Extracellular matrix remodelling in obesity and metabolic disorders

Vishal Musale, David H. Wasserman, Li Kang

PDF(1263 KB)
PDF(1263 KB)
Life Metabolism ›› 2023, Vol. 2 ›› Issue (4) : 155-169. DOI: 10.1093/lifemeta/load021
Minireview

Extracellular matrix remodelling in obesity and metabolic disorders

Author information +
History +

Abstract

Obesity causes extracellular matrix (ECM) remodelling which can develop into serious pathology and fibrosis, having metabolic effects in insulin-sensitive tissues. The ECM components may be increased in response to overnutrition. This review will focus on specific obesity-associated molecular and pathophysiological mechanisms of ECM remodelling and the impact of specific interactions on tissue metabolism. In obesity, a complex network of signalling molecules such as cytokines and growth factors has been implicated in fibrosis. Increased ECM deposition contributes to the pathogenesis of insulin resistance at least in part through the activation of cell surface integrin receptors and CD44 signalling cascades. These cell surface receptors transmit signals to the cell adhesome which orchestrates an intracellular response that adapts to the extracellular environment. Matrix proteins, glycoproteins, and poly-saccharides interact through ligand-specific cell surface receptors that interact with the cytosolic adhesion proteins to elicit specific actions. Cell adhesion proteins may have catalytic activity or serve as scaffolds. The vast number of cell surface receptors and the complexity of the cell adhesome have made study of their roles challenging in health and disease. Further complicating the role of ECM-cell receptor interactions is the variation between cell types. This review will focus on recent insights gained from studies of two highly conserved, ubiquitous axes and how they contribute to insulin resistance and metabolic dysfunction in obesity. These are the collagen-integrin receptor-IPP (ILK-PINCH-Parvin) axis and the hyaluronan-CD44 interaction. We speculate that targeting ECM components or their receptor-mediated cell signalling may provide novel insights into the treatment of obesity-associated cardiometabolic complications.

Keywords

extracellular matrix / fibrosis / insulin resistance / obesity / metabolism

Cite this article

Download citation ▾
Vishal Musale, David H. Wasserman, Li Kang. Extracellular matrix remodelling in obesity and metabolic disorders. Life Metabolism, 2023, 2(4): 155‒169 https://doi.org/10.1093/lifemeta/load021

