiomyocyte, with a focus on therapies implemented late in life. These strategies include 1) established systemic approaches (caloric restriction, exercise), 2) pharmacologic approaches (mTOR, AMPK, SIRT1, and autophagy-targeting molecules), and 3) emerging rejuvenation approaches (partial reprogramming, parabiosis/modulation of circulating factors, targeting endogenous stem cell populations, and senotherapeutics). Collectively, these studies demonstrate the exciting potential and limitations of current rejuvenation strategies and highlight future areas of investigation that will contribute to the development of rejuvenation therapies for the aged heart.
Collapse
Affiliation(s)
- Faisal J Alibhai
- Toronto General Research Hospital Institute, University Health Network, Toronto, Ontario, Canada
| | - Ren-Ke Li
- Toronto General Research Hospital Institute, University Health Network, Toronto, Ontario, Canada; Department of Surgery, Division of Cardiovascular Surgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Zheng LY, Da YX, Luo X, Zhang X, Sun ZJ, Dong DL. Sorafenib extends the lifespan of C. elegans through mitochondrial uncoupling mechanism. Free Radic Biol Med 2024; 214:101-113. [PMID: 38360276 DOI: 10.1016/j.freeradbiomed.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024]
Abstract
Sorafenib is a targeted anticancer drug in clinic. Low-dose sorafenib has been reported to activate AMPK through inducing mitochondrial uncoupling without detectable toxicities. AMPK activation has been the approach for extending lifespan, therefore, we investigated the effect of sorafenib on lifespan and physical activity of C. elegans and the underlying mechanisms. In the present study, we found that the effect of sorafenib on C. elegans lifespan was typically hermetic. Sorafenib treatment at higher concentrations (100 μM) was toxic but at lower concentrations (1, 2.5, 5 μM) was beneficial to C. elegans. Sorafenib (1 μM) treatment for whole-life period extended C. elegans lifespan and improved C. elegans physical activity as manifested by increasing pharyngeal pumping and body movement, preserving intestinal barrier integrity, muscle fibers organization and mitochondrial morphology. In addition, sorafenib (1 μM) treatment enhanced C. elegans stress resistance. Sorafenib activated AMPK through inducing mitochondrial uncoupling in C. elegans. Sorafenib treatment activated DAF-16, SKN-1, and increased SOD-3, HSP-16.2, GST-4 expression in C. elegans. Sorafenib treatment induced AMPK-dependent autophagy in C. elegans. We conclude that low-dose sorafenib protects C. elegans against aging through activating AMPK/DAF-16 dependent anti-oxidant pathways and stimulating autophagy responses. Low-dose sorafenib could be a strategy for treating aging and aging-related diseases.
Collapse
Affiliation(s)
- Lu-Yao Zheng
- Department of Pharmacology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yan-Xin Da
- Department of Pharmacology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Xiu Luo
- Department of Pharmacology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Xiao Zhang
- Department of Pharmacology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Zhi-Jie Sun
- Department of Pharmacology, China Pharmaceutical University, Nanjing, People's Republic of China.
| | - De-Li Dong
- Department of Pharmacology, China Pharmaceutical University, Nanjing, People's Republic of China.
| |
Collapse
|
3
|
Nir Sade A, Levy G, Schokoroy Trangle S, Elad Sfadia G, Bar E, Ophir O, Fischer I, Rokach M, Atzmon A, Parnas H, Rosenberg T, Marco A, Elroy Stein O, Barak B. Neuronal Gtf2i deletion alters mitochondrial and autophagic properties. Commun Biol 2023; 6:1269. [PMID: 38097729 PMCID: PMC10721858 DOI: 10.1038/s42003-023-05612-5] [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: 01/10/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023] Open
Abstract
Gtf2i encodes the general transcription factor II-I (TFII-I), with peak expression during pre-natal and early post-natal brain development stages. Because these stages are critical for proper brain development, we studied at the single-cell level the consequences of Gtf2i's deletion from excitatory neurons, specifically on mitochondria. Here we show that Gtf2i's deletion resulted in abnormal morphology, disrupted mRNA related to mitochondrial fission and fusion, and altered autophagy/mitophagy protein expression. These changes align with elevated reactive oxygen species levels, illuminating Gtf2i's importance in neurons mitochondrial function. Similar mitochondrial issues were demonstrated by Gtf2i heterozygous model, mirroring the human condition in Williams syndrome (WS), and by hemizygous neuronal Gtf2i deletion model, indicating Gtf2i's dosage-sensitive role in mitochondrial regulation. Clinically relevant, we observed altered transcript levels related to mitochondria, hypoxia, and autophagy in frontal cortex tissue from WS individuals. Our study reveals mitochondrial and autophagy-related deficits shedding light on WS and other Gtf2i-related disorders.
Collapse
Affiliation(s)
- Ariel Nir Sade
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Gilad Levy
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Sari Schokoroy Trangle
- The School of Psychological Sciences, Faculty of Social Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Galit Elad Sfadia
- The School of Psychological Sciences, Faculty of Social Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ela Bar
- The School of Psychological Sciences, Faculty of Social Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Omer Ophir
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Inbar Fischer
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - May Rokach
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Andrea Atzmon
- The Shmunis School of Biomedicine & Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Hadar Parnas
- Neuro-Epigenetics Laboratory, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Tali Rosenberg
- Neuro-Epigenetics Laboratory, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Asaf Marco
- Neuro-Epigenetics Laboratory, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Orna Elroy Stein
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- The Shmunis School of Biomedicine & Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Boaz Barak
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
- The School of Psychological Sciences, Faculty of Social Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
4
|
Li A, Shami GJ, Griffiths L, Lal S, Irving H, Braet F. Giant mitochondria in cardiomyocytes: cellular architecture in health and disease. Basic Res Cardiol 2023; 118:39. [PMID: 37775647 PMCID: PMC10541842 DOI: 10.1007/s00395-023-01011-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/29/2023] [Accepted: 09/15/2023] [Indexed: 10/01/2023]
Abstract
Giant mitochondria are frequently observed in different disease models within the brain, kidney, and liver. In cardiac muscle, these enlarged organelles are present across diverse physiological and pathophysiological conditions including in ageing and exercise, and clinically in alcohol-induced heart disease and various cardiomyopathies. This mitochondrial aberration is widely considered an early structural hallmark of disease leading to adverse organ function. In this thematic paper, we discuss the current state-of-knowledge on the presence, structure and functional implications of giant mitochondria in heart muscle. Despite its demonstrated reoccurrence in different heart diseases, the literature on this pathophysiological phenomenon remains relatively sparse since its initial observations in the early 60s. We review historical and contemporary investigations from cultured cardiomyocytes to human tissue samples to address the role of giant mitochondria in cardiac health and disease. Finally, we discuss their significance for the future development of novel mitochondria-targeted therapies to improve cardiac metabolism and functionality.
Collapse
Affiliation(s)
- Amy Li
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, Bendigo, VIC, Australia.
- Centre for Healthy Futures, Torrens University Australia, Surry Hills, NSW, Australia.
- School of Medical Sciences, The University of Sydney, Camperdown, NSW, Australia.
| | - Gerald J Shami
- School of Medical Sciences (Molecular and Cellular Biomedicine), The University of Sydney, Camperdown, NSW, Australia
- Australian Centre for Microscopy and Microanalysis, The University of Sydney, Camperdown, NSW, Australia
| | - Lisa Griffiths
- Anatomical Pathology, PathWest, QEII Medical Centre, Nedlands, WA, Australia
| | - Sean Lal
- School of Medical Sciences, The University of Sydney, Camperdown, NSW, Australia
| | - Helen Irving
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, Bendigo, VIC, Australia
| | - Filip Braet
- School of Medical Sciences (Molecular and Cellular Biomedicine), The University of Sydney, Camperdown, NSW, Australia.
- Australian Centre for Microscopy and Microanalysis, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
5
|
Shang Y, Li Z, Cai P, Li W, Xu Y, Zhao Y, Xia S, Shao Q, Wang H. Megamitochondria plasticity: function transition from adaption to disease. Mitochondrion 2023:S1567-7249(23)00053-3. [PMID: 37276954 DOI: 10.1016/j.mito.2023.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/08/2023] [Accepted: 06/02/2023] [Indexed: 06/07/2023]
Abstract
As the cell's energy factory and metabolic hub, mitochondria are critical for ATP synthesis to maintain cellular function. Mitochondria are highly dynamic organelles that continuously undergo fusion and fission to alter their size, shape, and position, with mitochondrial fusion and fission being interdependent to maintain the balance of mitochondrial morphological changes. However, in response to metabolic and functional damage, mitochondria can grow in size, resulting in a form of abnormal mitochondrial morphology known as megamitochondria. Megamitochondria are characterized by their considerably larger size, pale matrix, and marginal cristae structure and have been observed in various human diseases. In energy-intensive cells like hepatocytes or cardiomyocytes, the pathological process can lead to the growth of megamitochondria, which can further cause metabolic disorders, cell damage and aggravates the progression of the disease. Nonetheless, megamitochondria can also form in response to short-term environmental stimulation as a compensatory mechanism to support cell survival. However, extended stimulation can reverse the benefits of megamitochondria leading to adverse effects. In this review, we will focus on the findings of the different roles of megamitochondria, and their link to disease development to identify promising clinical therapeutic targets.
Collapse
Affiliation(s)
- Yuxing Shang
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Zhanghui Li
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Peiyang Cai
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Wuhao Li
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Ye Xu
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Yangjing Zhao
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Sheng Xia
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Qixiang Shao
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China; Institute of Medical Genetics and Reproductive Immunity, School of Medical Science and Laboratory Medicine, Jiangsu College of Nursing, Huai'an 223002, Jiangsu, PR China.
| | - Hui Wang
- Reproductive Sciences Institute, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China.
| |
Collapse
|
6
|
Voglhuber J, Holzer M, Radulović S, Thai PN, Djalinac N, Matzer I, Wallner M, Bugger H, Zirlik A, Leitinger G, Dedkova EN, Bers DM, Ljubojevic-Holzer S. Functional remodelling of perinuclear mitochondria alters nucleoplasmic Ca 2+ signalling in heart failure. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210320. [PMID: 36189813 PMCID: PMC9527904 DOI: 10.1098/rstb.2021.0320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/23/2022] [Indexed: 11/12/2022] Open
Abstract
Mitochondrial dysfunction in cardiomyocytes is a hallmark of heart failure development. Although initial studies recognized the importance of different mitochondrial subpopulations, there is a striking lack of direct comparison of intrafibrillar (IF) versus perinuclear (PN) mitochondria during the development of HF. Here, we use multiple approaches to examine the morphology and functional properties of IF versus PN mitochondria in pressure overload-induced cardiac remodelling in mice, and in non-failing and failing human cardiomyocytes. We demonstrate that PN mitochondria from failing cardiomyocytes are more susceptible to depolarization of mitochondrial membrane potential, reactive oxygen species generation and impairment in Ca2+ uptake compared with IF mitochondria at baseline and under physiological stress protocol. We also demonstrate, for the first time to our knowledge, that under normal conditions PN mitochondrial Ca2+ uptake shapes nucleoplasmic Ca2+ transients (CaTs) and limits nucleoplasmic Ca2+ loading. The loss of PN mitochondrial Ca2+ buffering capacity translates into increased nucleoplasmic CaTs and may explain disproportionate rise in nucleoplasmic [Ca2+] in failing cardiomyocytes at increased stimulation frequencies. Therefore, a previously unidentified benefit of restoring the mitochondrial Ca2+ uptake may be normalization of nuclear Ca2+ signalling and alleviation of altered excitation-transcription, which could be an important therapeutic approach to prevent adverse cardiac remodelling. This article is part of the theme issue 'The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease'.
