Hostname: page-component-8448b6f56d-mp689 Total loading time: 0 Render date: 2024-04-19T16:11:22.179Z Has data issue: false hasContentIssue false

Autophagy of naïve CD4+ T cells in aging – the role of body adiposity and physical fitness

Published online by Cambridge University Press:  19 January 2023

Camila S. Padilha*
Affiliation:
Exercise and Immunometabolism Research Group, Post-graduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, São Paulo, Brazil
Mehdi Kushkestani
Affiliation:
Wellness and Lifestyle Science Initiative Group, PhD Student of Kinesiology and Health, School of art and science, Rutgers University New Brunswick, New Jersey, USA
Liliana P. Baptista
Affiliation:
Department of Medicine, Division of Gerontology, Geriatrics, and Palliative Care Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, USA Research Center in Physical Activity, Health, and Leisure (CIAFEL), Faculty of Sports, University of Porto (FADEUP), Portugal Laboratory for Integrative and Translational Research in Population Health (ITR), Porto, Portugal
Karsten Krüger
Affiliation:
Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, University of Giessen, Giessen, Germany
Fábio Santos Lira*
Affiliation:
Exercise and Immunometabolism Research Group, Post-graduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, São Paulo, Brazil Research Center for Sport and Physical Activity, Faculty of Sports Science and Physical Education, University of Coimbra, Coimbra, Portugal
*
Authors for correspondence: Camila S. Padilha, E-mail: cs.padilha@unesp.br; Fábio Santos Lira, E-mail: fabio.lira@unesp.br
Authors for correspondence: Camila S. Padilha, E-mail: cs.padilha@unesp.br; Fábio Santos Lira, E-mail: fabio.lira@unesp.br

Abstract

Life expectancy has increased exponentially in the last century accompanied by disability, poor quality of life, and all-cause mortality in older age due to the high prevalence of obesity and physical inactivity in older people. Biologically, the aging process reduces the cell’s metabolic and functional efficiency, and disrupts the cell’s anabolic and catabolic homeostasis, predisposing older people to many dysfunctional conditions such as cardiovascular disease, neurodegenerative disorders, cancer, and diabetes. In the immune system, aging also alters cells' metabolic and functional efficiency, a process known as ‘immunosenescence’, where cells become more broadly inflammatory and their functionality is altered. Notably, autophagy, the conserved and important cellular process that maintains the cell’s efficiency and functional homeostasis may protect the immune system from age-associated dysfunctional changes by regulating cell death in activated CD4+ T cells. This regulatory process increases the delivery of the dysfunctional cytoplasmic material to lysosomal degradation while increasing cytokine production, proliferation, and differentiation of CD4+ T cell-mediated immune responses. Poor proliferation and diminished responsiveness to cytokines appear to be ubiquitous features of aged T cells and may explain the delayed peak in T cell expansion and cytotoxic activity commonly observed in the ‘immunosenescence’ phenotype in the elderly. On the other hand, physical exercise stimulates the expression of crucial nutrient sensors and inhibits the mechanistic target of the rapamycin (mTOR) signaling cascade which increases autophagic activity in cells. Therefore, in this perspective review, we will first contextualize the overall view of the autophagy process and then, we will discuss how body adiposity and physical fitness may counteract autophagy in naïve CD4+ T cells in aging.

Type
Review
Copyright
Copyright © The Author(s), 2023. Published by Cambridge University Press

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

Footnotes

*

CSP and MK contributed equally to this work.