References

[1]
Loganathan R, Rongish BJ, Smith CM et al. Extracellular matrix motion and early morphogenesis. Development 2016;143:2056–65.
CrossRef Google scholar
[2]
Kang L, Ayala JE, Lee-Young RS et al. Diet-induced muscle insulin resistance is associated with extracellular matrix remodeling and interaction with integrin α2β 1in mice. Diabetes 2011;60:416–26.
CrossRef Google scholar
[3]
Kang L, Lantier L, Kennedy A et al. Hyaluronan accumulates with high-fat feeding and contributes to insulin resistance. Diabetes 2013;62:1888–96.
CrossRef Google scholar
[4]
Kang L, Mayes WH, James FD et al. Matrix metalloproteinase 9 opposes diet-induced muscle insulin resistance in mice. Diabetologia 2014;57:603–13.
CrossRef Google scholar
[5]
Williams AS, Kang L, Zheng J et al. Integrin α1-null mice exhibit improved fatty liver when fed a high fat diet despite severe hepatic insulin resistance. J Biol Chem 2015;290:6546–57.
CrossRef Google scholar
[6]
Kang L, Mokshagundam S, Reuter B et al. Integrin-linked kinase in muscle is necessary for the development of insulin resistance in diet-induced obese mice. Diabetes 2016;65:1590–600.
CrossRef Google scholar
[7]
Hasib A, Hennayake CK, Bracy DP et al. CD44 contributes to hyaluronan-mediated insulin resistance in skeletal muscle of high-fat-fed C57BL/6 mice. Am J Physiol Endocrinol Metab 2019;317:E973–83.
CrossRef Google scholar
[8]
Bugler-Lamb AR, Hasib A, Weng X et al. Adipocyte integrin-linked kinase plays a key role in the development of diet-induced adipose insulin resistance in male mice. Mol Metab 2021;49:101197.
CrossRef Google scholar
[9]
Schuppan D, Ashfaq-Khan M, Yang AT et al. Liver fibrosis: direct antifibrotic agents and targeted therapies. Matrix Biol 2018;68–69:435–51.
CrossRef Google scholar
[10]
Cooper J, Giancotti FG. Integrin signaling in cancer: mechanotransduction, stemness, epithelial plasticity, and therapeutic resistance. Cancer Cell 2019;35:347–67.
CrossRef Google scholar
[11]
Slack RJ, Macdonald SJF, Roper JA et al. Emerging therapeutic opportunities for integrin inhibitors. Nat Rev Drug Discov 2022;21:60–78.
CrossRef Google scholar
[12]
Bonnans C, Chou J, Werb Z. Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol 2014;15:786–801.
CrossRef Google scholar
[13]
Porter KE, Turner NA. Cardiac fibroblasts: at the heart of myocardial remodeling. Pharmacol Ther 2009;123:255–78.
CrossRef Google scholar
[14]
Datta R, Podolsky MJ, Atabai K. Fat fibrosis: friend or foe? JCI Insight 2018;3:e122289.
CrossRef Google scholar
[15]
Pompili S, Latella G, Gaudio E et al. The charming world of the extracellular matrix: a dynamic and protective network of the intestinal wall. Front Med (Lausanne) 2021;8:610189.
CrossRef Google scholar
[16]
Frantz C, Stewart KM, Weaver VM. The extracellular matrix at a glance. J Cell Sci 2010;123:4195–200.
CrossRef Google scholar
[17]
Plotnikov SV, Pasapera AM, Sabass B et al. Force fluctuations within focal adhesions mediate ECM-rigidity sensing to guide directed cell migration. Cell 2012;151:1513–27.
CrossRef Google scholar
[18]
Ginsberg MH, Partridge A, Shattil SJ. Integrin regulation. Curr Opin Cell Biol 2005;17:509–16.
CrossRef Google scholar
[19]
Hynes RO. The extracellular matrix: not just pretty fibrils. Science 2009;326:1216–9.
CrossRef Google scholar
[20]
Cox TR, Erler JT. Remodeling and homeostasis of the extracellular matrix: implications for fibrotic diseases and cancer. DMM Disease Models Mech 2011;4:165–78.
CrossRef Google scholar
[21]
Lu P, Takai K, Weaver VM et al. Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harbor Perspect Biol 2011;3:a005058.
CrossRef Google scholar
[22]
Rozario T, DeSimone DW. The extracellular matrix in development and morphogenesis: a dynamic view. Dev Biol 2010;341:126–40.
CrossRef Google scholar
[23]
Cox TR, Bird D, Baker AM et al. LOX-mediated collagen crosslinking is responsible for fibrosis-enhanced metastasis. Cancer Res 2013;73:1721–32.
CrossRef Google scholar
[24]
Kagan HM, Li W. Lysyl oxidase: properties, specificity, and biological roles inside and outside of the cell. J Cell Biochem 2003;88:660–72.
CrossRef Google scholar
[25]
Facchini FS, Hua N, Abbasi F et al. Insulin resistance as a predictor of age-related diseases. J Clin Endocrinol Metab 2001;86:3574–8.
CrossRef Google scholar
[26]
Lin D, Chun TH, Kang L. Adipose extracellular matrix remodelling in obesity and insulin resistance. Biochem Pharmacol 2016;119:8–16.
CrossRef Google scholar
[27]
Cavalera M, Wang J, Frangogiannis NG. Obesity, metabolic dysfunction, and cardiac fibrosis: pathophysiological pathways, molecular mechanisms, and therapeutic opportunities. Transl Res 2014;164:323–35.
CrossRef Google scholar
[28]
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.
CrossRef Google scholar
[29]
Lawler HM, Underkofler CM, Kern PA et al. Adipose tissue hypoxia, inflammation, and fibrosis in obese insulin-sensitive and obese insulin-resistant subjects. J Clin Endocrinol Metab 2016;101:1422–8.
CrossRef Google scholar
[30]
Tam CS, Covington JD, Bajpeyi S et al. Weight gain reveals dramatic increases in skeletal muscle extracellular matrix remodeling. J Clin Endocrinol Metab 2014;99:1749–57.
CrossRef Google scholar
[31]
Richardson DK, Kashyap S, Bajaj M et al. Lipid infusion decreases the expression of nuclear encoded mitochondrial genes and increases the expression of extracellular matrix genes in human skeletal muscle. J Biol Chem 2005;280:10290–7.
CrossRef Google scholar
[32]
