Mitochondria and Ageing: Comparison
Please note this is a comparison between Version 2 by Vicky Zhou and Version 1 by Michael Webb.

Declining mitochondrial function, reflected in defects in ATP synthesis and increased generation of toxic reactive oxygen species is a universal feature of natural ageing. It accompanies the other hallmarks of ageing which include progressive loss of function in multiple organs, sarcopenia and increasing maladaptive low-grade inflammation. These end in death, which is a cumulative result of loss of function, leading to either increased vulnerability to environmental hazards such as predation and disease or to failure of critical organ systems such as the heart, liver or kidney. Several processes that may contribute mechanistically to age related degeneration have been identified, including oxidative damage, accumulation of toxic protein aggregates, autoinflammatory processes, loss of stem cell populations and an increasing load of malfunctional senescent cells.  Mitochondrial dysfunction has connections with each of these processes.  The following is a brief overview of some of these connections.

  • Mitochondria
  • Ageing
  • Biochemistry
  • Molecular Biology
  • Mitochondrial Quality Control
  • Free Radical
  • Mutation
  • Cell Fate
  • Senesence
Please wait, diff process is still running!

References

  1. Taanman, J.W. The mitochondrial genome: Structure, transcription, translation and replication. Biochim. Biophys. Acta 1999, 1410, 103–123.
  2. Chinnery, P.F.; Hudson, G. Mitochondrial genetics. Br. Med. Bull. 2013, 106, 135–159.
  3. Short, K.R.; Bigelow, M.L.; Kahl, J.; Singh, R.; Coenen-Schimke, J.; Raghavakaimal, S.; Nair, K.S. Decline in skeletal muscle mitochondrial function with aging in humans. Proc. Natl. Acad. Sci. USA 2005, 102, 5618–15623.
  4. Stauch, K.L.; Purnell, P.R.; Fox, H.S. Aging synaptic mitochondria exhibit dynamic proteomic changes while maintaining bioenergetic function. Aging (Albany NY) 2014, 6, 320–334.
  5. Zahn, J.M.; Sonu, R.; Vogel, H.; Crane, E.; Mazan-Mamczarz, K.; Rabkin, R.; Davis, R.W.; Becker, K.G.; Owen, A.B.; Kim, S.K. Transcriptional profiling of aging in human muscle reveals a common aging signature. PLoS Genet. 2006, 2, e115.
  6. Terman, A. Catabolic insufficiency and aging. Ann. NY Acad. Sci. 2006, 1067, 27–36.
  7. Beckman, K.B.; Ames, B.N. Endogenous oxidative damage of mtDNA. Mutat. Res. 1999, 424, 51–58.
  8. Radzvilavicius, A.L.; Hadjivasiliou, Z.; Pomiankowski, A.; Lane, N. Selection for Mitochondrial Quality Drives Evolution of the Germline. PLoS Biol. 2016, 14, e2000410.
  9. Ziada, A.S.; Lu, M.Y.; Ignas-Menzies, J.; Paintsil, E.; Li, M.; Ogbuagu, O.; Saberi, S.; Hsieh, A.Y.Y.; Sattha, B.; Harrigan, P.R.; et al. Mitochondrial DNA somatic mutation burden and heteroplasmy are associated with chronological age, smoking, and HIV infection. Aging Cell 2019, 18, e13018.
  10. Reichart, G.; Mayer, J.; Tokay, T.; Lange, F.; Johne, C.; Baltrusch, S.; Tiedge, M.; Fuellen, G.; Ibrahim, S.; Köhling, R. Combination of mitochondrial tRNA and OXPHOS mutation reduces lifespan and physical condition in aged mice. bioRxiv 2017, 233593, doi:10.1101/233593.
  11. Fan, W.; Evans, R. PPARs and ERRs: Molecular mediators of mitochondrial metabolism. Curr. Opin. Cell Biol. 2015, 33, 49–54.
  12. Li, F.; Wang, Y.; Zeller, K.I.; Potter, J.J.; Wonsey, D.R.; OʼDonnell, K.A.; Kim, J.W.; Yustein, J.T.; Lee, L.A.; Dang, C.V. Myc stimulates nuclearly encoded mitochondrial genes and mitochondrial biogenesis. Mol. Cell Biol. 2005, 25, 6225–6234.
