Skip to main content
Top
Published in: Reviews in Endocrine and Metabolic Disorders 1/2022

Open Access 01-02-2022 | Obesity

Adipogenic progenitors in different organs: Pathophysiological implications

Authors: Francesca Favaretto, Silvia Bettini, Luca Busetto, Gabriella Milan, Roberto Vettor

Published in: Reviews in Endocrine and Metabolic Disorders | Issue 1/2022

Login to get access

Abstract

In physiological conditions, the adipose organ resides in well-defined areas, where it acts providing an energy supply and as an endocrine organ involved in the control of whole-body energy metabolism. Adipose tissue adipokines connect the body’s nutritional status to the regulation of energy balance. When it surrounds organs, it provides also for mechanical protection. Adipose tissue has a complex and heterogenous cellular composition that includes adipocytes, adipose tissue-derived stromal and stem cells (ASCs) which are mesenchymal stromal cells, and endothelial and immune cells, which signal to each other and to other tissues to maintain homeostasis. In obesity and in other nutrition related diseases, as well as in age-related diseases, biological and functional changes of adipose tissue give rise to several complications. Obesity triggers alterations of ASCs, impairing adipose tissue remodeling and adipose tissue function, which induces low-grade systemic inflammation, progressive insulin resistance and other metabolic disorders. Adipose tissue grows by hyperplasia recruiting new ASCs and by hypertrophy, up to its expandability limit. To overcome this limitation and to store the excess of nutrients, adipose tissue develops ectopically, involving organs such as muscle, bone marrow and the heart. The origin of ectopic adipose organ is not clearly elucidated, and a possible explanation lies in the stimulation of the adipogenic differentiation of mesenchymal precursor cells which normally differentiate toward a lineage specific for the organ in which they reside. The chronic exposition of these newly-formed adipose depots to the pathological environment, will confer to them all the phenotypic characteristics of a dysfunctional adipose tissue, perpetuating the organ alterations. Visceral fat, but also ectopic fat, either in the liver, muscle or heart, can increase the risk of developing insulin resistance, type 2 diabetes, and cardiovascular diseases. Being able to prevent and to target dysfunctional adipose tissue will avoid the progression towards the complications of obesity and other nutrition-related diseases. The aim of this review is to summarize some of the knowledge regarding the presence of adipose tissue in particular tissues (where it is not usually present), describing the composition of its adipogenic precursors, and the interactions responsible for the development of organ pathologies.
Literature
1.
go back to reference Choe SS, Huh JY, Hwang IJ, Kim JI, Kim JB. Adipose tissue remodeling: its role in energy metabolism and metabolic disorders. Front Endocrinol. 2016;7:30. Choe SS, Huh JY, Hwang IJ, Kim JI, Kim JB. Adipose tissue remodeling: its role in energy metabolism and metabolic disorders. Front Endocrinol. 2016;7:30.
3.
go back to reference Kahn CR, Wang G, Lee KY. Altered adipose tissue and adipocyte function in the pathogenesis of metabolic syndrome. J Clin Invest. 2019;129:3990–4000.PubMedCentralPubMed Kahn CR, Wang G, Lee KY. Altered adipose tissue and adipocyte function in the pathogenesis of metabolic syndrome. J Clin Invest. 2019;129:3990–4000.PubMedCentralPubMed
4.
go back to reference Pillon NJ, Loos RJF, Marshall SM, Zierath JR. Metabolic consequences of obesity and type 2 diabetes: balancing genes and environment for personalized care. Cell. 2021;184:1530–44.PubMed Pillon NJ, Loos RJF, Marshall SM, Zierath JR. Metabolic consequences of obesity and type 2 diabetes: balancing genes and environment for personalized care. Cell. 2021;184:1530–44.PubMed
5.
go back to reference Qasim A, Turcotte M, de Souza RJ, Samaan MC, Champredon D, Dushoff J, et al. On the origin of obesity: identifying the biological, environmental and cultural drivers of genetic risk among human populations. Obes Rev. 2018;19:121–49.PubMed Qasim A, Turcotte M, de Souza RJ, Samaan MC, Champredon D, Dushoff J, et al. On the origin of obesity: identifying the biological, environmental and cultural drivers of genetic risk among human populations. Obes Rev. 2018;19:121–49.PubMed
6.
go back to reference Kim K-S, Seeley RJ, Sandoval DA. Signalling from the periphery to the brain that regulates energy homeostasis. Nat Rev Neurosci. 2018;19:185–96.PubMed Kim K-S, Seeley RJ, Sandoval DA. Signalling from the periphery to the brain that regulates energy homeostasis. Nat Rev Neurosci. 2018;19:185–96.PubMed
7.
go back to reference Schwartz MW, Porte D. Diabetes, obesity, and the brain. Science. 2005;307:375–9.PubMed Schwartz MW, Porte D. Diabetes, obesity, and the brain. Science. 2005;307:375–9.PubMed
8.
go back to reference GBD 2015 Obesity Collaborators, Afshin A, Forouzanfar MH, Reitsma MB, Sur P, Estep K, et al. Health effects of overweight and obesity in 195 countries over 25 years. N Engl J Med. 2017;377:13–27. GBD 2015 Obesity Collaborators, Afshin A, Forouzanfar MH, Reitsma MB, Sur P, Estep K, et al. Health effects of overweight and obesity in 195 countries over 25 years. N Engl J Med. 2017;377:13–27.
9.
go back to reference Longo M, Zatterale F, Naderi J, Parrillo L, Formisano P, Raciti GA, et al. Adipose tissue dysfunction as determinant of obesity-associated metabolic complications. Int J Mol Sci. 2019;20:E2358.PubMed Longo M, Zatterale F, Naderi J, Parrillo L, Formisano P, Raciti GA, et al. Adipose tissue dysfunction as determinant of obesity-associated metabolic complications. Int J Mol Sci. 2019;20:E2358.PubMed
10.
go back to reference Schoettl T, Fischer IP, Ussar S. Heterogeneity of adipose tissue in development and metabolic function. J Exp Biol. 2018;221:jeb162958.PubMed Schoettl T, Fischer IP, Ussar S. Heterogeneity of adipose tissue in development and metabolic function. J Exp Biol. 2018;221:jeb162958.PubMed
12.
go back to reference Heilbronn L, Smith SR, Ravussin E. Failure of fat cell proliferation, mitochondrial function and fat oxidation results in ectopic fat storage, insulin resistance and type II diabetes mellitus. Int J Obes Relat Metab Disord. 2004;28(Suppl 4):S12-21.PubMed Heilbronn L, Smith SR, Ravussin E. Failure of fat cell proliferation, mitochondrial function and fat oxidation results in ectopic fat storage, insulin resistance and type II diabetes mellitus. Int J Obes Relat Metab Disord. 2004;28(Suppl 4):S12-21.PubMed
13.
go back to reference Zwick RK, Guerrero-Juarez CF, Horsley V, Plikus MV. Anatomical, physiological, and functional diversity of adipose tissue. Cell Metab. 2018;27:68–83.PubMedCentralPubMed Zwick RK, Guerrero-Juarez CF, Horsley V, Plikus MV. Anatomical, physiological, and functional diversity of adipose tissue. Cell Metab. 2018;27:68–83.PubMedCentralPubMed
14.
go back to reference Ghaben AL, Scherer PE. Adipogenesis and metabolic health. Nat Rev Mol Cell Biol. 2019;20:242–58.PubMed Ghaben AL, Scherer PE. Adipogenesis and metabolic health. Nat Rev Mol Cell Biol. 2019;20:242–58.PubMed
15.
go back to reference Paavonsalo S, Hariharan S, Lackman MH, Karaman S. Capillary rarefaction in obesity and metabolic diseases-organ-specificity and possible mechanisms. Cells. 2020;9. Paavonsalo S, Hariharan S, Lackman MH, Karaman S. Capillary rarefaction in obesity and metabolic diseases-organ-specificity and possible mechanisms. Cells. 2020;9.