Berria R, Wang L, Richardson DK et al. Increased collagen content in insulin-resistant skeletal muscle. Am J Physiol Endocrinol Metab 2006;290:560–5.
CrossRef Google scholar
[33]
Dantas WS, Roschel H, Murai IH et al. Exercise-induced increases in insulin sensitivity after bariatric surgery are mediated by muscle extracellular matrix remodeling. Diabetes 2020;69:1675–91.
CrossRef Google scholar
[34]
Crujeiras AB, Diaz-Lagares A, Moreno-Navarrete JM et al. Genome-wide DNA methylation pattern in visceral adipose tissue differentiates insulin-resistant from insulin-sensitive obese subjects. Transl Res 2016;178:13–24.e5.
CrossRef Google scholar
[35]
Crewe C, An YA, Scherer PE. The ominous triad of adipose tissue dysfunction: inflammation, fibrosis, and impaired angiogenesis. J Clin Investig 2017;127:74–82.
CrossRef Google scholar
[36]
Marcelin G, Gautier EL, Clement K. Adipose tissue fibrosis in obesity: etiology and challenges. Annu Rev Physiol 2022;84:135–55.
CrossRef Google scholar
[37]
Reilly SM, Saltiel AR. Adapting to obesity with adipose tissue inflammation. Nat Publish Group 2017;13:633–43.
CrossRef Google scholar
[38]
Verboven K, Wouters K, Gaens K et al. Abdominal subcutaneous and visceral adipocyte size, lipolysis and inflammation relate to insulin resistance in male obese humans. Sci Rep 2018;8:4677.
CrossRef Google scholar
[39]
Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest 2007;117:175–84.
CrossRef Google scholar
[40]
Haase J, Weyer U, Immig K et al. Local proliferation of macro-phages in adipose tissue during obesity-induced inflammation. Diabetologia 2014;57:562–71.
CrossRef Google scholar
[41]
Sun K, Li X, Scherer PE. Extracellular matrix (ECM) and fibrosis in adipose tissue: overview and perspectives. Compr Physiol 2023;13:4387–407.
CrossRef Google scholar
[42]
Williams AS, Trefts E, Lantier L et al. Integrin-linked kinase is necessary for the development of diet-induced hepatic insulin resistance. Diabetes 2017;66:325–34.
CrossRef Google scholar
[43]
Kodama K, Horikoshi M, Toda K et al. Expression-based genome-wide association study links the receptor CD44 in adipose tissue with type 2 diabetes. Proc Natl Acad Sci USA 2012;109:7049–54.
CrossRef Google scholar
[44]
Kodama K, Toda K, Morinaga S et al. Anti-CD44 antibody treatment lowers hyperglycemia and improves insulin resistance, adipose inflammation, and hepatic steatosis in diet-induced obese mice. Diabetes 2015;64:867–75.
CrossRef Google scholar
[45]
Reggio S, Pellegrinelli V, Clément K et al. Fibrosis as a cause or a consequence of white adipose tissue inflammation in obesity. Curr Obes Rep 2013;2:1–9.
CrossRef Google scholar
[46]
Rabhi N, Desevin K, Belkina AC et al. Obesity-induced senescent macrophages activate a fibrotic transcriptional program in adipocyte progenitors. Life Sci Alliance 2022;5:e202101286.
CrossRef Google scholar
[47]
Spencer M, Yao-Borengasser A, Unal R et al. Adipose tissue macrophages in insulin-resistant subjects are associated with collagen VI and fibrosis and demonstrate alternative activation. Am J Physiol Endocrinol Metab 2010;299:E1016–27.
CrossRef Google scholar
[48]
Galli SJ, Borregaard N, Wynn TA. Phenotypic and functional plasticity of cells of innate immunity: macrophages, mast cells and neutrophils. Nat Immunol 2011;12:1035–44.
CrossRef Google scholar
[49]
Martinez-Santibanez G, Lumeng CN. Macrophages and the regulation of adipose tissue remodeling. Annu Rev Nutr 2014;34:57–76.
CrossRef Google scholar
[50]
Marcelin G, Silveira ALM, Martins LB et al. Deciphering the cellular interplays underlying obesity-induced adipose tissue fibrosis. J Clin Invest 2019;129:4032–40.
CrossRef Google scholar
[51]
Bertola A, Deveaux V, Bonnafous S et al. Elevated expression of osteopontin may be related to adipose tissue macrophage accumulation and liver steatosis in morbid obesity. Diabetes 2009;58:125–33.
CrossRef Google scholar
[52]
Kiefer FW, Neschen S, Pfau B et al. Osteopontin deficiency protects against obesity-induced hepatic steatosis and attenuates glucose production in mice. Diabetologia 2011;54:2132–42.
CrossRef Google scholar
[53]
Paulus WJ, Tschöpe C. A novel paradigm for heart failure with preserved ejection fraction: Comorbidities drive myocardial dysfunction and remodeling through coronary microvascular endothelial inflammation. J Am Coll Cardiol 2013;62:263–71.
CrossRef Google scholar
[54]
Sabbah MS, Fayyaz AU, de Denus S et al. Obese-inflammatory phenotypes in heart failure with preserved ejection fraction. Circ Heart Fail 2020;13:e006414.
CrossRef Google scholar
[55]
Westermann D, Lindner D, Kasner M et al. Cardiac inflammation contributes to changes in the extracellular matrix in patients with heart failure and normal ejection fraction. Circ Heart Fail 2011;4:44–52.
CrossRef Google scholar
[56]
Lee HC, Shiou YL, Jhuo SJ et al. The sodium-glucose co-transporter 2 inhibitor empagliflozin attenuates cardiac fibrosis and improves ventricular hemodynamics in hypertensive heart failure rats. Cardiovasc Diabetol 2019;18:45.
CrossRef Google scholar
[57]
Wandrer F, Liebig S, Marhenke S et al. TNF-Receptor-1 inhibition reduces liver steatosis, hepatocellular injury and fibrosis in NAFLD mice. Cell Death Dis 2020;11:212.
CrossRef Google scholar
[58]
Tarrats N, Moles A, Morales A et al. Critical role of tumor necrosis factor receptor 1, but not 2, in hepatic stellate cell proliferation, extracellular matrix remodeling, and liver fibrogenesis. Hepatology 2011;54:319–27.
CrossRef Google scholar
[59]
