Horm Metab Res 2022; 54(12): 785-794
DOI: 10.1055/a-1922-7052
Review

Adipose Tissue Dysfunction: Impact on Metabolic Changes?

Jennifer Beatriz Silva Morais
1   Nutrition, Universidade Federal do Piaui, Teresina, Brazil
,
Thaline Millany da Silva Dias
1   Nutrition, Universidade Federal do Piaui, Teresina, Brazil
,
Bruna Emanuele Pereira Cardoso
1   Nutrition, Universidade Federal do Piaui, Teresina, Brazil
,
Mickael de Paiva Sousa
1   Nutrition, Universidade Federal do Piaui, Teresina, Brazil
,
Thayanne Gabryelle Visgueira de Sousa
1   Nutrition, Universidade Federal do Piaui, Teresina, Brazil
,
Diana Stefany Cardoso de Araújo
1   Nutrition, Universidade Federal do Piaui, Teresina, Brazil
,
Dilina do Nascimento Marreiro
1   Nutrition, Universidade Federal do Piaui, Teresina, Brazil
› Author Affiliations

Abstract

Adipose tissue is a metabolically dynamic organ that is the primary site of storage for excess energy, but it serves as an endocrine organ capable of synthesizing a number of biologically active compounds that regulate metabolic homeostasis. However, when the capacity of expansion of this tissue exceeds, dysfunction occurs, favoring ectopic accumulation of fat in the visceral, which has been implicated in several disease states, most notably obesity. This review highlights the mechanisms involved in the structure of adipose tissue, tissue expandability, adipocyte dysfunction, as well as the impact of these events on the manifestation of important metabolic disorders associated with adipose tissue dysfunction. A literature search using Pubmed, Web of Science, Scopus, and Cochrane databases were used to identify relevant studies, using clinical trials, experimental studies in animals and humans, case-control studies, case series, letters to the editor, and review articles published in English, without restrictions on year of publication. The excessive ectopic lipid accumulation leads to local inflammation and insulin resistance. Indeed, overnutrition triggers uncontrolled inflammatory responses white adipose tissue, leading to chronic low-grade inflammation, therefore fostering the progression of important metabolic disorders. Thus, it is essential to advance the understanding of the molecular mechanisms involved in adipose tissue dysfunction in order to mitigate the negative metabolic consequences of obesity.