16.
go back to reference Guilherme A, Henriques F, Bedard AH, Czech MP. Molecular pathways linking adipose innervation to insulin action in obesity and diabetes mellitus. Nat Rev Endocrinol. 2019;15:207–25.PubMedCentralPubMed Guilherme A, Henriques F, Bedard AH, Czech MP. Molecular pathways linking adipose innervation to insulin action in obesity and diabetes mellitus. Nat Rev Endocrinol. 2019;15:207–25.PubMedCentralPubMed
17.
go back to reference Liu R, Nikolajczyk BS. Tissue immune cells fuel obesity-associated inflammation in adipose tissue and beyond. Front Immunol. 2019;10:1587.PubMedCentralPubMed Liu R, Nikolajczyk BS. Tissue immune cells fuel obesity-associated inflammation in adipose tissue and beyond. Front Immunol. 2019;10:1587.PubMedCentralPubMed
18.
go back to reference Ferrero R, Rainer P, Deplancke B. Toward a consensus view of mammalian adipocyte stem and progenitor cell heterogeneity. Trends Cell Biol. 2020;30:937–50.PubMed Ferrero R, Rainer P, Deplancke B. Toward a consensus view of mammalian adipocyte stem and progenitor cell heterogeneity. Trends Cell Biol. 2020;30:937–50.PubMed
19.
go back to reference Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7:211–28.PubMed Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7:211–28.PubMed
20.
go back to reference Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13:4279–95.PubMedCentralPubMed Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13:4279–95.PubMedCentralPubMed
21.
go back to reference Zimmerlin L, Donnenberg VS, Rubin JP, Donnenberg AD. Mesenchymal markers on human adipose stem/progenitor cells. Cytometry A. 2013;83:134–40.PubMed Zimmerlin L, Donnenberg VS, Rubin JP, Donnenberg AD. Mesenchymal markers on human adipose stem/progenitor cells. Cytometry A. 2013;83:134–40.PubMed
22.
go back to reference Ceccarelli S, Pontecorvi P, Anastasiadou E, Napoli C, Marchese C. Immunomodulatory effect of adipose-derived stem cells: the cutting edge of clinical application. Front Cell Dev Biol. 2020;8:236.PubMedCentralPubMed Ceccarelli S, Pontecorvi P, Anastasiadou E, Napoli C, Marchese C. Immunomodulatory effect of adipose-derived stem cells: the cutting edge of clinical application. Front Cell Dev Biol. 2020;8:236.PubMedCentralPubMed
23.
go back to reference Strioga M, Viswanathan S, Darinskas A, Slaby O, Michalek J. Same or not the same? Comparison of adipose tissue-derived versus bone marrow-derived mesenchymal stem and stromal cells. Stem Cells Dev. 2012;21:2724–52.PubMed Strioga M, Viswanathan S, Darinskas A, Slaby O, Michalek J. Same or not the same? Comparison of adipose tissue-derived versus bone marrow-derived mesenchymal stem and stromal cells. Stem Cells Dev. 2012;21:2724–52.PubMed
24.
go back to reference Guimarães-Camboa N, Evans SM. Are perivascular adipocyte progenitors mural cells or adventitial fibroblasts? Cell Stem Cell. 2017;20:587–9.PubMedCentralPubMed Guimarães-Camboa N, Evans SM. Are perivascular adipocyte progenitors mural cells or adventitial fibroblasts? Cell Stem Cell. 2017;20:587–9.PubMedCentralPubMed
25.
go back to reference Tchkonia T, Giorgadze N, Pirtskhalava T, Thomou T, DePonte M, Koo A, et al. Fat depot-specific characteristics are retained in strains derived from single human preadipocytes. Diabetes. 2006;55:2571–8.PubMed Tchkonia T, Giorgadze N, Pirtskhalava T, Thomou T, DePonte M, Koo A, et al. Fat depot-specific characteristics are retained in strains derived from single human preadipocytes. Diabetes. 2006;55:2571–8.PubMed
26.
go back to reference Sanchez-Gurmaches J, Guertin DA. Adipocytes arise from multiple lineages that are heterogeneously and dynamically distributed. Nat Commun. 2014;5:4099.PubMed Sanchez-Gurmaches J, Guertin DA. Adipocytes arise from multiple lineages that are heterogeneously and dynamically distributed. Nat Commun. 2014;5:4099.PubMed
27.
go back to reference Scheja L, Heeren J. The endocrine function of adipose tissues in health and cardiometabolic disease. Nat Rev Endocrinol. 2019;15:507–24.PubMed Scheja L, Heeren J. The endocrine function of adipose tissues in health and cardiometabolic disease. Nat Rev Endocrinol. 2019;15:507–24.PubMed
28.
go back to reference Morigny P, Boucher J, Arner P, Langin D. Lipid and glucose metabolism in white adipocytes: pathways, dysfunction and therapeutics. Nat Rev Endocrinol. 2021;17:276–95.PubMed Morigny P, Boucher J, Arner P, Langin D. Lipid and glucose metabolism in white adipocytes: pathways, dysfunction and therapeutics. Nat Rev Endocrinol. 2021;17:276–95.PubMed
29.
go back to reference Vishvanath L, Gupta RK. Contribution of adipogenesis to healthy adipose tissue expansion in obesity. J Clin Invest. 2019;129:4022–31.PubMedCentralPubMed Vishvanath L, Gupta RK. Contribution of adipogenesis to healthy adipose tissue expansion in obesity. J Clin Invest. 2019;129:4022–31.PubMedCentralPubMed
30.
go back to reference Marcelin G, Silveira ALM, Martins LB, Ferreira AV, Clément K. Deciphering the cellular interplays underlying obesity-induced adipose tissue fibrosis. J Clin Invest. 2019;129:4032–40.PubMedCentralPubMed Marcelin G, Silveira ALM, Martins LB, Ferreira AV, Clément K. Deciphering the cellular interplays underlying obesity-induced adipose tissue fibrosis. J Clin Invest. 2019;129:4032–40.PubMedCentralPubMed
31.
go back to reference Caslin HL, Bhanot M, Bolus WR, Hasty AH. Adipose tissue macrophages: unique polarization and bioenergetics in obesity. Immunol Rev. 2020;295:101–13.PubMedCentralPubMed Caslin HL, Bhanot M, Bolus WR, Hasty AH. Adipose tissue macrophages: unique polarization and bioenergetics in obesity. Immunol Rev. 2020;295:101–13.PubMedCentralPubMed
32.
go back to reference Belligoli A, Compagnin C, Sanna M, Favaretto F, Fabris R, Busetto L, et al. Characterization of subcutaneous and omental adipose tissue in patients with obesity and with different degrees of glucose impairment. Sci Rep. 2019;9:11333.PubMedCentralPubMed Belligoli A, Compagnin C, Sanna M, Favaretto F, Fabris R, Busetto L, et al. Characterization of subcutaneous and omental adipose tissue in patients with obesity and with different degrees of glucose impairment. Sci Rep. 2019;9:11333.PubMedCentralPubMed
33.
go back to reference Liu F, He J, Wang H, Zhu D, Bi Y. Adipose morphology: a critical factor in regulation of human metabolic diseases and adipose tissue dysfunction. Obes Surg. 2020;30:5086–100.PubMedCentralPubMed Liu F, He J, Wang H, Zhu D, Bi Y. Adipose morphology: a critical factor in regulation of human metabolic diseases and adipose tissue dysfunction. Obes Surg. 2020;30:5086–100.PubMedCentralPubMed
34.
go back to reference Barazzoni R, Bischoff S, Boirie Y, Busetto L, Cederholm T, Dicker D, et al. Sarcopenic obesity: time to meet the challenge. Obes Facts. 2018;11:294–305.PubMedCentralPubMed Barazzoni R, Bischoff S, Boirie Y, Busetto L, Cederholm T, Dicker D, et al. Sarcopenic obesity: time to meet the challenge. Obes Facts. 2018;11:294–305.PubMedCentralPubMed
35.
go back to reference Cruz-Jentoft AJ, Baeyens JP, Bauer JM, Boirie Y, Cederholm T, Landi F, et al. Sarcopenia: European consensus on definition and diagnosis: report of the European working group on Sarcopenia in older people. Age Ageing. 2010;39:412–23.PubMedCentralPubMed Cruz-Jentoft AJ, Baeyens JP, Bauer JM, Boirie Y, Cederholm T, Landi F, et al. Sarcopenia: European consensus on definition and diagnosis: report of the European working group on Sarcopenia in older people. Age Ageing. 2010;39:412–23.PubMedCentralPubMed
36.
go back to reference Donini LM, Busetto L, Bauer JM, Bischoff S, Boirie Y, Cederholm T, et al. Critical appraisal of definitions and diagnostic criteria for sarcopenic obesity based on a systematic review. Clin Nutr. 2020;39:2368–88.PubMed Donini LM, Busetto L, Bauer JM, Bischoff S, Boirie Y, Cederholm T, et al. Critical appraisal of definitions and diagnostic criteria for sarcopenic obesity based on a systematic review. Clin Nutr. 2020;39:2368–88.PubMed
37.
go back to reference Mingrone G, Rosa G, Di Rocco P, Manco M, Capristo E, Castagneto M, et al. Skeletal muscle triglycerides lowering is associated with net improvement of insulin sensitivity, TNF-alpha reduction and GLUT4 expression enhancement. Int J Obes Relat Metab Disord. 2002;26:1165–72.PubMed Mingrone G, Rosa G, Di Rocco P, Manco M, Capristo E, Castagneto M, et al. Skeletal muscle triglycerides lowering is associated with net improvement of insulin sensitivity, TNF-alpha reduction and GLUT4 expression enhancement. Int J Obes Relat Metab Disord. 2002;26:1165–72.PubMed
38.