  13. Dhar, S.S.; Ongwijitwat, S.; Wong-Riley, M.T. Nuclear respiratory factor 1 regulates all ten nuclear-encoded subunits of cytochrome c oxidase in neurons. J. Biol. Chem. 2008, 283, 3120–3129.
  14. Salminen, A.; Kaarniranta, K. AMP- activated protein kinase (AMPK) controls the aging process via an integrated signaling network. Ageing Res. Rev. 2012, 11, 230–241.
  15. Salminen, A.; Kaarniranta, K.; Kauppinen, A. Age-related changes in AMPK activation: Role for AMPK phosphatases and inhibitory phosphorylation by upstream signaling pathways. Ageing Res. Rev. 2016, 28, 15–26.
  16. Qiang, W.; Weiqiang, K.; Qing, Z.; Pengju, Z.; Yi, L. Aging impairs insulin-stimulated glucose uptake in rat skeletal muscle via suppressing AMPKalpha. Exp. Mol. Med. 2007, 39, 53543.
  17. Reznick, R.M.; Zong, H.; Li, J.; Morino, K.; Moore, I.K.; Yu, H.J.; Liu, Z.X.; Dong, J.; Mustard, K.J.; Hawley, S.A.; et al. Aging-associated reductions in AMP-activated protein kinase activity and mitochondrial biogenesis. Cell Metab. 2007, 5, 151–156.
  18. Beyer, R.E.; Burnett, B.A.; Cartwright, K.J.; Edington, D.W.; Falzon, M.J.; Kreitman, K.R.; Kuhn, T.W.; Ramp, B.J.; Rhee, S.Y.; Rosenwasser, M.J.; et al. Tissue coenzyme Q (ubiquinone) and protein concentrations over the life span of the laboratory rat. Mech. Ageing Dev. 1985, 32, 267–281.
  19. Kalen, A.; Appelkvist, E.L.; Dallner, G. Age-related changes in the lipid compositions of rat and human tissues. Lipids 1989, 24, 579–584.
  20. Mailloux, R.J.; Beriault, R.; Lemire, J.; Singh, R.; Chenier, D.R.; Hamel, R.D.; Appanna, V.D. The tricarboxylic acid cycle, an ancient metabolic network with a novel twist. PLoS ONE 2007, 2, e690.
  21. Zdzisinska, B.; Zurek, A.; Kandefer-Szerszen, M. Alpha-Ketoglutarate as a Molecule with Pleiotropic Activity: Well-Known and Novel Possibilities of Therapeutic Use. Arch. Immunol. Ther. Exp. (Warsz.) 2017, 65, 21–36.
  22. Salminen, A.; Kauppinen, A.; Hiltunen, M.; Kaarniranta, K. Krebs cycle intermediates regulate DNA and histone methylation: Epigenetic impact on the aging process. Ageing Res. Rev. 2014, 16, 45–65.
  23. He, W.; Miao, F.J.; Lin, D.C.; Schwandner, R.T.; Wang, Z.; Gao, J.; Chen, J.L.; Tian, H.; Ling, L. Citric acid cycle intermediates as ligands for orphan G-protein-coupled receptors. Nature 2004, 429, 188–193.
  24. Gilissen, J.; Jouret, F.; Pirotte, B.; Hanson, J. Insight into SUCNR1 (GPR91) structure and function. Pharmacol. Ther. 2016, 159, 56–65.
  25. Hu, J.; Li, T.; Du, X.; Wu, Q.; Le, Y.Z. G protein-coupled receptor 91 signaling in diabetic retinopathy and hypoxic retinal diseases. Vis. Res. 2017, 139, 59–64.
  26. Zhu, X.H.; Lu, M.; Lee, B.Y.; Ugurbil, K.; Chen, W. In vivo NAD assay reveals the intracellular NAD contents and redox state in healthy human brain and their age dependences. Proc. Natl. Acad. Sci. USA 2015, 112, 2876–2881.