Li YY, Feng YQ, Kadokami T et al. Myocardial extracellular matrix remodeling in transgenic mice overexpressing tumor necrosis factor α can be modulated by anti-tumor necrosis factor α therapy. Proc Natl Acad Sci USA 2000;97:12746–51.
CrossRef Google scholar
[60]
Zhang Y, Wang JH, Zhang YY et al. Deletion of interleukin-6 alleviated interstitial fibrosis in streptozotocin-induced diabetic cardiomyopathy of mice through affecting TGFβ1 and miR-29 pathways. Sci Rep 2016;6:23010.
CrossRef Google scholar
[61]
Debari MK, Abbott RD. Adipose tissue fibrosis: mechanisms, models, and importance. Int J Mol Sci 2020;21:6030.
CrossRef Google scholar
[62]
Rausch ME, Weisberg S, Vardhana P et al. Obesity in C57BL/6J mice is characterized by adipose tissue hypoxia and cytotoxic T-cell infiltration. Int J Obes 2008;32:451–63.
CrossRef Google scholar
[63]
Todorčević M, Manuel AR, Austen L et al. Markers of adipose tissue hypoxia are elevated in subcutaneous adipose tissue of severely obese patients with obesity hypoventilation syndrome but not in the moderately obese. Int J Obes 2021;45:1618–22.
CrossRef Google scholar
[64]
Halberg N, Khan T, Trujillo ME et al. Hypoxia-inducible factor 1α induces fibrosis and insulin resistance in white adipose tissue. Mol Cell Biol 2009;29:4467–83.
CrossRef Google scholar
[65]
Anvari G, Bellas E. Hypoxia induces stress fiber formation in adipocytes in the early stage of obesity. Sci Rep 2021;11:21473.
CrossRef Google scholar
[66]
Hammarstedt A, Gogg S, Hedjazifar S et al. Impaired adipogenesis and dysfunctional adipose tissue in human hypertrophic obesity. Physiol Rev 2018;98:1911–41.
CrossRef Google scholar
[67]
Khan T, Muise ES, Iyengar P et al. Metabolic dysregulation and adipose tissue fibrosis: role of collagen VI. Mol Cell Biol 2009;29:1575–91.
CrossRef Google scholar
[68]
Engin A. Adipose tissue hypoxia in obesity and its impact on preadipocytes and macrophages: hypoxia hypothesis. Adv Exp Med Biol 2017;960:305–26.
CrossRef Google scholar
[69]
Herold J, Kalucka J. Angiogenesis in adipose tissue: the interplay between adipose and endothelial cells. Front Physiol 2021;11:624903.
CrossRef Google scholar
[70]
Stockmann C, Kerdiles Y, Nomaksteinsky M et al. Loss of myeloid cell-derived vascular endothelial growth factor accelerates fibrosis. Proc Natl Acad Sci USA 2010;107:4329–34.
CrossRef Google scholar
[71]
Elias I, Franckhauser S, Ferré T et al. Adipose tissue over-expression of vascular endothelial growth factor protects against diet-induced obesity and insulin resistance. Diabetes 2012;61:1801–13.
CrossRef Google scholar
[72]
Sun K, Wernstedt Asterholm I, Kusminski CM et al. Dichotomous effects of VEGF-A on adipose tissue dysfunction. Proc Natl Acad Sci U S A 2012;109:5874–9.
CrossRef Google scholar
[73]
Sun K, Kusminski CM, Luby-Phelps K et al. Brown adipose tissue derived VEGF-A modulates cold tolerance and energy expenditure. Mol Metab 2014;3:474–83.
CrossRef Google scholar
[74]
Abdel-Misih SR, Bloomston M. Liver anatomy. Surg Clin North Am 2010;90:643–53.
CrossRef Google scholar
[75]
Gutiérrez-Cuevas J, Sandoval-Rodriguez A, Meza-Rios A et al. Molecular mechanisms of obesity-linked cardiac dysfunction: an up-date on current knowledge. Cells 2021;10:629.
CrossRef Google scholar
[76]
Siddesha JM, Valente AJ, Sakamuri SS et al. Angiotensin II stimulates cardiac fibroblast migration via the differential regulation of matrixins and RECK. J Mol Cell Cardiol 2013;65:9–18.
CrossRef Google scholar
[77]
Schorb W, Booz GW, Dostal DE et al. Angiotensin II is mitogenic in neonatal rat cardiac fibroblasts. Circ Res 1993;72:1245–54.
CrossRef Google scholar
[78]
Sadoshima JI, Izumo S. Molecular characterization of angiotensin II-induced hypertrophy of cardiac myocytes and hyperplasia of cardiac fibroblasts critical role of the AT1 receptor subtype. Circ Res 1993;73:413–23.
CrossRef Google scholar
[79]
Ohkubo N, Matsubara H, Nozawa Y et al. Angiotensin type 2 receptors are reexpressed by cardiac fibroblasts from failing myopathic hamster hearts and inhibit cell growth and fibrillar collagen metabolism. Circulation 1997;96:3954–62.
CrossRef Google scholar
[80]
Gao X, He X, Luo B et al. Angiotensin II increases collagen I expression via transforming growth factor-beta1 and extra-cellular signal-regulated kinase in cardiac fibroblasts. Eur J Pharmacol 2009;606:115–20.
CrossRef Google scholar
[81]
Toblli JE, Cao G, DeRosa G et al. Reduced cardiac expression of plasminogen activator inhibitor 1 and transforming growth factor β1 in obese Zucker rats by perindopril. Heart 2005;91:80–6.
CrossRef Google scholar
[82]
Sano M, Fukuda K, Sato T et al. ERK and p38 MAPK, but not NF-κB, are critically involved in reactive oxygen species-mediated induction of IL-6 by angiotensin II in cardiac fibroblasts. Circ Res 2001;89:661–9.
CrossRef Google scholar
[83]
Toblli JE, Mũoz MC, Cao G et al. ACE inhibition and AT1 receptor blockade prevent fatty liver and fibrosis in obese zucker rats. Obesity 2008;16:770–6.
CrossRef Google scholar
[84]
Azushima K, Ohki K, Wakui H et al. Adipocyte-specific enhancement of angiotensin II type 1 receptor-associated protein ameliorates diet-induced visceral obesity and insulin resistance. J Am Heart Assoc 2017;6:e004488.
CrossRef Google scholar
[85]
Menikdiwela KR, Ramalingam L, Allen L et al. Angiotensin II increases endoplasmic reticulum stress in adipose tissue and adipocytes. Sci Rep 2019;9:8481.
CrossRef Google scholar
[86]
