Skip to main content
Top
Published in: BMC Medicine 1/2022

Open Access 01-12-2022 | Obesity | Review

Obese visceral fat tissue inflammation: from protective to detrimental?

Author: Hubert Kolb

Published in: BMC Medicine | Issue 1/2022

Login to get access

Abstract

Obesity usually is accompanied by inflammation of fat tissue, with a prominent role of visceral fat. Chronic inflammation in obese fat tissue is of a lower grade than acute immune activation for clearing the tissue from an infectious agent. It is the loss of adipocyte metabolic homeostasis that causes activation of resident immune cells for supporting tissue functions and regaining homeostasis. Initially, the excess influx of lipids and glucose in the context of overnutrition is met by adipocyte growth and proliferation. Eventual lipid overload of hypertrophic adipocytes leads to endoplasmic reticulum stress and the secretion of a variety of signals causing increased sympathetic tone, lipolysis by adipocytes, lipid uptake by macrophages, matrix remodeling, angiogenesis, and immune cell activation. Pro-inflammatory signaling of adipocytes causes the resident immune system to release increased amounts of pro-inflammatory and other mediators resulting in enhanced tissue-protective responses. With chronic overnutrition, these protective actions are insufficient, and death of adipocytes as well as senescence of several tissue cell types is seen. This structural damage causes the expression or release of immunostimulatory cell components resulting in influx and activation of monocytes and many other immune cell types, with a contribution of stromal cells. Matrix remodeling and angiogenesis is further intensified as well as possibly detrimental fibrosis. The accumulation of senescent cells also may be detrimental via eventual spread of senescence state from affected to neighboring cells by the release of microRNA-containing vesicles. Obese visceral fat inflammation can be viewed as an initially protective response in order to cope with excess ambient nutrients and restore tissue homeostasis but may contribute to tissue damage at a later stage.
Literature
1.
go back to reference Chen L, Deng H, Cui H, Fang J, Zuo Z, Deng J, et al. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget. 2018;9:7204.CrossRef Chen L, Deng H, Cui H, Fang J, Zuo Z, Deng J, et al. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget. 2018;9:7204.CrossRef
2.
go back to reference Medzhitov R. The spectrum of inflammatory responses. Science. 2021;374:1070.CrossRef Medzhitov R. The spectrum of inflammatory responses. Science. 2021;374:1070.CrossRef
3.
go back to reference Psaila AM, Vohralik EJ, Quinlan KGR. Shades of white: new insights into tissue-resident leukocyte heterogeneity. FEBS J. 2022;289:308.CrossRef Psaila AM, Vohralik EJ, Quinlan KGR. Shades of white: new insights into tissue-resident leukocyte heterogeneity. FEBS J. 2022;289:308.CrossRef
4.
go back to reference Weinstock A, Brown EJ, Garabedian ML, Pena S, Sharma M, Lafaille J, et al. Single-Cell RNA Sequencing of Visceral Adipose Tissue Leukocytes Reveals that Caloric Restriction Following Obesity Promotes the Accumulation of a Distinct Macrophage Population with Features of Phagocytic Cells. Immunometabolism. 2019;1:e190008. Weinstock A, Brown EJ, Garabedian ML, Pena S, Sharma M, Lafaille J, et al. Single-Cell RNA Sequencing of Visceral Adipose Tissue Leukocytes Reveals that Caloric Restriction Following Obesity Promotes the Accumulation of a Distinct Macrophage Population with Features of Phagocytic Cells. Immunometabolism. 2019;1:e190008.
5.
go back to reference Schipper HS, Prakken B, Kalkhoven E, Boes M. Adipose tissue-resident immune cells: key players in immunometabolism. Trends Endocrinol Metab. 2012;23:407.CrossRef Schipper HS, Prakken B, Kalkhoven E, Boes M. Adipose tissue-resident immune cells: key players in immunometabolism. Trends Endocrinol Metab. 2012;23:407.CrossRef
6.
go back to reference Xu X, Grijalva A, Skowronski A, van EM SMJ, Ferrante AW Jr. Obesity activates a program of lysosomal-dependent lipid metabolism in adipose tissue macrophages independently of classic activation. Cell Metab. 2013;18:816.CrossRef Xu X, Grijalva A, Skowronski A, van EM SMJ, Ferrante AW Jr. Obesity activates a program of lysosomal-dependent lipid metabolism in adipose tissue macrophages independently of classic activation. Cell Metab. 2013;18:816.CrossRef
7.
go back to reference Jaitin DA, Adlung L, Thaiss CA, Weiner A, Li B, Descamps H, et al. Lipid-Associated Macrophages Control Metabolic Homeostasis in a Trem2-Dependent Manner. Cell. 2019;178:686.CrossRef Jaitin DA, Adlung L, Thaiss CA, Weiner A, Li B, Descamps H, et al. Lipid-Associated Macrophages Control Metabolic Homeostasis in a Trem2-Dependent Manner. Cell. 2019;178:686.CrossRef
8.
go back to reference Wang Q, Li D, Cao G, Shi Q, Zhu J, Zhang M, et al. IL-27 signalling promotes adipocyte thermogenesis and energy expenditure. Nature. 2021;600:314.CrossRef Wang Q, Li D, Cao G, Shi Q, Zhu J, Zhang M, et al. IL-27 signalling promotes adipocyte thermogenesis and energy expenditure. Nature. 2021;600:314.CrossRef
9.
go back to reference Rahman MS, Jun H. The Adipose Tissue Macrophages Central to Adaptive Thermoregulation. Front Immunol. 2022;13:884126.CrossRef Rahman MS, Jun H. The Adipose Tissue Macrophages Central to Adaptive Thermoregulation. Front Immunol. 2022;13:884126.CrossRef
10.
go back to reference Backdahl J, Franzen L, Massier L, Li Q, Jalkanen J, Gao H, et al. Spatial mapping reveals human adipocyte subpopulations with distinct sensitivities to insulin. Cell Metab. 2021;33:1869.CrossRef Backdahl J, Franzen L, Massier L, Li Q, Jalkanen J, Gao H, et al. Spatial mapping reveals human adipocyte subpopulations with distinct sensitivities to insulin. Cell Metab. 2021;33:1869.CrossRef
11.
go back to reference Brestoff JR, Kim BS, Saenz SA, Stine RR, Monticelli LA, Sonnenberg GF, et al. Group 2 innate lymphoid cells promote beiging of white adipose tissue and limit obesity. Nature. 2015;519:242.CrossRef Brestoff JR, Kim BS, Saenz SA, Stine RR, Monticelli LA, Sonnenberg GF, et al. Group 2 innate lymphoid cells promote beiging of white adipose tissue and limit obesity. Nature. 2015;519:242.CrossRef
12.
go back to reference Painter JD, Akbari O. Type 2 Innate Lymphoid Cells: Protectors in Type 2 Diabetes. Front Immunol. 2021;12:727008.CrossRef Painter JD, Akbari O. Type 2 Innate Lymphoid Cells: Protectors in Type 2 Diabetes. Front Immunol. 2021;12:727008.CrossRef
13.
go back to reference Macdougall CE, Wood EG, Loschko J, Scagliotti V, Cassidy FC, Robinson ME, et al. Visceral Adipose Tissue Immune Homeostasis Is Regulated by the Crosstalk between Adipocytes and Dendritic Cell Subsets. Cell Metab. 2018;27:588.CrossRef Macdougall CE, Wood EG, Loschko J, Scagliotti V, Cassidy FC, Robinson ME, et al. Visceral Adipose Tissue Immune Homeostasis Is Regulated by the Crosstalk between Adipocytes and Dendritic Cell Subsets. Cell Metab. 2018;27:588.CrossRef
14.
go back to reference Feuerer M, Herrero L, Cipolletta D, Naaz A, Wong J, Nayer A, et al. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med. 2009;15:930.CrossRef Feuerer M, Herrero L, Cipolletta D, Naaz A, Wong J, Nayer A, et al. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med. 2009;15:930.CrossRef
15.
