Skip to main content
Top
Published in: Molecular and Cellular Pediatrics 1/2023

Open Access 14-11-2023 | Obesity | Review

Relevance and consequence of chronic inflammation for obesity development

Authors: Lisa Ruck, Susanna Wiegand, Peter Kühnen

Published in: Molecular and Cellular Pediatrics | Issue 1/2023

Login to get access

Abstract

Background

Increasing prevalence of morbid obesity accompanied by comorbidities like type 2 diabetes mellitus (T2DM) led to a demand for improving therapeutic strategies and pharmacological intervention options. Apart from genetics, inflammation processes have been hypothesized to be of importance for the development of obesity and related aspects like insulin resistance.

Main text

Within this review, we provide an overview of the intricate interplay between chronic inflammation of the adipose tissue and the hypothalamus and the development of obesity. Further understanding of this relationship might improve the understanding of the underlying mechanism and may be of relevance for the establishment of new treatment strategies.
Literature
1.
go back to reference Seki H, Tani Y, Arita M (2009) Omega-3 PUFA derived anti-inflammatory lipid mediator resolvin E1. Prostaglandins Other Lipid Mediat 89(3–4):126–130PubMedCrossRef Seki H, Tani Y, Arita M (2009) Omega-3 PUFA derived anti-inflammatory lipid mediator resolvin E1. Prostaglandins Other Lipid Mediat 89(3–4):126–130PubMedCrossRef
2.
go back to reference Lafontan M (2005) Fat cells: afferent and efferent messages define new approaches to treat obesity. Annu Rev Pharmacol Toxicol 45:119–146PubMedCrossRef Lafontan M (2005) Fat cells: afferent and efferent messages define new approaches to treat obesity. Annu Rev Pharmacol Toxicol 45:119–146PubMedCrossRef
3.
go back to reference Winer S, Chan Y, Paltser G, Truong D, Tsui H, Bahrami J et al (2009) Normalization of obesity-associated insulin resistance through immunotherapy. Nat Med 15(8):921–929PubMedPubMedCentralCrossRef Winer S, Chan Y, Paltser G, Truong D, Tsui H, Bahrami J et al (2009) Normalization of obesity-associated insulin resistance through immunotherapy. Nat Med 15(8):921–929PubMedPubMedCentralCrossRef
4.
5.
go back to reference Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW Jr (2003) Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest 112(12):1796–1808PubMedPubMedCentralCrossRef Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW Jr (2003) Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest 112(12):1796–1808PubMedPubMedCentralCrossRef
6.
go back to reference Winer DA, Winer S, Shen L, Wadia PP, Yantha J, Paltser G et al (2011) B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med 17(5):610–617PubMedPubMedCentralCrossRef Winer DA, Winer S, Shen L, Wadia PP, Yantha J, Paltser G et al (2011) B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med 17(5):610–617PubMedPubMedCentralCrossRef
7.
go back to reference Yang H, Youm YH, Vandanmagsar B, Ravussin A, Gimble JM, Greenway F et al (2010) Obesity increases the production of proinflammatory mediators from adipose tissue T cells and compromises TCR repertoire diversity: implications for systemic inflammation and insulin resistance. J Immunol 185(3):1836–1845PubMedCrossRef Yang H, Youm YH, Vandanmagsar B, Ravussin A, Gimble JM, Greenway F et al (2010) Obesity increases the production of proinflammatory mediators from adipose tissue T cells and compromises TCR repertoire diversity: implications for systemic inflammation and insulin resistance. J Immunol 185(3):1836–1845PubMedCrossRef