Collapse
Affiliation(s)
- Julia Voglhuber
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Michael Holzer
- BioTechMed-Graz, Graz, Austria
- Division of Pharmacology, Otto-Loewi Research Centre, Medical University of Graz, Graz, Austria
| | - Snježana Radulović
- Research Unit Electron Microscopic Techniques, Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Phung N. Thai
- Department of Internal Medicine, Cardiovascular Medicine, University of California Davis, Davis, CA, USA
| | - Natasa Djalinac
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Ingrid Matzer
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Markus Wallner
- Department of Cardiology, Medical University of Graz, Graz, Austria
- Lewis Katz School of Medicine, Temple University, Cardiovascular Research Center, Philadelphia, PA, USA
| | - Heiko Bugger
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Andreas Zirlik
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Gerd Leitinger
- Research Unit Electron Microscopic Techniques, Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Elena N. Dedkova
- Department of Pharmacology, University of California Davis, Davis, CA, USA
- Department of Molecular Biosciences, University of California Davis, Davis, CA, USA
| | - Donald M. Bers
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Senka Ljubojevic-Holzer
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| |
Collapse
|
7
|
Messerer J, Wrede C, Schipke J, Brandenberger C, Abdellatif M, Eisenberg T, Madeo F, Sedej S, Mühlfeld C. Spermidine supplementation influences mitochondrial number and morphology in the heart of aged mice. J Anat 2021; 242:91-101. [PMID: 34958481 PMCID: PMC9773166 DOI: 10.1111/joa.13618] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/10/2021] [Accepted: 12/15/2021] [Indexed: 01/18/2023] Open
Abstract
Aging is associated with cardiac hypertrophy and progressive decline in heart function. One of the hallmarks of cellular aging is the dysfunction of mitochondria. These organelles occupy around 1/4 to 1/3 of the cardiomyocyte volume. During cardiac aging, the removal of defective or dysfunctional mitochondria by mitophagy as well as the dynamic equilibrium between mitochondrial fusion and fission is distorted. Here, we hypothesized that these changes affect the number of mitochondria and alter their three-dimensional (3D) characteristics in aged mouse hearts. The polyamine spermidine stimulates both mitophagy and mitochondrial biogenesis, and these are associated with improved cardiac function and prolonged lifespan. Therefore, we speculated that oral spermidine administration normalizes the number of mitochondria and their 3D morphology in aged myocardium. Young (4-months old) and old (24-months old) mice, treated or not treated with spermidine, were used in this study (n = 10 each). The number of mitochondria in the left ventricles was estimated by design-based stereology using the Euler-Poincaré characteristic based on a disector at the transmission electron microscopic level. The 3D morphology of mitochondria was investigated by 3D reconstruction (using manual contour drawing) from electron microscopic z-stacks obtained by focused ion beam scanning electron microscopy. The volume of the left ventricle and cardiomyocytes were significantly increased in aged mice with or without spermidine treatment. Although the number of mitochondria was similar in young and old control mice, it was significantly increased in aged mice treated with spermidine. The interfibrillar mitochondria from old mice exhibited a lower degree of organization and a greater variation in shape and size compared to young animals. The mitochondrial alignment along the myofibrils in the spermidine-treated mice appeared more regular than in control aged mice, however, old mitochondria from animals fed spermidine also showed a greater diversity of shape and size than young mitochondria. In conclusion, mitochondria of the aged mouse left ventricle exhibited changes in number and 3D ultrastructure that is likely the structural correlate of dysfunctional mitochondrial dynamics. Spermidine treatment reduced, at least in part, these morphological changes, indicating a beneficial effect on cardiac mitochondrial alterations associated with aging.
Collapse
Affiliation(s)
- Jil Messerer
- Institute of Functional and Applied AnatomyHannover Medical SchoolHannoverGermany
| | - Christoph Wrede
- Institute of Functional and Applied AnatomyHannover Medical SchoolHannoverGermany,Research Core Unit Electron MicroscopyHannover Medical SchoolHannoverGermany
| | - Julia Schipke
- Institute of Functional and Applied AnatomyHannover Medical SchoolHannoverGermany,Biomedical Research in Endstage and Obstructive Lung Disease HannoverMember of the German Center for Lung Research (DZL)HannoverGermany
| | - Christina Brandenberger
- Institute of Functional and Applied AnatomyHannover Medical SchoolHannoverGermany,Biomedical Research in Endstage and Obstructive Lung Disease HannoverMember of the German Center for Lung Research (DZL)HannoverGermany
| | | | - Tobias Eisenberg
- BioTechMed GrazGrazAustria,Institute of Molecular BiosciencesNAWI GrazUniversity of GrazGrazAustria,Field of Excellence BioHealth—University of GrazGrazAustria
| | - Frank Madeo
- BioTechMed GrazGrazAustria,Institute of Molecular BiosciencesNAWI GrazUniversity of GrazGrazAustria,Field of Excellence BioHealth—University of GrazGrazAustria
| | - Simon Sedej
- Department of CardiologyMedical University of GrazGrazAustria,BioTechMed GrazGrazAustria,Faculty of MedicineUniversity of MariborMariborSlovenia
| | - Christian Mühlfeld
- Institute of Functional and Applied AnatomyHannover Medical SchoolHannoverGermany,Research Core Unit Electron MicroscopyHannover Medical SchoolHannoverGermany,Biomedical Research in Endstage and Obstructive Lung Disease HannoverMember of the German Center for Lung Research (DZL)HannoverGermany
| |
Collapse
|
8
|
Zhang C, Tao L, Yue Y, Ren L, Zhang Z, Wang X, Tian J, An L. Mitochondrial transfer from induced pluripotent stem cells rescues developmental potential of in vitro fertilized embryos from aging females†. Biol Reprod 2021; 104:1114-1125. [PMID: 33511405 DOI: 10.1093/biolre/ioab009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 09/09/2020] [Accepted: 01/21/2021] [Indexed: 11/14/2022] Open
Abstract
Conventional heterologous mitochondrial replacement therapy is clinically complicated by "tri-parental" ethical concerns and limited source of healthy donor oocytes or zygotes. Autologous mitochondrial transfer is a promising alternative in rescuing poor oocyte quality and impaired embryo developmental potential associated with mitochondrial disorders, including aging. However, the efficacy and safety of mitochondrial transfer from somatic cells remains largely controversial, and unsatisfying outcomes may be due to distinct mitochondrial state in somatic cells from that in oocytes. Here, we propose a potential strategy for improving in vitro fertilization (IVF) outcomes of aging female patients via mitochondrial transfer from induced pluripotent stem (iPS) cells. Using naturally aging mice and well-established cell lines as models, we found iPS cells and oocytes share similar mitochondrial morphology and functions, whereas the mitochondrial state in differentiated somatic cells is substantially different. By microinjection of isolated mitochondria into fertilized oocytes following IVF, our results indicate that mitochondrial transfer from iPS, but not MEF cells, can rescue the impaired developmental potential of embryos from aging female mice and obtain an enhanced implantation rate following embryo transfer. The beneficial effect may be explained by the fact that mitochondrial transfer from iPS cells not only compensates for aging-associated loss of mtDNA, but also rescues mitochondrial metabolism of subsequent preimplantation embryos. Using mitochondria from iPS cells as the donor, our study not only proposes a promising strategy for improving IVF outcomes of aging females, but also highlights the importance of synchronous mitochondrial state in supporting embryo developmental potential.
Collapse
Affiliation(s)
- Chao Zhang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs; College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Li Tao
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs; College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Yuan Yue
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs; College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Likun Ren
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs; College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Zhenni Zhang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs; College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Xiaodong Wang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs; College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Jianhui Tian
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs; College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Lei An
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs; College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| |
Collapse
|
9
|
Debska-Vielhaber G, Miller I, Peeva V, Zuschratter W, Walczak J, Schreiber S, Petri S, Machts J, Vogt S, Szczepanowska J, Gellerich FN, Hermann A, Vielhaber S, Kunz WS. Impairment of mitochondrial oxidative phosphorylation in skin fibroblasts of SALS and FALS patients is rescued by in vitro treatment with ROS scavengers. Exp Neurol 2021; 339:113620. [PMID: 33497646 DOI: 10.1016/j.expneurol.2021.113620] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 12/22/2020] [Accepted: 01/19/2021] [Indexed: 11/19/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating, rapidly progressive, neurodegenerative disorder affecting upper and lower motor neurons. Approximately 10% of patients suffer from familial ALS (FALS) with mutations in different ubiquitously expressed genes including SOD1, C9ORF72, TARDBP, and FUS. There is compelling evidence for mitochondrial involvement in the pathogenic mechanisms of FALS and sporadic ALS (SALS), which is believed to be relevant for disease. Owing to the ubiquitous expression of relevant disease-associated genes, mitochondrial dysfunction is also detectable in peripheral patient tissue. We here report results of a detailed investigation of the functional impairment of mitochondrial oxidative phosphorylation (OXPHOS) in cultured skin fibroblasts from 23 SALS and 17 FALS patients, harboring pathogenic mutations in SOD1, C9ORF72, TARDBP and FUS. A considerable functional and structural mitochondrial impairment was detectable in fibroblasts from patients with SALS. Similarly, fibroblasts from patients with FALS, harboring pathogenic mutations in TARDBP, FUS and SOD1, showed mitochondrial defects, while fibroblasts from C9ORF72 associated FALS showed a very mild impairment detectable in mitochondrial ATP production rates only. While we could not detect alterations in the mtDNA copy number in the SALS or FALS fibroblast cultures, the impairment of OXPHOS in SALS fibroblasts and SOD1 or TARDBP FALS could be rescued by in vitro treatments with CoQ10 (5 μM for 3 weeks) or Trolox (300 μM for 5 days). This underlines the role of elevated oxidative stress as a potential cause for the observed functional effects on mitochondria, which might be relevant disease modifying factors.