go back to reference Shao Q, Gu J, Zhou J, Wang Q, Li X, Deng Z, et al. Tissue Tregs and Maintenance of Tissue Homeostasis. Front Cell Dev Biol. 2021;9:717903.CrossRef Shao Q, Gu J, Zhou J, Wang Q, Li X, Deng Z, et al. Tissue Tregs and Maintenance of Tissue Homeostasis. Front Cell Dev Biol. 2021;9:717903.CrossRef
16.
go back to reference Kolodin D, van PN LC, Magnuson AM, Cipolletta D, Miller CM, et al. Antigen- and cytokine-driven accumulation of regulatory T cells in visceral adipose tissue of lean mice. Cell Metab. 2015;21:543.CrossRef Kolodin D, van PN LC, Magnuson AM, Cipolletta D, Miller CM, et al. Antigen- and cytokine-driven accumulation of regulatory T cells in visceral adipose tissue of lean mice. Cell Metab. 2015;21:543.CrossRef
17.
go back to reference Harmon DB, Srikakulapu P, Kaplan JL, Oldham SN, McSkimming C, Garmey JC, et al. Protective Role for B-1b B Cells and IgM in Obesity-Associated Inflammation, Glucose Intolerance, and Insulin Resistance. Arterioscler Thromb Vasc Biol. 2016;36:682.CrossRef Harmon DB, Srikakulapu P, Kaplan JL, Oldham SN, McSkimming C, Garmey JC, et al. Protective Role for B-1b B Cells and IgM in Obesity-Associated Inflammation, Glucose Intolerance, and Insulin Resistance. Arterioscler Thromb Vasc Biol. 2016;36:682.CrossRef
18.
go back to reference Rangel-Moreno J, Moyron-Quiroz JE, Carragher DM, Kusser K, Hartson L, Moquin A, et al. Omental milky spots develop in the absence of lymphoid tissue-inducer cells and support B and T cell responses to peritoneal antigens. Immunity. 2009;30:731.CrossRef Rangel-Moreno J, Moyron-Quiroz JE, Carragher DM, Kusser K, Hartson L, Moquin A, et al. Omental milky spots develop in the absence of lymphoid tissue-inducer cells and support B and T cell responses to peritoneal antigens. Immunity. 2009;30:731.CrossRef
19.
go back to reference Moro K, Yamada T, Tanabe M, Takeuchi T, Ikawa T, Kawamoto H, et al. Innate production of T(H)2 cytokines by adipose tissue-associated c-Kit(+)Sca-1(+) lymphoid cells. Nature. 2010;463:540.CrossRef Moro K, Yamada T, Tanabe M, Takeuchi T, Ikawa T, Kawamoto H, et al. Innate production of T(H)2 cytokines by adipose tissue-associated c-Kit(+)Sca-1(+) lymphoid cells. Nature. 2010;463:540.CrossRef
20.
go back to reference Benezech C, Luu NT, Walker JA, Kruglov AA, Loo Y, Nakamura K, et al. Inflammation-induced formation of fat-associated lymphoid clusters. Nat Immunol. 2015;16:819.CrossRef Benezech C, Luu NT, Walker JA, Kruglov AA, Loo Y, Nakamura K, et al. Inflammation-induced formation of fat-associated lymphoid clusters. Nat Immunol. 2015;16:819.CrossRef
21.
go back to reference Trim WV, Lynch L. Immune and non-immune functions of adipose tissue leukocytes. Nat Rev Immunol. 2022;22:371.CrossRef Trim WV, Lynch L. Immune and non-immune functions of adipose tissue leukocytes. Nat Rev Immunol. 2022;22:371.CrossRef
22.
go back to reference Hu B, Jin C, Zeng X, Resch JM, Jedrychowski MP, Yang Z, et al. gammadelta T cells and adipocyte IL-17RC control fat innervation and thermogenesis. Nature. 2020;578:610.CrossRef Hu B, Jin C, Zeng X, Resch JM, Jedrychowski MP, Yang Z, et al. gammadelta T cells and adipocyte IL-17RC control fat innervation and thermogenesis. Nature. 2020;578:610.CrossRef
23.
go back to reference Pirzgalska RM, Seixas E, Seidman JS, Link VM, Sanchez NM, Mahu I, et al. Sympathetic neuron-associated macrophages contribute to obesity by importing and metabolizing norepinephrine. Nat Med. 2017;23:1309.CrossRef Pirzgalska RM, Seixas E, Seidman JS, Link VM, Sanchez NM, Mahu I, et al. Sympathetic neuron-associated macrophages contribute to obesity by importing and metabolizing norepinephrine. Nat Med. 2017;23:1309.CrossRef
24.
go back to reference Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007;117:175.CrossRef Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007;117:175.CrossRef
25.
go back to reference Lee BC, Lee J. Cellular and molecular players in adipose tissue inflammation in the development of obesity-induced insulin resistance. Biochim Biophys Acta. 2014;1842:446.CrossRef Lee BC, Lee J. Cellular and molecular players in adipose tissue inflammation in the development of obesity-induced insulin resistance. Biochim Biophys Acta. 2014;1842:446.CrossRef
26.
go back to reference Hotamisligil GS. Inflammation, metaflammation and immunometabolic disorders. Nature. 2017;542:177.CrossRef Hotamisligil GS. Inflammation, metaflammation and immunometabolic disorders. Nature. 2017;542:177.CrossRef
27.
go back to reference Schmidleithner L, Thabet Y, Schonfeld E, Kohne M, Sommer D, Abdullah Z, et al. Enzymatic Activity of HPGD in Treg Cells Suppresses Tconv Cells to Maintain Adipose Tissue Homeostasis and Prevent Metabolic Dysfunction. Immunity. 2019;50:1232.CrossRef Schmidleithner L, Thabet Y, Schonfeld E, Kohne M, Sommer D, Abdullah Z, et al. Enzymatic Activity of HPGD in Treg Cells Suppresses Tconv Cells to Maintain Adipose Tissue Homeostasis and Prevent Metabolic Dysfunction. Immunity. 2019;50:1232.CrossRef
28.
go back to reference Kane H, Lynch L. Innate Immune Control of Adipose Tissue Homeostasis. Trends Immunol. 2019;40:857.CrossRef Kane H, Lynch L. Innate Immune Control of Adipose Tissue Homeostasis. Trends Immunol. 2019;40:857.CrossRef
29.
go back to reference Ahmed DS, Isnard S, Lin J, Routy B, Routy JP. GDF15/GFRAL Pathway as a Metabolic Signature for Cachexia in Patients with Cancer. J Cancer. 2021;12:1125.CrossRef Ahmed DS, Isnard S, Lin J, Routy B, Routy JP. GDF15/GFRAL Pathway as a Metabolic Signature for Cachexia in Patients with Cancer. J Cancer. 2021;12:1125.CrossRef
30.
go back to reference Saitoh S, Van WK, Nakajima O. Crosstalk between Metabolic Disorders and Immune Cells. Int J Mol Sci. 2021;22:10017. Saitoh S, Van WK, Nakajima O. Crosstalk between Metabolic Disorders and Immune Cells. Int J Mol Sci. 2021;22:10017.