  27. Zhang, H.; Ryu, D.; Wu, Y.; Gariani, K.; Wang, X.; Luan, P.; DʼAmico, D.; Ropelle, E.R.; Lutolf, M.P.; Aebersold, R.; et al. NAD(+) repletion improves mitochondrial and stem cell function and enhances life span in mice. Science 2016, 352, 1436–1443.
  28. Mota, R.A.; Sanchez-Bueno, F.; Saenz, L.; Hernandez-Espinosa, D.; Jimeno, J.; Tornel, P.L.; Martinez-Torrano, A.; Ramirez, P.; Parrilla, P.; Yelamos, J. Inhibition of poly(ADP-ribose) polymerase attenuates the severity of acute pancreatitis and associated lung injury. Lab. Investig. 2005, 85, 1250–1262.
  29. Grube, K.; Burkle, A. Poly(ADP-ribose) polymerase activity in mononuclear leukocytes of 13 mammalian species correlates with species-specific life span. Proc. Natl. Acad. Sci. USA 1992, 89, 11759–11763.
  30. Muiras, M.L.; Muller, M.; Schachter, F.; Burkle, A. Increased poly(ADP-ribose) polymerase activity in lymphoblastoid cell lines from centenarians. J. Mol. Med. (Berl.) 1998, 76, 346–354.
  31. Chini, E.N. CD38 as a regulator of cellular NAD: a novel potential pharmacological target for metabolic conditions. Curr. Pharm. Des. 2009, 15, 57–63.
  32. Amici, S.A.; Young, N.A.; Narvaez-Miranda, J.; Jablonski, K.A.; Arcos, J.; Rosas, L.; Papenfuss, T.L.; Torrelles, J.B.; Jarjour, W.N.; Guerau-De-Arellano, M. CD38 Is Robustly Induced in Human Macrophages and Monocytes in Inflammatory Conditions. Front. Immunol. 2018, 9, 1593.
  33. Braidy, N.; Poljak, A.; Grant, R.; Jayasena, T.; Mansour, H.; Chan-Ling, T.; Guillemin, G.J.; Smythe, G.; Sachdev, P.S. Mapping NAD(+) metabolism in the brain of ageing Wistar rats: Potential targets for influencing brain senescence. Biogerontology 2014, 15, 177–198.
  34. Camacho-Pereira, J.; Tarrago, M.G.; Chini, C.C.S.; Nin, V.; Escande, C.; Warner, G.M.; Puranik, A.S.; Schoon, R.A.; Reid, J.M.; Galina, A.; et al. CD38 Dictates Age-Related NAD Decline and Mitochondrial Dysfunction through an SIRT3-Dependent Mechanism. Cell Metab. 2016, 23, 1127–1139.
  35. Kim, H.J.; Kim, K.W.; Yu, B.P.; Chung, H.Y. The effect of age on cyclooxygenase-2 gene expression: NF-kappaB activation and IkappaBalpha degradation. Free. Radic. Biol. Med. 2000, 28, 683–692.
  36. Lumeng, C.N.; Liu, J.; Geletka, L.; Delaney, C.; DelProposto, J.; Desai, A.; Oatmen, K.; Martinez-Santibanez, G.; Julius, A.; Garg, S.; et al. Aging Is Associated with an Increase in T Cells and Inflammatory Macrophages in Visceral Adipose Tissue. J. Immunol. 2011, 187, 6208–6216.
  37. Hearps, A.C.; Martin, G.E.; Angelovich, T.A.; Cheng, W.J.; Maisa, A.; Landay, A.L.; Jaworowski, A.; Crowe, S.M. Aging is associated with chronic innate immune activation and dysregulation of monocyte phenotype and function. Aging Cell 2012, 11, 867–875.
  38. Jurk, D.; Wilson, C.; Passos, J.F.; Oakley, F.; Correia-Melo, C.; Greaves, L.; Saretzki, G.; Fox, C.; Lawless, C.; Anderson, R.; et al. Chronic inflammation induces telomere dysfunction and accelerates ageing in mice. Nat. Commun. 2014, 2, 4172.