Collapse
Affiliation(s)
- Grazyna Debska-Vielhaber
- Department of Neurology, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Irina Miller
- Department of Neurology, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany; Department of Neurosurgery, University Hospital Carl Gustav Carus Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Viktoriya Peeva
- Institute of Experimental Epileptology and Cognition Research, Life & Brain Center, University of Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | - Werner Zuschratter
- Leibniz Institute for Neurobiology, Laboratory for Electron- and Laserscanning- Microscopy, Brenneckestr.6, 39118 Magdeburg, Germany
| | - Jaroslaw Walczak
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St, 02-993 Warsaw, Poland; Institute of Fundamental Technological Research, Polish Academy of Sciences, 5b Pawińskiego St, 02-106 Warsaw, Poland
| | - Stefanie Schreiber
- Department of Neurology, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany; German Centre for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str.44, 39120 Magdeburg, Germany
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Judith Machts
- Department of Neurology, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany; German Centre for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str.44, 39120 Magdeburg, Germany
| | - Susanne Vogt
- Department of Neurology, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Joanna Szczepanowska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St, 02-993 Warsaw, Poland
| | - Frank N Gellerich
- Department of Neurology, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Andreas Hermann
- Translational Neurodegeneration Section "Albrecht-Kossel", Department of Neurology and Center for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Gehlsheimer Straße 20, 18147 Rostock, Germany; German Centre for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Gehlsheimer Straße 20, 18147 Rostock, Germany
| | - Stefan Vielhaber
- Department of Neurology, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany; German Centre for Neurodegenerative Diseases (DZNE) Magdeburg, Leipziger Str.44, 39120 Magdeburg, Germany.
| | - Wolfram S Kunz
- Institute of Experimental Epileptology and Cognition Research, Life & Brain Center, University of Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| |
Collapse
|
10
|
Machado-Oliveira G, Ramos C, Marques ARA, Vieira OV. Cell Senescence, Multiple Organelle Dysfunction and Atherosclerosis. Cells 2020; 9:E2146. [PMID: 32977446 PMCID: PMC7598292 DOI: 10.3390/cells9102146] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/19/2020] [Accepted: 09/20/2020] [Indexed: 01/10/2023] Open
Abstract
Atherosclerosis is an age-related disorder associated with long-term exposure to cardiovascular risk factors. The asymptomatic progression of atherosclerotic plaques leads to major cardiovascular diseases (CVD), including acute myocardial infarctions or cerebral ischemic strokes in some cases. Senescence, a biological process associated with progressive structural and functional deterioration of cells, tissues and organs, is intricately linked to age-related diseases. Cell senescence involves coordinated modifications in cellular compartments and has been demonstrated to contribute to different stages of atheroma development. Senescence-based therapeutic strategies are currently being pursued to treat and prevent CVD in humans in the near-future. In addition, distinct experimental settings allowed researchers to unravel potential approaches to regulate anti-apoptotic pathways, facilitate excessive senescent cell clearance and eventually reverse atherogenesis to improve cardiovascular function. However, a deeper knowledge is required to fully understand cellular senescence, to clarify senescence and atherogenesis intertwining, allowing researchers to establish more effective treatments and to reduce the cardiovascular disorders' burden. Here, we present an objective review of the key senescence-related alterations of the major intracellular organelles and analyze the role of relevant cell types for senescence and atherogenesis. In this context, we provide an updated analysis of therapeutic approaches, including clinically relevant experiments using senolytic drugs to counteract atherosclerosis.
Collapse
Affiliation(s)
- Gisela Machado-Oliveira
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (C.R.); (A.R.A.M.)
| | | | | | - Otília V. Vieira
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (C.R.); (A.R.A.M.)
| |
Collapse
|
11
|
Bornstein R, Gonzalez B, Johnson SC. Mitochondrial pathways in human health and aging. Mitochondrion 2020; 54:72-84. [PMID: 32738358 PMCID: PMC7508824 DOI: 10.1016/j.mito.2020.07.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/20/2020] [Accepted: 07/27/2020] [Indexed: 12/27/2022]
Abstract
Mitochondria are eukaryotic organelles known best for their roles in energy production and metabolism. While often thought of as simply the 'powerhouse of the cell,' these organelles participate in a variety of critical cellular processes including reactive oxygen species (ROS) production, regulation of programmed cell death, modulation of inter- and intracellular nutrient signaling pathways, and maintenance of cellular proteostasis. Disrupted mitochondrial function is a hallmark of eukaryotic aging, and mitochondrial dysfunction has been reported to play a role in many aging-related diseases. While mitochondria are major players in human diseases, significant questions remain regarding their precise mechanistic role. In this review, we detail mechanisms by which mitochondrial dysfunction participate in disease and aging based on findings from model organisms and human genetics studies.
Collapse
Affiliation(s)
| | - Brenda Gonzalez
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Simon C Johnson
- Department of Neurology, University of Washington, Seattle, WA, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA.
| |
Collapse
|
12
|
Pu X, Luo A, Su H, Zhang K, Tian C, Chen B, Chai P, Xia X. Optimization and mechanism of postponing aging of polysaccharides from Chinese herbal medicine formula. Toxicol Res (Camb) 2020; 9:239-248. [PMID: 32670555 DOI: 10.1093/toxres/tfaa020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/03/2020] [Indexed: 11/14/2022] Open
Abstract
To study the extraction technology of polysaccharides (AAP) from Chinese herbal medicine formula and its mechanism of delaying aging. First, L9(3)4 orthogonal test was used to optimize the optimal enzyme-assisted extraction parameters of polysaccharides. And the anti-aging effects was evaluated by detecting mitochondrial function, protein, DNA, adhesion molecules and cell cycle in aging rats. The optimal extraction process parameters were the cellulase concentration of 1.5%, the pH at 5, the enzyme temperature at 50°C and the extraction time of 180 min. The anti-aging results showed that AAP can effectively increase the activities of malate dehydrogenase, succinate dehydrogenase and superoxide dismutase. It also can decrease the activity of monoamine oxidase and methane dicarboxylic aldehyde levels in the brain tissue. Meanwhile, the polysaccharides enhanced telomerase activity while reduced p16 protein expression of the brain mitochondria. In addition, the polysaccharides continued to improve heart damage and significantly lessen mitochondrial DNA concentrations. For a certain period of time, it also enhanced the activity of superoxide dismutase, reduced glutathione, glutathione peroxidase and decreased protein carbonyl and methane dicarboxylic aldehyde content of kidney in D-galactose-induced aging rats. Furthermore, the polysaccharides restored the number of cells in the peripheral blood lines and BMNC through inhibiting the drop of the number of red blood cells, white blood cells, platelets in the peripheral blood and bone marrow mononuclear cell of the aging rats. At the same time, AAP accelerated G1 phase cell to enter S phase in cell cycle in aging rats. Our research suggests that the polysaccharides may be a potential anti-aging agent and can be further developed as a functional food or new drug to delay aging or treat aging-related diseases.
Collapse
Affiliation(s)
- Xiuying Pu
- School of Life Science and Engineering, Lanzhou University of Technology, The Key Lab of Screening, Evaluation and Advanced Processing of TCM and Tibetan Medicine, Gansu Educational Department, No. 287, Langongping Road, Qilihe District, Lanzhou 730050, Gansu, China
| | - Amiao Luo
- School of Life Science and Engineering, Lanzhou University of Technology, The Key Lab of Screening, Evaluation and Advanced Processing of TCM and Tibetan Medicine, Gansu Educational Department, No. 287, Langongping Road, Qilihe District, Lanzhou 730050, Gansu, China
| | - Hui Su
- School of Life Science and Engineering, Lanzhou University of Technology, The Key Lab of Screening, Evaluation and Advanced Processing of TCM and Tibetan Medicine, Gansu Educational Department, No. 287, Langongping Road, Qilihe District, Lanzhou 730050, Gansu, China
| | - Kaili Zhang
- School of Life Science and Engineering, Lanzhou University of Technology, The Key Lab of Screening, Evaluation and Advanced Processing of TCM and Tibetan Medicine, Gansu Educational Department, No. 287, Langongping Road, Qilihe District, Lanzhou 730050, Gansu, China
| | - Changyi Tian
- School of Life Science and Engineering, Lanzhou University of Technology, The Key Lab of Screening, Evaluation and Advanced Processing of TCM and Tibetan Medicine, Gansu Educational Department, No. 287, Langongping Road, Qilihe District, Lanzhou 730050, Gansu, China
| | - Bo Chen
- School of Life Science and Engineering, Lanzhou University of Technology, The Key Lab of Screening, Evaluation and Advanced Processing of TCM and Tibetan Medicine, Gansu Educational Department, No. 287, Langongping Road, Qilihe District, Lanzhou 730050, Gansu, China
| | - Pengdi Chai
- School of Life Science and Engineering, Lanzhou University of Technology, The Key Lab of Screening, Evaluation and Advanced Processing of TCM and Tibetan Medicine, Gansu Educational Department, No. 287, Langongping Road, Qilihe District, Lanzhou 730050, Gansu, China
| | - Xiaoyu Xia
- School of Life Science and Engineering, Lanzhou University of Technology, The Key Lab of Screening, Evaluation and Advanced Processing of TCM and Tibetan Medicine, Gansu Educational Department, No. 287, Langongping Road, Qilihe District, Lanzhou 730050, Gansu, China
| |
Collapse
|
13
|
Nixon RA. The aging lysosome: An essential catalyst for late-onset neurodegenerative diseases. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140443. [PMID: 32416272 DOI: 10.1016/j.bbapap.2020.140443] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/04/2020] [Accepted: 05/07/2020] [Indexed: 01/13/2023]
Abstract
Lysosomes figure prominently in theories of aging as the proteolytic system most responsible for eliminating growing burdens of damaged proteins and organelles in aging neurons and other long lived cells. Newer evidence shows that diverse experimental measures known to extend lifespan in invertebrate aging models share the property of boosting lysosomal clearance of substrates through the autophagy pathway. Maintaining an optimal level of lysosome acidification is particularly crucial for these anti-aging effects. The exceptional dependence of neurons on fully functional lysosomes is reflected by the neurological phenotypes that develop in congenital lysosomal storage disorders, which commonly present as severe neurodevelopmental or neurodegenerative conditions even though the lysosomal deficit maybe systemic. Similar connections are now being appreciated between primary lysosomal deficit and the risk for late age-onset neurodegenerative disorders. In diseases such as Alzheimer's and Parkinson's, as in aging alone, primary lysosome dysfunction due to acidification impairment is emerging as a frequent theme, supported by the growing list of familial neurodegenerative disorders that involve primary vATPase dysfunction. The additional cellular roles played by intraluminal pH in sensing nutrient and stress and modulating cellular signaling have further expanded the possible ways that lysosomal pH dysregulation in aging and disease can disrupt neuronal function. Here, we consider the impact of cellular aging on lysosomes and how the changes during aging may create the tipping point for disease emergence in major late-age onset neurodegenerative disorders.
Collapse
Affiliation(s)
- Ralph A Nixon
- Center for Dementia Research, Nathan S. Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA; Department of Psychiatry, New York University Langone Medical Center, 550 First Ave, New York, NY 10016, USA; Department of Cell Biology, New York University Langone Medical Center, 550 First Ave, New York, NY 10016, USA; Department of NYU Neuroscience Institute, New York University Langone Medical Center, 550 First Ave, New York, NY 10016, USA.
| |
Collapse
|
14
|
Sánchez-Díaz M, Nicolás-Ávila JÁ, Cordero MD, Hidalgo A. Mitochondrial Adaptations in the Growing Heart. Trends Endocrinol Metab 2020; 31:308-319. [PMID: 32035734 DOI: 10.1016/j.tem.2020.01.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/22/2019] [Accepted: 01/09/2020] [Indexed: 12/14/2022]
Abstract
The heart pumps blood throughout the whole life of an organism, without rest periods during which to replenish energy or detoxify. Hence, cardiomyocytes, the working units of the heart, have mechanisms to ensure constitutive production of energy and detoxification to preserve fitness and function for decades. Even more challenging, the heart must adapt to the varying conditions of the organism from fetal life to adulthood, old age, and pathological stress. Mitochondria are at the nexus of these processes by producing not only energy but also metabolites and oxidative byproducts that can activate alarm signals and be toxic to the cell. We review basic concepts about cardiac mitochondria with a focus on their remarkable adaptations, including elimination, throughout the mammalian lifetime.