31.
go back to reference Toubal A, Lehuen A. Role of MAIT cells in metabolic diseases. Mol Immunol. 2021;130:142.CrossRef Toubal A, Lehuen A. Role of MAIT cells in metabolic diseases. Mol Immunol. 2021;130:142.CrossRef
32.
go back to reference Munoz-Rojas AR, Mathis D. Tissue regulatory T cells: regulatory chameleons. Nat Rev Immunol. 2021;21:597.CrossRef Munoz-Rojas AR, Mathis D. Tissue regulatory T cells: regulatory chameleons. Nat Rev Immunol. 2021;21:597.CrossRef
33.
go back to reference Stevens HY, Bowles AC, Yeago C, Roy K. Molecular Crosstalk Between Macrophages and Mesenchymal Stromal Cells. Front Cell Dev Biol. 2020;8:600160.CrossRef Stevens HY, Bowles AC, Yeago C, Roy K. Molecular Crosstalk Between Macrophages and Mesenchymal Stromal Cells. Front Cell Dev Biol. 2020;8:600160.CrossRef
34.
go back to reference Davidson S, Coles M, Thomas T, Kollias G, Ludewig B, Turley S, et al. Fibroblasts as immune regulators in infection, inflammation and cancer. Nat Rev Immunol. 2021;21:704.CrossRef Davidson S, Coles M, Thomas T, Kollias G, Ludewig B, Turley S, et al. Fibroblasts as immune regulators in infection, inflammation and cancer. Nat Rev Immunol. 2021;21:704.CrossRef
35.
go back to reference van der Poll T, Jansen J, Levi M, ten Cate H, ten Cate JW, van Deventer SJ. Regulation of interleukin 10 release by tumor necrosis factor in humans and chimpanzees. J Exp Med. 1994;180:1985.CrossRef van der Poll T, Jansen J, Levi M, ten Cate H, ten Cate JW, van Deventer SJ. Regulation of interleukin 10 release by tumor necrosis factor in humans and chimpanzees. J Exp Med. 1994;180:1985.CrossRef
36.
go back to reference Li B, Hao J, Zeng J. Sauter ER. SnapShot: FABP Functions. Cell. 2020;182:1066.CrossRef Li B, Hao J, Zeng J. Sauter ER. SnapShot: FABP Functions. Cell. 2020;182:1066.CrossRef
37.
go back to reference Fain JN. Release of interleukins and other inflammatory cytokines by human adipose tissue is enhanced in obesity and primarily due to the nonfat cells. Vitam Horm. 2006;74:443.CrossRef Fain JN. Release of interleukins and other inflammatory cytokines by human adipose tissue is enhanced in obesity and primarily due to the nonfat cells. Vitam Horm. 2006;74:443.CrossRef
38.
go back to reference Fain JN. Release of inflammatory mediators by human adipose tissue is enhanced in obesity and primarily by the nonfat cells: a review. Mediators Inflamm. 2010;2010:513948.CrossRef Fain JN. Release of inflammatory mediators by human adipose tissue is enhanced in obesity and primarily by the nonfat cells: a review. Mediators Inflamm. 2010;2010:513948.CrossRef
39.
go back to reference Kolb H, Stumvoll M, Kramer W, Kempf K, Martin S. Insulin translates unfavourable lifestyle into obesity. BMC Med. 2018;16:232.CrossRef Kolb H, Stumvoll M, Kramer W, Kempf K, Martin S. Insulin translates unfavourable lifestyle into obesity. BMC Med. 2018;16:232.CrossRef
40.
go back to reference Stenkula KG, Erlanson-Albertsson C. Adipose cell size: importance in health and disease. Am J Physiol Regul Integr Comp Physiol. 2018;315:R284–95.CrossRef Stenkula KG, Erlanson-Albertsson C. Adipose cell size: importance in health and disease. Am J Physiol Regul Integr Comp Physiol. 2018;315:R284–95.CrossRef
41.
go back to reference Pellegrinelli V, Carobbio S, Vidal-Puig A. Adipose tissue plasticity: how fat depots respond differently to pathophysiological cues. Diabetologia. 2016;59:1075.CrossRef Pellegrinelli V, Carobbio S, Vidal-Puig A. Adipose tissue plasticity: how fat depots respond differently to pathophysiological cues. Diabetologia. 2016;59:1075.CrossRef
42.
go back to reference Marques BG, Hausman DB, Martin RJ. Association of fat cell size and paracrine growth factors in development of hyperplastic obesity. Am J Physiol. 1998;275:R1898–908. Marques BG, Hausman DB, Martin RJ. Association of fat cell size and paracrine growth factors in development of hyperplastic obesity. Am J Physiol. 1998;275:R1898–908.
43.
go back to reference Haczeyni F, Bell-Anderson KS, Farrell GC. Causes and mechanisms of adipocyte enlargement and adipose expansion. Obes Rev. 2018;19:406.CrossRef Haczeyni F, Bell-Anderson KS, Farrell GC. Causes and mechanisms of adipocyte enlargement and adipose expansion. Obes Rev. 2018;19:406.CrossRef
44.
go back to reference Iwayama T, Steele C, Yao L, Dozmorov MG, Karamichos D, Wren JD, et al. PDGFRalpha signaling drives adipose tissue fibrosis by targeting progenitor cell plasticity. Genes Dev. 2015;29:1106.CrossRef Iwayama T, Steele C, Yao L, Dozmorov MG, Karamichos D, Wren JD, et al. PDGFRalpha signaling drives adipose tissue fibrosis by targeting progenitor cell plasticity. Genes Dev. 2015;29:1106.CrossRef
45.
go back to reference Wernstedt AI, Tao C, Morley TS, Wang QA, Delgado-Lopez F, Wang ZV, et al. Adipocyte inflammation is essential for healthy adipose tissue expansion and remodeling. Cell Metab. 2014;20:103.CrossRef Wernstedt AI, Tao C, Morley TS, Wang QA, Delgado-Lopez F, Wang ZV, et al. Adipocyte inflammation is essential for healthy adipose tissue expansion and remodeling. Cell Metab. 2014;20:103.CrossRef
46.
go back to reference Crewe C, An YA, Scherer PE. The ominous triad of adipose tissue dysfunction: inflammation, fibrosis, and impaired angiogenesis. J Clin Invest. 2017;127:74.CrossRef Crewe C, An YA, Scherer PE. The ominous triad of adipose tissue dysfunction: inflammation, fibrosis, and impaired angiogenesis. J Clin Invest. 2017;127:74.CrossRef
47.
go back to reference Zhu Q, An YA, Kim M, Zhang Z, Zhao S, Zhu Y, et al. Suppressing adipocyte inflammation promotes insulin resistance in mice. Mol Metab. 2020;39:101010.CrossRef Zhu Q, An YA, Kim M, Zhang Z, Zhao S, Zhu Y, et al. Suppressing adipocyte inflammation promotes insulin resistance in mice. Mol Metab. 2020;39:101010.CrossRef
48.
go back to reference Vinaik R, Barayan D, Jeschke MG. NLRP3 Inflammasome in Inflammation and Metabolism: Identifying Novel Roles in Postburn Adipose Dysfunction. Endocrinology. 2020;161:bqaa116. Vinaik R, Barayan D, Jeschke MG. NLRP3 Inflammasome in Inflammation and Metabolism: Identifying Novel Roles in Postburn Adipose Dysfunction. Endocrinology. 2020;161:bqaa116.