8.
go back to reference Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M et al (2009) CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med 15(8):914–920PubMedCrossRef Nishimura S, Manabe I, Nagasaki M, Eto K, Yamashita H, Ohsugi M et al (2009) CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat Med 15(8):914–920PubMedCrossRef
10.
go back to reference Yang H, Lang S, Zhai Z, Li L, Kahr WH, Chen P et al (2009) Fibrinogen is required for maintenance of platelet intracellular and cell-surface P-selectin expression. Blood 114(2):425–436PubMedCrossRef Yang H, Lang S, Zhai Z, Li L, Kahr WH, Chen P et al (2009) Fibrinogen is required for maintenance of platelet intracellular and cell-surface P-selectin expression. Blood 114(2):425–436PubMedCrossRef
11.
go back to reference Smith AG, Sheridan PA, Harp JB, Beck MA (2007) Diet-induced obese mice have increased mortality and altered immune responses when infected with influenza virus. J Nutr 137(5):1236–1243PubMedCrossRef Smith AG, Sheridan PA, Harp JB, Beck MA (2007) Diet-induced obese mice have increased mortality and altered immune responses when infected with influenza virus. J Nutr 137(5):1236–1243PubMedCrossRef
12.
go back to reference Karlsson EA, Sheridan PA, Beck MA (2010) Diet-induced obesity impairs the T cell memory response to influenza virus infection. J Immunol 184(6):3127–3133PubMedCrossRef Karlsson EA, Sheridan PA, Beck MA (2010) Diet-induced obesity impairs the T cell memory response to influenza virus infection. J Immunol 184(6):3127–3133PubMedCrossRef
13.
go back to reference Stienstra R, Tack CJ, Kanneganti TD, Joosten LA, Netea MG (2012) The inflammasome puts obesity in the danger zone. Cell Metab 15(1):10–18PubMedCrossRef Stienstra R, Tack CJ, Kanneganti TD, Joosten LA, Netea MG (2012) The inflammasome puts obesity in the danger zone. Cell Metab 15(1):10–18PubMedCrossRef
15.
go back to reference Farooqi IS, O’Rahilly S (2008) Mutations in ligands and receptors of the leptin-melanocortin pathway that lead to obesity. Nat Clin Pract Endocrinol Metab 4(10):569–577PubMedCrossRef Farooqi IS, O’Rahilly S (2008) Mutations in ligands and receptors of the leptin-melanocortin pathway that lead to obesity. Nat Clin Pract Endocrinol Metab 4(10):569–577PubMedCrossRef
16.
go back to reference Montague CT, Farooqi IS, Whitehead JP, Soos MA, Rau H, Wareham NJ et al (1997) Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 387(6636):903–908PubMedCrossRef Montague CT, Farooqi IS, Whitehead JP, Soos MA, Rau H, Wareham NJ et al (1997) Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 387(6636):903–908PubMedCrossRef
17.
go back to reference Mattioli B, Straface E, Quaranta MG, Giordani L, Viora M (2005) Leptin promotes differentiation and survival of human dendritic cells and licenses them for Th1 priming. J Immunol 174(11):6820–6828PubMedCrossRef Mattioli B, Straface E, Quaranta MG, Giordani L, Viora M (2005) Leptin promotes differentiation and survival of human dendritic cells and licenses them for Th1 priming. J Immunol 174(11):6820–6828PubMedCrossRef
18.
go back to reference Lam QL, Liu S, Cao X, Lu L (2006) Involvement of leptin signaling in the survival and maturation of bone marrow-derived dendritic cells. Eur J Immunol 36(12):3118–3130PubMedCrossRef Lam QL, Liu S, Cao X, Lu L (2006) Involvement of leptin signaling in the survival and maturation of bone marrow-derived dendritic cells. Eur J Immunol 36(12):3118–3130PubMedCrossRef