  39. Rodier, F.; Coppé, J.P.; Patil, C.K.; Hoeijmakers, W.A.; Muñoz, D.P.; Raza, S.R.; Freund, A.; Campeau, E.; Davalos, A.R.; Campisi, J. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat. Cell Biol. 2009, 11, 973–979.
  40. Tarragó, M.G.; Chini, C.C.S.; Kanamori, K.S.; Warner, G.M.; Caride, A.; De Oliveira, G.C.; Rud, M.; Samani, A.; Hein, K.Z.; Huang, R.; et al. A Potent and Specific CD38 Inhibitor Ameliorates Age-Related Metabolic Dysfunction by Reversing Tissue NAD+ Decline. Cell Metab. 2018, 27, 1081–1095.e10.
  41. Schlame, M.; Greenberg, M.L. Biosynthesis, remodeling and turnover of mitochondrial cardiolipin. Biochim. Biophys. Acta 2016, 1, 3–7, doi:10.1016/j.bbalip.2016.08.010.
  42. Kagan, V.E.; Chu, C.T.; Tyurina, Y.Y.; Cheikhi, A.; Bayir, H. Cardiolipin asymmetry, oxidation and signaling. Chem. Phys. Lipids 2014, 179, 64–69, doi:10.1016/j.chemphyslip.2013.11.010.
  43. Paradies, G.; Petrosillo, G.; Pistolese, M.; Ruggiero, F.M. The effect of reactive oxygen species generated from the mitochondrial electron transport chain on the cytochrome c oxidase activity and on the cardiolipin content in bovine heart submitochondrial particles. FEBS Lett. 2000, 466, 323–326.
  44. Paradies, G.; Petrosillo, G.; Pistolese, M.; Ruggiero, F.M. Reactive oxygen species generated by the mitochondrial respiratory chain affect the complex III activity via cardiolipin peroxidation in beef-heart submitochondrial particles. Mitochondrion 2001, 1, 151–159.
  45. Chu, C.T.; Ji, J.; Dagda, R.K.; Jiang, J.F.; Tyurina, Y.Y.; Kapralov, A.A.; Tyurin, V.A.; Yanamala, N.; Shrivastava, I.H.; Mohammadyani, D.; et al. Cardiolipin externalization to the outer mitochondrial membrane acts as an elimination signal for mitophagy in neuronal cells. Nature 2013, 15, 1197–1205.
  46. Petrosillo, G.; Casanova, G.; Matera, M.; Ruggiero, F.M.; Paradies, G. Interaction of peroxidized cardiolipin with rat-heart mitochondrial membranes: Induction of permeability transition and cytochrome c release. FEBS Lett. 2006, 580, 6311–6316.
  47. Schug, Z.T.; Gottlieb, E. Cardiolipin acts as a mitochondrial signaling platform to launch apoptosis. Biochim. Biophys. Acta (BBA) Biomembr. 2009, 1788, 2022–2031.
  48. Sen, T.; Sen, N.; Jana, S.; Khan, F.H.; Chatterjee, U.; Chakrabarti, S. Depolarization and cardiolipin depletion in aged rat brain mitochondria: Relationship with oxidative stress and electron transport chain activity. Neurochem. Int. 2007, 50, 719–725.
  49. Vorbeck, M.L.; Martin, A.P.; Long, J.W., Jr.; Smith, J.M.; Orr, R.R., Jr. Aging-dependent modification of lipid composition and lipid structural order parameter of hepatic mitochondria. Arch. Biochem. Biophys. 1982, 217, 351–361.
  50. Paradies, G.; Ruggiero. F.M.; Petrosillo, G.; Quagliariello, E. Age-dependent decline in the cytochrome c oxidase activity in rat heart mitochondria: Role of cardiolipin. FEBS Lett. 1997, 406, 136–138.
  51. Harman, D. Aging: A theory based on free radical and radiation chemistry. J. Gerontol. 1956, 11, 298–300.
  52. Harman, D. The biologic clock: The mitochondria? J. Am. Geriatr. Soc. 1972, 20, 145–147.
  53. Reutzel, M.; Grewal, R.; Dilberger, B.; Silaidos, C.; Joppe, A.; Eckert, G. P. Cerebral Mitochondrial Function and Cognitive Performance during Aging: A Longitudinal Study in NMRI Mice. Oxid. Med. Cell Longev. 2020, 4060769.