Collapse
Affiliation(s)
- María Sánchez-Díaz
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid 28029, Spain
| | | | - Mario D Cordero
- Cátedra de Reproducción y Genética Humana del Instituto para el Estudio de la Biología de la Reproducción Humana (INEBIR), 41009 Sevilla, Spain; Universidad Europea del Atlántico (UNEATLANTICO), and Fundación Universitaria Iberoamericana (FUNIBER), 39011 Santander, Spain.
| | - Andrés Hidalgo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid 28029, Spain; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität, Munich, Germany.
| |
Collapse
|
15
|
Wang Y, Li Y, He C, Gou B, Song M. Mitochondrial regulation of cardiac aging. Biochim Biophys Acta Mol Basis Dis 2018; 1865:1853-1864. [PMID: 30593894 DOI: 10.1016/j.bbadis.2018.12.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 12/13/2022]
Abstract
Aging is associated with progressive decline in cardiac structure and function. Accumulating evidence in model organisms and humans links cardiac aging to mitochondrial regulation, encompassing a complex interplay of mitochondrial morphology, mitochondrial ROS, mitochondrial DNA mutations, mitochondrial unfolded protein response, nicotinamide adenine dinucleotide levels and sirtuins, as well as mitophagy. This review summarizes the recent discoveries on the mitochondrial regulation of cardiac aging and the possible molecular mechanisms underlying the anti-aging effects, as well as the potential interventions that alleviate aging-related cardiac diseases and attenuate cardiac aging via the regulation of mitochondria.
Collapse
Affiliation(s)
- Yuhan Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Forestry University, Beijing 100083, China
| | - Yujing Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Science and Technology of China, Anhui 230026, China
| | - Chuting He
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bo Gou
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
16
|
Dorsch LM, Schuldt M, Knežević D, Wiersma M, Kuster DWD, van der Velden J, Brundel BJJM. Untying the knot: protein quality control in inherited cardiomyopathies. Pflugers Arch 2018; 471:795-806. [PMID: 30109411 PMCID: PMC6475634 DOI: 10.1007/s00424-018-2194-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 08/06/2018] [Indexed: 12/27/2022]
Abstract
Mutations in genes encoding sarcomeric proteins are the most important causes of inherited cardiomyopathies, which are a major cause of mortality and morbidity worldwide. Although genetic screening procedures for early disease detection have been improved significantly, treatment to prevent or delay mutation-induced cardiac disease onset is lacking. Recent findings indicate that loss of protein quality control (PQC) is a central factor in the disease pathology leading to derailment of cellular protein homeostasis. Loss of PQC includes impairment of heat shock proteins, the ubiquitin-proteasome system, and autophagy. This may result in accumulation of misfolded and aggregation-prone mutant proteins, loss of sarcomeric and cytoskeletal proteins, and, ultimately, loss of cardiac function. PQC derailment can be a direct effect of the mutation-induced activation, a compensatory mechanism due to mutation-induced cellular dysfunction or a consequence of the simultaneous occurrence of the mutation and a secondary hit. In this review, we discuss recent mechanistic findings on the role of proteostasis derailment in inherited cardiomyopathies, with special focus on sarcomeric gene mutations and possible therapeutic applications.
Collapse
Affiliation(s)
- Larissa M Dorsch
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands.
| | - Maike Schuldt
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands.
| | - Dora Knežević
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Marit Wiersma
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Diederik W D Kuster
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Jolanda van der Velden
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Bianca J J M Brundel
- Amsterdam UMC, Department of Physiology, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, O2 building 11W53, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Abstract
Changes in mitochondrial capacity and quality play a critical role in skeletal and cardiac muscle dysfunction. In vivo measurements of mitochondrial capacity provide a clear link between physical activity and mitochondrial function in aging and heart failure, although the cause and effect relationship remains unclear. Age-related decline in mitochondrial quality leads to mitochondrial defects that affect redox, calcium, and energy-sensitive signaling by altering the cellular environment that can result in skeletal muscle dysfunction independent of reduced mitochondrial capacity. This reduced mitochondrial quality with age is also likely to sensitize skeletal muscle mitochondria to elevated angiotensin or beta-adrenergic signaling associated with heart failure. This synergy between aging and heart failure could further disrupt cell energy and redox homeostasis and contribute to exercise intolerance in this patient population. Therefore, the interaction between aging and heart failure, particularly with respect to mitochondrial dysfunction, should be a consideration when developing strategies to improve quality of life in heart failure patients. Given the central role of the mitochondria in skeletal and cardiac muscle dysfunction, mitochondrial quality may provide a common link for targeted interventions in these populations.
Collapse
Affiliation(s)
- Sophia Z Liu
- Department of Radiology, University of Washington, Box 358050, Seattle, WA, 98109, USA
| | - David J Marcinek
- Department of Radiology, University of Washington, Box 358050, Seattle, WA, 98109, USA. .,Department of Pathology, University of Washington, Seattle, WA, 98109, USA. .,Department of Bioengineering, University of Washington, Seattle, WA, 98109, USA.
| |
Collapse
|
18
|
Yang YH, Liu JB, Gui Y, Lei LL, Zhang SJ. Relationship between autophagy and perineural invasion, clinicopathological features, and prognosis in pancreatic cancer. World J Gastroenterol 2017; 23:7232-7241. [PMID: 29142470 PMCID: PMC5677195 DOI: 10.3748/wjg.v23.i40.7232] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 09/13/2017] [Accepted: 09/19/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the relationship between autophagy and perineural invasion (PNI), clinical features, and prognosis in patients with pancreatic cancer.
METHODS Clinical and pathological data were retrospectively collected from 109 patients with pancreatic ductal adenocarcinoma who underwent radical resection at the First Affiliated Hospital of Zhengzhou University from January 2011 to August 2016. Expression levels of the autophagy-related protein microtubule-associated protein 1A/1B-light chain 3 (LC3) and PNI marker ubiquitin carboxy-terminal hydrolase (UCH) in pancreatic cancer tissues were detected by immunohistochemistry. The correlations among LC3 expression, PNI, and clinical pathological features in pancreatic cancer were analyzed. The patients were followed for further survival analysis.
RESULTS In 109 cases of pancreatic cancer, 68.8% (75/109) had evidence of PNI and 61.5% (67/109) had high LC3 expression. PNI was associated with lymph node metastasis, pancreatitis, and CA19-9 levels (P < 0.05). LC3 expression was related to lymph node metastasis (P < 0.05) and was positively correlated with neural invasion (P < 0.05, r = 0.227). Multivariate logistic regression analysis indicated that LC3 expression, lymph node metastasis, pancreatitis, and CA19-9 level were factors that influenced neural invasion, whereas only neural invasion itself was an independent factor for high LC3 expression. Univariate analysis showed that LC3 expression, neural invasion, and CA19-9 level were related to the overall survival of pancreatic cancer patients (P < 0.05). Multivariate COX regression analysis indicated that PNI and LC3 expression were independent risk factors for poor prognosis in pancreatic cancer (P < 0.05).
CONCLUSION PNI in patients with pancreatic cancer is positively related to autophagy. Neural invasion and LC3 expression are independent risk factors for pancreatic cancer with a poor prognosis.
Collapse
Affiliation(s)
- Yan-Hui Yang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
- Department of Hepatobiliary Surgery, First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan Province, China
| | - Jiang-Bo Liu
- Department of General Surgery, First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan Province, China
| | - Yang Gui
- Department of Hepatobiliary Surgery, First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan Province, China
| | - Liang-Liang Lei
- Department of General Surgery, First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan Province, China
| | - Shui-Jun Zhang
- Henan Key Laboratory of Digestive Organ Transplantation, Open and Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou Key Laboratory of Hepatobiliary Pancreatic Diseases and Organ Transplantation, Zhengzhou 450052, Henan Province, China
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| |
Collapse
|
19
|
Sheng Y, Lv S, Huang M, Lv Y, Yu J, Liu J, Tang T, Qi H, Di W, Ding G. Opposing effects on cardiac function by calorie restriction in different-aged mice. Aging Cell 2017; 16:1155-1167. [PMID: 28799249 PMCID: PMC5595678 DOI: 10.1111/acel.12652] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2017] [Indexed: 12/25/2022] Open
Abstract
Calorie restriction (CR) increases average and maximum lifespan and exhibits an apparent beneficial impact on age‐related diseases. Several studies have shown that CR initiated either in middle or old age could improve ischemic tolerance and rejuvenate the aging heart; however, the data are not uniform when initiated in young. The accurate time to initiate CR providing maximum benefits for cardiac remodeling and function during aging remains unclear. Thus, whether a similar degree of CR initiated in mice of different ages could exert a similar effect on myocardial protection was investigated in this study. C57BL/6 mice were subjected to a calorically restricted diet (40% less than the ad libitum diet) for 3 months initiated in 3, 12, and 19 months. It was found that CR significantly reversed the aging phenotypes of middle‐aged and old mice including cardiac remodeling (cardiomyocyte hypertrophy and cardiac fibrosis), inflammation, mitochondrial damage, telomere shortening, as well as senescence‐associated markers but accelerated in young mice. Furthermore, whole‐genome microarray demonstrated that the AMP‐activated protein kinase (AMPK)–Forkhead box subgroup ‘O’ (FOXO) pathway might be a major contributor to contrasting regulation by CR initiated in different ages; thus, increased autophagy was seen in middle‐aged and old mice but decreased in young mice. Together, the findings demonstrated promising myocardial protection by 40% CR should be initiated in middle or old age that may have vital implications for the practical nutritional regimen.
Collapse
Affiliation(s)
- Yunlu Sheng
- Department of Geratology; The First Hospital Affiliated to Nanjing Medical University; 300 Guangzhou Road Nanjing 210029 China
| | - Shan Lv
- Department of Geratology; The First Hospital Affiliated to Nanjing Medical University; 300 Guangzhou Road Nanjing 210029 China
| | - Min Huang
- Department of Geratology; The First Hospital Affiliated to Nanjing Medical University; 300 Guangzhou Road Nanjing 210029 China
| | - Yifan Lv
- Department of Geratology; The First Hospital Affiliated to Nanjing Medical University; 300 Guangzhou Road Nanjing 210029 China
| | - Jing Yu
- Department of Geratology; The First Hospital Affiliated to Nanjing Medical University; 300 Guangzhou Road Nanjing 210029 China
| | - Juan Liu
- Department of Geratology; The First Hospital Affiliated to Nanjing Medical University; 300 Guangzhou Road Nanjing 210029 China
| | - Tingting Tang
- Department of Geratology; The First Hospital Affiliated to Nanjing Medical University; 300 Guangzhou Road Nanjing 210029 China
| | - Hanmei Qi
- Department of Geratology; The First Hospital Affiliated to Nanjing Medical University; 300 Guangzhou Road Nanjing 210029 China
| | - Wenjuan Di
- Department of Geratology; The First Hospital Affiliated to Nanjing Medical University; 300 Guangzhou Road Nanjing 210029 China
| | - Guoxian Ding
- Department of Geratology; The First Hospital Affiliated to Nanjing Medical University; 300 Guangzhou Road Nanjing 210029 China
| |
Collapse
|
20
|
Impact of Aging and Exercise on Mitochondrial Quality Control in Skeletal Muscle. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3165396. [PMID: 28656072 PMCID: PMC5471566 DOI: 10.1155/2017/3165396] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/03/2017] [Indexed: 12/17/2022]
Abstract
Mitochondria are characterized by its pivotal roles in managing energy production, reactive oxygen species, and calcium, whose aging-related structural and functional deteriorations are observed in aging muscle. Although it is still unclear how aging alters mitochondrial quality and quantity in skeletal muscle, dysregulation of mitochondrial biogenesis and dynamic controls has been suggested as key players for that. In this paper, we summarize current understandings on how aging regulates muscle mitochondrial biogenesis, while focusing on transcriptional regulations including PGC-1α, AMPK, p53, mtDNA, and Tfam. Further, we review current findings on the muscle mitochondrial dynamic systems in aging muscle: fusion/fission, autophagy/mitophagy, and protein import. Next, we also discuss how endurance and resistance exercises impact on the mitochondrial quality controls in aging muscle, suggesting possible effective exercise strategies to improve/maintain mitochondrial health.