19.
go back to reference Caldefie-Chezet F, Poulin A, Vasson MP (2003) Leptin regulates functional capacities of polymorphonuclear neutrophils. Free Radic Res 37(8):809–814PubMedCrossRef Caldefie-Chezet F, Poulin A, Vasson MP (2003) Leptin regulates functional capacities of polymorphonuclear neutrophils. Free Radic Res 37(8):809–814PubMedCrossRef
20.
go back to reference Zhao Y, Sun R, You L, Gao C, Tian Z (2003) Expression of leptin receptors and response to leptin stimulation of human natural killer cell lines. Biochem Biophys Res Commun 300(2):247–252PubMedCrossRef Zhao Y, Sun R, You L, Gao C, Tian Z (2003) Expression of leptin receptors and response to leptin stimulation of human natural killer cell lines. Biochem Biophys Res Commun 300(2):247–252PubMedCrossRef
21.
go back to reference Tian Z, Sun R, Wei H, Gao B (2002) Impaired natural killer (NK) cell activity in leptin receptor deficient mice: leptin as a critical regulator in NK cell development and activation. Biochem Biophys Res Commun 298(3):297–302PubMedCrossRef Tian Z, Sun R, Wei H, Gao B (2002) Impaired natural killer (NK) cell activity in leptin receptor deficient mice: leptin as a critical regulator in NK cell development and activation. Biochem Biophys Res Commun 298(3):297–302PubMedCrossRef
22.
go back to reference Otero M, Lago R, Lago F, Casanueva FF, Dieguez C, Gomez-Reino JJ et al (2005) Leptin, from fat to inflammation: old questions and new insights. FEBS Lett 579(2):295–301PubMedCrossRef Otero M, Lago R, Lago F, Casanueva FF, Dieguez C, Gomez-Reino JJ et al (2005) Leptin, from fat to inflammation: old questions and new insights. FEBS Lett 579(2):295–301PubMedCrossRef
23.
go back to reference Sarraf P, Frederich RC, Turner EM, Ma G, Jaskowiak NT, Rivet DJ 3rd et al (1997) Multiple cytokines and acute inflammation raise mouse leptin levels: potential role in inflammatory anorexia. J Exp Med 185(1):171–175PubMedPubMedCentralCrossRef Sarraf P, Frederich RC, Turner EM, Ma G, Jaskowiak NT, Rivet DJ 3rd et al (1997) Multiple cytokines and acute inflammation raise mouse leptin levels: potential role in inflammatory anorexia. J Exp Med 185(1):171–175PubMedPubMedCentralCrossRef
24.
go back to reference Fraser DA, Thoen J, Reseland JE, Forre O, Kjeldsen-Kragh J (1999) Decreased CD4+ lymphocyte activation and increased interleukin-4 production in peripheral blood of rheumatoid arthritis patients after acute starvation. Clin Rheumatol 18(5):394–401PubMedCrossRef Fraser DA, Thoen J, Reseland JE, Forre O, Kjeldsen-Kragh J (1999) Decreased CD4+ lymphocyte activation and increased interleukin-4 production in peripheral blood of rheumatoid arthritis patients after acute starvation. Clin Rheumatol 18(5):394–401PubMedCrossRef
25.
go back to reference Lord GM, Matarese G, Howard JK, Baker RJ, Bloom SR, Lechler RI (1998) Leptin modulates the T-cell immune response and reverses starvation-induced immunosuppression. Nature 394(6696):897–901PubMedCrossRef Lord GM, Matarese G, Howard JK, Baker RJ, Bloom SR, Lechler RI (1998) Leptin modulates the T-cell immune response and reverses starvation-induced immunosuppression. Nature 394(6696):897–901PubMedCrossRef
26.
go back to reference Lam QL, Lu L (2007) Role of leptin in immunity. Cell Mol Immunol 4(1):1–13PubMed Lam QL, Lu L (2007) Role of leptin in immunity. Cell Mol Immunol 4(1):1–13PubMed
27.