References

Partridge, L, Deelen, J and Slagboom, PE (2018) Facing up to the global challenges of ageing. Nature 561, 4556.CrossRefGoogle Scholar
Burman, M et al. (2022) Prevalence of obesity and malnutrition in four cohorts of very old adults, 2000–2017. The Journal of Nutrition, Health & Aging 26, 706713.CrossRefGoogle ScholarPubMed
Kaushik, S and Cuervo, AM (2015) Proteostasis and aging. Nature Medicine 21, 14061415.CrossRefGoogle ScholarPubMed
Suominen, A et al. (2022) Physical fitness and frailty in males after allogeneic hematopoietic stem cell transplantation in childhood: a long-term follow-up study. Cancers (Basel) 14.CrossRefGoogle ScholarPubMed
Fridh, MK et al. (2021) Cardiorespiratory fitness and physical performance after childhood hematopoietic stem cell transplantation: a systematic review and meta-analysis. Bone Marrow Transplantation 56, 20632078.CrossRefGoogle ScholarPubMed
Guseman, EH et al. (2017) The association between measures of fitness and metabolic health in treatment-seeking youth with obesity. Metabolic Syndrome and Related Disorders 15, 107111.CrossRefGoogle ScholarPubMed
Carnethon, MR, Gulati, M and Greenland, P (2005) Prevalence and cardiovascular disease correlates of low cardiorespiratory fitness in adolescents and adults. JAMA 294, 29812988.CrossRefGoogle ScholarPubMed
López-Otín, C et al. (2013) The hallmarks of aging. Cell 153, 11941217.CrossRefGoogle ScholarPubMed
Liu, JK (2022) Antiaging agents: safe interventions to slow aging and healthy life span extension. Natural Products and Bioprospecting 12, 18.CrossRefGoogle ScholarPubMed
Franceschi, C et al. (2018) Inflammaging: a new immune-metabolic viewpoint for age-related diseases. Nature Reviews. Endocrinology 14, 576590.CrossRefGoogle ScholarPubMed
Franceschi, C et al. (2000) Inflamm-aging. An evolutionary perspective on immunosenescence. Annals of the New York Academy of Sciences 908, 244254.CrossRefGoogle ScholarPubMed
Franceschi, C, Bonafè, M and Valensin, S (2000) Human immunosenescence: the prevailing of innate immunity, the failing of clonotypic immunity, and the filling of immunological space. Vaccine 18, 17171720.CrossRefGoogle ScholarPubMed
Santoro, A et al. (2020) Inflammaging, hormesis and the rationale for anti-aging strategies. Ageing Research Reviews 64, 101142.CrossRefGoogle ScholarPubMed
Pirola, L and Ferraz, JC (2017) Role of pro- and anti-inflammatory phenomena in the physiopathology of type 2 diabetes and obesity. World Journal of Biological Chemistry 8, 120128.CrossRefGoogle ScholarPubMed
Connaughton, RM et al. (2016) Impact of anti-inflammatory nutrients on obesity-associated metabolic-inflammation from childhood through to adulthood. Proceedings of the Nutrition Society 75, 115124.CrossRefGoogle ScholarPubMed
Shirakawa, K et al. (2016) Obesity accelerates T cell senescence in murine visceral adipose tissue. Journal of Clinical Investigation 126, 46264639.CrossRefGoogle ScholarPubMed
Salvestrini, V, Sell, C and Lorenzini, A (2019) Obesity may accelerate the aging process. Frontiers in Endocrinology (Lausanne) 10, 266.CrossRefGoogle ScholarPubMed
Deretic, V (2021) Autophagy in inflammation, infection, and immunometabolism. Immunity 54, 437453.CrossRefGoogle ScholarPubMed
Levine, B, Mizushima, N and Virgin, HW (2011) Autophagy in immunity and inflammation. Nature 469, 323335.CrossRefGoogle ScholarPubMed
Lopez-Otin, C et al. (2016) Metabolic control of longevity. In Cell. USA: Elsevier Inc, pp. 802821.Google Scholar
Merkley, SD et al. (2018) Modulating T cell responses via autophagy: the intrinsic influence controlling the function of both antigen-presenting cells and T cells. Frontiers in Immunology 9, 2914.CrossRefGoogle ScholarPubMed
Jeong, J, Choi, YJ and Lee, HK (2022) The role of autophagy in the function of CD4(+) T cells and the development of chronic inflammatory diseases. Frontiers in Pharmacology 13, 860146.CrossRefGoogle ScholarPubMed
Watanabe, R et al. (2014) Autophagy plays a protective role as an anti-oxidant system in human T cells and represents a novel strategy for induction of T-cell apoptosis. European Journal of Immunology 44, 25082520.CrossRefGoogle Scholar