  54. Cand, F.; Verdetti, J. Superoxide dismutase, glutathione peroxidase, catalase, and lipid peroxidation in the major organs of the aging rats. Free Radic. Biol. Med. 1989, 7, 59–63.
  55. Lewis, K.N.; Andziak, B.; Yang, T.; Buffenstein, R. The naked mole-rat response to oxidative stress: Just deal with it. Antioxid. Redox Signal. 2013, 19, 1388–1399.
  56. Yang, W.; Li, J.; Hekimi, S. A Measurable increase in oxidative damage due to reduction in superoxide detoxification fails to shorten the life span of long-lived mitochondrial mutants of Caenorhabditis elegans. Genetics 2007, 177, 2063–2074.
  57. Van Raamsdonk, J.M.; Hekimi, S. Superoxide dismutase is dispensable for normal animal lifespan. Proc. Natl. Acad. Sci. USA 2012, 109, 5785–5790.
  58. Doonan, R.; McElwee, J.J.; Matthijssens, F.; Walker, G.A.; Houthoofd, K.; Back, P.; Matscheski, A.; Vanfleteren, J.R.; Gems, D. Against the oxidative damage theory of aging: superoxide dismutases protect against oxidative stress but have little or no effect on life span in Caenorhabditis elegans. Genes Dev. 2008, 22, 3236–3241.
  59. Ristow, M.; Schmeisser, S. Extending life span by increasing oxidative stress. Free Radic. Biol. Med. 2011, 51, 327–336.
  60. Yang, W.; Hekimi, S. Two modes of mitochondrial dysfunction lead independently to lifespan extension in Caenorhabditis elegans. Aging Cell 2010, 9, 433–447.
  61. Schulz, T.J.; Zarse, K.; Voigt, A.; Urban, N.; Birringer, M.; Ristow, M. Glucose restriction extends Caenorhabditis elegans life span by inducing mitochondrial respiration and increasing oxidative stress. Cell Metab. 2007, 6, 280–293.
  62. Fouquerel, E.; Barnes, R.P.; Uttam, S.; Watkins, S.C.; Bruchez, M.P.; Opresko, P.L. Targeted and Persistent 8-Oxoguanine Base Damage at Telomeres Promotes Telomere Loss and Crisis. Mol. Cell 2019, 75, 117e6–130e6.
  63. Passos, J.F.; Saretzki, G.; Ahmed, S.; Nelson, G.; Richter, T.; Peters, H.; Wappler, I.; Birket, M.J.; Harold, G.; Schaeuble, K.; et al. Mitochondrial dysfunction accounts for the stochastic heterogeneity in telomere-dependent senescence. PLoS Biol. 2007, 5, e110.
  64. Bazopoulou, D.; Knoefler, D.; Zheng, Y.; Ulrich, K.; Oleson, B.J.; Xie, L.; Kim, M.; Kaufmann, A.; Lee, Y.T.; Dou, Y.; et al. Developmental ROS individualizes organismal stress resistance and lifespan. Nature 2019, 576, 301–305.
  65. Tapia, P.C. Sublethal mitochondrial stress with an attendant stoichiometric augmentation of reactive oxygen species may precipitate many of the beneficial alterations in cellular physiology produced by caloric restriction, intermittent fasting, exercise and dietary phytonutrients: “Mitohormesis” for health and vitality. Med. Hypotheses 2006, 66, 832–843.
  66. Van Zant, G.; Liang, Y. The role of stem cells in aging. Exp. Hematol. 2003, 31, 659–672.
  67. Anso, E.; Weinberg, S.E.; Diebold, L.P.; Thompson, B.J.; Malinge, S.; Schumacker, P.T.; Liu, X.; Zhang, Y.; Shao, Z.; Steadman, M.; et al. The mitochondrial respiratory chain is essential for haematopoietic stem cell function. Nat. Cell Biol. 2017, 19, 614–625.