go back to reference Farooqi IS, Matarese G, Lord GM, Keogh JM, Lawrence E, Agwu C et al (2002) Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency. J Clin Invest 110(8):1093–1103PubMedPubMedCentralCrossRef Farooqi IS, Matarese G, Lord GM, Keogh JM, Lawrence E, Agwu C et al (2002) Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency. J Clin Invest 110(8):1093–1103PubMedPubMedCentralCrossRef
28.
go back to reference Baumann H, Morella KK, White DW, Dembski M, Bailon PS, Kim H et al (1996) The full-length leptin receptor has signaling capabilities of interleukin 6-type cytokine receptors. Proc Natl Acad Sci U S A 93(16):8374–8378PubMedPubMedCentralCrossRef Baumann H, Morella KK, White DW, Dembski M, Bailon PS, Kim H et al (1996) The full-length leptin receptor has signaling capabilities of interleukin 6-type cytokine receptors. Proc Natl Acad Sci U S A 93(16):8374–8378PubMedPubMedCentralCrossRef
29.
go back to reference Tartaglia LA, Dembski M, Weng X, Deng N, Culpepper J, Devos R et al (1995) Identification and expression cloning of a leptin receptor. OB-R Cell 83(7):1263–1271PubMedCrossRef Tartaglia LA, Dembski M, Weng X, Deng N, Culpepper J, Devos R et al (1995) Identification and expression cloning of a leptin receptor. OB-R Cell 83(7):1263–1271PubMedCrossRef
30.
go back to reference Guan XM, Hess JF, Yu H, Hey PJ, van der Ploeg LH (1997) Differential expression of mRNA for leptin receptor isoforms in the rat brain. Mol Cell Endocrinol 133(1):1–7PubMedCrossRef Guan XM, Hess JF, Yu H, Hey PJ, van der Ploeg LH (1997) Differential expression of mRNA for leptin receptor isoforms in the rat brain. Mol Cell Endocrinol 133(1):1–7PubMedCrossRef
31.
go back to reference Takaya K, Ogawa Y, Isse N, Okazaki T, Satoh N, Masuzaki H et al (1996) Molecular cloning of rat leptin receptor isoform complementary DNAs–identification of a missense mutation in Zucker fatty (fa/fa) rats. Biochem Biophys Res Commun 225(1):75–83PubMedCrossRef Takaya K, Ogawa Y, Isse N, Okazaki T, Satoh N, Masuzaki H et al (1996) Molecular cloning of rat leptin receptor isoform complementary DNAs–identification of a missense mutation in Zucker fatty (fa/fa) rats. Biochem Biophys Res Commun 225(1):75–83PubMedCrossRef
32.
go back to reference Bjorbaek C, Uotani S, da Silva B, Flier JS (1997) Divergent signaling capacities of the long and short isoforms of the leptin receptor. J Biol Chem 272(51):32686–32695PubMedCrossRef Bjorbaek C, Uotani S, da Silva B, Flier JS (1997) Divergent signaling capacities of the long and short isoforms of the leptin receptor. J Biol Chem 272(51):32686–32695PubMedCrossRef
33.
go back to reference Howard JK, Lord GM, Matarese G, Vendetti S, Ghatei MA, Ritter MA et al (1999) Leptin protects mice from starvation-induced lymphoid atrophy and increases thymic cellularity in ob/ob mice. J Clin Invest 104(8):1051–1059PubMedPubMedCentralCrossRef Howard JK, Lord GM, Matarese G, Vendetti S, Ghatei MA, Ritter MA et al (1999) Leptin protects mice from starvation-induced lymphoid atrophy and increases thymic cellularity in ob/ob mice. J Clin Invest 104(8):1051–1059PubMedPubMedCentralCrossRef
34.
go back to reference Chandra RK (1980) Cell-mediated immunity in genetically obese C57BL/6J ob/ob) mice. Am J Clin Nutr 33(1):13–16PubMedCrossRef Chandra RK (1980) Cell-mediated immunity in genetically obese C57BL/6J ob/ob) mice. Am J Clin Nutr 33(1):13–16PubMedCrossRef
35.
go back to reference Mandel MA, Mahmoud AA (1978) Impairment of cell-mediated immunity in mutation diabetic mice (db/db). J Immunol 120(4):1375–1377PubMedCrossRef Mandel MA, Mahmoud AA (1978) Impairment of cell-mediated immunity in mutation diabetic mice (db/db). J Immunol 120(4):1375–1377PubMedCrossRef