Levine, B, Packer, M and Codogno, P (2015) Development of autophagy inducers in clinical medicine. Journal of Clinical Investigation 125, 1424.CrossRefGoogle ScholarPubMed
Kovacs, JR et al. (2012) Autophagy promotes T-cell survival through degradation of proteins of the cell death machinery. Cell Death & Differentiation 19, 144152.CrossRefGoogle ScholarPubMed
Fitzwalter, BE and Thorburn, A (2015) Recent insights into cell death and autophagy. FEBS Journal 282, 42794288.CrossRefGoogle ScholarPubMed
Liu, Y and Levine, B (2015) Autosis and autophagic cell death: the dark side of autophagy. Cell Death & Differentiation 22, 367376.CrossRefGoogle ScholarPubMed
Madrigal-Matute, J and Cuervo, AM (2016) Regulation of liver metabolism by autophagy. Gastroenterology 150, 328339.CrossRefGoogle ScholarPubMed
Cheng, Y et al. (2013) Therapeutic targeting of autophagy in disease: biology and pharmacology. Pharmacological Reviews 65, 11621197.CrossRefGoogle ScholarPubMed
Russell, RC et al. (2013) ULK1 induces autophagy by phosphorylating Beclin-1 and activating VPS34 lipid kinase. Nature Cell Biology 15, 741750.CrossRefGoogle ScholarPubMed
Macian, F (2019) Autophagy in T cell function and aging. Frontiers in Cell and Developmental Biology 7, 213.CrossRefGoogle Scholar
Parzych, KR and Klionsky, DJ (2014) An overview of autophagy: morphology, mechanism, and regulation. Antioxidants & Redox Signaling 20, 460473.CrossRefGoogle ScholarPubMed
Kimura, T, Mandell, M and Deretic, V (2016) Precision autophagy directed by receptor regulators - emerging examples within the TRIM family. Journal of Cell Science 129, 881891.Google ScholarPubMed
Longatti, A and Tooze, SA (2009) Vesicular trafficking and autophagosome formation. Cell Death & Differentiation 16, 956965.CrossRefGoogle ScholarPubMed
Mejlvang, J et al. (2018) Starvation induces rapid degradation of selective autophagy receptors by endosomal microautophagy. Journal of Cell Biology 217, 36403655.CrossRefGoogle ScholarPubMed
Mizushima, N et al. (2004) In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. Molecular Biology of the Cell 15, 11011111.CrossRefGoogle ScholarPubMed
Wang, L, Klionsky, DJ and Shen, H-M (2022) The emerging mechanisms and functions of microautophagy. Nature Reviews Molecular Cell Biology 4, 740.Google Scholar
Li, WW, Li, J and Bao, JK (2012) Microautophagy: lesser-known self-eating. Cellular and Molecular Life Sciences 69, 11251136.CrossRefGoogle ScholarPubMed
Schuck, S, Gallagher, CM and Walter, P (2014) ER-phagy mediates selective degradation of endoplasmic reticulum independently of the core autophagy machinery. Journal of Cell Science 127, 40784088.Google ScholarPubMed
Dice, JF (2007) Chaperone-mediated autophagy. Autophagy 3, 295299.CrossRefGoogle ScholarPubMed
Cuervo, AM and Wong, E (2014) Chaperone-mediated autophagy: roles in disease and aging. Cell Research 24, 92104.CrossRefGoogle ScholarPubMed
Majeski, AE and Dice, JF (2004) Mechanisms of chaperone-mediated autophagy. Int J Biochem Cell Biol 36, 24352444.CrossRefGoogle ScholarPubMed
Wang, D et al. (2017) Roles of autophagy in ischemic heart diseases and the modulatory effects of Chinese herbal medicine. American Journal of Chinese Medicine 45, 14011419.CrossRefGoogle ScholarPubMed
Rockel, JS and Kapoor, M (2016) Autophagy: controlling cell fate in rheumatic diseases. Nature Reviews Rheumatology 12, 517531.CrossRefGoogle ScholarPubMed
Hernandez-Segura, A, Nehme, J and Demaria, M (2018) Hallmarks of cellular senescence. Trends in Cell Biology 28, 436453.CrossRefGoogle ScholarPubMed
Kaushik, S et al. (2021) Autophagy and the hallmarks of aging. Ageing Research Reviews 72, 101468.CrossRefGoogle ScholarPubMed
Kuballa, P et al. (2012) Autophagy and the immune system. Annual Review of Immunology 30, 611646.CrossRefGoogle ScholarPubMed
Harris, J (2011) Autophagy and cytokines. Cytokine 56, 140144.CrossRefGoogle ScholarPubMed
Jia, W and He, YW (2011) Temporal regulation of intracellular organelle homeostasis in T lymphocytes by autophagy. Journal of Immunology 186, 53135322.CrossRefGoogle Scholar
Linton, P and Thoman, ML (2001) T cell senescence. Frontiers in Bioscience 6, D248D261.CrossRefGoogle ScholarPubMed