Kakutani N, Takada S, Nambu H et al. Angiotensin-convertingenzyme inhibitor prevents skeletal muscle fibrosis in myocardial infarction mice. Skelet Muscle 2020;10:11.
CrossRef Google scholar
[87]
MacLaren RE, Cui W, Lu H et al. Association of adipocyte genes with ASP expression: a microarray analysis of subcutaneous and omental adipose tissue in morbidly obese subjects. BMC Med Genomics 2010;3:3.
CrossRef Google scholar
[88]
Watanabe Y, Nagai Y, Honda H et al. Isoliquiritigenin attenuates adipose tissue inflammation in vitro and adipose tissue fibrosis through inhibition of innate immune responses in mice. Sci Rep 2016;6:4–6.
CrossRef Google scholar
[89]
Yadav H, Quijano C, Kamaraju AK et al. Protection from obesity and diabetes by blockade of TGF-β/Smad3 signaling. Cell Metab 2011;14:67–79.
CrossRef Google scholar
[90]
Ulmasov B, Noritake H, Carmichael P et al. An inhibitor of arginine-glycine-aspartate-binding integrins reverses fibrosis in a mouse model of nonalcoholic steatohepatitis. Hepatol Commun 2019;3:246–61.
CrossRef Google scholar
[91]
Hermida N, López B, González A et al. A synthetic peptide from transforming growth factor-β1 type III receptor prevents myocardial fibrosis in spontaneously hypertensive rats. Cardiovasc Res 2009;81:601–9.
CrossRef Google scholar
[92]
Kanzler S, Lohse AW, Keil A et al. TGF-β1 in liver fibrosis: An inducible transgenic mouse model to study liver fibrogenesis. Am J Physiol 1999;276:G1059-68.
CrossRef Google scholar
[93]
Dobaczewski M, Bujak M, Li N et al. Smad3 signaling critically regulates fibroblast phenotype and function in healing myocardial infarction. Circ Res 2010;107:418–28.
CrossRef Google scholar
[94]
Frangogiannis NG. Cardiac fibrosis: cell biological mechanisms, molecular pathways and therapeutic opportunities. Mol Aspects Med 2019;65:70–99.
CrossRef Google scholar
[95]
Hu HH, Chen DQ, Wang YN et al. New insights into TGF-beta/Smad signaling in tissue fibrosis. Chem Biol Interact 2018;292:76–83.
CrossRef Google scholar
[96]
Tzavlaki K, Moustakas A. TGF-β signaling. Biomolecules 2020;10:487.
CrossRef Google scholar
[97]
Zhang L, Liu C, Meng XM et al. Smad2 protects against TGF-β1/Smad3-mediated collagen synthesis in human hepatic stellate cells during hepatic fibrosis. Mol Cell Biochem 2015;400:17–28.
CrossRef Google scholar
[98]
Piera-Velazquez S, Mendoza FA, Jimenez SA. Endothelial to Mesenchymal Transition (EndoMT) in the pathogenesis of human fibrotic diseases. J Clin Med 2016;5:45.
CrossRef Google scholar
[99]
Li Z, Jimenez SA. Protein kinase Cdelta and c-Abl kinase are required for transforming growth factor beta induction of endothelial-mesenchymal transition in vitro. Arthritis Rheum 2011;63:2473–83.
CrossRef Google scholar
[100]
Fadok VA, Bratton DL, Konowal A et al. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J Clin Invest 1998;101:890–8.
CrossRef Google scholar
[101]
Naiki Y, Michelsen KS, Zhang W et al. Transforming growth factor-beta differentially inhibits MyD88-dependent, but not TRAM- and TRIF-dependent, lipopolysaccharide-induced TLR4 signaling. J Biol Chem 2005;280:5491–5.
CrossRef Google scholar
[102]
Chen B, Huang S, Su Y et al. Macrophage Smad3 protects the infarcted heart, stimulating phagocytosis and regulating inflammation. Circ Res 2019;125:55–70.
CrossRef Google scholar
[103]
Wynn TA, Vannella KM. Macrophages in tissue repair, regeneration, and fibrosis. Immunity 2016;44:450–62.
CrossRef Google scholar
[104]
Frangogiannis N. Transforming growth factor-β in tissue fibrosis. J Exp Med 2020;217:e20190103.
CrossRef Google scholar
[105]
Noeman SA, Hamooda HE, Baalash AA. Biochemical study of oxidative stress markers in the liver, kidney and heart of high fat diet induced obesity in rats. Diabetol Metab Syndr 2011;3:17.
CrossRef Google scholar
[106]
Luangmonkong T, Suriguga S, Mutsaers HAM et al. Targeting oxidative stress for the treatment of liver fibrosis. Rev Physiol Biochem Pharmacol 2018;175:71–102.
CrossRef Google scholar
[107]
Cucoranu I, Clempus R, Dikalova A et al. NAD(P)H oxidase 4 mediates transforming growth factor-β1-induced differentiation of cardiac fibroblasts into myofibroblasts. Circ Res 2005;97:900–7.
CrossRef Google scholar
[108]
Wang M, Murdoch CE, Brewer AC et al. Endothelial NADPH oxidase 4 protects against angiotensin II-induced cardiac fibrosis and inflammation. ESC Heart Fail 2021;8:1427–37.
CrossRef Google scholar
[109]
Murdoch CE, Chaubey S, Zeng L et al. Endothelial NADPH oxidase- 2 promotes interstitial cardiac fibrosis and diastolic dysfunction through proinflammatory effects and endothelial-mesenchymal transition. J Am Coll Cardiol 2014;63:2734–41.
CrossRef Google scholar
[110]
Alcalá M, Sánchez-Vera I, Sevillano J et al. Vitamin E reduces adipose tissue fibrosis, inflammation, and oxidative stress and improves metabolic profile in obesity. Obesity 2015;23:1598–606.
CrossRef Google scholar
[111]
Kim KY, Choi I, Kim SS. Progression of hepatic stellate cell activation is associated with the level of oxidative stress rather than cytokines during CCl4-induced fibrogenesis. Mol Cells 2000;10:289–300.
CrossRef Google scholar
[112]
Jiang JX, Chen X, Serizawa N et al. Liver fibrosis and hepatocyte apoptosis are attenuated by GKT137831, a novel NOX4/NOX1 inhibitor in vivo. Free Radic Biol Med 2012;53:289–96.
CrossRef Google scholar
[113]
Lan T, Kisseleva T, Brenner DA. Deficiency of NOX1 or NOX4 prevents liver inflammation and fibrosis in mice through inhibition of hepatic stellate cell activation. PLoS One 2015;10:e0129743.
CrossRef Google scholar