go back to reference Barazzoni R, Zanetti M, Gortan Cappellari G, Semolic A, Boschelle M, Codarin E, et al. Fatty acids acutely enhance insulin-induced oxidative stress and cause insulin resistance by increasing mitochondrial reactive oxygen species (ROS) generation and nuclear factor-κB inhibitor (IκB)-nuclear factor-κB (NFκB) activation in rat muscle, in the absence of mitochondrial dysfunction. Diabetologia. 2012;55:773–82.PubMed Barazzoni R, Zanetti M, Gortan Cappellari G, Semolic A, Boschelle M, Codarin E, et al. Fatty acids acutely enhance insulin-induced oxidative stress and cause insulin resistance by increasing mitochondrial reactive oxygen species (ROS) generation and nuclear factor-κB inhibitor (IκB)-nuclear factor-κB (NFκB) activation in rat muscle, in the absence of mitochondrial dysfunction. Diabetologia. 2012;55:773–82.PubMed
39.
go back to reference Lipina C, Hundal HS. Lipid modulation of skeletal muscle mass and function. J Cachexia Sarcopenia Muscle. 2017;8:190–201.PubMed Lipina C, Hundal HS. Lipid modulation of skeletal muscle mass and function. J Cachexia Sarcopenia Muscle. 2017;8:190–201.PubMed
40.
go back to reference Tardif N, Salles J, Guillet C, Tordjman J, Reggio S, Landrier JF, et al. Muscle ectopic fat deposition contributes to anabolic resistance in obese sarcopenic old rats through eIF2α activation. Aging Cell. 2014;13:1001–11.PubMedCentralPubMed Tardif N, Salles J, Guillet C, Tordjman J, Reggio S, Landrier JF, et al. Muscle ectopic fat deposition contributes to anabolic resistance in obese sarcopenic old rats through eIF2α activation. Aging Cell. 2014;13:1001–11.PubMedCentralPubMed
41.
go back to reference Vettor R, Milan G, Franzin C, Sanna M, De Coppi P, Rizzuto R, et al. The origin of intermuscular adipose tissue and its pathophysiological implications. Am J Physiol Endocrinol Metab. 2009;297:E987–998.PubMed Vettor R, Milan G, Franzin C, Sanna M, De Coppi P, Rizzuto R, et al. The origin of intermuscular adipose tissue and its pathophysiological implications. Am J Physiol Endocrinol Metab. 2009;297:E987–998.PubMed
42.
go back to reference De Coppi P, Milan G, Scarda A, Boldrin L, Centobene C, Piccoli M, et al. Rosiglitazone modifies the adipogenic potential of human muscle satellite cells. Diabetologia. 2006;49:1962–73.PubMed De Coppi P, Milan G, Scarda A, Boldrin L, Centobene C, Piccoli M, et al. Rosiglitazone modifies the adipogenic potential of human muscle satellite cells. Diabetologia. 2006;49:1962–73.PubMed
43.
go back to reference Rossi CA, Pozzobon M, Ditadi A, Archacka K, Gastaldello A, Sanna M, et al. Clonal characterization of rat muscle satellite cells: proliferation, metabolism and differentiation define an intrinsic heterogeneity. PLoS One. 2010;5:e8523.PubMedCentralPubMed Rossi CA, Pozzobon M, Ditadi A, Archacka K, Gastaldello A, Sanna M, et al. Clonal characterization of rat muscle satellite cells: proliferation, metabolism and differentiation define an intrinsic heterogeneity. PLoS One. 2010;5:e8523.PubMedCentralPubMed
44.
go back to reference Scarda A, Franzin C, Milan G, Sanna M, Dal Prà C, Pagano C, et al. Increased adipogenic conversion of muscle satellite cells in obese Zucker rats. Int J Obes. 2010;34:1319–27. Scarda A, Franzin C, Milan G, Sanna M, Dal Prà C, Pagano C, et al. Increased adipogenic conversion of muscle satellite cells in obese Zucker rats. Int J Obes. 2010;34:1319–27.
45.
go back to reference Aguiari P, Leo S, Zavan B, Vindigni V, Rimessi A, Bianchi K, et al. High glucose induces adipogenic differentiation of muscle-derived stem cells. Proc Natl Acad Sci U S A. 2008;105:1226–31.PubMedCentralPubMed Aguiari P, Leo S, Zavan B, Vindigni V, Rimessi A, Bianchi K, et al. High glucose induces adipogenic differentiation of muscle-derived stem cells. Proc Natl Acad Sci U S A. 2008;105:1226–31.PubMedCentralPubMed
46.
go back to reference Thornell LE. Sarcopenic obesity: satellite cells in the aging muscle. Curr Opin Clin Nutr Metab Care. 2011;14:22–7.PubMed Thornell LE. Sarcopenic obesity: satellite cells in the aging muscle. Curr Opin Clin Nutr Metab Care. 2011;14:22–7.PubMed
47.
go back to reference Wang L, Shan T. Factors inducing transdifferentiation of myoblasts into adipocytes. J Cell Physiol. 2021;236:2276–89.PubMed Wang L, Shan T. Factors inducing transdifferentiation of myoblasts into adipocytes. J Cell Physiol. 2021;236:2276–89.PubMed
48.
go back to reference Camps J, Breuls N, Sifrim A, Giarratana N, Corvelyn M, Danti L, et al. Interstitial cell remodeling promotes aberrant adipogenesis in dystrophic muscles. Cell Rep. 2020;31:107597.PubMed Camps J, Breuls N, Sifrim A, Giarratana N, Corvelyn M, Danti L, et al. Interstitial cell remodeling promotes aberrant adipogenesis in dystrophic muscles. Cell Rep. 2020;31:107597.PubMed
49.
go back to reference Uezumi A, Fukada S, Yamamoto N, Takeda S, Tsuchida K. Mesenchymal progenitors distinct from satellite cells contribute to ectopic fat cell formation in skeletal muscle. Nat Cell Biol. 2010;12:143–52.PubMed Uezumi A, Fukada S, Yamamoto N, Takeda S, Tsuchida K. Mesenchymal progenitors distinct from satellite cells contribute to ectopic fat cell formation in skeletal muscle. Nat Cell Biol. 2010;12:143–52.PubMed
50.
go back to reference Uezumi A, Fukada S, Yamamoto N, Ikemoto-Uezumi M, Nakatani M, Morita M, et al. Identification and characterization of PDGFRα+ mesenchymal progenitors in human skeletal muscle. Cell Death Dis. 2014;5:e1186.PubMedCentralPubMed Uezumi A, Fukada S, Yamamoto N, Ikemoto-Uezumi M, Nakatani M, Morita M, et al. Identification and characterization of PDGFRα+ mesenchymal progenitors in human skeletal muscle. Cell Death Dis. 2014;5:e1186.PubMedCentralPubMed
51.
go back to reference Hogarth MW, Defour A, Lazarski C, Gallardo E, Diaz Manera J, Partridge TA, et al. Fibroadipogenic progenitors are responsible for muscle loss in limb girdle muscular dystrophy 2B. Nat Commun. 2019;10:2430.PubMedCentralPubMed Hogarth MW, Defour A, Lazarski C, Gallardo E, Diaz Manera J, Partridge TA, et al. Fibroadipogenic progenitors are responsible for muscle loss in limb girdle muscular dystrophy 2B. Nat Commun. 2019;10:2430.PubMedCentralPubMed
52.
go back to reference Gil-Ortega M, Garidou L, Barreau C, Maumus M, Breasson L, Tavernier G, et al. Native adipose stromal cells egress from adipose tissue in vivo: evidence during lymph node activation. Stem Cells. 2013;31:1309–20.PubMed Gil-Ortega M, Garidou L, Barreau C, Maumus M, Breasson L, Tavernier G, et al. Native adipose stromal cells egress from adipose tissue in vivo: evidence during lymph node activation. Stem Cells. 2013;31:1309–20.PubMed
53.
go back to reference Girousse A, Gil-Ortega M, Bourlier V, Bergeaud C, Sastourné-Arrey Q, Moro C, et al. The release of adipose stromal cells from subcutaneous adipose tissue regulates ectopic intramuscular adipocyte deposition. Cell Rep. 2019;27:323-333.e5.PubMed Girousse A, Gil-Ortega M, Bourlier V, Bergeaud C, Sastourné-Arrey Q, Moro C, et al. The release of adipose stromal cells from subcutaneous adipose tissue regulates ectopic intramuscular adipocyte deposition. Cell Rep. 2019;27:323-333.e5.PubMed
54.
go back to reference Scheller EL, Cawthorn WP, Burr AA, Horowitz MC, MacDougald OA. Marrow adipose tissue: trimming the fat. Trends Endocrinol Metab. 2016;27:392–403.PubMedCentralPubMed Scheller EL, Cawthorn WP, Burr AA, Horowitz MC, MacDougald OA. Marrow adipose tissue: trimming the fat. Trends Endocrinol Metab. 2016;27:392–403.PubMedCentralPubMed
55.