Collapse
|
21
|
Singh S, Sharma S. Dynamin-related protein-1 as potential therapeutic target in various diseases. Inflammopharmacology 2017; 25:383-392. [PMID: 28409390 DOI: 10.1007/s10787-017-0347-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 03/31/2017] [Indexed: 12/19/2022]
Abstract
Mitochondria can interchange morphology due to their dynamic nature. It can exist in either fragmented disconnected arrangement or elongated interconnected mitochondrial networks due to fission and fusion, respectively. The recent studies have revealed the remarkable and unexpected insights into the physiological impact and molecular regulation of mitochondrial morphology. The balance between fission and fusion governs the faith of the cell. The active targeting of DRP 1 to the outer mitochondrial membrane (OMM) is done by non-GTPase receptor proteins such as mitochondrial fission factor, mitochondrial fission protein 1 and mitochondrial elongation factor 1. The active targeting of DRP 1 to OMM leads to the fission of mitochondria. However, the imbalance of DRP 1-dependent mitochondrial fission and modulation of equilibrium of fission and fusion has been documented to be involved in several cardiovascular and neurodegenerative disorders. In this review, we are focusing on the active participation of DRP 1 in various diseases and also the factors responsible for the activation of DRP 1 for its action.
Collapse
Affiliation(s)
- Surinder Singh
- Cardiovascular Division, Department of Pharmacology, I.S.F. College of Pharmacy, Moga, 142001, Punjab, India
| | - Saurabh Sharma
- Cardiovascular Division, Department of Pharmacology, I.S.F. College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
22
|
McCulloch K, Litherland GJ, Rai TS. Cellular senescence in osteoarthritis pathology. Aging Cell 2017; 16:210-218. [PMID: 28124466 PMCID: PMC5334539 DOI: 10.1111/acel.12562] [Citation(s) in RCA: 236] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2016] [Indexed: 12/19/2022] Open
Abstract
Cellular senescence is a state of stable proliferation arrest of cells. The senescence pathway has many beneficial effects and is seen to be activated in damaged/stressed cells, as well as during embryonic development and wound healing. However, the persistence and accumulation of senescent cells in various tissues can also impair function and have been implicated in the pathogenesis of many age‐related diseases. Osteoarthritis (OA), a severely debilitating chronic condition characterized by progressive tissue remodeling and loss of joint function, is the most prevalent disease of the synovial joints, and increasing age is the primary OA risk factor. The profile of inflammatory and catabolic mediators present during the pathogenesis of OA is strikingly similar to the secretory profile observed in ‘classical’ senescent cells. During OA, chondrocytes (the sole cell type present within articular cartilage) exhibit increased levels of various senescence markers, such as senescence‐associated beta‐galactosidase (SAβGal) activity, telomere attrition, and accumulation of p16ink4a. This suggests the hypothesis that senescence of cells within joint tissues may play a pathological role in the causation of OA. In this review, we discuss the mechanisms by which senescent cells may predispose synovial joints to the development and/or progression of OA, as well as touching upon various epigenetic alterations associated with both OA and senescence.
Collapse
Affiliation(s)
- Kendal McCulloch
- Institute of Biomedical and Environmental Health Research; University of the West of Scotland; Paisley PA1 2BE UK
| | - Gary J. Litherland
- Institute of Biomedical and Environmental Health Research; University of the West of Scotland; Paisley PA1 2BE UK
| | - Taranjit Singh Rai
- Institute of Biomedical and Environmental Health Research; University of the West of Scotland; Paisley PA1 2BE UK
| |
Collapse
|
23
|
Parry TL, Willis MS. Cardiac ubiquitin ligases: Their role in cardiac metabolism, autophagy, cardioprotection and therapeutic potential. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1862:2259-2269. [PMID: 27421947 PMCID: PMC5159290 DOI: 10.1016/j.bbadis.2016.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/05/2016] [Accepted: 07/11/2016] [Indexed: 12/19/2022]
Abstract
Both the ubiquitin-proteasome system (UPS) and the lysosomal autophagy system have emerged as complementary key players responsible for the turnover of cellular proteins. The regulation of protein turnover is critical to cardiomyocytes as post-mitotic cells with very limited regenerative capacity. In this focused review, we describe the emerging interface between the UPS and autophagy, with E3's regulating autophagy at two critical points through multiple mechanisms. Moreover, we discuss recent insights in how both the UPS and autophagy can alter metabolism at various levels, to present new ways to think about therapeutically regulating autophagy in a focused manner to optimize disease-specific cardioprotection, without harming the overall homeostasis of protein quality control. This article is part of a Special Issue entitled: The role of post-translational protein modifications on heart and vascular metabolism edited by Jason R.B. Dyck & Jan F.C. Glatz.
Collapse
Affiliation(s)
- Traci L Parry
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Monte S Willis
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA; Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
24
|
Yang Y, Zhao C, Yang P, Wang X, Wang L, Chen A. Autophagy in cardiac metabolic control: Novel mechanisms for cardiovascular disorders. Cell Biol Int 2016; 40:944-54. [PMID: 27191043 DOI: 10.1002/cbin.10626] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/10/2016] [Accepted: 05/13/2016] [Indexed: 01/01/2023]
Abstract
As an extensively studied quality control system, autophagy is responsible for clearance of dysfunctional organelles and damaged marcomolecules in cells. In addition to its biological recycling function, autophagy plays a significant role in the pathogenesis of metabolic syndromes such as obesity and diabetes. In particular, metabolic disorders contribute to cardiovascular disease development. As energy required to maintain cardiac cells functional is immense, disturbances in the balance between anabolic and catabolic metabolism possibly contribute to cardiovascular disorders. Therefore, an urgent need to expand our knowledge on the role of autophagy on the metabolic regulation of hearts emerges. In this review, the potential relationship between autophagic activity and cardiac metabolism is explored and we also discuss how dysregulated autophagy leads to severe cardiac disorders from the perspective of metabolic control.
Collapse
Affiliation(s)
- Yufei Yang
- Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou City, China
| | - Cong Zhao
- Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou City, China
| | - Pingzhen Yang
- Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou City, China
| | - Xianbao Wang
- Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou City, China
| | - Lizi Wang
- Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou City, China
| | - Aihua Chen
- Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou City, China
| |
Collapse
|
25
|
ATP synthase deficiency due to TMEM70 mutation leads to ultrastructural mitochondrial degeneration and is amenable to treatment. BIOMED RESEARCH INTERNATIONAL 2015; 2015:462592. [PMID: 26550569 PMCID: PMC4621340 DOI: 10.1155/2015/462592] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/29/2015] [Indexed: 12/21/2022]
Abstract
TMEM70 is involved in the biogenesis of mitochondrial ATP synthase and mutations in the TMEM70 gene impair oxidative phosphorylation. Herein, we report on pathology and treatment of ATP synthase deficiency in four siblings. A consanguineous family of Roma (Gipsy) ethnic origin gave birth to 6 children of which 4 were affected presenting with dysmorphic features, failure to thrive, cardiomyopathy, metabolic crises, and 3-methylglutaconic aciduria as clinical symptoms. Genetic testing revealed a homozygous mutation (c.317-2A>G) in the TMEM70 gene. While light microscopy was unremarkable, ultrastructural investigation of muscle tissue revealed accumulation of swollen degenerated mitochondria with lipid crystalloid inclusions, cristae aggregation, and exocytosis of mitochondrial material. Biochemical analysis of mitochondrial complexes showed an almost complete ATP synthase deficiency. Despite harbouring the same mutation, the clinical outcome in the four siblings was different. Two children died within 60 h after birth; the other two had recurrent life-threatening metabolic crises but were successfully managed with supplementation of anaplerotic amino acids, lipids, and symptomatic treatment during metabolic crisis. In summary, TMEM70 mutations can cause distinct ultrastructural mitochondrial degeneration and almost complete deficiency of ATP synthase but are still amenable to treatment.
Collapse
|
26
|
Allen SP, Duffy LM, Shaw PJ, Grierson AJ. Altered age-related changes in bioenergetic properties and mitochondrial morphology in fibroblasts from sporadic amyotrophic lateral sclerosis patients. Neurobiol Aging 2015; 36:2893-903. [DOI: 10.1016/j.neurobiolaging.2015.07.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 06/19/2015] [Accepted: 07/01/2015] [Indexed: 12/11/2022]
|
27
|
Masotti A, Celluzzi A, Petrini S, Bertini E, Zanni G, Compagnucci C. Aged iPSCs display an uncommon mitochondrial appearance and fail to undergo in vitro neurogenesis. Aging (Albany NY) 2015; 6:1094-108. [PMID: 25567319 PMCID: PMC4298368 DOI: 10.18632/aging.100708] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Reprogramming of human fibroblasts into induced pluripotent stem cells (iPSCs) leads to mitochondrial rejuvenation, making iPSCs a candidate model to study the mitochondrial biology during stemness and differentiation. At present, it is generally accepted that iPSCs can be maintained and propagated indefinitely in culture, but no specific studies have addressed this issue. In our study, we investigated features related to the 'biological age' of iPSCs, culturing and analyzing iPSCs kept for prolonged periods in vitro. We have demonstrated that aged iPSCs present an increased number of mitochondria per cell with an altered mitochondrial membrane potential and fail to properly undergo in vitro neurogenesis. In aged iPSCs we have also found an altered expression of genes relevant to mitochondria biogenesis. Overall, our results shed light on the mitochondrial biology of young and aged iPSCs and explore how an altered mitochondrial status may influence neuronal differentiation. Our work suggests to deepen the understanding of the iPSCs biology before considering their use in clinical applications.
Collapse
|
28
|
Tocchi A, Quarles EK, Basisty N, Gitari L, Rabinovitch PS. Mitochondrial dysfunction in cardiac aging. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2015; 1847:1424-33. [PMID: 26191650 DOI: 10.1016/j.bbabio.2015.07.009] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 07/06/2015] [Accepted: 07/09/2015] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases are the leading cause of death in most developed nations. While it has received the least public attention, aging is the dominant risk factor for developing cardiovascular diseases, as the prevalence of cardiovascular diseases increases dramatically with increasing age. Cardiac aging is an intrinsic process that results in impaired cardiac function, along with cellular and molecular changes. Mitochondria play a great role in these processes, as cardiac function is an energetically demanding process. In this review, we examine mitochondrial dysfunction in cardiac aging. Recent research has demonstrated that mitochondrial dysfunction can disrupt morphology, signaling pathways, and protein interactions; conversely, mitochondrial homeostasis is maintained by mechanisms that include fission/fusion, autophagy, and unfolded protein responses. Finally, we describe some of the recent findings in mitochondrial targeted treatments to help meet the challenges of mitochondrial dysfunction in aging.