Publication History

Received: 31 May 2022

Accepted after revision: 11 August 2022

Accepted Manuscript online:
11 August 2022

Article published online:
29 September 2022

© 2022. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Murawska-Ciałowicz E.. Adipose tissue – morphological and biochemical characteristic of different depots. Postepy Hig Med Dosw (Online) 2017; 8: 466-484
  • 2 Chait A, Hartigh LJ.. Adipose tissue distribution, inflammation and its metabolic consequences, including diabetes and cardiovascular disease. Front Cardiovasc Med 2020; 7: 22
  • 3 Bartelt A, Heeren J.. Adipose tissue browning and metabolic health. Nat Rev Endocrinol 2014; 10: 24-36
  • 4 Czech MP.. Mechanisms of insulin resistance related to white, beige, and brown adipocytes. Mol Metab 2020; 34: 27-42
  • 5 Sakers A, De Siqueira MK, Seale P, Villanueva CJ.. Adipose-tissue plasticity in health and disease. Cell 2022; 185: 419-446
  • 6 Tilg H, Moschen AR.. Adipocytokines: mediators linking adipose tissue, inflammation, and immunity. Nat Rev Immunol 2006; 6: 772-783
  • 7 Bray GA, Heisel WE, Afshin A. et al. The science of obesity management: an endocrine society scientific statement. Endocr Rev 2018; 39: 79-132
  • 8 Lee MW, Lee M, Oh KJ.. Adipose tissue-derived signatures for obesity and type 2 diabetes: adipokines, batokines and microRNAs. J Clin Med 2019; 8: 854
  • 9 Mittal B.. Subcutaneous adipose tissue & visceral adipose tissue. Indian J Med Res 2019; 149: 571-573
  • 10 Vishvanath L, Gupta RK.. Contribution of adipogenesis to healthy adipose tissue expansion in obesity. J Clin Invest 2019; 129: 4022-4031
  • 11 Ghezzi AC.. Modulation of macrophage infiltration in brown adipose tissue and white adipose tissue of obese rats: effects of treatment of GLP-1 analogue, DPP-4 inhibitor and physical exercise. PhD Thesis. 2017 Faculdade Estadual de Campinas
  • 12 Silva NSB, Castro CFB.. O papel das adipocitocinas leptina e adiponectina no desenvolvimento da obesidade. REBES 2019; 9: 70-76
  • 13 Silva NI, Sobrinho HMR, Blanch GT. et al. Adipocinas e sua relação com a obesidade. Revista EVS 2019; 46: 53-64
  • 14 Becher T, Palanisamy S, Kramer DJ. et al. Brown adipose tissue is associated with cardiometabolic health. Nat Med 2021; 27: 58-65
  • 15 Sanchez-Gurmaches J, Guertin DA.. Adipocytes arise from multiple lineages that are heterogeneously and dynamically distributed. Nat Commun 2014; 5: 4099
  • 16 Shireesh S, Veech RL.. The fat soldiers in the anti-obesity fight. Front Physiol 2019; 10: 38
  • 17 Ghaben AL, Scherer PE.. Adipogenesis and metabolic health. Nat Ver. Mol Cell Biol 2019; 20: 242-258
  • 18 Parrenttini S, Cavallo M, Gaggia F. et al. Adipokines: a rainbow of proteins with metabolic and endocrine functions. Protein Pept Lett 2020; 27: 1204-1230
  • 19 Guha D, Mukherjee R, Ainch P.. O macrófago desempenha um papel importante na adipogênese induzida pelo cortisol e serotonina in vitro. In Vitro Cell Dev Biol Anim 2020; 56: 511-521
  • 20 Martus-Rus C, Katz-Greenberg G, Lin Z. et al. Macrophage and adipocyte interaction as a source of inflammation in kidney disease. Sci Rep 2021; 11: 2974
  • 21 Sárvári AK, Hauwaert ELV, Markussen LK. et al. Plasticity of epididymal adipose tissue in response to diet-induced obesity at single-nucleus resolution. Cell Metab 2021; 33: 437-453
  • 22 He Y, Xia J, Chen H. et al. Human adipose liquid extract induces angiogenesis and adipogenesis: a novel cell-free therapeutic agent. Stem Cell Res Ther 2019; 10: 1-14
  • 23 Torres-Perez E, Valero M, Garcia-Rodriguez B. et al. The FAT expandability (FATe) project: biomarkers to determine the limit of expansion and the complications of obesity. Cardiovasc Diabetol 2015; 14: 40
  • 24 Hammarstedt A, Gogg S, Hedjazifar S. et al. Impaired adipogenesis and dysfunctional adipose tissue in human hypertrophic obesity. Physiol Rev 2018; 98: 1911-1941