go back to reference Suchacki KJ, Tavares AAS, Mattiucci D, Scheller EL, Papanastasiou G, Gray C, et al. Bone marrow adipose tissue is a unique adipose subtype with distinct roles in glucose homeostasis. Nat Commun. 2020;11:3097.PubMedCentralPubMed Suchacki KJ, Tavares AAS, Mattiucci D, Scheller EL, Papanastasiou G, Gray C, et al. Bone marrow adipose tissue is a unique adipose subtype with distinct roles in glucose homeostasis. Nat Commun. 2020;11:3097.PubMedCentralPubMed
56.
go back to reference Cawthorn WP, Scheller EL, Learman BS, Parlee SD, Simon BR, Mori H, et al. Bone marrow adipose tissue is an endocrine organ that contributes to increased circulating adiponectin during caloric restriction. Cell Metab. 2014;20:368–75.PubMedCentralPubMed Cawthorn WP, Scheller EL, Learman BS, Parlee SD, Simon BR, Mori H, et al. Bone marrow adipose tissue is an endocrine organ that contributes to increased circulating adiponectin during caloric restriction. Cell Metab. 2014;20:368–75.PubMedCentralPubMed
57.
go back to reference Sulston RJ, Cawthorn WP. Bone marrow adipose tissue as an endocrine organ: close to the bone? Hormone molecular biology and clinical investigation. De Gruyter. 2016;28:21–38. Sulston RJ, Cawthorn WP. Bone marrow adipose tissue as an endocrine organ: close to the bone? Hormone molecular biology and clinical investigation. De Gruyter. 2016;28:21–38.
58.
go back to reference Scheller EL, Khandaker S, Learman BS, Cawthorn WP, Anderson LM, Pham HA, et al. Bone marrow adipocytes resist lipolysis and remodeling in response to β-adrenergic stimulation. Bone. 2019;118:32–41.PubMed Scheller EL, Khandaker S, Learman BS, Cawthorn WP, Anderson LM, Pham HA, et al. Bone marrow adipocytes resist lipolysis and remodeling in response to β-adrenergic stimulation. Bone. 2019;118:32–41.PubMed
59.
go back to reference Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–7.PubMed Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143–7.PubMed
60.
go back to reference Tikhonova AN, Dolgalev I, Hu H, Sivaraj KK, Hoxha E, Cuesta-Domínguez Á, et al. The bone marrow microenvironment at single-cell resolution. Nature. 2019;569:222–8.PubMedCentralPubMed Tikhonova AN, Dolgalev I, Hu H, Sivaraj KK, Hoxha E, Cuesta-Domínguez Á, et al. The bone marrow microenvironment at single-cell resolution. Nature. 2019;569:222–8.PubMedCentralPubMed
61.
go back to reference Yue R, Zhou BO, Shimada IS, Zhao Z, Morrison SJ. Leptin receptor promotes adipogenesis and reduces osteogenesis by regulating mesenchymal stromal cells in adult bone marrow. Cell Stem Cell. 2016;18:782–96.PubMed Yue R, Zhou BO, Shimada IS, Zhao Z, Morrison SJ. Leptin receptor promotes adipogenesis and reduces osteogenesis by regulating mesenchymal stromal cells in adult bone marrow. Cell Stem Cell. 2016;18:782–96.PubMed
62.
go back to reference Mizoguchi T, Pinho S, Ahmed J, Kunisaki Y, Hanoun M, Mendelson A, et al. Osterix marks distinct waves of primitive and definitive stromal progenitors during bone marrow development. Dev Cell. 2014;29:340–9.PubMedCentralPubMed Mizoguchi T, Pinho S, Ahmed J, Kunisaki Y, Hanoun M, Mendelson A, et al. Osterix marks distinct waves of primitive and definitive stromal progenitors during bone marrow development. Dev Cell. 2014;29:340–9.PubMedCentralPubMed
63.
go back to reference Ambrosi TH, Scialdone A, Graja A, Gohlke S, Jank AM, Bocian C, et al. Adipocyte accumulation in the bone marrow during obesity and aging impairs stem cell-based hematopoietic and bone regeneration. Cell Stem Cell. 2017;20:771-784.e6.PubMedCentralPubMed Ambrosi TH, Scialdone A, Graja A, Gohlke S, Jank AM, Bocian C, et al. Adipocyte accumulation in the bone marrow during obesity and aging impairs stem cell-based hematopoietic and bone regeneration. Cell Stem Cell. 2017;20:771-784.e6.PubMedCentralPubMed
64.
go back to reference Zhou BO, Yu H, Yue R, Zhao Z, Rios JJ, Naveiras O, et al. Bone marrow adipocytes promote the regeneration of stem cells and hematopoiesis by secreting SCF. Nat Cell Biol. 2017;19:891–903.PubMedCentralPubMed Zhou BO, Yu H, Yue R, Zhao Z, Rios JJ, Naveiras O, et al. Bone marrow adipocytes promote the regeneration of stem cells and hematopoiesis by secreting SCF. Nat Cell Biol. 2017;19:891–903.PubMedCentralPubMed
65.
go back to reference Bowers E, Singer K. Obesity-induced inflammation: the impact of the hematopoietic stem cell niche. JCI Insight. 2021;6:145295.PubMed Bowers E, Singer K. Obesity-induced inflammation: the impact of the hematopoietic stem cell niche. JCI Insight. 2021;6:145295.PubMed
66.
go back to reference Naveiras O, Nardi V, Wenzel PL, Hauschka PV, Fahey F, Daley GQ. Bone-marrow adipocytes as negative regulators of the haematopoietic microenvironment. Nature. 2009;460:259–63.PubMedCentralPubMed Naveiras O, Nardi V, Wenzel PL, Hauschka PV, Fahey F, Daley GQ. Bone-marrow adipocytes as negative regulators of the haematopoietic microenvironment. Nature. 2009;460:259–63.PubMedCentralPubMed
67.
go back to reference Bilwani FA, Knight KL. Adipocyte-derived soluble factor(s) inhibits early stages of B lymphopoiesis. J Immunol. 2012;189:4379–86.PubMed Bilwani FA, Knight KL. Adipocyte-derived soluble factor(s) inhibits early stages of B lymphopoiesis. J Immunol. 2012;189:4379–86.PubMed
68.
go back to reference Kennedy DE, Knight KL. Inhibition of B lymphopoiesis by adipocytes and IL-1–producing myeloid-derived suppressor cells. J Immunol. 2015;195:2666–74.PubMed Kennedy DE, Knight KL. Inhibition of B lymphopoiesis by adipocytes and IL-1–producing myeloid-derived suppressor cells. J Immunol. 2015;195:2666–74.PubMed
69.
go back to reference Kennedy DE, Knight KL. Inflammatory changes in bone marrow microenvironment associated with declining B lymphopoiesis. JI. 2017;198:3471–9. Kennedy DE, Knight KL. Inflammatory changes in bone marrow microenvironment associated with declining B lymphopoiesis. JI. 2017;198:3471–9.
70.
go back to reference Griffin C, Eter L, Lanzetta N, Abrishami S, Varghese M, McKernan K, et al. TLR4, TRIF, and MyD88 are essential for myelopoiesis and CD11c+ adipose tissue macrophage production in obese mice. J Biol Chem. 2018;293:8775–86.PubMedCentralPubMed Griffin C, Eter L, Lanzetta N, Abrishami S, Varghese M, McKernan K, et al. TLR4, TRIF, and MyD88 are essential for myelopoiesis and CD11c+ adipose tissue macrophage production in obese mice. J Biol Chem. 2018;293:8775–86.PubMedCentralPubMed
71.
go back to reference Bredella MA, Gill CM, Gerweck AV, Landa MG, Kumar V, Daley SM, et al. Ectopic and serum lipid levels are positively associated with bone marrow fat in obesity. Radiology. 2013;269:534–41.PubMedCentralPubMed Bredella MA, Gill CM, Gerweck AV, Landa MG, Kumar V, Daley SM, et al. Ectopic and serum lipid levels are positively associated with bone marrow fat in obesity. Radiology. 2013;269:534–41.PubMedCentralPubMed
72.
go back to reference Bredella MA, Torriani M, Ghomi RH, Thomas BJ, Brick DJ, Gerweck AV, et al. Vertebral bone marrow fat is positively associated with visceral fat and inversely associated with IGF-1 in obese women. Obesity. 2011;19:49–53.PubMed Bredella MA, Torriani M, Ghomi RH, Thomas BJ, Brick DJ, Gerweck AV, et al. Vertebral bone marrow fat is positively associated with visceral fat and inversely associated with IGF-1 in obese women. Obesity. 2011;19:49–53.PubMed
73.
go back to reference Hasic D, Lorbeer R, Bertheau RC, Machann J, Rospleszcz S, Nattenmüller J, et al. Vertebral bone marrow fat is independently associated to VAT but not to SAT: KORA FF4—whole-body MR imaging in a population-based cohort. Nutrients. 2020;12:1527.PubMedCentral Hasic D, Lorbeer R, Bertheau RC, Machann J, Rospleszcz S, Nattenmüller J, et al. Vertebral bone marrow fat is independently associated to VAT but not to SAT: KORA FF4—whole-body MR imaging in a population-based cohort. Nutrients. 2020;12:1527.PubMedCentral
74.
go back to reference Fazeli PK, Bredella MA, Pachon-Peña G, Zhao W, Zhang X, Faje AT, Resulaj M, Polineni SP, Holmes TM, Lee H, O'Donnell EK, MacDougald OA, Horowitz MC, Rosen CJ, Klibanski A. The dynamics of human bone marrow adipose tissue in response to feeding and fasting. JCI Insight. 22 Jun 2021;6(12):e138636. Fazeli PK, Bredella MA, Pachon-Peña G, Zhao W, Zhang X, Faje AT, Resulaj M, Polineni SP, Holmes TM, Lee H, O'Donnell EK, MacDougald OA, Horowitz MC, Rosen CJ, Klibanski A. The dynamics of human bone marrow adipose tissue in response to feeding and fasting. JCI Insight. 22 Jun 2021;6(12):e138636.