49.
go back to reference Smith GI, Mittendorfer B, Klein S. Metabolically healthy obesity: facts and fantasies. J Clin Invest. 2019;129:3978.CrossRef Smith GI, Mittendorfer B, Klein S. Metabolically healthy obesity: facts and fantasies. J Clin Invest. 2019;129:3978.CrossRef
50.
go back to reference Vishvanath L, Gupta RK. Contribution of adipogenesis to healthy adipose tissue expansion in obesity. J Clin Invest. 2019;129:4022.CrossRef Vishvanath L, Gupta RK. Contribution of adipogenesis to healthy adipose tissue expansion in obesity. J Clin Invest. 2019;129:4022.CrossRef
51.
go back to reference Boulet N, Esteve D, Bouloumie A, Galitzky J. Cellular heterogeneity in superficial and deep subcutaneous adipose tissues in overweight patients. J Physiol Biochem. 2013;69:575.CrossRef Boulet N, Esteve D, Bouloumie A, Galitzky J. Cellular heterogeneity in superficial and deep subcutaneous adipose tissues in overweight patients. J Physiol Biochem. 2013;69:575.CrossRef
52.
go back to reference Kosaka K, Kubota Y, Adachi N, Akita S, Sasahara Y, Kira T, et al. Human adipocytes from the subcutaneous superficial layer have greater adipogenic potential and lower PPAR-gamma DNA methylation levels than deep layer adipocytes. Am J Physiol Cell Physiol. 2016;311:C322–9.CrossRef Kosaka K, Kubota Y, Adachi N, Akita S, Sasahara Y, Kira T, et al. Human adipocytes from the subcutaneous superficial layer have greater adipogenic potential and lower PPAR-gamma DNA methylation levels than deep layer adipocytes. Am J Physiol Cell Physiol. 2016;311:C322–9.CrossRef
53.
go back to reference Lemmer IL, Willemsen N, Hilal N, Bartelt A. A guide to understanding endoplasmic reticulum stress in metabolic disorders. Mol Metab. 2021;47:101169.CrossRef Lemmer IL, Willemsen N, Hilal N, Bartelt A. A guide to understanding endoplasmic reticulum stress in metabolic disorders. Mol Metab. 2021;47:101169.CrossRef
54.
go back to reference Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu Rev Immunol. 2011;29:415.CrossRef Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu Rev Immunol. 2011;29:415.CrossRef
55.
go back to reference Reilly SM, Saltiel AR. Adapting to obesity with adipose tissue inflammation. Nat Rev Endocrinol. 2017;13:633.CrossRef Reilly SM, Saltiel AR. Adapting to obesity with adipose tissue inflammation. Nat Rev Endocrinol. 2017;13:633.CrossRef
56.
go back to reference Korbecki J, Bajdak-Rusinek K. The effect of palmitic acid on inflammatory response in macrophages: an overview of molecular mechanisms. Inflamm Res. 2019;68:915.CrossRef Korbecki J, Bajdak-Rusinek K. The effect of palmitic acid on inflammatory response in macrophages: an overview of molecular mechanisms. Inflamm Res. 2019;68:915.CrossRef
57.
go back to reference Prentice KJ, Saksi J, Hotamisligil GS. Adipokine FABP4 integrates energy stores and counterregulatory metabolic responses. J Lipid Res. 2019;60:734.CrossRef Prentice KJ, Saksi J, Hotamisligil GS. Adipokine FABP4 integrates energy stores and counterregulatory metabolic responses. J Lipid Res. 2019;60:734.CrossRef
58.
go back to reference Li HL, Wu X, Xu A, Hoo RL. A-FABP in Metabolic Diseases and the Therapeutic Implications: An Update. Int J Mol Sci. 2021;22:9386. Li HL, Wu X, Xu A, Hoo RL. A-FABP in Metabolic Diseases and the Therapeutic Implications: An Update. Int J Mol Sci. 2021;22:9386.
59.
go back to reference Zhou H, Urso CJ, Jadeja V. Saturated Fatty Acids in Obesity-Associated Inflammation. J Inflamm Res. 2020;13:1.CrossRef Zhou H, Urso CJ, Jadeja V. Saturated Fatty Acids in Obesity-Associated Inflammation. J Inflamm Res. 2020;13:1.CrossRef
60.
go back to reference Lancaster GI, Langley KG, Berglund NA, Kammoun HL, Reibe S, Estevez E, et al. Evidence that TLR4 Is Not a Receptor for Saturated Fatty Acids but Mediates Lipid-Induced Inflammation by Reprogramming Macrophage Metabolism. Cell Metab. 2018;27:1096.CrossRef Lancaster GI, Langley KG, Berglund NA, Kammoun HL, Reibe S, Estevez E, et al. Evidence that TLR4 Is Not a Receptor for Saturated Fatty Acids but Mediates Lipid-Induced Inflammation by Reprogramming Macrophage Metabolism. Cell Metab. 2018;27:1096.CrossRef
61.
go back to reference Moon JS, da Cunha FF, Huh JY, Andreyev AY, Lee J, Mahata SK, et al. ANT2 drives proinflammatory macrophage activation in obesity. JCI. Insight. 2021;6:e147033. Moon JS, da Cunha FF, Huh JY, Andreyev AY, Lee J, Mahata SK, et al. ANT2 drives proinflammatory macrophage activation in obesity. JCI. Insight. 2021;6:e147033.
62.
go back to reference Kiernan K, MacIver NJ. The Role of the Adipokine Leptin in Immune Cell Function in Health and Disease. Front Immunol. 2020;11:622468.CrossRef Kiernan K, MacIver NJ. The Role of the Adipokine Leptin in Immune Cell Function in Health and Disease. Front Immunol. 2020;11:622468.CrossRef
63.
go back to reference Cao Y. Angiogenesis modulates adipogenesis and obesity. J Clin Invest. 2007;117:2362.CrossRef Cao Y. Angiogenesis modulates adipogenesis and obesity. J Clin Invest. 2007;117:2362.CrossRef
64.
go back to reference Trayhurn P. Hypoxia and adipose tissue function and dysfunction in obesity. Physiol Rev. 2013;93:1.CrossRef Trayhurn P. Hypoxia and adipose tissue function and dysfunction in obesity. Physiol Rev. 2013;93:1.CrossRef
65.
go back to reference Hosogai N, Fukuhara A, Oshima K, Miyata Y, Tanaka S, Segawa K, et al. Adipose tissue hypoxia in obesity and its impact on adipocytokine dysregulation. Diabetes. 2007;56:901.CrossRef Hosogai N, Fukuhara A, Oshima K, Miyata Y, Tanaka S, Segawa K, et al. Adipose tissue hypoxia in obesity and its impact on adipocytokine dysregulation. Diabetes. 2007;56:901.CrossRef
66.
go back to reference Skurk T, Mack I, Kempf K, Kolb H, Hauner H, Herder C. Expression and secretion of RANTES (CCL5) in human adipocytes in response to immunological stimuli and hypoxia. Horm Metab Res. 2009;41:183.CrossRef Skurk T, Mack I, Kempf K, Kolb H, Hauner H, Herder C. Expression and secretion of RANTES (CCL5) in human adipocytes in response to immunological stimuli and hypoxia. Horm Metab Res. 2009;41:183.CrossRef
67.
go back to reference Trayhurn P, Alomar SY. Oxygen deprivation and the cellular response to hypoxia in adipocytes - perspectives on white and brown adipose tissues in obesity. Front Endocrinol (Lausanne). 2015;6:19.CrossRef Trayhurn P, Alomar SY. Oxygen deprivation and the cellular response to hypoxia in adipocytes - perspectives on white and brown adipose tissues in obesity. Front Endocrinol (Lausanne). 2015;6:19.CrossRef
68.
go back to reference Mori MA, Ludwig RG, Garcia-Martin R, Brandao BB, Kahn CR. Extracellular miRNAs: From Biomarkers to Mediators of Physiology and Disease. Cell Metab. 2019;30:656.CrossRef Mori MA, Ludwig RG, Garcia-Martin R, Brandao BB, Kahn CR. Extracellular miRNAs: From Biomarkers to Mediators of Physiology and Disease. Cell Metab. 2019;30:656.CrossRef
69.
go back to reference Cho CH, Koh YJ, Han J, Sung HK, Jong LH, Morisada T, et al. Angiogenic role of LYVE-1-positive macrophages in adipose tissue. Circ Res. 2007;100:e47–57.CrossRef Cho CH, Koh YJ, Han J, Sung HK, Jong LH, Morisada T, et al. Angiogenic role of LYVE-1-positive macrophages in adipose tissue. Circ Res. 2007;100:e47–57.CrossRef
70.