  68. Khacho, M.; Clark, A.; Svoboda, D.S.; Azzi, J.; MacLaurin, J.G.; Meghaizel, C.; Sesaki, H.; Lagace, D.C.; Germain, M.; Harper, M.E.; et al. Mitochondrial Dynamics Impacts Stem Cell Identity and Fate Decisions by Regulating a Nuclear Transcriptional Program. Cell Stem Cell 2016, 19, 232–247.
  69. Mohrin, M.; Shin, J.; Liu, Y.; Brown, K.; Luo, H.; Xi, Y.; Haynes, C.M.; Chen, D. Stem cell aging. A mitochondrial UPR-mediated metabolic checkpoint regulates hematopoietic stem cell aging. Science 2015, 347, 1374–1377.
  70. Williams, N.C.; O'Neill, L.A.J. A Role for the Krebs Cycle Intermediate Citrate in Metabolic Reprogramming in Innate Immunity and Inflammation. Front. Immunol. 2018, 9, 141.
  71. Rafalski, V.A.; Mancini, E.; Brunet, A. Energy metabolism and energy-sensing pathways in mammalian embryonic and adult stem cell fate. J. Cell Sci. 2012, 125, 5597–608.
  72. Zheng, X.; Boyer, L.; Jin, M.; Mertens, J.; Kim, Y.; Ma, L.; Ma, L.; Hamm, M.; Gage, F.H.; Hunter, T. Metabolic reprogramming during neuronal differentiation from aerobic glycolysis to neuronal oxidative phosphorylation. Elife 2016, 5, e13374.
  73. Inoue, S.; Noda, S.; Kashima, K.; Nakada, K.; Hayashi, J.; Miyoshi, H. Mitochondrial respiration defects modulate differentiation but not proliferation ofhematopoietic stem and progenitor cells. FEBS Lett. 2010, 584, 3402–3409.
  74. Katajisto, P.; Döhla, J.; Chaffer, C.L.; Pentinmikko, N.; Marjanovic, N.; Iqbal, S.; Zoncu, R.; Chen, W.W.; Weinberg, R.A.; Sabatini, D.M. Asymmetric apportioning of aged mitochondria between daughter cells is required for stemness. Science 2015, 348, 340–343.
  75. Prattichizzo, F.; Giuliani, A.; Recchioni, R.; Bonafè, M.; Marcheselli, F.; De Carolis, S.; Campanati, A.; Giuliodori, K.; Rippo, M.R.; Brugè, F.; et al. Anti-TNF-α treatment modulates SASP and SASP-related microRNAs in endothelial cells and in circulating angiogenic cells. Oncotarget 2016, 7, 11945–11958.
  76. Rea, I.M.; Gibson, D.S.; McGilligan, V.; McNerlan, S.E.; Alexander, H.D.; Ross, O.A. Age and Age-Related Diseases: Role of Inflammation Triggers and Cytokines. Front. Immunol. 2018, 9, 586.
  77. Hernandez-Segura, A.; Brandenburg, S.; Demaria, M. Induction and Validation of Cellular Senescence in Primary Human Cells. J. Vis. Exp. 2018, 136, 55782.
  78. Millerand, M.; Berenbaum, F.; Jacques, C. Danger signals and inflammaging in osteoarthritis. Clin. Exp. Rheumatol. 2019, 3, 48–56.
  79. Nie, L.; Zhang, P.; Wang, Q.; Zhou, X.; Wang, Q. lncRNA—Triggered Macrophage Inflammaging Deteriorates Age-Related Diseases. Mediat. Inflamm. 2019, 2019, 4260309.
  80. Yoon, Y.S.; Byun, H.O.; Cho, H.; Kim, B.K.; Yoon, G. Complex II defect via down-regulation of iron-sulfur subunit induces mitochondrial dysfunction and cell cycle delay in iron chelation-induce senescence—Associated growth arrest. J. Biol. Chem. 2003, 278, 51577–51586.