[114]
Graae AS, Grarup N, Ribel-Madsen R et al. ADAMTS9 regulates skeletal muscle insulin sensitivity through extracellular matrix alterations. Diabetes 2019;68:502–14.
CrossRef Google scholar
[115]
Liu LF, Kodama K, Wei K et al. The receptor CD44 is associated with systemic insulin resistance and proinflammatory macrophages in human adipose tissue. Diabetologia 2015;58:1579–86.
CrossRef Google scholar
[116]
Datta R, Lizama CO, Soltani AK et al. Autoregulation of insulin receptor signaling through MFGE8 and the αvβ5 integrin. Proc Natl Acad Sci USA 2021;118:e2102171118.
CrossRef Google scholar
[117]
Kitzman DW, Lam CSP. Obese heart failure with preserved ejection fraction phenotype: from pariah to central player. Circulation 2017;136:20–3.
CrossRef Google scholar
[118]
Paulus WJ, Zile MR. From systemic inflammation to myocardial fibrosis: the heart failure with preserved ejection fraction paradigm revisited. Circ Res 2021;128:1451–67.
CrossRef Google scholar
[119]
Witteles RM, Fowler MB. Insulin-resistant cardiomyopathy clinical evidence, mechanisms, and treatment options. J Am Coll Cardiol 2008;51:93–102.
CrossRef Google scholar
[120]
McQueen AP, Zhang D, Hu P et al. Contractile dysfunction in hypertrophied hearts with deficient insulin receptor signaling: possible role of reduced capillary density. J Mol Cell Cardiol 2005;39:882–92.
CrossRef Google scholar
[121]
Domenighetti AA, Danes VR, Curl CL et al. Targeted GLUT-4 deficiency in the heart induces cardiomyocyte hypertrophy and impaired contractility linked with Ca2+ and proton flux dysregulation. J Mol Cell Cardiol 2010;48:663–72.
CrossRef Google scholar
[122]
Qi Y, Xu Z, Zhu Q et al. Myocardial loss of IRS1 and IRS2 causes heart failure and is controlled by p38α MAPK during insulin resistance. Diabetes 2013;62:3887–900.
CrossRef Google scholar
[123]
Riehle C, Wende AR, Sena S et al. Insulin receptor substrate signaling suppresses neonatal autophagy in the heart. J Clin Investig 2013;123:5319–33.
CrossRef Google scholar
[124]
Fu F, Zhao K, Li J et al. Direct evidence that myocardial insulin resistance following myocardial ischemia contributes to post-ischemic heart failure. Sci Rep 2015;5:17927.
CrossRef Google scholar
[125]
Zou T, Zhu M, Ma YC et al. MicroRNA-410-5p exacerbates high-fat diet-induced cardiac remodeling in mice in an endocrine fashion. Sci Rep 2018;8:8780.
CrossRef Google scholar
[126]
Shi Q, Liu X, Bai Y et al. In vitro effects of pirfenidone on cardiac fibroblasts: proliferation, myofibroblast differentiation, migration and cytokine secretion. PLoS One 2011;6:e28134.
CrossRef Google scholar
[127]
Chen G, Ni Y, Nagata N et al. Pirfenidone prevents and reverses hepatic insulin resistance and steatohepatitis by polarizing M2 macrophages. Lab Invest 2019;99:1335–48.
CrossRef Google scholar
[128]
Sandoval-Rodriguez A, Monroy-Ramirez HC, Meza-Rios A et al. Pirfenidone is an agonistic ligand for PPARalpha and improves NASH by activation of SIRT1/LKB1/pAMPK. Hepatol Commun 2020;4:434–49.
CrossRef Google scholar
[129]
Wang Z, Li L, Zhao H et al. Chronic high fat diet induces cardiac hypertrophy and fibrosis in mice. Metabolism 2015;64:917–25.
CrossRef Google scholar
[130]
McCurdy S, Baicu CF, Heymans S et al. Cardiac extracellular matrix remodeling: fibrillar collagens and Secreted Protein Acidic and Rich in Cysteine (SPARC). J Mol Cell Cardiol 2010;48:544–9.
CrossRef Google scholar
[131]
Chowdhury B, Xiang B, Liu M et al. Hyaluronidase 2 deficiency causes increased mesenchymal cells, congenital heart defects, and heart failure. Circul Cardiovasc Genet 2017;10:e001598.
CrossRef Google scholar
[132]
Muggenthaler MMA, Chowdhury B, Hasan SN et al. Mutations in HYAL2, encoding hyaluronidase 2, cause a syndrome of orofacial clefting and cor triatriatum sinister in humans and mice. PLoS Genet 2017;13:e1006470.
CrossRef Google scholar
[133]
Valiente-Alandi I, Potter SJ, Salvador AM et al. Inhibiting fibronectin attenuates fibrosis and improves cardiac function in a model of heart failure. Circulation 2018;138:1236–52.
CrossRef Google scholar
[134]
Konstandin MH, Völkers M, Collins B et al. Fibronectin contributes to pathological cardiac hypertrophy but not physiological growth. Basic Res Cardiol 2013;108:375.
CrossRef Google scholar
[135]
Huber J, Löffler M, Bilban M et al. Prevention of high-fat diet-induced adipose tissue remodeling in obese diabetic mice by n-3 polyunsaturated fatty acids. Int J Obes 2007;31:1004–13.
CrossRef Google scholar
[136]
Weng X, Lin D, Huang JTJ et al. Collagen 24 α1 is increased in insulin-resistant skeletal muscle and adipose tissue. Int J Mol Sci 2020;21:5738.
CrossRef Google scholar
[137]
Suganami T, Tanaka M, Ogawa Y. Adipose tissue inflammation and ectopic lipid accumulation. Endocr J 2012;59:849–57.
CrossRef Google scholar
[138]
Buechler C, Krautbauer S, Eisinger K. Adipose tissue fibrosis. World J Diabetes 2015;6:548–53.
CrossRef Google scholar
[139]
Sun K, Tordjman J, Clement K et al. Fibrosis and adipose tissue dysfunction. Cell Metab 2013;18:470–7.
CrossRef Google scholar
[140]
Kos K, Wilding JP. SPARC: a key player in the pathologies associated with obesity and diabetes. Nat Rev Endocrinol 2010;6:225–35.
CrossRef Google scholar
[141]
Li Y, Tong X, Rumala C et al. Thrombospondin1 deficiency reduces obesity-associated inflammation and improves insulin sensitivity in a diet-induced obese mouse model. PLoS One 2011;6:e26656.
CrossRef Google scholar
[142]