75.
go back to reference Justesen J, Stenderup K, Ebbesen EN, Mosekilde L, Steiniche T, Kassem M. Adipocyte tissue volume in bone marrow is increased with aging and in patients with osteoporosis. Biogerontology. 2001;2:165–71.PubMed Justesen J, Stenderup K, Ebbesen EN, Mosekilde L, Steiniche T, Kassem M. Adipocyte tissue volume in bone marrow is increased with aging and in patients with osteoporosis. Biogerontology. 2001;2:165–71.PubMed
76.
go back to reference Shen W, Chen J, Gantz M, Punyanitya M, Heymsfield SB, Gallagher D, et al. MRI-measured pelvic bone marrow adipose tissue is inversely related to DXA-measured bone mineral in younger and older adults. Eur J Clin Nutr. 2012;66:983–8.PubMedCentralPubMed Shen W, Chen J, Gantz M, Punyanitya M, Heymsfield SB, Gallagher D, et al. MRI-measured pelvic bone marrow adipose tissue is inversely related to DXA-measured bone mineral in younger and older adults. Eur J Clin Nutr. 2012;66:983–8.PubMedCentralPubMed
77.
go back to reference Bani Hassan E, Demontiero O, Vogrin S, Ng A, Duque G. Marrow adipose tissue in older men: association with visceral and subcutaneous fat, bone volume, metabolism, and inflammation. Calcif Tissue Int. 2018;103:164–74.PubMed Bani Hassan E, Demontiero O, Vogrin S, Ng A, Duque G. Marrow adipose tissue in older men: association with visceral and subcutaneous fat, bone volume, metabolism, and inflammation. Calcif Tissue Int. 2018;103:164–74.PubMed
78.
go back to reference Fazeli PK, Faje AT, Bredella MA, Polineni S, Russell S, Resulaj M, et al. Changes in marrow adipose tissue with short-term changes in weight in premenopausal women with anorexia nervosa. Eur J Endocrinol. 2019;180:189–99.PubMedCentralPubMed Fazeli PK, Faje AT, Bredella MA, Polineni S, Russell S, Resulaj M, et al. Changes in marrow adipose tissue with short-term changes in weight in premenopausal women with anorexia nervosa. Eur J Endocrinol. 2019;180:189–99.PubMedCentralPubMed
79.
go back to reference Madonna R, Massaro M, Scoditti E, Pescetelli I, De Caterina R. The epicardial adipose tissue and the coronary arteries: dangerous liaisons. Cardiovasc Res. 2019;115:1013–25.PubMed Madonna R, Massaro M, Scoditti E, Pescetelli I, De Caterina R. The epicardial adipose tissue and the coronary arteries: dangerous liaisons. Cardiovasc Res. 2019;115:1013–25.PubMed
80.
go back to reference Ayton SL, Gulsin GS, McCann GP, Moss AJ. Epicardial adipose tissue in obesity-related cardiac dysfunction. Heart. 2021;heartjnl-2020-318242. Ayton SL, Gulsin GS, McCann GP, Moss AJ. Epicardial adipose tissue in obesity-related cardiac dysfunction. Heart. 2021;heartjnl-2020-318242.
81.
go back to reference Rosito GA, Massaro JM, Hoffmann U, Ruberg FL, Mahabadi AA, Vasan RS, et al. Pericardial fat, visceral abdominal fat, cardiovascular disease risk factors, and vascular calcification in a community-based sample: the framingham heart study. Circulation. 2008;117:605–13.PubMed Rosito GA, Massaro JM, Hoffmann U, Ruberg FL, Mahabadi AA, Vasan RS, et al. Pericardial fat, visceral abdominal fat, cardiovascular disease risk factors, and vascular calcification in a community-based sample: the framingham heart study. Circulation. 2008;117:605–13.PubMed
82.
go back to reference Kim BJ, Kang JG, Lee SH, Lee JY, Sung KC, Kim BS, et al. Relationship of echocardiographic epicardial fat thickness and epicardial fat volume by computed tomography with coronary artery calcification: data from the CAESAR study. Arch Med Res. 2017;48:352–9.PubMed Kim BJ, Kang JG, Lee SH, Lee JY, Sung KC, Kim BS, et al. Relationship of echocardiographic epicardial fat thickness and epicardial fat volume by computed tomography with coronary artery calcification: data from the CAESAR study. Arch Med Res. 2017;48:352–9.PubMed
83.
go back to reference Alexopoulos N, McLean DS, Janik M, Arepalli CD, Stillman AE, Raggi P. Epicardial adipose tissue and coronary artery plaque characteristics. Atherosclerosis. 2010;210:150–4.PubMed Alexopoulos N, McLean DS, Janik M, Arepalli CD, Stillman AE, Raggi P. Epicardial adipose tissue and coronary artery plaque characteristics. Atherosclerosis. 2010;210:150–4.PubMed
84.
go back to reference Powell-Wiley TM, Poirier P, Burke LE, Després JP, Gordon-Larsen P, Lavie CJ, et al. Obesity and cardiovascular disease: a scientific statement from the American heart association. Circulation. 2021;143:e984-1010.PubMedCentralPubMed Powell-Wiley TM, Poirier P, Burke LE, Després JP, Gordon-Larsen P, Lavie CJ, et al. Obesity and cardiovascular disease: a scientific statement from the American heart association. Circulation. 2021;143:e984-1010.PubMedCentralPubMed
85.
go back to reference Rabkin SW, Campbell H. Comparison of reducing epicardial fat by exercise, diet or bariatric surgery weight loss strategies: a systematic review and meta-analysis. Obes Rev. 2015;16:406–15.PubMed Rabkin SW, Campbell H. Comparison of reducing epicardial fat by exercise, diet or bariatric surgery weight loss strategies: a systematic review and meta-analysis. Obes Rev. 2015;16:406–15.PubMed
86.
go back to reference Goeller M, Achenbach S, Cadet S, Kwan AC, Commandeur F, Slomka PJ, et al. Pericoronary adipose tissue computed tomography attenuation and high-risk plaque characteristics in acute coronary syndrome compared with stable coronary artery disease. JAMA Cardiol. 2018;3:858–63.PubMedCentralPubMed Goeller M, Achenbach S, Cadet S, Kwan AC, Commandeur F, Slomka PJ, et al. Pericoronary adipose tissue computed tomography attenuation and high-risk plaque characteristics in acute coronary syndrome compared with stable coronary artery disease. JAMA Cardiol. 2018;3:858–63.PubMedCentralPubMed
87.
go back to reference Patel VB, Shah S, Verma S, Oudit GY. Epicardial adipose tissue as a metabolic transducer: role in heart failure and coronary artery disease. Heart Fail Rev. 2017;22:889–902.PubMed Patel VB, Shah S, Verma S, Oudit GY. Epicardial adipose tissue as a metabolic transducer: role in heart failure and coronary artery disease. Heart Fail Rev. 2017;22:889–902.PubMed
88.
go back to reference Yudkin JS, Eringa E, Stehouwer CDA. Vasocrine signalling from perivascular fat: a mechanism linking insulin resistance to vascular disease. Lancet. 2005;365:1817–20.PubMed Yudkin JS, Eringa E, Stehouwer CDA. Vasocrine signalling from perivascular fat: a mechanism linking insulin resistance to vascular disease. Lancet. 2005;365:1817–20.PubMed
89.
go back to reference Vilahur G, Ben-Aicha S, Badimon L. New insights into the role of adipose tissue in thrombosis. Cardiovasc Res. 2017;113:1046–54.PubMed Vilahur G, Ben-Aicha S, Badimon L. New insights into the role of adipose tissue in thrombosis. Cardiovasc Res. 2017;113:1046–54.PubMed
90.
go back to reference Jiang Z, Feng T, Lu Z, Wei Y, Meng J, Lin CP, et al. PDGFRb+ mesenchymal cells, but not NG2+ mural cells, contribute to cardiac fat. Cell Rep. 2021;34:108697.PubMed Jiang Z, Feng T, Lu Z, Wei Y, Meng J, Lin CP, et al. PDGFRb+ mesenchymal cells, but not NG2+ mural cells, contribute to cardiac fat. Cell Rep. 2021;34:108697.PubMed
91.