  81. Passos, J.F.; Nelson, G.; Wang, C.; Richter, T.; Simillion, C.; Proctor, C.J.; Miwa, S.; Olijslagers, S.; Hallinan, J.; Wipat, A.; et al. Feedback between p21 and reactive oxygen production is necessary for cell senescence. Mol. Syst. Biol. 2010, 6, 347.
  82. Zheng, H.; Huang, Q.; Huang, S.; Yang, X.; Zhu, T.; Wang, W.; Wang, H.; He, S.; Ji, L.; Wang, Y.; et al. Senescence Inducer Shikonin ROS-Dependently Suppressed Lung Cancer Progression. Front. Pharmacol. 2018, 9, 519.
  83. Aarreberg, L.D.; Esser-Nobis, K.; Driscoll, C.; Shuvarikov, A.; Roby, J.A.; Gale, M., Jr. Interleukin-1beta Induces mtDNA Release to Activate Innate Immune Signaling via cGAS-STING. Mol Cell 2019, 74, 801 e6–815 e6.
  84. Pinti, M.; Cevenini, E.; Nasi, M.; De Biasi, S.; Salvioli, S.; Monti, D.; Benatti, S.; Gibellini, L.; Cotichini, R.; Stazi, M.A.; et al. Circulating mitochondrial DNA increases with age and is a familiar trait: Implications for “inflamm-aging”. Eur. J. Immunol. 2014, 44, 1552–1162.
  85. Jylhava, J.; Nevalainen, T.; Marttila, S.; Jylha, M.; Hervonen, A.; Hurme, M. Characterization of the role of distinct plasma cell-free DNA species in age-associated inflammation and frailty. Aging Cell 2013, 12, 388–397.
  86. Loson, O.C.; Song, Z.; Chen, H.; Chan, D.C. Fis1, Mff, MiD49, and MiD51 mediate Drp1 recruitment in mitochondrial fission. Mol. Biol. Cell 2013, 24, 659–667.
  87. Liu, R.; Chan, D.C. The mitochondrial fission receptor Mff selectively recruits oligomerized Drp1. Mol. Biol. Cell 2015, 26, 4466–4477.
  88. Hall, A.R.; Burke, N.; Dongworth, R.K.; Hausenloy, D.J. Mitochondrial fusion and fission proteins: Novel therapeutic targets for combating cardiovascular disease. Br. J. Pharmacol. 2014, 171, 1890–1906.
  89. Byrne, J.J.; Soh, M.S.; Chandhok, G.; Vijayaraghavan, T.; Teoh, J.S.; Crawford, S.; Cobham, A.E.; Yapa, N.M.B.; Mirth, C.K.; Neumann, B. Disruption of mitochondrial dynamics affects behaviour and lifespan in Caenorhabditis elegans. Cell Mol. Life Sci. 2019, 76, 1967–1985.
  90. Bernhardt, D.; Müller, M.; Reichert, A.S.; Osiewacz, H.D. Simultaneous impairment of mitochondrial fission and fusion reduces mitophagy and shortens replicative lifespan. Sci. Rep. 2015, 5, 7885.
  91. Youle, R.J.; Narendra, D.P. Mechanisms of mitophagy. Nat. Rev. Mol. Cell Biol. 2011, 12, 9–14.
  92. Sun, N.; Yun, J.; Liu, J.; Malide, D.; Liu, C.; Rovira, II; Holmstrom, K.M.; Fergusson, M.M.; Yoo, Y.H.; Combs, C.A.; et al. Measuring In Vivo Mitophagy. Mol. Cell 2015, 60, 685–696.
  93. McWilliams, T.G.; Prescott, A.R.; Allen, G.F.; Tamjar, J.; Munson, M.J.; Thomson, C.; Muqit, M.M.; Ganley, I.G. mito-QC illuminates mitophagy and mitochondrial architecture in vivo. J. Cell Biol. 2016, 214, 333–345.
  94. Song, M.; Franco, A.; Fleischer, J.A.; Zhang, L.; Dorn, G.W. 2nd, Abrogating Mitochondrial Dynamics in Mouse Hearts Accelerates Mitochondrial Senescence. Cell Metab. 2017, 26, 872 e5–883 e5.
More
Video Production Service