go back to reference Rausch ME, Weisberg S, Vardhana P, Tortoriello DV. Obesity in C57BL/6J mice is characterized by adipose tissue hypoxia and cytotoxic T-cell infiltration. Int J Obes (Lond). 2008;32:451.CrossRef Rausch ME, Weisberg S, Vardhana P, Tortoriello DV. Obesity in C57BL/6J mice is characterized by adipose tissue hypoxia and cytotoxic T-cell infiltration. Int J Obes (Lond). 2008;32:451.CrossRef
71.
go back to reference Song J, Deng T. The Adipocyte and Adaptive Immunity. Front Immunol. 2020;11:593058.CrossRef Song J, Deng T. The Adipocyte and Adaptive Immunity. Front Immunol. 2020;11:593058.CrossRef
72.
go back to reference Fruhbeck G, Fernandez-Quintana B, Paniagua M, Hernandez-Pardos AW, Valenti V, Moncada R, et al. FNDC4, a novel adipokine that reduces lipogenesis and promotes fat browning in human visceral adipocytes. Metabolism. 2020;108:154261.CrossRef Fruhbeck G, Fernandez-Quintana B, Paniagua M, Hernandez-Pardos AW, Valenti V, Moncada R, et al. FNDC4, a novel adipokine that reduces lipogenesis and promotes fat browning in human visceral adipocytes. Metabolism. 2020;108:154261.CrossRef
73.
go back to reference Bosma M, Gerling M, Pasto J, Georgiadi A, Graham E, Shilkova O, et al. FNDC4 acts as an anti-inflammatory factor on macrophages and improves colitis in mice. Nat Commun. 2016;7:11314.CrossRef Bosma M, Gerling M, Pasto J, Georgiadi A, Graham E, Shilkova O, et al. FNDC4 acts as an anti-inflammatory factor on macrophages and improves colitis in mice. Nat Commun. 2016;7:11314.CrossRef
74.
go back to reference Meizlish ML, Franklin RA, Zhou X, Medzhitov R. Tissue Homeostasis and Inflammation. Annu Rev Immunol. 2021;39:557.CrossRef Meizlish ML, Franklin RA, Zhou X, Medzhitov R. Tissue Homeostasis and Inflammation. Annu Rev Immunol. 2021;39:557.CrossRef
75.
go back to reference Fuchs A, Samovski D, Smith GI, Cifarelli V, Farabi SS, Yoshino J, et al. Associations Among Adipose Tissue Immunology, Inflammation, Exosomes and Insulin Sensitivity in People With Obesity and Nonalcoholic Fatty Liver Disease. Gastroenterology. 2021;161:968.CrossRef Fuchs A, Samovski D, Smith GI, Cifarelli V, Farabi SS, Yoshino J, et al. Associations Among Adipose Tissue Immunology, Inflammation, Exosomes and Insulin Sensitivity in People With Obesity and Nonalcoholic Fatty Liver Disease. Gastroenterology. 2021;161:968.CrossRef
76.
go back to reference Hong S, Song W, Zushin PH, Liu B, Jedrychowski MP, Mina AI, et al. Phosphorylation of Beta-3 adrenergic receptor at serine 247 by ERK MAP kinase drives lipolysis in obese adipocytes. Mol Metab. 2018;12:25.CrossRef Hong S, Song W, Zushin PH, Liu B, Jedrychowski MP, Mina AI, et al. Phosphorylation of Beta-3 adrenergic receptor at serine 247 by ERK MAP kinase drives lipolysis in obese adipocytes. Mol Metab. 2018;12:25.CrossRef
77.
go back to reference Foley KP, Chen Y, Barra NG, Heal M, Kwok K, Tamrakar AK, et al. Inflammation promotes adipocyte lipolysis via IRE1 kinase. J Biol Chem. 2021;296:100440.CrossRef Foley KP, Chen Y, Barra NG, Heal M, Kwok K, Tamrakar AK, et al. Inflammation promotes adipocyte lipolysis via IRE1 kinase. J Biol Chem. 2021;296:100440.CrossRef
78.
go back to reference Xu L, Liu W, Bai F, Xu Y, Liang X, Ma C, et al. Hepatic Macrophage as a Key Player in Fatty Liver Disease. Front Immunol. 2021;12:708978.CrossRef Xu L, Liu W, Bai F, Xu Y, Liang X, Ma C, et al. Hepatic Macrophage as a Key Player in Fatty Liver Disease. Front Immunol. 2021;12:708978.CrossRef
79.
go back to reference Huang Z, Xu A. Adipose Extracellular Vesicles in Intercellular and Inter-Organ Crosstalk in Metabolic Health and Diseases. Front Immunol. 2021;12:608680.CrossRef Huang Z, Xu A. Adipose Extracellular Vesicles in Intercellular and Inter-Organ Crosstalk in Metabolic Health and Diseases. Front Immunol. 2021;12:608680.CrossRef
80.
go back to reference Cai Z, Huang Y, He B. New Insights into Adipose Tissue Macrophages in Obesity and Insulin Resistance. Cells. 2022;11:1424. Cai Z, Huang Y, He B. New Insights into Adipose Tissue Macrophages in Obesity and Insulin Resistance. Cells. 2022;11:1424.
81.
go back to reference Strissel KJ, Stancheva Z, Miyoshi H, Perfield JW, DeFuria J, Jick Z, et al. Adipocyte death, adipose tissue remodeling, and obesity complications. Diabetes. 2007;56:2910.CrossRef Strissel KJ, Stancheva Z, Miyoshi H, Perfield JW, DeFuria J, Jick Z, et al. Adipocyte death, adipose tissue remodeling, and obesity complications. Diabetes. 2007;56:2910.CrossRef
82.
go back to reference Gong T, Liu L, Jiang W, Zhou R. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat Rev Immunol. 2020;20:95.CrossRef Gong T, Liu L, Jiang W, Zhou R. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat Rev Immunol. 2020;20:95.CrossRef
83.
go back to reference Guzman-Ruiz R, Tercero-Alcazar C, Lopez-Alcala J, Sanchez-Ceinos J, Malagon MM, Gordon A. The potential role of the adipokine HMGB1 in obesity and insulin resistance. Novel effects on adipose tissue biology. Mol Cell Endocrinol. 2021;536:111417.CrossRef Guzman-Ruiz R, Tercero-Alcazar C, Lopez-Alcala J, Sanchez-Ceinos J, Malagon MM, Gordon A. The potential role of the adipokine HMGB1 in obesity and insulin resistance. Novel effects on adipose tissue biology. Mol Cell Endocrinol. 2021;536:111417.CrossRef
84.
go back to reference Haase J, Weyer U, Immig K, Kloting N, Bluher M, Eilers J, et al. Local proliferation of macrophages in adipose tissue during obesity-induced inflammation. Diabetologia. 2014;57:562.CrossRef Haase J, Weyer U, Immig K, Kloting N, Bluher M, Eilers J, et al. Local proliferation of macrophages in adipose tissue during obesity-induced inflammation. Diabetologia. 2014;57:562.CrossRef
85.
go back to reference Hill DA, Lim HW, Kim YH, Ho WY, Foong YH, Nelson VL, et al. Distinct macrophage populations direct inflammatory versus physiological changes in adipose tissue. Proc Natl Acad Sci U S A. 2018;115:E5096–105.CrossRef Hill DA, Lim HW, Kim YH, Ho WY, Foong YH, Nelson VL, et al. Distinct macrophage populations direct inflammatory versus physiological changes in adipose tissue. Proc Natl Acad Sci U S A. 2018;115:E5096–105.CrossRef
86.
go back to reference Vandanmagsar B, Youm YH, Ravussin A, Galgani JE, Stadler K, Mynatt RL, et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med. 2011;17:179.CrossRef Vandanmagsar B, Youm YH, Ravussin A, Galgani JE, Stadler K, Mynatt RL, et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med. 2011;17:179.CrossRef
87.
go back to reference Kanda H, Tateya S, Tamori Y, Kotani K, Hiasa K, Kitazawa R, et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest. 2006;116:1494.CrossRef Kanda H, Tateya S, Tamori Y, Kotani K, Hiasa K, Kitazawa R, et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest. 2006;116:1494.CrossRef
88.
go back to reference Dommel S, Bluher M. Does C-C Motif Chemokine Ligand 2 (CCL2) Link Obesity to a Pro-Inflammatory State? Int J Mol Sci. 2021;22:1500. Dommel S, Bluher M. Does C-C Motif Chemokine Ligand 2 (CCL2) Link Obesity to a Pro-Inflammatory State? Int J Mol Sci. 2021;22:1500.