Goronzy, JJ and Weyand, CM (2017) Successful and maladaptive T cell aging. Immunity 46, 364378.CrossRefGoogle ScholarPubMed
Chang, C, Jensen, LE and Hurley, JH (2021) Autophagosome biogenesis comes out of the black box. Nature Cell Biology 23, 450456.CrossRefGoogle ScholarPubMed
Goronzy, JJ et al. (2012) Signaling pathways in aged T cells - a reflection of T cell differentiation, cell senescence and host environment. Seminars in Immunology 24, 365372.CrossRefGoogle ScholarPubMed
Cambier, J (2005) Immunosenescence: a problem of lymphopoiesis, homeostasis, microenvironment, and signaling. Immunological Reviews 205, 56.CrossRefGoogle ScholarPubMed
Clise-Dwyer, K et al. (2007) Environmental and intrinsic factors lead to antigen unresponsiveness in CD4(+) recent thymic emigrants from aged mice. Journal of Immunology 178, 13211331.CrossRefGoogle ScholarPubMed
Dobber, R et al. (1992) The involvement of the intestinal microflora in the expansion of CD4 + T cells with a naive phenotype in the periphery. Developmental Immunology 2, 141150.CrossRefGoogle ScholarPubMed
Haynes, L, Linton, PJ and Swain, SL (1997) Age-related changes in CD4 T cells of T cell receptor transgenic mice. Mechanisms of Ageing and Development 93, 95105.CrossRefGoogle ScholarPubMed
Haynes, L et al. (1999) Interleukin 2, but not other common gamma chain-binding cytokines, can reverse the defect in generation of CD4 effector T cells from naive T cells of aged mice. Journal of Experimental Medicine 190, 10131024.CrossRefGoogle Scholar
Hale, JS et al. (2006) Thymic output in aged mice. Proceedings of the National Academy of Sciences of the USA 103, 84478452.CrossRefGoogle ScholarPubMed
Fülöp, T et al. (2016) The role of immunosenescence in the development of age-related diseases. Revista de Investigacion Clinica 68, 8491.Google ScholarPubMed
Eisenbraun, MD, Tamir, A and Miller, RA (2000) Altered composition of the immunological synapse in an anergic, age-dependent memory T cell subset. Journal of Immunology 164, 61056112.CrossRefGoogle Scholar
Gupta, S and Gollapudi, S (2006) Molecular mechanisms of TNF-alpha-induced apoptosis in naïve and memory T cell subsets. Autoimmunity Reviews 5, 264268.CrossRefGoogle ScholarPubMed
Gupta, S, Bi, R and Gollapudi, S (2005) Central memory and effector memory subsets of human CD4(+) and CD8(+) T cells display differential sensitivity to TNF-{alpha}-induced apoptosis. Annals of the New York Academy of Sciences 1050, 108114.CrossRefGoogle ScholarPubMed
Sharma, K et al. (2000) Death the Fas way: regulation and pathophysiology of CD95 and its ligand. Pharmacology & Therapeutics 88, 333347.CrossRefGoogle ScholarPubMed
Suda, T et al. (1993) Molecular cloning and expression of the Fas ligand, a novel member of the tumor necrosis factor family. Cell 75, 11691178.CrossRefGoogle ScholarPubMed
Locksley, RM, Killeen, N and Lenardo, MJ (2001) The TNF and TNF receptor superfamilies: integrating mammalian biology. Cell 104, 487501.CrossRefGoogle ScholarPubMed
Paulsen, M and Janssen, O (2011) Pro- and anti-apoptotic CD95 signaling in T cells. Cell Communication and Signaling: CCS 9, 7.CrossRefGoogle ScholarPubMed
Strasser, A, Jost, PJ and Nagata, S (2009) The many roles of FAS receptor signaling in the immune system. Immunity 30, 180192.CrossRefGoogle ScholarPubMed
Magnusson, C and Vaux, DL (1999) Signalling by CD95 and TNF receptors: not only life and death. Immunology and Cell Biology 77, 4146.CrossRefGoogle Scholar
Marsters, SA et al. (1998) Identification of a ligand for the death-domain-containing receptor Apo3. Current Biology 8, 525528.CrossRefGoogle ScholarPubMed
Boatright, KM and Salvesen, GS (2003) Mechanisms of caspase activation. Current Opinion in Cell Biology 15, 725731.CrossRefGoogle ScholarPubMed
Perl, M et al. (2005) Silencing of Fas, but not caspase-8, in lung epithelial cells ameliorates pulmonary apoptosis, inflammation, and neutrophil influx after hemorrhagic shock and sepsis. American Journal of Pathology 167, 15451559.CrossRefGoogle Scholar
Gupta, S and Gollapudi, S (2008) CD95-mediated apoptosis in naïve, central and effector memory subsets of CD4 + and CD8 + T cells in aged humans. Experimental Gerontology 43, 266274.CrossRefGoogle ScholarPubMed