  • 25 Pellegrinelli V, Carobbio S, Vidal-Puig A.. Adipose tissue plasticity: how fat depots respond differently to pathophysiological cues. Diabetologia 2016; 59: 1075-1088
  • 26 Bruning U, Morales-Rodriguez F, Kalucka J. et al. O comprometimento da angiogênese pela inibição da ácido graxo sintase envolve a malonilação de mTOR. Metab Cell 2018; 28: 866-880
  • 27 Viallard C, Larrivée B. Tumor angiogenesis and vascular normalization: alternative therapeutic targets. Angiogenesis 2017; 20: 409-426
  • 28 Cao Y.. Angiogenesis modulates adipogenesis and obesity. J Clin Invest 2007; 117: 2362-2368
  • 29 Matulewicz N, Stefanowicz M, Nikolajuk A. et al. Markers of adipogenesis, but not inflammation, in adipose tissue are independently related to insulin sensitivity. J Clin Endocrinol Metab 2017; 102: 3040-3049
  • 30 Ruiz-Ojeda FJ, Méndez-Gutiérrez A, Aguilera CM. et al. Extracellular matrix remodeling of adipose tissue in obesity and metabolic diseases. Int J Mol Sci 2019; 20: 4888
  • 31 Corrêa LH, Heyn GS, Magalhaes KG. The impact of the adipose organ plasticity on inflammation and cancer progression. Cells 2019; 8: 662
  • 32 Schoettl T, Fischer IP, Ussar S.. Heterogeneity of adipose tissue in development and metabolic function. J Exp Biol 2018; 221: jeb162958
  • 33 Yu-Kyoung P, Byeong-Churl J.. Cryptotanshinone inhibits lipid accumulation in differentiating 3T3-L1 preadipocytes by down-regulating C/EBP-α, PPAR-γ, FAS, Perilipin A, and STAT-3. Keimyung Med J 2019; 1-10
  • 34 Sztalryd C, Brasaemle DL.. The perilipin family of lipid droplet proteins: gatekeepers of intracellular lipolysis. Biochim Biophys Acta Mol Cell Biol Lipids 1862; 1221-1232
  • 35 Illesca P, Valenzuela R, Espinosa A. et al. Hydroxytyrosol supplementation ameliorates the metabolic disturbances in white adipose tissue from mice fed a high-fat diet through recovery of transcription factors Nrf2, SREBP-1c, PPAR-γ and NF-κB.. Biomed Pharmacother 2019; 109: 2472-2481
  • 36 Morigny P, Houssier M, Mairal A. et al. Interaction between hormone-sensitive lipase and ChREBP in fat cells controls insulin sensitivity. Nat Metab 2019; 1: 133-146
  • 37 Maximus PS, Achkar ZA, Hamid PF. et al. Adipocytokines: are they the theory of everything?. Cytokine 2020; 133: 155144
  • 38 Exley MA, Hand L. O’Shea et al. Interplay between the imune system and adipose tissue in obesity. J Endocrinol 2014; 223: R41-R48
  • 39 Chung KJ, Nati M, Chavakis T. et al. Innate immune cells in the adipose tissue. Rev Endocr Metab Disord 2018; 19: 283-292
  • 40 McLaughlin T, Lamendola C, Liu A. et al. Preferential fat deposition in subcutaneous versus visceral depots is associated with insulin sensitivity. J Clin Endocrinol Metab 2011; 96: E1756-E1760
  • 41 Li C, Xu MM, Wang K. et al. Macrophage polarization and meta-inflammation. Transl Res 2018; 191: 29-44
  • 42 Mescher AL.. Macrophages and fibroblasts during inflammation and tissue repair in models of organ regeneration. Regeneration 2017; 4: 39-53
  • 43 Remmerie A, Martens L, Scott CL.. Macrophage subsets in obesity, aligning the liver and adipose tissue. Front Endocrinol 2020; 11: 259
  • 44 Chistiakov DA, Myasoedova VA, Revin VV. et al. The impact of interferon-regulatory factors to macrophage differentiation and polarization into M1 and M2. Immunobiology 2018; 223: 101-111
  • 45 Hume DA.. The many alternative faces of macrophage activation. Front Immunol 2015; 22: 370
  • 46 Saha S, Shalova IN, Biswas SK.. Metabolic regulation of macrophage phenotype and function. Immunol Rev 2017; 280: 102-111
  • 47 Villarroya F, Cereijo R, Gavaldà-Navarro A. et al. Inflammation of brown/beige adipose tissues in obesity and metabolic disease. J Intern Med 2018; 284: 492-504
  • 48 Gomes BF, Accardo CM.. Mediadores imunoinflamatórios na patogênese do diabetes mellitus. Einstein 2019; 17: eRB4596
  • 49 Silva-Jr AJ.. Adipocinas: A relação endócrina entre obesidade e diabetes tipo II. RBONE 2017; 11: 135-145