89.
go back to reference Palmer AK, Xu M, Zhu Y, Pirtskhalava T, Weivoda MM, Hachfeld CM, et al. Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell. 2019;18:e12950.CrossRef Palmer AK, Xu M, Zhu Y, Pirtskhalava T, Weivoda MM, Hachfeld CM, et al. Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell. 2019;18:e12950.CrossRef
90.
go back to reference Wang L, Wang B, Gasek NS, Zhou Y, Cohn RL, Martin DE, et al. Targeting p21(Cip1) highly expressing cells in adipose tissue alleviates insulin resistance in obesity. Cell Metab. 2022;34:75.CrossRef Wang L, Wang B, Gasek NS, Zhou Y, Cohn RL, Martin DE, et al. Targeting p21(Cip1) highly expressing cells in adipose tissue alleviates insulin resistance in obesity. Cell Metab. 2022;34:75.CrossRef
91.
go back to reference Matacchione G, Perugini J, Di ME, Sabbatinelli J, Prattichizzo F, Senzacqua M, et al. Senescent macrophages in the human adipose tissue as a source of inflammaging. Geroscience. 2022;44:1941-60. Matacchione G, Perugini J, Di ME, Sabbatinelli J, Prattichizzo F, Senzacqua M, et al. Senescent macrophages in the human adipose tissue as a source of inflammaging. Geroscience. 2022;44:1941-60.
92.
go back to reference Sun K, Park J, Gupta OT, Holland WL, Auerbach P, Zhang N, et al. Endotrophin triggers adipose tissue fibrosis and metabolic dysfunction. Nat Commun. 2014;5:3485.CrossRef Sun K, Park J, Gupta OT, Holland WL, Auerbach P, Zhang N, et al. Endotrophin triggers adipose tissue fibrosis and metabolic dysfunction. Nat Commun. 2014;5:3485.CrossRef
93.
go back to reference Marcelin G, Silveira ALM, Martins LB, Ferreira AV, Clement K. Deciphering the cellular interplays underlying obesity-induced adipose tissue fibrosis. J Clin Invest. 2019;129:4032.CrossRef Marcelin G, Silveira ALM, Martins LB, Ferreira AV, Clement K. Deciphering the cellular interplays underlying obesity-induced adipose tissue fibrosis. J Clin Invest. 2019;129:4032.CrossRef
94.
go back to reference Qi Y, Hui X. The shades of grey in adipose tissue reprogramming. Biosci Rep. 2022;42:BSR20212358. Qi Y, Hui X. The shades of grey in adipose tissue reprogramming. Biosci Rep. 2022;42:BSR20212358.
95.
go back to reference Winer DA, Winer S, Shen L, Wadia PP, Yantha J, Paltser G, et al. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med. 2011;17:610.CrossRef Winer DA, Winer S, Shen L, Wadia PP, Yantha J, Paltser G, et al. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med. 2011;17:610.CrossRef
96.
go back to reference Brestoff JR, Artis D. Immune regulation of metabolic homeostasis in health and disease. Cell. 2015;161:146.CrossRef Brestoff JR, Artis D. Immune regulation of metabolic homeostasis in health and disease. Cell. 2015;161:146.CrossRef
97.
go back to reference Wensveen FM, Jelencic V, Valentic S, Sestan M, Wensveen TT, Theurich S, et al. NK cells link obesity-induced adipose stress to inflammation and insulin resistance. Nat Immunol. 2015;16:376.CrossRef Wensveen FM, Jelencic V, Valentic S, Sestan M, Wensveen TT, Theurich S, et al. NK cells link obesity-induced adipose stress to inflammation and insulin resistance. Nat Immunol. 2015;16:376.CrossRef
98.
go back to reference McLaughlin T, Ackerman SE, Shen L, Engleman E. Role of innate and adaptive immunity in obesity-associated metabolic disease. J Clin Invest. 2017;127:5.CrossRef McLaughlin T, Ackerman SE, Shen L, Engleman E. Role of innate and adaptive immunity in obesity-associated metabolic disease. J Clin Invest. 2017;127:5.CrossRef
99.
go back to reference Hildreth AD, Ma F, Wong YY, Sun R, Pellegrini M, O'Sullivan TE. Single-cell sequencing of human white adipose tissue identifies new cell states in health and obesity. Nat Immunol. 2021;22:639.CrossRef Hildreth AD, Ma F, Wong YY, Sun R, Pellegrini M, O'Sullivan TE. Single-cell sequencing of human white adipose tissue identifies new cell states in health and obesity. Nat Immunol. 2021;22:639.CrossRef
100.
go back to reference Kratz M, Coats BR, Hisert KB, Hagman D, Mutskov V, Peris E, et al. Metabolic dysfunction drives a mechanistically distinct proinflammatory phenotype in adipose tissue macrophages. Cell Metab. 2014;20:614.CrossRef Kratz M, Coats BR, Hisert KB, Hagman D, Mutskov V, Peris E, et al. Metabolic dysfunction drives a mechanistically distinct proinflammatory phenotype in adipose tissue macrophages. Cell Metab. 2014;20:614.CrossRef
101.
go back to reference Snodgrass RG, Boss M, Zezina E, Weigert A, Dehne N, Fleming I, et al. Hypoxia Potentiates Palmitate-induced Pro-inflammatory Activation of Primary Human Macrophages. J Biol Chem. 2016;291:413.CrossRef Snodgrass RG, Boss M, Zezina E, Weigert A, Dehne N, Fleming I, et al. Hypoxia Potentiates Palmitate-induced Pro-inflammatory Activation of Primary Human Macrophages. J Biol Chem. 2016;291:413.CrossRef
102.
go back to reference Boutens L, Hooiveld GJ, Dhingra S, Cramer RA, Netea MG, Stienstra R. Unique metabolic activation of adipose tissue macrophages in obesity promotes inflammatory responses. Diabetologia. 2018;61:942.CrossRef Boutens L, Hooiveld GJ, Dhingra S, Cramer RA, Netea MG, Stienstra R. Unique metabolic activation of adipose tissue macrophages in obesity promotes inflammatory responses. Diabetologia. 2018;61:942.CrossRef
103.
go back to reference Lee YS, Olefsky J. Chronic tissue inflammation and metabolic disease. Genes Dev. 2021;35:307.CrossRef Lee YS, Olefsky J. Chronic tissue inflammation and metabolic disease. Genes Dev. 2021;35:307.CrossRef
104.
go back to reference Lee BC, Kim MS, Pae M, Yamamoto Y, Eberle D, Shimada T, et al. Adipose Natural Killer Cells Regulate Adipose Tissue Macrophages to Promote Insulin Resistance in Obesity. Cell Metab. 2016;23:685.CrossRef Lee BC, Kim MS, Pae M, Yamamoto Y, Eberle D, Shimada T, et al. Adipose Natural Killer Cells Regulate Adipose Tissue Macrophages to Promote Insulin Resistance in Obesity. Cell Metab. 2016;23:685.CrossRef
105.
go back to reference Li C, Menoret A, Farragher C, Ouyang Z, Bonin C, Holvoet P, et al. Single cell transcriptomics based-MacSpectrum reveals novel macrophage activation signatures in diseases. JCI. Insight. 2019;5:e126453. Li C, Menoret A, Farragher C, Ouyang Z, Bonin C, Holvoet P, et al. Single cell transcriptomics based-MacSpectrum reveals novel macrophage activation signatures in diseases. JCI. Insight. 2019;5:e126453.