Lee SH, Park HS, Lee JA et al. Fibronectin gene expression in human adipose tissue and its associations with obesity-related genes and metabolic parameters. Obes Surg 2013;23:554–60.
CrossRef Google scholar
[143]
Gayral S, Garnotel R, Castaing-Berthou A et al. Elastin-derived peptides potentiate atherosclerosis through the immune Neu1-PI3Kgamma pathway. Cardiovasc Res 2014;102:118–27.
CrossRef Google scholar
[144]
Li X, Zhao Y, Chen C et al. Critical role of matrix metalloproteinase 14 in adipose tissue remodeling during obesity. Mol Cell Biol 2020;40:e00564-19.
CrossRef Google scholar
[145]
Sun K, Park J, Gupta OT et al. Endotrophin triggers adipose tissue fibrosis and metabolic dysfunction. Nat Commun 2014;5:3485.
CrossRef Google scholar
[146]
Lemoinne S, Cadoret A, El Mourabit H et al. Origins and functions of liver myofibroblasts. Biochim Biophys Acta 2013;1832:948–54.
CrossRef Google scholar
[147]
Chiang DJ, Pritchard MT, Nagy LE. Obesity, diabetes mellitus, and liver fibrosis. Am J Physiol Gastrointest Liver Physiol 2011;300:G697–702.
CrossRef Google scholar
[148]
Hoffmann C, Djerir NEH, Danckaert A et al. Hepatic stellate cell hypertrophy is associated with metabolic liver fibrosis. Sci Rep 2020;10:3850.
CrossRef Google scholar
[149]
Lin HY, Wang FS, Yang YL et al. MicroRNA-29a suppresses CD36 to ameliorate high fat diet-induced steatohepatitis and liver fibrosis in mice. Cells 2019;8:1298.
CrossRef Google scholar
[150]
Kundu A, Dey P, Park JH et al. EX-527 Prevents the progression of high-fat diet-induced hepatic steatosis and fibrosis by upregulating SIRT4 in Zucker rat. Cells 2020;9:1101.
CrossRef Google scholar
[151]
Jaskiewicz K, Rzepko R, Sledzinski Z. Fibrogenesis in fatty liver associated with obesity and diabetes mellitus type 2. Dig Dis Sci 2008;53:785–8.
CrossRef Google scholar
[152]
Ivaska J, Reunanen H, Westermarck J et al. Integrin α2β1 mediates isoform-specific activation of p38 and upregulation of collagen gene transcription by a mechanism involving the α2 cytoplasmic tail. J Cell Biol 1999;147:401–16.
CrossRef Google scholar
[153]
Pozzi A, Moberg PE, Miles LA et al. Elevated matrix metalloprotease and angiostatin levels in integrin α1 knockout mice cause reduced tumor vascularization. Proc Natl Acad Sci USA 2000;97:2202–7.
CrossRef Google scholar
[154]
Chen X, Moeckel G, Morrow JD et al. Lack of integrin α1β1 leads to severe glomerulosclerosis after glomerular injury. Am J Pathol 2004;165:617–30.
CrossRef Google scholar
[155]
Zhang Z, Ramirez NE, Yankeelov TE et al. Α2Β1 integrin expression in the tumor microenvironment enhances tumor angiogenesis in a tumor cell-specific manner. Blood 2008;111:1980–8.
CrossRef Google scholar
[156]
Kalluri R. Basement membranes: structure, assembly and role in tumour angiogenesis. Nat Rev Cancer 2003;3:422–33.
CrossRef Google scholar
[157]
Daoud J, Petropavlovskaia M, Rosenberg L et al. The effect of extracellular matrix components on the preservation of human islet function in vitro. Biomaterials 2010;31:1676–82.
CrossRef Google scholar
[158]
Bogdani M, Korpos E, Simeonovic CJ et al. Extracellular matrix components in the pathogenesis of type 1 diabetes. Curr Diab Rep 2014;14:552.
CrossRef Google scholar
[159]
Ferdek PE, Krzysztofik D, Stopa KB et al. When healing turns into killing - the pathophysiology of pancreatic and hepatic fibrosis. J Physiol 2022;600:2579–612.
CrossRef Google scholar
[160]
Zhang W, Zhang S, Zhang W et al. Matrix stiffness and its influence on pancreatic diseases. Biochim Biophys Acta Rev Cancer 2021;1876:188583.
CrossRef Google scholar
[161]
Huang C, Iovanna J, Santofimia-Castano P. Targeting fibrosis: the bridge that connects pancreatitis and pancreatic cancer. Int J Mol Sci 2021;22:4970.
CrossRef Google scholar
[162]
Böni-Schnetzler M, Meier DT. Islet inflammation in type 2 diabetes. Semin Immunopathol 2019;41:501–13.
CrossRef Google scholar
[163]
Hayden MR, Karuparthi PR, Habibi J et al. Ultrastructure of islet microcirculation, pericytes and the islet exocrine interface in the HIP rat model of diabetes. Exp Biol Med 2008;233:1109–23.
CrossRef Google scholar
[164]
Zhou YP, Madjidi A, Wilson ME et al. Matrix metalloproteinases contribute to insulin insufficiency in zucker diabetic fatty rats. Diabetes 2005;54:2612–9.
CrossRef Google scholar
[165]
Mateus Gonçalves L, Pereira E, Werneck de Castro JP et al. Islet pericytes convert into profibrotic myofibroblasts in a mouse model of islet vascular fibrosis. Diabetologia 2020;63:1564–75.
CrossRef Google scholar
[166]
Hull RL, Bogdani M, Nagy N et al. Hyaluronan: a mediator of islet dysfunction and destruction in diabetes? J Histochem Cytochem 2015;63:592–603.
CrossRef Google scholar
[167]
Eguchi K, Nagai R. Islet inflammation in type 2 diabetes and physiology. J Clin Invest 2017;127:14–23.
CrossRef Google scholar
[168]
Salah MM, Ashour AA, Abdelghany TM et al. Pirfenidone alleviates concanavalin A-induced liver fibrosis in mice. Life Sci 2019;239:116982.
CrossRef Google scholar
[169]
Seniutkin O, Furuya S, Luo YS et al. Effects of pirfenidone in acute and sub-chronic liver fibrosis, and an initiation-promotion cancer model in the mouse. Toxicol Appl Pharmacol 2018;339:1–9.
CrossRef Google scholar
[170]
García L, Hernández I, Sandoval A et al. Pirfenidone effectively reverses experimental liver fibrosis. J Hepatol 2002;37:797–805.
CrossRef Google scholar
[171]
Peng Y, Li L, Zhang X et al. Fluorofenidone affects hepatic stellate cell activation in hepatic fibrosis by targeting the TGF-β1/Smad and MAPK signaling pathways. Exp Ther Med 2019;18:41–48.