Conte, M et al. (2020) Mitochondria, immunosenescence and inflammaging: a role for mitokines? Seminars in Immunopathology 42, 607617.CrossRefGoogle ScholarPubMed
Mattoo, H et al. (2009) Naive CD4 T cells from aged mice show enhanced death upon primary activation. International Immunology 21, 12771289.CrossRefGoogle ScholarPubMed
Bharath, LP et al. (2020) Metformin enhances autophagy and normalizes mitochondrial function to alleviate aging-associated inflammation. Cell Metabolism 32, 4455.e6.CrossRefGoogle ScholarPubMed
Geisler, S et al. (2010) PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1. Nature Cell Biology 12, 119131.CrossRefGoogle ScholarPubMed
Saisho, Y (2015) Metformin and inflammation: its potential beyond glucose-lowering effect. Endocrine Metabolic Immune Disorders-Drug Targets 15, 196205.CrossRefGoogle ScholarPubMed
Malínská, H et al. (2016) Effects of metformin on tissue oxidative and dicarbonyl stress in transgenic spontaneously hypertensive rats expressing human C-reactive protein. PLoS ONE 11, e0150924.CrossRefGoogle ScholarPubMed
Cameron, AR et al. (2016) Anti-inflammatory effects of metformin irrespective of diabetes status. Circulation Research 119, 652665.CrossRefGoogle ScholarPubMed
Lochner, M, Berod, L and Sparwasser, T (2015) Fatty acid metabolism in the regulation of T cell function. Trends in Immunology 36, 8191.CrossRefGoogle ScholarPubMed
Weitz-Schmidt, G et al. (2001) Statins selectively inhibit leukocyte function antigen-1 by binding to a novel regulatory integrin site. Nature Medicine 7, 687692.CrossRefGoogle ScholarPubMed
Ghittoni, R et al. (2007) T lymphocytes as targets of statins: molecular mechanisms and therapeutic perspectives. Inflammation and Allergy - Drug Targets 6, 316.CrossRefGoogle ScholarPubMed
van Seventer, GA et al. (1998) Integrins and T helper cell activation. Transplantation proceedings 30, 42704274.CrossRefGoogle ScholarPubMed
Evans, DS et al. (2011) TOR signaling never gets old: aging, longevity and TORC1 activity. Ageing Research Reviews 10, 225237.CrossRefGoogle ScholarPubMed
Robida-Stubbs, S et al. (2012) TOR signaling and rapamycin influence longevity by regulating SKN-1/Nrf and DAF-16/FoxO. Cell Metabolism 15, 713724.CrossRefGoogle ScholarPubMed
Harrison, DE et al. (2009) Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 460, 392395.CrossRefGoogle ScholarPubMed
Galluzzi, L et al. (2017) Pharmacological modulation of autophagy: therapeutic potential and persisting obstacles. Nature Reviews Drug Discovery 16, 487511.CrossRefGoogle ScholarPubMed
Kim, YC and Guan, KL (2015) mTOR: a pharmacologic target for autophagy regulation. The Journal of Clinical Investigation 125, 2532.CrossRefGoogle ScholarPubMed
Perkey, E et al. (2013) Increased mammalian target of rapamycin complex 2 signaling promotes age-related decline in CD4 T cell signaling and function. The Journal of Immunology 191, 46484655.CrossRefGoogle ScholarPubMed
Powell, JD et al. (2012) Regulation of immune responses by mTOR. The Annual Review of Immunology 30, 3968.CrossRefGoogle ScholarPubMed
Colman, RJ et al. (2009) Caloric restriction delays disease onset and mortality in rhesus monkeys. Science 325, 201204.CrossRefGoogle ScholarPubMed
Colman, RJ et al. (2014) Caloric restriction reduces age-related and all-cause mortality in rhesus monkeys. Nature Communications 5, 3557.CrossRefGoogle ScholarPubMed
Kemnitz, JW (2011) Calorie restriction and aging in nonhuman primates. ILAR Journal 52, 6677.CrossRefGoogle ScholarPubMed
Liang, H et al. (2019) Mammalian target of rapamycin at the crossroad between Alzheimer's disease and diabetes. Advances in Experimental Medicine and Biology 1128, 185225.CrossRefGoogle ScholarPubMed
Farazi, M et al. (2014) Caloric restriction maintains OX40 agonist-mediated tumor immunity and CD4 T cell priming during aging. Cancer Immunology, Immunotherapy 63, 615626.CrossRefGoogle ScholarPubMed
Wang, J et al. (2013) Caloric restriction favorably impacts metabolic and immune/inflammatory profiles in obese mice but curcumin/piperine consumption adds no further benefit. Nutrition and Metabolism (Lond) 10, 29.CrossRefGoogle ScholarPubMed