  • 50 Cuthbertson DJ, Steele T, Wilding JP. et al. What have human experimental overfeeding studies taught us about adipose tissue expansion and susceptibility to obesity and metabolic complications?. Int J Obes 2017; 41: 853-865
  • 51 Caprio S, Pierpont B, Kursawe R. The “adipose tissue expandability” hypothesis: a potential mechanism for insulin resistance in obese youth. Horm Mol Biol Clin Investig 2018; 33: 20180005
  • 52 Carobbio S, Pellegrinelli V, Vidal-Puig A.. Adipose tissue function and expandability as determinants of lipotoxicity and the metabolic syndrome. Adv Exp Med Biol 2017; 960: 161-196
  • 53 Lempesis IG, Meijel RLJ, Manolopoulos KN. et al. Oxygenation of adipose tissue: a human perspective. Acta Physiol 2020; 228: e13298
  • 54 Crewe C, An YA, Scherer PE.. The ominous triad of adipose tissue dysfunction: inflammation, fibrosis, and impaired angiogenesis. J Clin Invest 2017; 127: 74-82
  • 55 Zhang J, Zhang L, Zhang S. et al. HMGB1, an innate alarmin, plays a critical role in chronic inflammation of adipose tissue in obesity. Mol Cell Endocrinol 2017; 454: 103-111
  • 56 Torres S, Fabersani E, Marquez A. et al. Adipose tissue inflammation and metabolic syndrome. The proactive role of probiotics. Eur J Nutr 2019; 58: 27-43
  • 57 Lin D, Chun TH, Kang L.. Adipose extracellular matrix remodelling in obesity and insulin resistance. Biochem Pharmacol 2016; 119: 8-16
  • 58 Lawler HM, Underkofler CM, Kern PA. et al. Adipose tissue hypoxia, inflammation, and fibrosis in oese insulin-sensitive and obese insulin-resistant subjects. J Clin Endocrinol Metab 2016; 101: 1422-1428
  • 59 Muir LA, Neeley CK, Meyer KA. et al. Adipose tissue fibrosis, hypertrophy, and hyperplasia: Correlations with diabetes in human obesity. Obesity 2016; 24: 597-605
  • 60 Lackey DE, Burk DH, Ali MR. et al. Contributions of adipose tissue architectural and tensile properties toward defining healthy and unhealthy obesity. Am J Physiol Endocrinol Metab 2014; 306: E233-E246
  • 61 Ye J.. Emerging role of adipose tissue hypoxia in obesity and insulin resistance. Int J Obes 2009; 33: 54-66
  • 62 Wang B, Wood IS, Trayhurn P.. Hypoxia induces leptin gene expression and secretion in human preadipocytes: differential effects of hypoxia on adipokine expression by preadipocytes. J Endocrinol 2008; 198: 127-134
  • 63 Patel PS, Buras ED, Balasubramanyam A.. The role of the immune system in obesity and insulin resistance. J Obes 2013; 616193
  • 64 Lauterbach MA, Wunderlich FT.. Macrophage function in obesity-induced inflammation and insulin resistance. Pflugers Arch 2017; 469: 385-396
  • 65 Russo L, Lumeng CN.. Properties and functions of adipose tissue macrophages in obesity. Immunology 2018; 155: 407-417
  • 66 Nishimura S, Manabe I, Nagasaki M. et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med 2009; 15: 914-920
  • 67 Alexopoulos N, Katritsis D, Raggi P.. Visceral adipose tissue as a source of inflammation and promoter of atherosclerosis. Atherosclerosis 2014; 233: 104-112
  • 68 Lovren F, Teoh H, Verma S.. Obesity and atherosclerosis: mechanistic insights. Can J Cardiol 2015; 31: 177-183
  • 69 Mansuy-Aubert V, Zhou QL, Xie X. et al. Imbalance between neutrophil elastase and its inhibitor alpha1-antitrypsin in obesity alters insulin sensitivity, inflammation, and energy expenditure. Cell Metab 2013; 17: 534-548
  • 70 Talukdar S, Oh DY, Bandyopadhyay G. et al. Neutrophils mediate insulin resistance in mice fed a high-fat diet through secreted elastase. Nat Med 2012; 18: 1407-1412
  • 71 Strzepa A, Pritchard KA, Dittel BN.. Myeloperoxidase: a new player in autoimmunity. Cell Immunol 2017; 317: 1-8
  • 72 Eberl G, Colonna M, Santo JP. et al. Innate lymphoid cells. Nnate lymphoid cells: a new paradigm in immunology. Science 2015; 348: aaa6566