go back to reference Lombardi R, Chen SN, Ruggiero A, Gurha P, Czernuszewicz GZ, Willerson JT, et al. Cardiac fibro-adipocyte progenitors express desmosome proteins and preferentially differentiate to adipocytes upon deletion of the desmoplakin gene. Circ Res. 2016;119:41–54.PubMedCentralPubMed Lombardi R, Chen SN, Ruggiero A, Gurha P, Czernuszewicz GZ, Willerson JT, et al. Cardiac fibro-adipocyte progenitors express desmosome proteins and preferentially differentiate to adipocytes upon deletion of the desmoplakin gene. Circ Res. 2016;119:41–54.PubMedCentralPubMed
92.
go back to reference Soliman H, Rossi FMV. Cardiac fibroblast diversity in health and disease. Matrix Biol. 2020;91–92:75–91.PubMed Soliman H, Rossi FMV. Cardiac fibroblast diversity in health and disease. Matrix Biol. 2020;91–92:75–91.PubMed
93.
go back to reference Moskalev A, Stambler I, Caruso C. Innate and adaptive immunity in aging and longevity: the foundation of resilience. Aging Dis. 2020;11:1363–73.PubMedCentralPubMed Moskalev A, Stambler I, Caruso C. Innate and adaptive immunity in aging and longevity: the foundation of resilience. Aging Dis. 2020;11:1363–73.PubMedCentralPubMed
94.
go back to reference Yang H, Youm YH, Vandanmagsar B, Rood J, Kumar KG, Butler AA, et al. Obesity accelerates thymic aging. Blood. 2009;114:3803–12.PubMedCentralPubMed Yang H, Youm YH, Vandanmagsar B, Rood J, Kumar KG, Butler AA, et al. Obesity accelerates thymic aging. Blood. 2009;114:3803–12.PubMedCentralPubMed
95.
go back to reference Sreenivasan J, Schlenner S, Franckaert D, Dooley J, Liston A. The thymoprotective function of leptin is indirectly mediated via suppression of obesity. Immunology. 2015;146:122–9.PubMedCentralPubMed Sreenivasan J, Schlenner S, Franckaert D, Dooley J, Liston A. The thymoprotective function of leptin is indirectly mediated via suppression of obesity. Immunology. 2015;146:122–9.PubMedCentralPubMed
96.
go back to reference Castro LL, Kitoko JZ, Xisto DG, Olsen PC, Guedes HLM, Morales MM, et al. Multiple doses of adipose tissue-derived mesenchymal stromal cells induce immunosuppression in experimental asthma. Stem Cells Transl Med. 2020;9:250–60.PubMed Castro LL, Kitoko JZ, Xisto DG, Olsen PC, Guedes HLM, Morales MM, et al. Multiple doses of adipose tissue-derived mesenchymal stromal cells induce immunosuppression in experimental asthma. Stem Cells Transl Med. 2020;9:250–60.PubMed
97.
go back to reference Lazennec G, Lam PY. Recent discoveries concerning the tumor - mesenchymal stem cell interactions. Biochim Biophys Acta. 2016;1866:290–9.PubMed Lazennec G, Lam PY. Recent discoveries concerning the tumor - mesenchymal stem cell interactions. Biochim Biophys Acta. 2016;1866:290–9.PubMed
98.
go back to reference Freese KE, Kokai L, Edwards RP, Philips BJ, Sheikh MA, Kelley J, et al. Adipose-derived stems cells and their role in human cancer development, growth, progression, and metastasis: a systematic review. Cancer Res. 2015;75:1161–8.PubMed Freese KE, Kokai L, Edwards RP, Philips BJ, Sheikh MA, Kelley J, et al. Adipose-derived stems cells and their role in human cancer development, growth, progression, and metastasis: a systematic review. Cancer Res. 2015;75:1161–8.PubMed
99.
go back to reference Strong AL, Burow ME, Gimble JM, Bunnell BA. Concise review: The obesity cancer paradigm: exploration of the interactions and crosstalk with adipose stem cells. Stem Cells. 2015;33:318–26.PubMedCentralPubMed Strong AL, Burow ME, Gimble JM, Bunnell BA. Concise review: The obesity cancer paradigm: exploration of the interactions and crosstalk with adipose stem cells. Stem Cells. 2015;33:318–26.PubMedCentralPubMed
100.
go back to reference Trevellin E, Scarpa M, Carraro A, Lunardi F, Kotsafti A, Porzionato A, et al. Esophageal adenocarcinoma and obesity: peritumoral adipose tissue plays a role in lymph node invasion. Oncotarget. 2015;6:11203–15.PubMedCentralPubMed Trevellin E, Scarpa M, Carraro A, Lunardi F, Kotsafti A, Porzionato A, et al. Esophageal adenocarcinoma and obesity: peritumoral adipose tissue plays a role in lymph node invasion. Oncotarget. 2015;6:11203–15.PubMedCentralPubMed
101.
go back to reference Colleluori G, Perugini J, Barbatelli G, Cinti S. Mammary gland adipocytes in lactation cycle, obesity and breast cancer. Rev Endocr Metab Disord. 2021;22:241–55.PubMedCentralPubMed Colleluori G, Perugini J, Barbatelli G, Cinti S. Mammary gland adipocytes in lactation cycle, obesity and breast cancer. Rev Endocr Metab Disord. 2021;22:241–55.PubMedCentralPubMed
102.
go back to reference Carraro A, Trevellin E, Fassan M, Kotsafti A, Lunardi F, Porzionato A, et al. Esophageal adenocarcinoma microenvironment: peritumoral adipose tissue effects associated with chemoresistance. Cancer Sci. 2017;108:2393–404.PubMedCentralPubMed Carraro A, Trevellin E, Fassan M, Kotsafti A, Lunardi F, Porzionato A, et al. Esophageal adenocarcinoma microenvironment: peritumoral adipose tissue effects associated with chemoresistance. Cancer Sci. 2017;108:2393–404.PubMedCentralPubMed
103.
go back to reference Wang Z, Aguilar EG, Luna JI, Dunai C, Khuat LT, Le CT, et al. Paradoxical effects of obesity on T cell function during tumor progression and PD-1 checkpoint blockade. Nat Med. 2019;25:141–51.PubMed Wang Z, Aguilar EG, Luna JI, Dunai C, Khuat LT, Le CT, et al. Paradoxical effects of obesity on T cell function during tumor progression and PD-1 checkpoint blockade. Nat Med. 2019;25:141–51.PubMed
104.
go back to reference Ringel AE, Drijvers JM, Baker GJ, Catozzi A, García-Cañaveras JC, Gassaway BM, et al. Obesity shapes metabolism in the tumor microenvironment to suppress anti-tumor immunity. Cell. 2020;183:1848-1866.e26.PubMedCentralPubMed Ringel AE, Drijvers JM, Baker GJ, Catozzi A, García-Cañaveras JC, Gassaway BM, et al. Obesity shapes metabolism in the tumor microenvironment to suppress anti-tumor immunity. Cell. 2020;183:1848-1866.e26.PubMedCentralPubMed
106.
go back to reference Brier S. Finding an information concept suited for a universal theory of information. Prog Biophys Mol Biol. 2015;119:622–33.PubMed Brier S. Finding an information concept suited for a universal theory of information. Prog Biophys Mol Biol. 2015;119:622–33.PubMed
108.
go back to reference Collins GS, Moons KGM. Reporting of artificial intelligence prediction models. Lancet. 2019;393:1577–9.PubMed Collins GS, Moons KGM. Reporting of artificial intelligence prediction models. Lancet. 2019;393:1577–9.PubMed
109.
go back to reference Alcocer-Cuarón C, Rivera AL, Castaño VM. Hierarchical structure of biological systems: a bioengineering approach. Bioengineered. 2014;5:73–9.PubMed Alcocer-Cuarón C, Rivera AL, Castaño VM. Hierarchical structure of biological systems: a bioengineering approach. Bioengineered. 2014;5:73–9.PubMed
110.
go back to reference Gesta S, Tseng Y-H, Kahn CR. Developmental origin of fat: tracking obesity to its source. Cell. 2007;131:242–56.PubMed Gesta S, Tseng Y-H, Kahn CR. Developmental origin of fat: tracking obesity to its source. Cell. 2007;131:242–56.PubMed
111.
go back to reference Pagano C, Dorigo A, Nisoli E, Tonello C, Calcagno A, Tami V, et al. Role of insulin and free fatty acids in the regulation of ob gene expression and plasma leptin in normal rats. Obes Res. 2004;12:2062–9.PubMed Pagano C, Dorigo A, Nisoli E, Tonello C, Calcagno A, Tami V, et al. Role of insulin and free fatty acids in the regulation of ob gene expression and plasma leptin in normal rats. Obes Res. 2004;12:2062–9.PubMed
112.