Johnson, DA et al. (2021) Understanding the determinants of circadian health disparities and cardiovascular disease. Chronobiology International 21, 18.CrossRefGoogle ScholarPubMed
Bastien, M et al. (2014) Overview of epidemiology and contribution of obesity to cardiovascular disease. Progress in Cardiovascular Diseases 56, 369381.CrossRefGoogle ScholarPubMed
Afshin, A et al. (2017) Health effects of overweight and obesity in 195 countries over 25 years. New England Journal of Medicine 377, 1327.Google ScholarPubMed
Hubler, MJ and Kennedy, AJ (2016) Role of lipids in the metabolism and activation of immune cells. Journal of Nutritional Biochemistry 34, 17.CrossRefGoogle Scholar
Ross, EA, Devitt, A and Johnson, JR (2021) Macrophages: the good, the bad, and the gluttony. Frontiers in Immunology 12, 708186.CrossRefGoogle ScholarPubMed
Lin, YW and Wei, LN (2017) Innate immunity orchestrates adipose tissue homeostasis. Hormone Molecular Biology and Clinical Investigation 31. EpubCrossRefGoogle ScholarPubMed
Zhang, Y, Sowers, JR and Ren, J (2018) Targeting autophagy in obesity: from pathophysiology to management. Nature Reviews Endocrinology 14, 356376.CrossRefGoogle ScholarPubMed
Katsi, V et al. (2022) Chrononutrition in cardiometabolic health. Journal of Clinical Medicine 11, 296.CrossRefGoogle ScholarPubMed
Katsi, VK et al. (2015) Anti-angiogenic therapy and cardiovascular diseases: current strategies and future perspectives. In Atta-ur-Rahman and Muhammad Iqbal Choudhary (eds), Anti-Angiogenesis Drug Discovery and Development. Elsevier, pp. 268308. https://doi.org/10.1016/B978-0-12-803963-2.50008-9Google Scholar
Mattson, MP, Longo, VD and Harvie, M (2017) Impact of intermittent fasting on health and disease processes. Ageing Research Reviews 39, 4658.CrossRefGoogle ScholarPubMed
Bellot, G et al. (2009) Hypoxia-induced autophagy is mediated through hypoxia-inducible factor induction of BNIP3 and BNIP3L via their BH3 domains. Molecular and Cellular Biology 29, 25702581.CrossRefGoogle ScholarPubMed
Soussi, H, Clément, K and Dugail, I (2016) Adipose tissue autophagy status in obesity: expression and flux--two faces of the picture. Autophagy 12, 588589.CrossRefGoogle ScholarPubMed
Haim, Y et al. (2015) Elevated autophagy gene expression in adipose tissue of obese humans: a potential non-cell-cycle-dependent function of E2F1. Autophagy 11, 20742088.CrossRefGoogle ScholarPubMed
Juárez-Rojas, JG et al. (2014) Autophagy and cardiometabolic risk factors. Reviews in Endocrine & Metabolic Disorders 15, 307315.CrossRefGoogle ScholarPubMed
Ignacio-Souza, LM et al. (2014) Defective regulation of the ubiquitin/proteasome system in the hypothalamus of obese male mice. Endocrinology 155, 28312844.CrossRefGoogle ScholarPubMed
Jaishy, B et al. (2015) Lipid-induced NOX2 activation inhibits autophagic flux by impairing lysosomal enzyme activity. Journal of Lipid Research 56, 546561.CrossRefGoogle ScholarPubMed
Lee, HY et al. (2016) Autophagy deficiency in myeloid cells increases susceptibility to obesity-induced diabetes and experimental colitis. Autophagy 12, 13901403.CrossRefGoogle ScholarPubMed
Kim, KH and Lee, MS (2014) Autophagy--a key player in cellular and body metabolism. Nature Reviews. Endocrinology 10, 322337.CrossRefGoogle ScholarPubMed
Oga, EA and Eseyin, OR (2016) The obesity paradox and heart failure: a systematic review of a decade of evidence. Journal of Obesity 2016, 9040248.CrossRefGoogle ScholarPubMed
He, C et al. (2013) Beclin 2 functions in autophagy, degradation of G protein-coupled receptors, and metabolism. Cell 154, 10851099.CrossRefGoogle ScholarPubMed
Liu, Y et al. (2016) Bif-1 deficiency impairs lipid homeostasis and causes obesity accompanied by insulin resistance. Scientific Reports 6, 20453.CrossRefGoogle ScholarPubMed
Jain, S and Chaves, SS (2011) Obesity and influenza. Clinical infectious Diseases. 53, 422424.Google Scholar
Duffaut, C et al. (2009) Interplay between human adipocytes and T lymphocytes in obesity: CCL20 as an adipochemokine and T lymphocytes as lipogenic modulators. Arteriosclerosis Thrombosis and Vascular Biology 29, 16081614.CrossRefGoogle Scholar