  • 73 Robinette ML, Fuchs A, Cortez VS. et al. Transcriptional programs define molecular characteristics of innate lymphoid cell classes and subsets. Nat Immunol 2015; 16: 306-317
  • 74 O‘Sullivan TE, Rapp M, Fan X. et al. Adipose-resident group 1 innate lymphoid cells promote obesity-associated insulin resistance. Immunity 2016; 45: 428-441
  • 75 Ballesteros-Pomar MD, Calleja S, Diez-Rodriguez R. et al. Inflammatory status is different in relationship to insulin resistance in severely obese people and changes after bariatric surgery or diet-induced weight loss. Exp Clin Endocrinol Diabetes 2014; 122: 592-596
  • 76 Simar D, Versteyhe S, Donkin I. et al. DNA methylation is altered in B and NK lymphocytes in obese and type 2 diabetic human. Metabolism 2014; 63: 1188-1197
  • 77 Bartelt A, Widenmaier SB, Schlein C. et al. Brown adipose tissue thermogenic adaptation requires Nrf1-mediated proteasomal activity. Nat Med 2018; 24: 292-303
  • 78 Matoušková P, Hanousková B, Skálová L.. MicroRNAs as potential regulators of glutathione peroxidases expression and their role in obesity and related pathologies. Int J Mol Sci 2018; 1199
  • 79 Fernández-Sánchez A, Madrigal-Santillán E, Bautista M. et al. Inflammation, oxidative stress, and obesity. Int J Mol Sci 2011; 12: 3117-3132
  • 80 Le Lay S, Simard G, Martinez MC. et al. Oxidative stress and metabolic pathologies: from an adipocentric point of view. Oxid Med Cell Longev 2014; 908539
  • 81 Tan BL, Norhaizan ME, Liew WP. et al. Antioxidant and oxidative stress: a mutual interplay in age-related diseases. Front Pharmacol 2018; 16: 1162
  • 82 Kalhara R, Menikdiwela JPTG, Ramalingam L. et al. Mechanisms linking endoplasmic reticulum (ER) stress and microRNAs to adipose tissue dysfunction in obesity. Crit Rev Biochem Mol Biol 2021; 56: 455-481
  • 83 Ghemrawi R, Battaglia-Hsu SF, Arnold C.. Endoplasmic reticulum stress in metabolic disorders. Cells 2018; 7: 63
  • 84 Lee BC, Kim MS, Pae M. et al. Adipose natural killer cells regulate adipose tissue macrophages to promote insulin resistance in obesity. Cell Metab 2016; 23: 685-698
  • 85 Wensveen FM, Jelencic V, Valentic S. et al. NK cells link obesity-induced adipose stress to inflammation and insulin resistance. Nat Immunol 2015; 16: 376-385
  • 86 Kim JI, Huh JY, Sohn JH. et al. Lipid-overloaded enlarged adipocytes provoke insulin resistance independent of inflammation. Mol Cell Biol 2015; 35: 1686-1699
  • 87 Longo M, Zatterale F, Naderi J. et al. Adipose tissue dysfunction as determinant of obesity-associated metabolic complications. Int J Mol Sci 2019; 20: 2358
  • 88 Castoldi A, Naffah de Souza C, Câmara NO. et al. The macrophage switch in obesity development. Front Immunol 2015; 6: 637
  • 89 Kilicarslan M, Weijer BA, Sjödin KS. et al. RBP4 increases lipolysis in human adipocytes and is associated with increased lipolysis and hepatic insulin resistance in obese women. FASEB J 2020; 34: 6099-6110
  • 90 Yang A, Motillo EP.. Adipocyte lipolysis: from molecular mechanisms of regulation to disease and therapeutics. Biochem J 2020; 477: 985-1008
  • 91 Britton KA, Fox CS.. Ectopic fat depots and cardiovascular disease. Circulation 2011; 124: e837-e841
  • 92 Sousa MP, Cruz KJC, Morais JBS. et al. Associação entre ingestão dietética de magnésio e parâmetros do perfil lipidico em mulheres obesas. Res Soc Dev 2019; 9: e53911592
  • 93 Vekic J, Zeljkovic A, Stefanovic A. et al. Obesity and dyslipidemia. Metabolism 2019; 92: 71-81
  • 94 Zhang T, Chen J, Tang X. et al. Interaction between adipocytes and high-density lipoprotein:new insights into the mechanism of obesity-induced dyslipidemia and atherosclerosis. Lipids Health Dis 2019; 18: 223
  • 95 Jung UJ, Choi MS.. Obesity and its metabolic complications: the role of adipokines and the relationship between obesity, inflammation, insulin resistance, dyslipidemia and nonalcoholic fatty liver disease. Int J Mol Sci 2014; 15: 6184-6223