go back to reference Zamboni M, Zoico E, Fantin F, Panourgia MP, Di Francesco V, Tosoni P, et al. Relation between leptin and the metabolic syndrome in elderly women. J Gerontol A Biol Sci Med Sci. 2004;59:396–400.PubMed Zamboni M, Zoico E, Fantin F, Panourgia MP, Di Francesco V, Tosoni P, et al. Relation between leptin and the metabolic syndrome in elderly women. J Gerontol A Biol Sci Med Sci. 2004;59:396–400.PubMed
113.
go back to reference Nisoli E, Carruba MO, Tonello C, Macor C, Federspil G, Vettor R. Induction of fatty acid translocase/CD36, peroxisome proliferator-activated receptor-gamma2, leptin, uncoupling proteins 2 and 3, and tumor necrosis factor-alpha gene expression in human subcutaneous fat by lipid infusion. Diabetes. 2000;49:319–24.PubMed Nisoli E, Carruba MO, Tonello C, Macor C, Federspil G, Vettor R. Induction of fatty acid translocase/CD36, peroxisome proliferator-activated receptor-gamma2, leptin, uncoupling proteins 2 and 3, and tumor necrosis factor-alpha gene expression in human subcutaneous fat by lipid infusion. Diabetes. 2000;49:319–24.PubMed
114.
go back to reference Fabris R, Nisoli E, Lombardi AM, Tonello C, Serra R, Granzotto M, et al. Preferential channeling of energy fuels toward fat rather than muscle during high free fatty acid availability in rats. Diabetes. 2001;50:601–8.PubMed Fabris R, Nisoli E, Lombardi AM, Tonello C, Serra R, Granzotto M, et al. Preferential channeling of energy fuels toward fat rather than muscle during high free fatty acid availability in rats. Diabetes. 2001;50:601–8.PubMed
115.
go back to reference Scheele C, Wolfrum C. Brown adipose crosstalk in tissue plasticity and human metabolism. Endocr Rev. 2020;41. Scheele C, Wolfrum C. Brown adipose crosstalk in tissue plasticity and human metabolism. Endocr Rev. 2020;41.
116.
go back to reference Pedersen BK, Febbraio MA. Muscles, exercise and obesity: skeletal muscle as a secretory organ. Nat Rev Endocrinol. 2012;8:457–65.PubMed Pedersen BK, Febbraio MA. Muscles, exercise and obesity: skeletal muscle as a secretory organ. Nat Rev Endocrinol. 2012;8:457–65.PubMed
117.
go back to reference Colaianni G, Cuscito C, Mongelli T, Pignataro P, Buccoliero C, Liu P, et al. The myokine irisin increases cortical bone mass. Proc Natl Acad Sci U S A. 2015;112:12157–62.PubMedCentralPubMed Colaianni G, Cuscito C, Mongelli T, Pignataro P, Buccoliero C, Liu P, et al. The myokine irisin increases cortical bone mass. Proc Natl Acad Sci U S A. 2015;112:12157–62.PubMedCentralPubMed
118.
go back to reference Colaianni G, Mongelli T, Cuscito C, Pignataro P, Lippo L, Spiro G, et al. Irisin prevents and restores bone loss and muscle atrophy in hind-limb suspended mice. Sci Rep. 2017;7:2811.PubMedCentralPubMed Colaianni G, Mongelli T, Cuscito C, Pignataro P, Lippo L, Spiro G, et al. Irisin prevents and restores bone loss and muscle atrophy in hind-limb suspended mice. Sci Rep. 2017;7:2811.PubMedCentralPubMed
119.
go back to reference Laurens C, Bergouignan A, Moro C. Exercise-released myokines in the control of energy metabolism. Front Physiol. 2020;11:91.PubMedCentralPubMed Laurens C, Bergouignan A, Moro C. Exercise-released myokines in the control of energy metabolism. Front Physiol. 2020;11:91.PubMedCentralPubMed
120.
go back to reference Vinel C, Lukjanenko L, Batut A, Deleruyelle S, Pradère J-P, Le Gonidec S, et al. The exerkine apelin reverses age-associated sarcopenia. Nat Med. 2018;24:1360–71.PubMed Vinel C, Lukjanenko L, Batut A, Deleruyelle S, Pradère J-P, Le Gonidec S, et al. The exerkine apelin reverses age-associated sarcopenia. Nat Med. 2018;24:1360–71.PubMed
121.
go back to reference McPherron AC, Lawler AM, Lee SJ. Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature. 1997;387:83–90.PubMed McPherron AC, Lawler AM, Lee SJ. Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature. 1997;387:83–90.PubMed
122.
go back to reference Palermo A, Strollo R, Maddaloni E, Tuccinardi D, D’Onofrio L, Briganti SI, et al. Irisin is associated with osteoporotic fractures independently of bone mineral density, body composition or daily physical activity. Clin Endocrinol. 2015;82:615–9. Palermo A, Strollo R, Maddaloni E, Tuccinardi D, D’Onofrio L, Briganti SI, et al. Irisin is associated with osteoporotic fractures independently of bone mineral density, body composition or daily physical activity. Clin Endocrinol. 2015;82:615–9.
123.
go back to reference Park HS, Kim HC, Zhang D, Yeom H, Lim SK. The novel myokine irisin: clinical implications and potential role as a biomarker for sarcopenia in postmenopausal women. Endocrine. 2019;64:341–8.PubMed Park HS, Kim HC, Zhang D, Yeom H, Lim SK. The novel myokine irisin: clinical implications and potential role as a biomarker for sarcopenia in postmenopausal women. Endocrine. 2019;64:341–8.PubMed
124.
go back to reference Ducy P, Desbois C, Boyce B, Pinero G, Story B, Dunstan C, et al. Increased bone formation in osteocalcin-deficient mice. Nature. 1996;382:448–52.PubMed Ducy P, Desbois C, Boyce B, Pinero G, Story B, Dunstan C, et al. Increased bone formation in osteocalcin-deficient mice. Nature. 1996;382:448–52.PubMed
125.
go back to reference Foresta C, Strapazzon G, De Toni L, Gianesello L, Calcagno A, Pilon C, et al. Evidence for osteocalcin production by adipose tissue and its role in human metabolism. J Clin Endocrinol Metab. 2010;95:3502–6.PubMed Foresta C, Strapazzon G, De Toni L, Gianesello L, Calcagno A, Pilon C, et al. Evidence for osteocalcin production by adipose tissue and its role in human metabolism. J Clin Endocrinol Metab. 2010;95:3502–6.PubMed
126.
go back to reference Lee NK, Sowa H, Hinoi E, Ferron M, Ahn JD, Confavreux C, et al. Endocrine regulation of energy metabolism by the skeleton. Cell. 2007;130:456–69.PubMedCentralPubMed Lee NK, Sowa H, Hinoi E, Ferron M, Ahn JD, Confavreux C, et al. Endocrine regulation of energy metabolism by the skeleton. Cell. 2007;130:456–69.PubMedCentralPubMed
127.
go back to reference Mera P, Laue K, Ferron M, Confavreux C, Wei J, Galán-Díez M, et al. Osteocalcin signaling in myofibers is necessary and sufficient for optimum adaptation to exercise. Cell Metab. 2016;23:1078–92.PubMedCentralPubMed Mera P, Laue K, Ferron M, Confavreux C, Wei J, Galán-Díez M, et al. Osteocalcin signaling in myofibers is necessary and sufficient for optimum adaptation to exercise. Cell Metab. 2016;23:1078–92.PubMedCentralPubMed
128.
go back to reference Mera P, Laue K, Wei J, Berger JM, Karsenty G. Osteocalcin is necessary and sufficient to maintain muscle mass in older mice. Mol Metab. 2016;5:1042–7.PubMedCentralPubMed Mera P, Laue K, Wei J, Berger JM, Karsenty G. Osteocalcin is necessary and sufficient to maintain muscle mass in older mice. Mol Metab. 2016;5:1042–7.PubMedCentralPubMed
129.
go back to reference Ponti F, Santoro A, Mercatelli D, Gasperini C, Conte M, Martucci M, et al. Aging and imaging assessment of body composition: from fat to facts. Front Endocrinol. 2019;10:861. Ponti F, Santoro A, Mercatelli D, Gasperini C, Conte M, Martucci M, et al. Aging and imaging assessment of body composition: from fat to facts. Front Endocrinol. 2019;10:861.
130.
go back to reference Hou J, He C, He W, Yang M, Luo X, Li C. Obesity and bone health: a complex link. Front Cell Dev Biol. 2018;8:600181. Hou J, He C, He W, Yang M, Luo X, Li C. Obesity and bone health: a complex link. Front Cell Dev Biol. 2018;8:600181.