106.
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.CrossRef Caslin HL, Bhanot M, Bolus WR, Hasty AH. Adipose tissue macrophages: Unique polarization and bioenergetics in obesity. Immunol Rev. 2020;295:101.CrossRef
107.
go back to reference Kintscher U, Hartge M, Hess K, Foryst-Ludwig A, Clemenz M, Wabitsch M, et al. T-lymphocyte infiltration in visceral adipose tissue: a primary event in adipose tissue inflammation and the development of obesity-mediated insulin resistance. Arterioscler Thromb Vasc Biol. 2008;28:1304.CrossRef Kintscher U, Hartge M, Hess K, Foryst-Ludwig A, Clemenz M, Wabitsch M, et al. T-lymphocyte infiltration in visceral adipose tissue: a primary event in adipose tissue inflammation and the development of obesity-mediated insulin resistance. Arterioscler Thromb Vasc Biol. 2008;28:1304.CrossRef
108.
go back to reference Duffaut C, Galitzky J, Lafontan M, Bouloumie A. Unexpected trafficking of immune cells within the adipose tissue during the onset of obesity. Biochem Biophys Res Commun. 2009;384:482.CrossRef Duffaut C, Galitzky J, Lafontan M, Bouloumie A. Unexpected trafficking of immune cells within the adipose tissue during the onset of obesity. Biochem Biophys Res Commun. 2009;384:482.CrossRef
109.
go back to reference Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med. 2009;15:914.CrossRef Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M, et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med. 2009;15:914.CrossRef
110.
go back to reference O'Rourke RW, White AE, Metcalf MD, Winters BR, Diggs BS, Zhu X, et al. Systemic inflammation and insulin sensitivity in obese IFN-gamma knockout mice. Metabolism. 2012;61:1152.CrossRef O'Rourke RW, White AE, Metcalf MD, Winters BR, Diggs BS, Zhu X, et al. Systemic inflammation and insulin sensitivity in obese IFN-gamma knockout mice. Metabolism. 2012;61:1152.CrossRef
111.
go back to reference Ferno J, Strand K, Mellgren G, Stiglund N, Bjorkstrom NK. Natural Killer Cells as Sensors of Adipose Tissue Stress. Trends Endocrinol Metab. 2020;31:3.CrossRef Ferno J, Strand K, Mellgren G, Stiglund N, Bjorkstrom NK. Natural Killer Cells as Sensors of Adipose Tissue Stress. Trends Endocrinol Metab. 2020;31:3.CrossRef
112.
go back to reference Toubal A, Kiaf B, Beaudoin L, Cagninacci L, Rhimi M, Fruchet B, et al. Mucosal-associated invariant T cells promote inflammation and intestinal dysbiosis leading to metabolic dysfunction during obesity. Nat Commun. 2020;11:3755.CrossRef Toubal A, Kiaf B, Beaudoin L, Cagninacci L, Rhimi M, Fruchet B, et al. Mucosal-associated invariant T cells promote inflammation and intestinal dysbiosis leading to metabolic dysfunction during obesity. Nat Commun. 2020;11:3755.CrossRef
113.
go back to reference Talukdar S, Oh DY, Bandyopadhyay G, Li D, Xu J, McNelis J, et al. Neutrophils mediate insulin resistance in mice fed a high-fat diet through secreted elastase. Nat Med. 2012;18:1407.CrossRef Talukdar S, Oh DY, Bandyopadhyay G, Li D, Xu J, McNelis J, et al. Neutrophils mediate insulin resistance in mice fed a high-fat diet through secreted elastase. Nat Med. 2012;18:1407.CrossRef
114.
go back to reference Freitas DF, Colon DF, Silva RL, Santos EM, Guimaraes VHD, Ribeiro GHM, et al. Neutrophil extracellular traps (NETs) modulate inflammatory profile in obese humans and mice: adipose tissue role on NETs levels. Mol Biol Rep. 2022;49:3225.CrossRef Freitas DF, Colon DF, Silva RL, Santos EM, Guimaraes VHD, Ribeiro GHM, et al. Neutrophil extracellular traps (NETs) modulate inflammatory profile in obese humans and mice: adipose tissue role on NETs levels. Mol Biol Rep. 2022;49:3225.CrossRef
115.
go back to reference Divoux A, Moutel S, Poitou C, Lacasa D, Veyrie N, Aissat A, et al. Mast cells in human adipose tissue: link with morbid obesity, inflammatory status, and diabetes. J Clin Endocrinol Metab. 2012;97:E1677–85.CrossRef Divoux A, Moutel S, Poitou C, Lacasa D, Veyrie N, Aissat A, et al. Mast cells in human adipose tissue: link with morbid obesity, inflammatory status, and diabetes. J Clin Endocrinol Metab. 2012;97:E1677–85.CrossRef
116.
go back to reference Goldstein N, Kezerle Y, Gepner Y, Haim Y, Pecht T, Gazit R, et al. Higher Mast Cell Accumulation in Human Adipose Tissues Defines Clinically Favorable Obesity Sub-Phenotypes. Cells. 2020;9:1508. Goldstein N, Kezerle Y, Gepner Y, Haim Y, Pecht T, Gazit R, et al. Higher Mast Cell Accumulation in Human Adipose Tissues Defines Clinically Favorable Obesity Sub-Phenotypes. Cells. 2020;9:1508.
117.
go back to reference Khan T, Muise ES, Iyengar P, Wang ZV, Chandalia M, Abate N, et al. Metabolic dysregulation and adipose tissue fibrosis: role of collagen VI. Mol Cell Biol. 2009;29:1575.CrossRef Khan T, Muise ES, Iyengar P, Wang ZV, Chandalia M, Abate N, et al. Metabolic dysregulation and adipose tissue fibrosis: role of collagen VI. Mol Cell Biol. 2009;29:1575.CrossRef
118.
go back to reference Datta R, Podolsky MJ, Atabai K. Fat fibrosis: friend or foe? JCI Insight. 2018;3:e122289. Datta R, Podolsky MJ, Atabai K. Fat fibrosis: friend or foe? JCI Insight. 2018;3:e122289.
119.
go back to reference Grosse L, Wagner N, Emelyanov A, Molina C, Lacas-Gervais S, Wagner KD, et al. Defined p16(High) Senescent Cell Types Are Indispensable for Mouse Healthspan. Cell Metab. 2020;32:87.CrossRef Grosse L, Wagner N, Emelyanov A, Molina C, Lacas-Gervais S, Wagner KD, et al. Defined p16(High) Senescent Cell Types Are Indispensable for Mouse Healthspan. Cell Metab. 2020;32:87.CrossRef
120.
go back to reference Demaria M, Ohtani N, Youssef SA, Rodier F, Toussaint W, Mitchell JR, et al. An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev Cell. 2014;31:722.CrossRef Demaria M, Ohtani N, Youssef SA, Rodier F, Toussaint W, Mitchell JR, et al. An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev Cell. 2014;31:722.CrossRef
121.
go back to reference Chaib S, Tchkonia T, Kirkland JL. Cellular senescence and senolytics: the path to the clinic. Nat Med. 2022;28:1556.CrossRef Chaib S, Tchkonia T, Kirkland JL. Cellular senescence and senolytics: the path to the clinic. Nat Med. 2022;28:1556.CrossRef
122.
go back to reference Fang J, Li L, Cao X, Yue H, Fu W, Chen Y, et al. Transmissible Endoplasmic Reticulum Stress Mediated by Extracellular Vesicles from Adipocyte Promoting the Senescence of Adipose-Derived Mesenchymal Stem Cells in Hypertrophic Obesity. Oxid Med Cell Longev. 2022;2022:7175027.CrossRef Fang J, Li L, Cao X, Yue H, Fu W, Chen Y, et al. Transmissible Endoplasmic Reticulum Stress Mediated by Extracellular Vesicles from Adipocyte Promoting the Senescence of Adipose-Derived Mesenchymal Stem Cells in Hypertrophic Obesity. Oxid Med Cell Longev. 2022;2022:7175027.CrossRef
123.