Collapse
Affiliation(s)
- Autumn Tocchi
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, USA.
| | - Ellen K Quarles
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, USA.
| | - Nathan Basisty
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, USA.
| | - Lemuel Gitari
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, USA.
| | - Peter S Rabinovitch
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, USA.
| |
Collapse
|
29
|
Tenenboim H, Smirnova J, Willmitzer L, Steup M, Brotman Y. VMP1-deficient Chlamydomonas exhibits severely aberrant cell morphology and disrupted cytokinesis. BMC PLANT BIOLOGY 2014; 14:121. [PMID: 24885763 PMCID: PMC4108031 DOI: 10.1186/1471-2229-14-121] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 04/28/2014] [Indexed: 05/20/2023]
Abstract
BACKGROUND The versatile Vacuole Membrane Protein 1 (VMP1) has been previously investigated in six species. It has been shown to be essential in macroautophagy, where it takes part in autophagy initiation. In addition, VMP1 has been implicated in organellar biogenesis; endo-, exo- and phagocytosis, and protein secretion; apoptosis; and cell adhesion. These roles underly its proven involvement in pancreatitis, diabetes and cancer in humans. RESULTS In this study we analyzed a VMP1 homologue from the green alga Chlamydomonas reinhardtii. CrVMP1 knockdown lines showed severe phenotypes, mainly affecting cell division as well as the morphology of cells and organelles. We also provide several pieces of evidence for its involvement in macroautophagy. CONCLUSION Our study adds a novel role to VMP1's repertoire, namely the regulation of cytokinesis. Though the directness of the observed effects and the mechanisms underlying them remain to be defined, the protein's involvement in macroautophagy in Chlamydomonas, as found by us, suggests that CrVMP1 shares molecular characteristics with its animal and protist counterparts.
Collapse
Affiliation(s)
- Hezi Tenenboim
- Institute of Biochemistry and Biology, Department of Plant Physiology, Universität Potsdam, Potsdam, Germany
- Max Planck Institute for Molecular Plant Physiology, Am Mühlenberg 1, 14476 Potsdam, Germany
| | - Julia Smirnova
- Max Planck Institute for Molecular Plant Physiology, Am Mühlenberg 1, 14476 Potsdam, Germany
| | - Lothar Willmitzer
- Max Planck Institute for Molecular Plant Physiology, Am Mühlenberg 1, 14476 Potsdam, Germany
| | - Martin Steup
- Institute of Biochemistry and Biology, Department of Plant Physiology, Universität Potsdam, Potsdam, Germany
| | - Yariv Brotman
- Max Planck Institute for Molecular Plant Physiology, Am Mühlenberg 1, 14476 Potsdam, Germany
| |
Collapse
|
30
|
Wiggins LM. Morphological changes and altered expression of antioxidant proteins in a heterozygous dynein mutant; a mouse model of spinal muscular atrophy. ACTA ACUST UNITED AC 2014; 3:161-173. [PMID: 25866698 DOI: 10.5455/oams.310714.or.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVE There is increased evidence that oxidative stress is involved in exacerbations of neurodegenerative diseases and spinal muscular atrophies. METHODS We examined changes in morphology and expression of antioxidant proteins and peroxiredoxins in motor neurons of lumbar spinal cord, dorsal root ganglion sensory neurons, macroglial cells and quadriceps muscles of newborn heterozygous Loa/+ mice ("legs at odd angles"), a mouse model for early onset of the spinal muscular atrophy with lower extremity predominance (SMA-LED). RESULTS Our data indicate that newborn Loa-mice develop: neuroinflammation of the sensory and motor neurons; muscular inflammation with atrophic and denervated myofibers; increased expression of neuronal mitochondrial peroxiredoxins (Prxs) 3, 5 and cytoplasmic Prx 6 in motor and sensory neurons, myofibers, fibroblasts of perimysium and chondrocytes of cartilage; and decreased expression of Prx 6 by glial cells and in extracellular space surrounding motor neurons. CONCLUSION The decrease in expression of Prx 6 by glial cells and extracellular Prx 6 secretion in early stages of the pathological conditions is consistent with the hypothesis that chronic oxidative stress may lead to neurodegeneration of motor neurons and exacerbation of the pathology.
Collapse
Affiliation(s)
- Larisa M Wiggins
- Department of Physiology and Cell Biology, University of Nevada, Reno
| |
Collapse
|
31
|
Short-term calorie restriction protects against renal senescence of aged rats by increasing autophagic activity and reducing oxidative damage. Mech Ageing Dev 2013; 134:570-9. [PMID: 24291536 DOI: 10.1016/j.mad.2013.11.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Revised: 08/21/2013] [Accepted: 11/16/2013] [Indexed: 11/23/2022]
Abstract
To explore the effect of short-term calorie restriction (CR) on renal aging, 8-week CR with 60% of the food intake of the ad libitum group was administered in 25-month-old male Sprague-Dawley rats. Aged rats subjected to short-term CR had lower body weight, level of triglycerides and ratio of urine protein to urine creatinine, respectively. Short-term CR blunted the increased glomerular volume, the degree of fibrosis, p16 and the positive rate of senescence-associated β-galactosidase staining of the kidneys in old ad libitum group. Light chain 3/Atg8 as an autophagy marker exhibited a marked decline in aged kidneys, which was increased by short-term CR. The levels of p62/SQSTM1 and polyubiquitin aggregates, which were increased in older kidneys, were blunted by short-term CR. Short-term CR retarded the level of 8-hydroxydeoxyguanosine, a marker of mitochondrial DNA oxidative damage. Moreover, we found an increased level of SIRT1 and AMPK, and a decreased level of mTOR in aged kidneys after short-term CR. These results suggested that short-term CR could be considered as a potential intervention for retardation of renal senescence by increasing autophagy and subsequently reducing oxidative damage. Three master regulators of energy metabolism, SIRT1, AMPK and mTOR are associated with these effects.
Collapse
|
32
|
Kim YY, Ku SY, Huh Y, Liu HC, Kim SH, Choi YM, Moon SY. Anti-aging effects of vitamin C on human pluripotent stem cell-derived cardiomyocytes. AGE (DORDRECHT, NETHERLANDS) 2013; 35:1545-1557. [PMID: 22843416 PMCID: PMC3776090 DOI: 10.1007/s11357-012-9457-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 07/06/2012] [Indexed: 06/01/2023]
Abstract
Human pluripotent stem cells (hPSCs) have arisen as a source of cells for biomedical research due to their developmental potential. Stem cells possess the promise of providing clinicians with novel treatments for disease as well as allowing researchers to generate human-specific cellular metabolism models. Aging is a natural process of living organisms, yet aging in human heart cells is difficult to study due to the ethical considerations regarding human experimentation as well as a current lack of alternative experimental models. hPSC-derived cardiomyocytes (CMs) bear a resemblance to human cardiac cells and thus hPSC-derived CMs are considered to be a viable alternative model to study human heart cell aging. In this study, we used hPSC-derived CMs as an in vitro aging model. We generated cardiomyocytes from hPSCs and demonstrated the process of aging in both human embryonic stem cell (hESC)- and induced pluripotent stem cell (hiPSC)-derived CMs. Aging in hESC-derived CMs correlated with reduced membrane potential in mitochondria, the accumulation of lipofuscin, a slower beating pattern, and the downregulation of human telomerase RNA (hTR) and cell cycle regulating genes. Interestingly, the expression of hTR in hiPSC-derived CMs was not significantly downregulated, unlike in hESC-derived CMs. In order to delay aging, vitamin C was added to the cultured CMs. When cells were treated with 100 μM of vitamin C for 48 h, anti-aging effects, specifically on the expression of telomere-related genes and their functionality in aging cells, were observed. Taken together, these results suggest that hPSC-derived CMs can be used as a unique human cardiomyocyte aging model in vitro and that vitamin C shows anti-aging effects in this model.
Collapse
Affiliation(s)
- Yoon Young Kim
- />Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, South Korea
| | - Seung-Yup Ku
- />Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, South Korea
- />Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul, 110-744 South Korea
| | - Yul Huh
- />Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, South Korea
| | - Hung-Ching Liu
- />Center for Reproductive Medicine and Infertility, Weill Cornell Medical College, New York, NY 10021 USA
| | - Seok Hyun Kim
- />Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, South Korea
- />Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul, 110-744 South Korea
| | - Young Min Choi
- />Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, South Korea
- />Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul, 110-744 South Korea
| | - Shin Yong Moon
- />Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, South Korea
- />Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul, 110-744 South Korea
| |
Collapse
|
33
|
Actions of 17β-estradiol and testosterone in the mitochondria and their implications in aging. Ageing Res Rev 2013; 12:907-17. [PMID: 24041489 DOI: 10.1016/j.arr.2013.09.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 09/06/2013] [Indexed: 02/02/2023]
Abstract
A decline in the mitochondrial functions and aging are two closely related processes. The presence of estrogen and androgen receptors and hormone-responsive elements in the mitochondria represents the starting point for the investigation of the effects of 17β-estradiol and testosterone on the mitochondrial functions and their relationships with aging. Both steroids trigger a complex molecular mechanism that involves crosstalk between the mitochondria, nucleus, and plasma membrane, and the cytoskeleton plays a key role in these interactions. The result of this signaling is mitochondrial protection. Therefore, the molecular components of the pathways activated by the sexual steroids could represent targets for anti-aging therapies. In this review, we discuss previous studies that describe the estrogen- and testosterone-dependent actions on the mitochondrial processes implicated in aging.
Collapse
|
34
|
Kim Y, Byun H, Jee BA, Cho H, Seo Y, Kim Y, Park MH, Chung H, Woo HG, Yoon G. Implications of time-series gene expression profiles of replicative senescence. Aging Cell 2013; 12:622-34. [PMID: 23590226 DOI: 10.1111/acel.12087] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2013] [Indexed: 12/21/2022] Open
Abstract
Although senescence has long been implicated in aging-associated pathologies, it is not clearly understood how senescent cells are linked to these diseases. To address this knowledge gap, we profiled cellular senescence phenotypes and mRNA expression patterns during replicative senescence in human diploid fibroblasts. We identified a sequential order of gain-of-senescence phenotypes: low levels of reactive oxygen species, cell mass/size increases with delayed cell growth, high levels of reactive oxygen species with increases in senescence-associated β-galactosidase activity (SA-β-gal), and high levels of SA-β-gal activity. Gene expression profiling revealed four distinct modules in which genes were prominently expressed at certain stages of senescence, allowing us to divide the process into four stages: early, middle, advanced, and very advanced. Interestingly, the gene expression modules governing each stage supported the development of the associated senescence phenotypes. Senescence-associated secretory phenotype-related genes also displayed a stage-specific expression pattern with three unique features during senescence: differential expression of interleukin isoforms, differential expression of interleukins and their receptors, and differential expression of matrix metalloproteinases and their inhibitory proteins. We validated these phenomena at the protein level using human diploid fibroblasts and aging Sprague-Dawley rat skin tissues. Finally, disease-association analysis of the modular genes also revealed stage-specific patterns. Taken together, our results reflect a detailed process of cellular senescence and provide diverse genome-wide information of cellular backgrounds for senescence.