131.
go back to reference Fassio A, Idolazzi L, Rossini M, Gatti D, Adami G, Giollo A, et al. The obesity paradox and osteoporosis. Eat Weight Disord. 2018;23:293–302.PubMed Fassio A, Idolazzi L, Rossini M, Gatti D, Adami G, Giollo A, et al. The obesity paradox and osteoporosis. Eat Weight Disord. 2018;23:293–302.PubMed
132.
go back to reference Lecka-Czernik B, Stechschulte LA, Czernik PJ, Dowling AR. High bone mass in adult mice with diet-induced obesity results from a combination of initial increase in bone mass followed by attenuation in bone formation; implications for high bone mass and decreased bone quality in obesity. Mol Cell Endocrinol. 2015;410:35–41.PubMed Lecka-Czernik B, Stechschulte LA, Czernik PJ, Dowling AR. High bone mass in adult mice with diet-induced obesity results from a combination of initial increase in bone mass followed by attenuation in bone formation; implications for high bone mass and decreased bone quality in obesity. Mol Cell Endocrinol. 2015;410:35–41.PubMed
133.
go back to reference Luo H, Mu WC, Karki R, Chiang H-H, Mohrin M, Shin JJ, et al. Mitochondrial stress-initiated aberrant activation of the NLRP3 inflammasome regulates the functional deterioration of hematopoietic stem cell aging. Cell Rep. 2019;26:945-954.e4.PubMedCentralPubMed Luo H, Mu WC, Karki R, Chiang H-H, Mohrin M, Shin JJ, et al. Mitochondrial stress-initiated aberrant activation of the NLRP3 inflammasome regulates the functional deterioration of hematopoietic stem cell aging. Cell Rep. 2019;26:945-954.e4.PubMedCentralPubMed
134.
go back to reference Mohrin M, Shin J, Liu Y, Brown K, Luo H, Xi Y, et al. Stem cell aging. A mitochondrial UPR-mediated metabolic checkpoint regulates hematopoietic stem cell aging. Science. 2015;347:1374–7.PubMedCentralPubMed Mohrin M, Shin J, Liu Y, Brown K, Luo H, Xi Y, et al. Stem cell aging. A mitochondrial UPR-mediated metabolic checkpoint regulates hematopoietic stem cell aging. Science. 2015;347:1374–7.PubMedCentralPubMed
135.
go back to reference Zhang H, Ryu D, Wu Y, Gariani K, Wang X, Luan P, et al. NAD+ repletion improves mitochondrial and stem cell function and enhances life span in mice. Science. 2016;352:1436–43.PubMed Zhang H, Ryu D, Wu Y, Gariani K, Wang X, Luan P, et al. NAD+ repletion improves mitochondrial and stem cell function and enhances life span in mice. Science. 2016;352:1436–43.PubMed
136.
go back to reference Yoshino M, Yoshino J, Kayser BD, Patti GJ, Franczyk MP, Mills KF, et al. Nicotinamide mononucleotide increases muscle insulin sensitivity in prediabetic women. Science. 2021;372:1224–9.PubMedCentralPubMed Yoshino M, Yoshino J, Kayser BD, Patti GJ, Franczyk MP, Mills KF, et al. Nicotinamide mononucleotide increases muscle insulin sensitivity in prediabetic women. Science. 2021;372:1224–9.PubMedCentralPubMed
137.
go back to reference Sanna M, Borgo C, Compagnin C, Favaretto F, Vindigni V, Trento M, et al. White adipose tissue expansion in multiple symmetric lipomatosis is associated with upregulation of CK2, AKT and ERK1/2. Int J Mol Sci. 2020;21(21):7933.PubMedCentral Sanna M, Borgo C, Compagnin C, Favaretto F, Vindigni V, Trento M, et al. White adipose tissue expansion in multiple symmetric lipomatosis is associated with upregulation of CK2, AKT and ERK1/2. Int J Mol Sci. 2020;21(21):7933.PubMedCentral
138.
go back to reference Borgo C, Milan G, Favaretto F, Stasi F, Fabris R, Salizzato V, et al. CK2 modulates adipocyte insulin-signaling and is up-regulated in human obesity. Sci Rep. 2017;7:17569.PubMedCentralPubMed Borgo C, Milan G, Favaretto F, Stasi F, Fabris R, Salizzato V, et al. CK2 modulates adipocyte insulin-signaling and is up-regulated in human obesity. Sci Rep. 2017;7:17569.PubMedCentralPubMed
139.
go back to reference Locke M, Windsor J, Dunbar PR. Human adipose-derived stem cells: isolation, characterization and applications in surgery. ANZ J Surg. 2009;79:235–44.PubMed Locke M, Windsor J, Dunbar PR. Human adipose-derived stem cells: isolation, characterization and applications in surgery. ANZ J Surg. 2009;79:235–44.PubMed
140.
go back to reference Badimon L, Cubedo J. Adipose tissue depots and inflammation: effects on plasticity and resident mesenchymal stem cell function. Cardiovasc Res. 2017;113:1064–73.PubMed Badimon L, Cubedo J. Adipose tissue depots and inflammation: effects on plasticity and resident mesenchymal stem cell function. Cardiovasc Res. 2017;113:1064–73.PubMed
141.
142.
go back to reference Hamrick MW, McGee-Lawrence ME, Frechette DM. Fatty infiltration of skeletal muscle: mechanisms and comparisons with bone marrow adiposity. Front Endocrinol. 2016;7:69. Hamrick MW, McGee-Lawrence ME, Frechette DM. Fatty infiltration of skeletal muscle: mechanisms and comparisons with bone marrow adiposity. Front Endocrinol. 2016;7:69.
143.
go back to reference Thompson D, Karpe F, Lafontan M, Frayn K. Physical activity and exercise in the regulation of human adipose tissue physiology. Physiol Rev. 2012;92:157–91.PubMed Thompson D, Karpe F, Lafontan M, Frayn K. Physical activity and exercise in the regulation of human adipose tissue physiology. Physiol Rev. 2012;92:157–91.PubMed
144.
go back to reference Addison O, Marcus RL, Lastayo PC, Ryan AS. Intermuscular fat: a review of the consequences and causes. Int J Endocrinol. 2014;2014:309570.PubMedCentralPubMed Addison O, Marcus RL, Lastayo PC, Ryan AS. Intermuscular fat: a review of the consequences and causes. Int J Endocrinol. 2014;2014:309570.PubMedCentralPubMed
145.
go back to reference Miller CT, Fraser SF, Levinger I, Straznicky NE, Dixon JB, Reynolds J, et al. The effects of exercise training in addition to energy restriction on functional capacities and body composition in obese adults during weight loss: a systematic review. PLoS One. 2013;8:e81692.PubMedCentralPubMed Miller CT, Fraser SF, Levinger I, Straznicky NE, Dixon JB, Reynolds J, et al. The effects of exercise training in addition to energy restriction on functional capacities and body composition in obese adults during weight loss: a systematic review. PLoS One. 2013;8:e81692.PubMedCentralPubMed
146.
147.
go back to reference Goloviznina NA, Xie N, Dandapat A, Iaizzo PA, Kyba M. Prospective isolation of human fibroadipogenic progenitors with CD73. Heliyon. 2020;6:e04503.PubMedCentralPubMed Goloviznina NA, Xie N, Dandapat A, Iaizzo PA, Kyba M. Prospective isolation of human fibroadipogenic progenitors with CD73. Heliyon. 2020;6:e04503.PubMedCentralPubMed
148.
go back to reference Suárez-Calvet X, Fernández-Simón E, Piñol-Jurado P, Alonso-Pérez J, Carrasco-Rozas A, Lleixà C, et al. Isolation of human fibroadipogenic progenitors and satellite cells from frozen muscle biopsies. FASEB J. 2021;35:e21819.PubMed Suárez-Calvet X, Fernández-Simón E, Piñol-Jurado P, Alonso-Pérez J, Carrasco-Rozas A, Lleixà C, et al. Isolation of human fibroadipogenic progenitors and satellite cells from frozen muscle biopsies. FASEB J. 2021;35:e21819.PubMed
149.
go back to reference Russo V, Yu C, Belliveau P, Hamilton A, Flynn LE. Comparison of human adipose-derived stem cells isolated from subcutaneous, omental, and intrathoracic adipose tissue depots for regenerative applications. Stem Cells Transl Med. 2014;3:206–17.PubMed Russo V, Yu C, Belliveau P, Hamilton A, Flynn LE. Comparison of human adipose-derived stem cells isolated from subcutaneous, omental, and intrathoracic adipose tissue depots for regenerative applications. Stem Cells Transl Med. 2014;3:206–17.PubMed
Metadata
Title
Adipogenic progenitors in different organs: Pathophysiological implications
Authors
Francesca Favaretto
Silvia Bettini
Luca Busetto
Gabriella Milan
Roberto Vettor
Publication date
01-02-2022
Publisher
Springer US
Keywords
Obesity
Obesity
Published in
Reviews in Endocrine and Metabolic Disorders / Issue 1/2022
Print ISSN: 1389-9155
Electronic ISSN: 1573-2606
DOI
https://doi.org/10.1007/s11154-021-09686-6

Other articles of this Issue 1/2022

Reviews in Endocrine and Metabolic Disorders 1/2022 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.