go back to reference Oh C, Koh D, Jeon HB, Kim KM. The Role of Extracellular Vesicles in Senescence. Mol Cells. 2022;45:603.CrossRef Oh C, Koh D, Jeon HB, Kim KM. The Role of Extracellular Vesicles in Senescence. Mol Cells. 2022;45:603.CrossRef
124.
go back to reference Li Q, Hagberg CE, Silva CH, Lang S, Hyvonen MT, Salehzadeh F, et al. Obesity and hyperinsulinemia drive adipocytes to activate a cell cycle program and senesce. Nat Med. 2021;27:1941.CrossRef Li Q, Hagberg CE, Silva CH, Lang S, Hyvonen MT, Salehzadeh F, et al. Obesity and hyperinsulinemia drive adipocytes to activate a cell cycle program and senesce. Nat Med. 2021;27:1941.CrossRef
125.
go back to reference Sierra-Ramirez A, Lopez-Aceituno JL, Costa-Machado LF, Plaza A, Barradas M, Fernandez-Marcos PJ. Transient metabolic improvement in obese mice treated with navitoclax or dasatinib/quercetin. Aging (Albany NY). 2020;12:11337.CrossRef Sierra-Ramirez A, Lopez-Aceituno JL, Costa-Machado LF, Plaza A, Barradas M, Fernandez-Marcos PJ. Transient metabolic improvement in obese mice treated with navitoclax or dasatinib/quercetin. Aging (Albany NY). 2020;12:11337.CrossRef
126.
go back to reference Roden M, Shulman GI. The integrative biology of type 2 diabetes. Nature. 2019;576:51.CrossRef Roden M, Shulman GI. The integrative biology of type 2 diabetes. Nature. 2019;576:51.CrossRef
127.
go back to reference Nogueiras R, Sabio G. Brain JNK and metabolic disease. Diabetologia. 2021;64:265.CrossRef Nogueiras R, Sabio G. Brain JNK and metabolic disease. Diabetologia. 2021;64:265.CrossRef
128.
go back to reference Regnier M, Van HM, Knauf C, Cani PD. Gut microbiome, endocrine control of gut barrier function and metabolic diseases. J Endocrinol. 2021;248:R67–82.CrossRef Regnier M, Van HM, Knauf C, Cani PD. Gut microbiome, endocrine control of gut barrier function and metabolic diseases. J Endocrinol. 2021;248:R67–82.CrossRef
129.
go back to reference Gallego-Duran R, Montero-Vallejo R, Maya-Miles D, Lucena A, Martin F, Ampuero J, et al. Analysis of Common Pathways and Markers From Non-Alcoholic Fatty Liver Disease to Immune-Mediated Diseases. Front Immunol. 2021;12:667354.CrossRef Gallego-Duran R, Montero-Vallejo R, Maya-Miles D, Lucena A, Martin F, Ampuero J, et al. Analysis of Common Pathways and Markers From Non-Alcoholic Fatty Liver Disease to Immune-Mediated Diseases. Front Immunol. 2021;12:667354.CrossRef
130.
go back to reference Barreby E, Chen P, Aouadi M. Macrophage functional diversity in NAFLD - more than inflammation. Nat Rev Endocrinol. 2022;18:461-72. Barreby E, Chen P, Aouadi M. Macrophage functional diversity in NAFLD - more than inflammation. Nat Rev Endocrinol. 2022;18:461-72.
131.
go back to reference Mashek DG. Hepatic lipid droplets: A balancing act between energy storage and metabolic dysfunction in NAFLD. Mol Metab. 2021;50:101115.CrossRef Mashek DG. Hepatic lipid droplets: A balancing act between energy storage and metabolic dysfunction in NAFLD. Mol Metab. 2021;50:101115.CrossRef
132.
go back to reference Flessa CM, Kyrou I, Nasiri-Ansari N, Kaltsas G, Kassi E, Randeva HS. Endoplasmic reticulum stress in nonalcoholic (metabolic associated) fatty liver disease (NAFLD/MAFLD). J Cell Biochem. 2022;123:1585-606. Flessa CM, Kyrou I, Nasiri-Ansari N, Kaltsas G, Kassi E, Randeva HS. Endoplasmic reticulum stress in nonalcoholic (metabolic associated) fatty liver disease (NAFLD/MAFLD). J Cell Biochem. 2022;123:1585-606.
133.
go back to reference Horn CL, Morales AL, Savard C, Farrell GC, Ioannou GN. Role of Cholesterol-Associated Steatohepatitis in the Development of NASH. Hepatol Commun. 2022;6:12.CrossRef Horn CL, Morales AL, Savard C, Farrell GC, Ioannou GN. Role of Cholesterol-Associated Steatohepatitis in the Development of NASH. Hepatol Commun. 2022;6:12.CrossRef
134.
go back to reference Huby T, Gautier EL. Immune cell-mediated features of non-alcoholic steatohepatitis. Nat Rev Immunol. 2022;22:429-43. Huby T, Gautier EL. Immune cell-mediated features of non-alcoholic steatohepatitis. Nat Rev Immunol. 2022;22:429-43.
135.
go back to reference Ramadori P, Kam S, Heikenwalder M. T cells: Friends and foes in NASH pathogenesis and hepatocarcinogenesis. Hepatology. 2022;75:1038.CrossRef Ramadori P, Kam S, Heikenwalder M. T cells: Friends and foes in NASH pathogenesis and hepatocarcinogenesis. Hepatology. 2022;75:1038.CrossRef
136.
go back to reference Shaker ME. The contribution of sterile inflammation to the fatty liver disease and the potential therapies. Biomed Pharmacother. 2022;148:112789.CrossRef Shaker ME. The contribution of sterile inflammation to the fatty liver disease and the potential therapies. Biomed Pharmacother. 2022;148:112789.CrossRef
137.
go back to reference Kolb H, Mandrup-Poulsen T. An immune origin of type 2 diabetes? Diabetologia. 2005;48:1038.CrossRef Kolb H, Mandrup-Poulsen T. An immune origin of type 2 diabetes? Diabetologia. 2005;48:1038.CrossRef
138.
go back to reference Goldfine AB, Shoelson SE. Therapeutic approaches targeting inflammation for diabetes and associated cardiovascular risk. J Clin Invest. 2017;127:83.CrossRef Goldfine AB, Shoelson SE. Therapeutic approaches targeting inflammation for diabetes and associated cardiovascular risk. J Clin Invest. 2017;127:83.CrossRef
139.
go back to reference da Cruz Nascimento SS, Carvalho de Queiroz JL, Fernandes de MA, de Franca Nunes AC, Piuvezam G, Lima Maciel BL, et al. Anti-inflammatory agents as modulators of the inflammation in adipose tissue: A systematic review. Plos one. 2022;17:e0273942.CrossRef da Cruz Nascimento SS, Carvalho de Queiroz JL, Fernandes de MA, de Franca Nunes AC, Piuvezam G, Lima Maciel BL, et al. Anti-inflammatory agents as modulators of the inflammation in adipose tissue: A systematic review. Plos one. 2022;17:e0273942.CrossRef
Metadata
Title
Obese visceral fat tissue inflammation: from protective to detrimental?
Author
Hubert Kolb
Publication date
01-12-2022
Publisher
BioMed Central
Published in
BMC Medicine / Issue 1/2022
Electronic ISSN: 1741-7015
DOI
https://doi.org/10.1186/s12916-022-02672-y

Other articles of this Issue 1/2022

BMC Medicine 1/2022 Go to the issue