Collapse
Affiliation(s)
- You‐Mie Kim
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine Suwon 443‐721 Korea
| | - Hae‐Ok Byun
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine Suwon 443‐721 Korea
| | | | | | - Yong‐Hak Seo
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine Suwon 443‐721 Korea
| | - You‐Sun Kim
- Institute for Medical Sciences Ajou University School of Medicine Suwon 443‐721Korea
| | - Min Hi Park
- College of Pharmacy Pusan National University Pusan 609‐735Korea
| | - Hae‐Young Chung
- College of Pharmacy Pusan National University Pusan 609‐735Korea
| | | | - Gyesoon Yoon
- Department of Biochemistry and Molecular BiologyAjou University School of Medicine Suwon 443‐721 Korea
| |
Collapse
|
35
|
Suh JH, Moreau R, Heath SHD, Hagen TM. Dietary supplementation with (R)-α-lipoic acid reverses the age-related accumulation of iron and depletion of antioxidants in the rat cerebral cortex. Redox Rep 2013; 10:52-60. [PMID: 15829111 DOI: 10.1179/135100005x21624] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Accumulation of divalent metal ions (e.g. iron and copper) has been proposed to contribute to heightened oxidative stress evident in aging and neurodegenerative disorders. To understand the extent of iron accumulation and its effect on antioxidant status, we monitored iron content in the cerebral cortex of F344 rats by inductively coupled plasma atomic emission spectrometry (ICP-AES) and found that the cerebral iron levels in 24-28-month-old rats were increased by 80% (p<0.01) relative to 3-month-old rats. Iron accumulation correlated with a decline in glutathione (GSH) and the GSH/GSSG ratio, indicating that iron accumulation altered antioxidant capacity and thiol redox state in aged animals. Because (R)-alpha-Lipoic acid (LA) is a potent chelator of divalent metal ions in vitro and also regenerates other antioxidants, we monitored whether feeding LA (0.2% [w/w]; 2 weeks) could lower cortical iron and improve antioxidant status. Results show that cerebral iron levels in old LA-fed animals were lower when compared to controls and were similar to levels seen in young rats. Antioxidant status and thiol redox state also improved markedly in old LA-fed rats versus controls. These results thus show that LA supplementation may be a means to modulate the age-related accumulation of cortical iron content, thereby lowering oxidative stress associated with aging.
Collapse
Affiliation(s)
- Jung H Suh
- Department Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, Corvallis, Oregon 97331, USA
| | | | | | | |
Collapse
|
36
|
Cho S, Hwang ES. Status of mTOR activity may phenotypically differentiate senescence and quiescence. Mol Cells 2012; 33:597-604. [PMID: 22570149 PMCID: PMC3887751 DOI: 10.1007/s10059-012-0042-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 03/26/2012] [Accepted: 03/26/2012] [Indexed: 10/28/2022] Open
Abstract
SA β-Gal activity is a key marker of cellular senescence. The origin of this activity is the lysosomal β-galactosidase, whose activity has increased high enough to be detected at suboptimal pH. SA β-Gal is also expressed in the cells in quiescence driven by serum-starvation or a high confluency, and it has been hypothesized that SA β-Gal positivity is rather a surrogate marker of high lysosome content or activity. In this study, it was determined how SA β-Gal activity is expressed in quiescence and how lysosome content and activities are differently maintained in senescence and quiescence using DNA damage-induced senescence and serum starvation-induced quiescence as study models. Lysosome content increased to facilitate SA β-Gal expression in both the conditions but with a big difference in the levels of the change. Lipofuscins whose accumulation leads to an increase in residual bodies also increased but with a smaller difference between the two conditions. Meanwhile, lysosome biogenesis was actively ongoing only in senescence progression, indicating that the difference in the lysosome contents may largely be due to lysosome biogenesis. Further, the cells undergoing senescence progression but not the ones in quiescence maintained high mTOR and low autophagy activities. Overall, the results indicate that, although SA β-Gal is expressed due to the elevated lysosome content in both cellular senescence and quiescence, senescence differs from quiescence with high lysosome biogenesis and low autophagy activity, and mTOR activity might be involved in these differences.
Collapse
Affiliation(s)
- Sohee Cho
- Department of Life Science, University of Seoul, Seoul 130-743,
Korea
| | - Eun Seong Hwang
- Department of Life Science, University of Seoul, Seoul 130-743,
Korea
| |
Collapse
|
37
|
Han J, Pan XY, Xu Y, Xiao Y, An Y, Tie L, Pan Y, Li XJ. Curcumin induces autophagy to protect vascular endothelial cell survival from oxidative stress damage. Autophagy 2012; 8:812-25. [PMID: 22622204 DOI: 10.4161/auto.19471] [Citation(s) in RCA: 207] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Our study first proposed that curcumin could protect human endothelial cells from the damage caused by oxidative stress via autophagy. Furthermore, our results revealed that curcumin causes some novel cellular mechanisms that promote autophagy as a protective effect. Pretreatment with curcumin remarkably improves the survival of human umbilical vein endothelial cells (HUVECs) from H 2O 2-induced viability loss, which specifically evokes an autophagic response. Exposed to H 2O 2, curcumin-treated HUVECs upregulate the level of microtubule-associated protein 1 light chain 3-II (LC3-II), the number of autophagosomes, and the degradation of p62. We show that this compound promotes BECN1 expression and inhibits the phosphatidylinositol 3-kinase (PtdIns3K)-AKT-mechanistic target of rapamycin (MTOR) signaling pathway. Curcumin can also reverse FOXO1 (a mediator of autophagy) nuclear localization along with causing an elevated level of cytoplasmic acetylation of FOXO1 and the interaction of acetylated FOXO1 and ATG7, under the circumstance of oxidative stress. Additionally, knockdown of FOXO1 by shRNA inhibits not only the protective effects that curcumin induced, but the autophagic process, from the quantity of LC3-II to the expression of RAB7. These results suggest that curcumin induces autophagy, indicating that curcumin has the potential for use as an autophagic-related antioxidant for prevention and treatment of oxidative stress. These data uncover a brand new protective mechanism involving FOXO1 as having a critical role in regulating autophagy in HUVECs, and suggest a novel role for curcumin in inducing a beneficial form of autophagy in HUVECs, which may be a potential multitargeted therapeutic avenue for the treatment of oxidative stress-related cardiovascular diseases.
Collapse
Affiliation(s)
- Jing Han
- State Key Laboratory of Natural and Biomimetic Drugs, and Department of Pharmacology. School of Basic Medical Sciences, Peking University, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Gómez LA, Hagen TM. Age-related decline in mitochondrial bioenergetics: does supercomplex destabilization determine lower oxidative capacity and higher superoxide production? Semin Cell Dev Biol 2012; 23:758-67. [PMID: 22521482 DOI: 10.1016/j.semcdb.2012.04.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 03/31/2012] [Accepted: 04/04/2012] [Indexed: 12/17/2022]
Abstract
Mitochondrial decay plays a central role in the aging process. Although certainly multifactorial in nature, defective operation of the electron transport chain (ETC) constitutes a key mechanism involved in the age-associated loss of mitochondrial energy metabolism. Primarily, mitochondrial dysfunction affects the aging animal by limiting bioenergetic reserve capacity and/or increasing oxidative stress via enhanced electron leakage from the ETC. Even though the important aging characteristics of mitochondrial decay are known, the molecular events underlying inefficient electron flux that ultimately leads to higher superoxide appearance and impaired respiration are not completely understood. This review focuses on the potential role(s) that age-associated destabilization of the macromolecular organization of the ETC (i.e. supercomplexes) may be important for development of the mitochondrial aging phenotype, particularly in post-mitotic tissues.
Collapse
Affiliation(s)
- Luis A Gómez
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | | |
Collapse
|
39
|
Cui J, Bai XY, Shi S, Cui S, Hong Q, Cai G, Chen X. Age-related changes in the function of autophagy in rat kidneys. AGE (DORDRECHT, NETHERLANDS) 2012; 34:329-39. [PMID: 21455601 PMCID: PMC3312632 DOI: 10.1007/s11357-011-9237-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2010] [Accepted: 03/10/2011] [Indexed: 05/14/2023]
Abstract
Autophagy is a highly regulated intracellular process for the degradation of cytoplasmic components, especially protein aggregates and damaged organelles. It is essential for maintaining healthy cells. Impaired or deficient autophagy is believed to cause or contribute to aging and age-related disease. In this study, we investigated the effects of age on autophagy in the kidneys of 3-, 12-, and 24-month-old Fischer 344 rats. The results revealed that autophagy-related gene (Atg)7 was significantly downregulated in kidneys of increasing age. The protein expression level of the autophagy marker light chain 3/Atg8 exhibited a marked decline in aged kidneys. The levels of p62/SQSTM1 and polyubiquitin aggregates, representing the function of autophagy and proteasomal degradation, increased in older kidneys. The level of 8-hydroxydeoxyguanosine, a marker of mitochondrial DNA oxidative damage, was also increased in older kidneys. Analysis by transmission electron microscope demonstrated swelling and disintegration of cristae in the mitochondria of aged kidneys. These results suggest that autophagic function decreases with age in the kidneys of Fischer 344 rats, and autophagy may mediate the process of kidney aging, leading to the accumulation of damaged mitochondria.
Collapse
Affiliation(s)
- Jing Cui
- Department of Nephrology, Chinese PLA Institute and Key Lab of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Xue-Yuan Bai
- Department of Nephrology, Chinese PLA Institute and Key Lab of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, Beijing, China
- Chinese PLA Institute of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, 28 Fuxing Road, Beijing, 100853 China
| | - Suozhu Shi
- Department of Nephrology, Chinese PLA Institute and Key Lab of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Shaoyuan Cui
- Department of Nephrology, Chinese PLA Institute and Key Lab of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Quan Hong
- Department of Nephrology, Chinese PLA Institute and Key Lab of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Guangyan Cai
- Department of Nephrology, Chinese PLA Institute and Key Lab of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, Beijing, China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA Institute and Key Lab of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, Beijing, China
- Chinese PLA Institute of Nephrology, Chinese PLA General Hospital and Military Medical Postgraduate College, 28 Fuxing Road, Beijing, 100853 China
| |
Collapse
|
40
|
Meng C, Zhou Z, Jiang K, Yu S, Jia L, Wu Y, Liu Y, Meng S, Ding C. Newcastle disease virus triggers autophagy in U251 glioma cells to enhance virus replication. Arch Virol 2012; 157:1011-8. [PMID: 22398914 PMCID: PMC7087167 DOI: 10.1007/s00705-012-1270-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2011] [Accepted: 01/23/2012] [Indexed: 12/23/2022]
Abstract
Newcastle disease virus (NDV) can replicate in tumor cells and induce apoptosis in late stages of infection. However, the interaction between NDV and cells in early stages of infection is not well understood. Here, we report that, shortly after infection, NDV triggers the formation of autophagosomes in U251 glioma cells, as demonstrated by an increased number of double-membrane vesicles, GFP-microtubule-associated protein 1 light chain 3 (GFP-LC3) a dot formations, and elevated production of LC3II. Moreover, modulation of NDV-induced autophagy by rapamycin, chloroquine or small interfering RNAs targeting the genes critical for autophagosome formation (Atg5 and Beclin-1) affects virus production, indicating that autophagy may be utilized by NDV to facilitate its own production. Furthermore, the class III phosphatidylinositol 3-kinase (PI3K)/Beclin-1 pathway plays a role in NDV-induced autophagy and virus production. Collectively, our data provide a unique example of a paramyxovirus that uses autophagy to enhance its production.