CrossRef Google scholar
[172]
Miric G, Dallemagne C, Endre Z et al. Reversal of cardiac and renal fibrosis by pirfenidone and spironolactone in streptozotocin- diabetic rats. Br J Pharmacol 2001;133:687–94.
CrossRef Google scholar
[173]
Nguyen DT, Ding C, Wilson E et al. Pirfenidone mitigates left ventricular fibrosis and dysfunction after myocardial infarction and reduces arrhythmias. Heart Rhythm 2010;7:1438–45.
CrossRef Google scholar
[174]
Mirkovic S, Seymour AML, Fenning A et al. Attenuation of cardiac fibrosis by pirfenidone and amiloride in DOCA-salt hypertensive rats. Br J Pharmacol 2002;135:961–8.
CrossRef Google scholar
[175]
Yamagami K, Oka T, Wang Q et al. Pirfenidone exhibits cardioprotective effects by regulating myocardial fibrosis and vascular permeability in pressure-overloaded hearts. Am J Physiol Heart Circ Physiol 2015;309:H512–22.
CrossRef Google scholar
[176]
Corona BT, Rivera JC, Dalske KA et al. Pharmacological mitigation of fibrosis in a porcine model of volumetric muscle loss injury. Tissue Eng Part A 2020;26:636–46.
CrossRef Google scholar
[177]
Provenzano PP, Cuevas C, Chang AE et al. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 2012;21:418–29.
CrossRef Google scholar
[178]
Flores-Contreras L, Sandoval-Rodríguez AS, Mena-Enriquez MG et al. Treatment with pirfenidone for two years decreases fibrosis, cytokine levels and enhances CB2 gene expression in patients with chronic hepatitis C. BMC Gastroenterol 2014;14:131.
CrossRef Google scholar
[179]
Poo JL, Torre A, Aguilar-Ramírez JR et al. Benefits of prolonged-release pirfenidone plus standard of care treatment in patients with advanced liver fibrosis: PROMETEO study. Hepatol Int 2020;14:817–27.
CrossRef Google scholar
[180]
Lewis GA, Dodd S, Clayton D et al. Pirfenidone in heart failure with preserved ejection fraction: a randomized phase 2 trial. Nat Med 2021;27:1477–82.
CrossRef Google scholar
[181]
van den Hoek AM, de Jong J, Worms N et al. Diet and exercise reduce pre-existing NASH and fibrosis and have additional beneficial effects on the vasculature, adipose tissue and skeletal muscle via organ-crosstalk. Metabolism 2021;124:154873.
CrossRef Google scholar
[182]
Wang W, Ishibashi J, Trefely S et al. A PRDM16-driven metabolic signal from adipocytes regulates precursor cell fate. Cell Metab 2019;30:174–89.e5.
CrossRef Google scholar
[183]
Oppedisano F, Mollace R, Tavernese A et al. PUFA supplementation and heart failure: effects on fibrosis and cardiac remodeling. Nutrients 2021;13:2965.
CrossRef Google scholar
[184]
Tacke F, Weiskirchen R. Non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH)-related liver fibrosis: mechanisms, treatment and prevention. Ann Transl Med 2021;9:729.
CrossRef Google scholar
[185]
Hannigan GE, Leung-Hagesteijn C, Fitz-Gibbon L et al. Regulation of cell adhesion and anchorage-dependent growth by a new beta 1-integrin-linked protein kinase. Nature 1996;379:91–6.
CrossRef Google scholar
[186]
Legate KR, Fässler R. Mechanisms that regulate adaptor binding to β-integrin cytoplasmic tails. J Cell Sci 2009;122:187–98.
CrossRef Google scholar
[187]
Wickström SA, Lange A, Montanez E et al. The ILK/PINCH/parvin complex: the kinase is dead, long live the pseudokinase! EMBO J 2010;29:281–91.
CrossRef Google scholar
[188]
Persad S, Attwell S, Gray V et al. Regulation of protein kinase B/Akt-serine 473 phosphorylation by integrin-linked kinase: critical roles for kinase activity and amino acids arginine 211 and serine 343. J Biol Chem 2001;276:27462–9.
CrossRef Google scholar
[189]
Hill MM, Feng J, Hemmings BA. Identification of a plasma membrane raft-associated PKB Ser473 kinase activity that is distinct from ILK and PDK1. Curr Biol 2002;12:1251–5.
CrossRef Google scholar
[190]
Idzerda RL, Carter WG, Nottenburg C et al. Isolation and DNA sequence of a cDNA clone encoding a lymphocyte adhesion receptor for high endothelium. Proc Natl Acad Sci USA 1989;86:4659–63.
CrossRef Google scholar
[191]
Chen C, Zhao S, Karnad A et al. The biology and role of CD44 in cancer progression: Therapeutic implications. J Hematol Oncol 2018;11:64.
CrossRef Google scholar
[192]
Faassen AE, Schrager JA, Klein DJ et al. A cell surface chondroitin sulfate proteoglycan, immunologically related to CD44, is involved in type I collagen-mediated melanoma cell motility and invasion. J Cell Biol 1992;116:521–31.
CrossRef Google scholar
[193]
Weber GF, Ashkar S, Glimcher MJ et al. Receptor-ligand interaction between CD44 and osteopontin (Eta-1). Science 1996;271:509–12.
CrossRef Google scholar
[194]
Ponta H, Sherman L, Herrlich PA. CD44: From adhesion molecules to signalling regulators. Nat Rev Mol Cell Biol 2003;4:33–45.
CrossRef Google scholar
[195]
Zöller M. CD44: can a cancer-initiating cell profit from an abundantly expressed molecule? Nat Rev Cancer 2011;11:254–67.
CrossRef Google scholar
[196]
Hollingsworth JW, Li Z, Brass DM et al. CD44 regulates macrophage recruitment to the lung in lipopolysaccharide-induced airway disease. Am J Respir Cell Mol Biol 2007;37:248–53.
CrossRef Google scholar
[197]
Weng X, Maxwell-Warburton S, Hasib A et al. The membrane receptor CD44: novel insights into metabolism. Trends Endocrinol Metab 2022;33:318–32.
CrossRef Google scholar

RIGHTS & PERMISSIONS

2023 The Author(s) 2023. Published by Oxford University Press on behalf of Higher Education Press.
AI Summary AI Mindmap
PDF(1263 KB)

Accesses

Citations

Detail

Sections
Recommended

/