Olson, NC et al. (2015) Associations of circulating lymphocyte subpopulations with type 2 diabetes: cross-sectional results from the multi-ethnic study of atherosclerosis (MESA). PLoS ONE 10, e0139962.CrossRefGoogle ScholarPubMed
Winer, DA et al. (2011) B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nature Medicine 17, 610617.CrossRefGoogle ScholarPubMed
McLaughlin, T et al. (2014) T-cell profile in adipose tissue is associated with insulin resistance and systemic inflammation in humans. Arteriosclerosis Thrombosis and Vascular Biology 34, 26372643.CrossRefGoogle ScholarPubMed
Gorjão, R et al. (2007) Regulation of interleukin-2 signaling by fatty acids in human lymphocytes. Journal of Lipid Research 48, 20092019.CrossRefGoogle ScholarPubMed
Niso-Santano, M et al. (2015) Unsaturated fatty acids induce non-canonical autophagy. EMBO Journal 34, 10251041.CrossRefGoogle ScholarPubMed
Koga, H, Kaushik, S and Cuervo, AM (2010) Altered lipid content inhibits autophagic vesicular fusion. FASEB Journal 24, 30523065.CrossRefGoogle ScholarPubMed
Mei, S et al. (2011) Differential roles of unsaturated and saturated fatty acids on autophagy and apoptosis in hepatocytes. Journal of Pharmacology and Experimental Therapeutics 339, 487498.CrossRefGoogle ScholarPubMed
Guerrero-Ros, I et al. (2020) The negative effect of lipid challenge on autophagy inhibits T cell responses. Autophagy 16, 223238.CrossRefGoogle ScholarPubMed
Wara, AK et al. (2020) KLF10 Deficiency in CD4(+) T cells triggers obesity, insulin resistance, and fatty liver. Cell Reports 33, 108550.CrossRefGoogle ScholarPubMed
Kalathookunnel Antony, A, Lian, Z and Wu, H (2018) T cells in adipose tissue in aging. Frontiers in Immunology 9, 2945.CrossRefGoogle ScholarPubMed
Misumi, I et al. (2019) Obesity Expands a Distinct Population of T Cells in Adipose Tissue and Increases Vulnerability to Infection. Cell Reports 27, 514524.e5.CrossRefGoogle ScholarPubMed
Ren, J, Sowers, JR and Zhang, Y (2018) Metabolic stress, autophagy, and cardiovascular aging: from pathophysiology to therapeutics. Trends in Endocrinology and Metabolism 29, 699711.CrossRefGoogle ScholarPubMed
He, C et al. (2012) Exercise-induced BCL2-regulated autophagy is required for muscle glucose homeostasis. Nature 481, 511515.CrossRefGoogle ScholarPubMed
Li, H et al. (2016) Acute exercise-induced mitochondrial stress triggers an inflammatory response in the myocardium via NLRP3 inflammasome activation with mitophagy. Oxidative Medicine and Cellular Longevity 2016, 1987149.Google ScholarPubMed
Wu, NN et al. (2019) Physical exercise and selective autophagy: benefit and risk on cardiovascular health. Cells 88, 1436.Google Scholar
Wu, NN, Zhang, Y and Ren, J (2019) Mitophagy, mitochondrial dynamics, and homeostasis in cardiovascular aging. Oxidative Medicine and Cellular Longevity 2019, 9825061.CrossRefGoogle ScholarPubMed
Biala, AK, Dhingra, R and Kirshenbaum, LA (2015) Mitochondrial dynamics: orchestrating the journey to advanced age. Journal of Molecular and Cellular Cardiology 83, 3743.CrossRefGoogle ScholarPubMed
Korolchuk, VI et al. (2017) Mitochondria in cell senescence: is mitophagy the weakest link? EBioMedicine 21, 713.CrossRefGoogle ScholarPubMed
Saito, T and Sadoshima, J (2015) Molecular mechanisms of mitochondrial autophagy/mitophagy in the heart. Circulation Research 116, 14771490.CrossRefGoogle ScholarPubMed
Antón, Z et al. (2016) Human Atg8-cardiolipin interactions in mitophagy: specific properties of LC3B, GABARAPL2 and GABARAP. Autophagy 12, 23862403.CrossRefGoogle ScholarPubMed
Chen, CCW, Erlich, AT and Hood, DA (2018) Role of Parkin and endurance training on mitochondrial turnover in skeletal muscle. Skeletal Muscle 8, 10.CrossRefGoogle ScholarPubMed
Fix, DK et al. (2018) Role of gp130 in basal and exercise-trained skeletal muscle mitochondrial quality control. Journal of Applied Physiology (1985) 124, 14561470.CrossRefGoogle ScholarPubMed
Lazarou, M et al. (2015) The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy. Nature 524, 309314.CrossRefGoogle ScholarPubMed