Collapse
Affiliation(s)
- Chunchun Meng
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Back JH, Zhu Y, Calabro A, Queenan C, Kim AS, Arbesman J, Kim AL. Resveratrol-mediated downregulation of Rictor attenuates autophagic process and suppresses UV-induced skin carcinogenesis. Photochem Photobiol 2012; 88:1165-72. [PMID: 22272775 DOI: 10.1111/j.1751-1097.2012.01097.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Macroautophagy is a cellular response to various environmental stresses that ensures lysosomal degradation of long-lived and damaged proteins and cellular organelles. It occurs through the formation of an autophagosome, which then fuses with a lysosome to form an autolysosome. Depending on the cellular context, autophagy may promote cancer cell survival or it may serve as a mechanism of tumor suppression. Herein, we show that resveratrol, a natural phytoalexin, induces premature senescence in human A431 SCC cells, and that resveratrol-induced premature senescence is associated with a blockade of autolysosome formation, as assessed by the absence of colocalization of LC3 and Lamp-2, markers for autophagosomes and lysosomes, respectively. Further, we show that resveratrol downregulates the level of Rictor, a component of mTORC2, leading to decreased RhoA-GTPase and altered actin cytoskeleton organization. Exogenous overexpression of Rictor restores RhoA-GTPase activity and actin cytoskeleton network, and decreases resveratrol-induced senescence-associated β-gal activity, indicating a direct role of Rictor in senescence induction. Rictor is overexpressed in UV-induced murine SCCs, whereas its expression is diminished by oral administration of resveratrol. These data indicate that resveratrol attenuates autophagic process via Rictor, and suggest that downregulation of Rictor may be a mechanism of tumor suppression associated with premature senescence.
Collapse
Affiliation(s)
- Jung H Back
- Department of Dermatology, Columbia University Medical Center, Russ Berrie Medical Science Pavilion, New York, NY, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Carreira RS, Lee P, Gottlieb RA. Mitochondrial therapeutics for cardioprotection. Curr Pharm Des 2012; 17:2017-35. [PMID: 21718247 DOI: 10.2174/138161211796904777] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 06/27/2011] [Indexed: 12/22/2022]
Abstract
Mitochondria represent approximately one-third of the mass of the heart and play a critical role in maintaining cellular function-however, they are also a potent source of free radicals and pro-apoptotic factors. As such, maintaining mitochondrial homeostasis is essential to cell survival. As the dominant source of ATP, continuous quality control is mandatory to ensure their ongoing optimal function. Mitochondrial quality control is accomplished by the dynamic interplay of fusion, fission, autophagy, and mitochondrial biogenesis. This review examines these processes in the heart and considers their role in the context of ischemia-reperfusion injury. Interventions that modulate mitochondrial turnover, including pharmacologic agents, exercise, and caloric restriction are discussed as a means to improve mitochondrial quality control, ameliorate cardiovascular dysfunction, and enhance longevity.
Collapse
Affiliation(s)
- Raquel S Carreira
- BioScience Center, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182-4650, USA
| | | | | |
Collapse
|
43
|
Abstract
Oxidative damage to cellular constituents has frequently been associated with aging in a wide range of organisms. The power of yeast genetics and biochemistry has provided the opportunity to analyse in some detail how reactive oxygen and nitrogen species arise in cells, how cells respond to the damage that these reactive species cause, and to begin to dissect how these species may be involved in the ageing process. This chapter reviews the major sources of reactive oxygen species that occur in yeast cells, the damage they cause and how cells sense and respond to this damage.
Collapse
Affiliation(s)
- May T Aung-Htut
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia,
| | | | | | | |
Collapse
|
44
|
Rezzani R, Stacchiotti A, Rodella LF. Morphological and biochemical studies on aging and autophagy. Ageing Res Rev 2012; 11:10-31. [PMID: 21939784 DOI: 10.1016/j.arr.2011.09.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 09/05/2011] [Accepted: 09/08/2011] [Indexed: 12/11/2022]
Abstract
To maintain health in the elderly is a crucial objective for modern medicine that involves both basic and clinical researches. Autophagy is a fundamental auto-cannibalizing process that preserves cellular homeostasis and, if altered, either by excess or defect, greatly changes cell fate and can result in incapacitating human diseases. Efficient autophagy may prolong lifespan, but unfortunately this process becomes less efficient with age. The present review is focused on the close relationship between autophagy and age-related disorders in different tissues/organs and in transgenic animal models. In particular, it comments on the up to date literature on mechanisms responsible for age-related impairment of autophagy. Moreover, before discussing about these mechanisms, it is necessary to describe the metabolic autophagic regulation of autophagy and the proteins involved in this process. At the end, these data would summarize the autophagic link with aging process, as important tools in the future biogerontology scenario.
Collapse
|
45
|
Abstract
During the development and normal function of T lymphocytes, the cells are subject to several checkpoints at which they must "decide" to live or die. At these critical times and during homeostasis, the molecules that regulate the classical apoptotic pathways and survival pathways such as autophagy have critical roles in controlling this decision. Our laboratory has focused on the roles of apoptotic and autophagic proteins in T lymphocyte development and function. Using genetic models in mice and in vitro analyses of T cell functions, we have outlined critical roles for the Bcl-2 family (regulators of the intrinsic pathway of apoptosis), c-FLIP (an anti-apoptotic protein in the extrinsic pathway of apoptosis), and autophagy in T lymphocytes.
Collapse
|
46
|
|
47
|
Shih H, Lee B, Lee RJ, Boyle AJ. The aging heart and post-infarction left ventricular remodeling. J Am Coll Cardiol 2011; 57:9-17. [PMID: 21185495 DOI: 10.1016/j.jacc.2010.08.623] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 08/17/2010] [Accepted: 08/24/2010] [Indexed: 12/28/2022]
Abstract
Aging is a risk factor for heart failure, which is a leading cause of death world-wide. Elderly patients are more likely than young patients to experience a myocardial infarction (MI) and are more likely to develop heart failure following MI. The poor clinical outcome of aging in cardiovascular disease is recapitulated on the cellular level. Increase in stress exposure and shifts in signaling pathways with age change the biology of cardiomyocytes. The progressive accumulation of metabolic waste and damaged organelles in cardiomyocytes blocks the intracellular recycling process of autophagy and increases the cell's propensity toward apoptosis. Additionally, the decreased cardiomyocyte renewal capacity in the elderly, due to reduction in cellular division and impaired stem cell function, leads to further cardiac dysfunction and maladaptive responses to disease or stress. We review the cellular and molecular aspects of post-infarction remodeling in the aged heart, and relate them to the clinical problem of post-infarction remodeling in elderly patients.
Collapse
Affiliation(s)
- Henry Shih
- Department of Medicine, Division of Cardiology, University of California San Francisco, San Francisco, California 94143, USA
| | | | | | | |
Collapse
|
48
|
Zanchetta LM, Garcia A, Lyng F, Walsh J, Murphy JEJ. Mitophagy and mitochondrial morphology in human melanoma-derived cells post exposure to simulated sunlight. Int J Radiat Biol 2011; 87:506-17. [PMID: 21381890 DOI: 10.3109/09553002.2011.556175] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE To assess changes in mitochondrial morphology and mitophagy induced by simulated sunlight irradiation (SSI) and how these changes are modulated by mitochondrial activity and energy source. MATERIALS AND METHODS Human malignant amelanotic melanoma A375 cells were pre-treated with either a mitochondrial activity enhancer, uncoupler or were either melanin or glutamine supplemented/starved for 4 hours pre-exposure to sunlight. A Q-Sun Solar Simulator (Q-Lab, Homestead, FL, USA) was employed to expose cells to simulated sunlight. Confocal microscopy imaging of A375 cells co-loaded with mitochondria and lysosome-specific fluorescent dyes was used to identify these organelles and predict mitophagic events. RESULTS SSI induces pronounced changes in mitochondrial dynamics and mitophagy in exposed skin cells compared to control and these effects were modified by both glutamine and melanin. CONCLUSIONS Mitochondrial dynamics and rate of mitophagy in melanoma cells are sensitive to even short bursts of environmentally relevant SSI. Mitochondrial dynamics, and its modulation, may also play a role in mitophagy regulation, cell survival and proliferation post SSI.
Collapse
Affiliation(s)
- Luciene M Zanchetta
- Mitochondrial Biology & Radiation Research, School of Science, Institute of Technology Sligo, Ash Lane, Sligo, Ireland.
| | | | | | | | | |
Collapse
|
49
|
Zhao H, Halicka HD, Traganos F, Jorgensen E, Darzynkiewicz Z. New biomarkers probing depth of cell senescence assessed by laser scanning cytometry. Cytometry A 2011; 77:999-1007. [PMID: 20939035 PMCID: PMC2977923 DOI: 10.1002/cyto.a.20983] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The imaging analytical capabilities of laser scanning cytometer (LSC) have been used to assess morphological features considered to be typical of the senescent phenotype. The characteristic “flattening” of senescent cells was reflected by the decline in the density of staining (intensity of maximal pixel) of DNA-associated fluorescence [4,6-diamidino-2-phenylindole (DAPI)] paralleled by an increase in nuclear size (area). The decrease in ratio of maximal pixel to nuclear area was even more sensitive senescence biomarker than the change in maximal pixel or nuclear area, each alone. The saturation cell density at plateau phase of growth recorded by LSC was found to be dramatically decreased in cultures of senescent cells, thereby also serving as an additional marker. The induction of cyclin dependent kinase inhibitors p21WAF1 and p27KIP1 and γH2AX and activation of ATM markers of DNA damage response were measured in parallel with DNA/DAPI maximal pixel and nuclear area. These biomarker indices were expressed in quantitative terms by reporting them as a fraction of the respective controls. The effect of treatment of A549 and WI-38 cells with different concentrations of mitoxantrone (Mxt) and trichostatin A for various time periods was studied to assess the degree (depth) of cell senescence. Also assessed was the effect of 2-deoxy-d-glucose, the agent attenuating metabolic cell activity, on the depth of senescence induced by Mxt. A relationship between the ability of cells to synthesize RNA (incorporate 5-ethynyluridine) that leads to growth imbalance and induction of cell senescence was also studied. The data show that morphometric analysis of cellular attributes by LSC offers an attractive tool to detect cell senescence and measure its degree particularly in assessing effects of the factors that enhance or attenuate this process. This methodology is of importance in light of the evidence that cellular senescence is not only a biological process that is fundamental for organismal aging but also impedes formation of induced-pluripotent stem cells providing the barrier for neoplastic transformation and is the major mechanism of induction of reproductive cell death during treatment of solid tumors. © 2010 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Hong Zhao
- Department of Pathology, Brander Cancer Research Institute, New York Medical College, Valhalla, New York 10595, USA
| | | | | | | | | |
Collapse
|
50
|
Abstract
Decreased expression of vascular endothelial growth factor (VEGF) in the renal tubules is thought to cause progressive loss of the renal microvasculature with age. Mitochondrial dysfunction may be a principal phenomenon underlying the process of aging. The relation between VEGF expression and mitochondrial dysfunction in aging is not fully understood. We hypothesized that mitochondrial dysfunction blocks VEGF expression and contributes to impaired angiogenesis in the aging kidney. The aim of this study was to assess the role of mitochondria in VEGF expression in the aging rat kidney. We evaluated the accumulation of 8-hydroxy-2'-deoxyguanosine in mitochondrial DNA, as well as mitochondrial dysfunction, as assessed by electron microscopy of mitochondrial structure and histochemical staining for respiratory chain complex IV, in aging rat kidney. An increase in hypoxic area and a decrease in peritubular capillaries were detected in the cortex of aging rat kidneys; however, upregulation of VEGF expression was not observed. The expression of VEGF in proximal tubular epithelial cells in response to hypoxia was suppressed by the mitochondrial electron transfer inhibitor myxothiazol. Mitochondrial DNA-deficient cells also failed to upregulate VEGF expression under hypoxic conditions. These results indicate that impairment of VEGF upregulation, possibly as a result of mitochondrial dysfunction, contributes to impaired angiogenesis, which in turn leads to renal injury in the aging rat kidney.
Collapse
|