Wang, JS et al. (2013) Effect of aerobic interval training on erythrocyte rheological and hemodynamic functions in heart failure patients with anemia. International Journal of Cardiology 168, 12431250.CrossRefGoogle ScholarPubMed
Fu, TC et al. (2013) Aerobic interval training improves oxygen uptake efficiency by enhancing cerebral and muscular hemodynamics in patients with heart failure. International Journal of Cardiology 167, 4150.CrossRefGoogle ScholarPubMed
Wang, JS, Chen, WL and Weng, TP (2011) Hypoxic exercise training reduces senescent T-lymphocyte subsets in blood. Brain Behavior and Immunity 25, 270278.CrossRefGoogle ScholarPubMed
Beyersdorf, N et al. (2007) Characterization of mouse CD4 T cell subsets defined by expression of KLRG1. European Journal of Immunology 37, 34453454.CrossRefGoogle ScholarPubMed
Weng, NP, Akbar, AN and Goronzy, J (2009) CD28(-) T cells: their role in the age-associated decline of immune function. Trends in Immunology 30, 306312.CrossRefGoogle ScholarPubMed
Weng, TP et al. (2013) Effects of interval and continuous exercise training on CD4 lymphocyte apoptotic and autophagic responses to hypoxic stress in sedentary men. PLoS ONE 8, e80248.CrossRefGoogle ScholarPubMed
Dutta, D et al. (2013) Upregulated autophagy protects cardiomyocytes from oxidative stress-induced toxicity. Autophagy 9, 328344.CrossRefGoogle ScholarPubMed
Hsieh, YC, Athar, M and Chaudry, IH (2009) When apoptosis meets autophagy: deciding cell fate after trauma and sepsis. Trends in Molecular Medicine 15, 129138.CrossRefGoogle ScholarPubMed
McCormick, JJ et al. (2019) Effect of acute aerobic exercise and rapamycin treatment on autophagy in peripheral blood mononuclear cells of adults with prediabetes. Canadian Journal of Diabetes 43, 457463.CrossRefGoogle ScholarPubMed
Jeon, SM (2016) Regulation and function of AMPK in physiology and diseases. Experimental and Molecular Medicine 48, e245.CrossRefGoogle ScholarPubMed
Rosa-Neto, JC et al. (2022) Immunometabolism-fit: How exercise and training can modify T cell and macrophage metabolism in health and disease. Exercise Immunology Review 28, 2946.Google ScholarPubMed
Day, EA, Ford, RJ and Steinberg, GR (2017) AMPK as a Therapeutic Target for Treating Metabolic Diseases. Trends in Endocrinology and Metabolism 28, 545560.CrossRefGoogle ScholarPubMed
Schwalm, C et al. (2015) Activation of autophagy in human skeletal muscle is dependent on exercise intensity and AMPK activation. FASEB Journal 29, 35153526.CrossRefGoogle ScholarPubMed
Mejías-Peña, Y et al. (2017) Impact of resistance training on the autophagy-inflammation-apoptosis crosstalk in elderly subjects. Aging (Albany NY) 9, 408418.CrossRefGoogle ScholarPubMed
Duggal, NA et al. (2018) Major features of immunesenescence, including reduced thymic output, are ameliorated by high levels of physical activity in adulthood. Aging Cell 17, e12750.CrossRefGoogle ScholarPubMed
Turner, JE (2016) Is immunosenescence influenced by our lifetime ‘dose’ of exercise? Biogerontology 17, 581602.CrossRefGoogle ScholarPubMed
Mathot, E et al. (2021) Systematic review on the effects of physical exercise on cellular immunosenescence-related markers - An update. Experimental Gerontology 149, 111318.CrossRefGoogle ScholarPubMed
Cao Dinh, H et al. (2017) Effects of physical exercise on markers of cellular immunosenescence: a systematic review. Calcified Tissue International 100, 193215.CrossRefGoogle ScholarPubMed
Philippe, M et al. (2019) Concentric and eccentric endurance exercise reverse hallmarks of T-cell senescence in pre-diabetic subjects. Frontiers in Physiology 10, 684.CrossRefGoogle ScholarPubMed
Lee, DE et al. (2019) Autophagy as a therapeutic target to enhance aged muscle regeneration. Cells 8, 183.CrossRefGoogle ScholarPubMed
Hansen, M, Rubinsztein, DC and Walker, DW (2018) Autophagy as a promoter of longevity: insights from model organisms. Nature Reviews Molecular Cell Biology 19, 579593.Google ScholarPubMed
Batatinha, HAP et al. (2019) Regulation of autophagy as a therapy for immunosenescence-driven cancer and neurodegenerative diseases: the role of exercise. Journal of Cellular Physiology 234, 1488314895.CrossRefGoogle ScholarPubMed