36.
go back to reference Webb SR, Loria RM, Madge GE, Kibrick S (1976) Susceptibility of mice to group B coxsackie virus is influenced by the diabetic gene. J Exp Med 143(5):1239–1248PubMedCrossRef Webb SR, Loria RM, Madge GE, Kibrick S (1976) Susceptibility of mice to group B coxsackie virus is influenced by the diabetic gene. J Exp Med 143(5):1239–1248PubMedCrossRef
37.
go back to reference Faggioni R, Fantuzzi G, Gabay C, Moser A, Dinarello CA, Feingold KR et al (1999) Leptin deficiency enhances sensitivity to endotoxin-induced lethality. Am J Physiol 276(1):R136–R142PubMed Faggioni R, Fantuzzi G, Gabay C, Moser A, Dinarello CA, Feingold KR et al (1999) Leptin deficiency enhances sensitivity to endotoxin-induced lethality. Am J Physiol 276(1):R136–R142PubMed
39.
go back to reference Hick RW, Gruver AL, Ventevogel MS, Haynes BF, Sempowski GD (2006) Leptin selectively augments thymopoiesis in leptin deficiency and lipopolysaccharide-induced thymic atrophy. J Immunol 177(1):169–176PubMedCrossRef Hick RW, Gruver AL, Ventevogel MS, Haynes BF, Sempowski GD (2006) Leptin selectively augments thymopoiesis in leptin deficiency and lipopolysaccharide-induced thymic atrophy. J Immunol 177(1):169–176PubMedCrossRef
40.
go back to reference Siegmund B, Lear-Kaul KC, Faggioni R, Fantuzzi G (2002) Leptin deficiency, not obesity, protects mice from Con A-induced hepatitis. Eur J Immunol 32(2):552–560PubMedCrossRef Siegmund B, Lear-Kaul KC, Faggioni R, Fantuzzi G (2002) Leptin deficiency, not obesity, protects mice from Con A-induced hepatitis. Eur J Immunol 32(2):552–560PubMedCrossRef
41.
go back to reference Tarzi RM, Cook HT, Jackson I, Pusey CD, Lord GM (2004) Leptin-deficient mice are protected from accelerated nephrotoxic nephritis. Am J Pathol 164(2):385–390PubMedPubMedCentralCrossRef Tarzi RM, Cook HT, Jackson I, Pusey CD, Lord GM (2004) Leptin-deficient mice are protected from accelerated nephrotoxic nephritis. Am J Pathol 164(2):385–390PubMedPubMedCentralCrossRef
42.
go back to reference Bennett BD, Solar GP, Yuan JQ, Mathias J, Thomas GR, Matthews W (1996) A role for leptin and its cognate receptor in hematopoiesis. Curr Biol 6(9):1170–1180PubMedCrossRef Bennett BD, Solar GP, Yuan JQ, Mathias J, Thomas GR, Matthews W (1996) A role for leptin and its cognate receptor in hematopoiesis. Curr Biol 6(9):1170–1180PubMedCrossRef
43.
go back to reference Farooqi IS, Wangensteen T, Collins S, Kimber W, Matarese G, Keogh JM et al (2007) Clinical and molecular genetic spectrum of congenital deficiency of the leptin receptor. N Engl J Med 356(3):237–247PubMedPubMedCentralCrossRef Farooqi IS, Wangensteen T, Collins S, Kimber W, Matarese G, Keogh JM et al (2007) Clinical and molecular genetic spectrum of congenital deficiency of the leptin receptor. N Engl J Med 356(3):237–247PubMedPubMedCentralCrossRef
45.
go back to reference Popa C, Netea MG, Radstake TR, van Riel PL, Barrera P, van der Meer JW (2005) Markers of inflammation are negatively correlated with serum leptin in rheumatoid arthritis. Ann Rheum Dis 64(8):1195–1198PubMedPubMedCentralCrossRef Popa C, Netea MG, Radstake TR, van Riel PL, Barrera P, van der Meer JW (2005) Markers of inflammation are negatively correlated with serum leptin in rheumatoid arthritis. Ann Rheum Dis 64(8):1195–1198PubMedPubMedCentralCrossRef
46.
go back to reference Rajora N, Ceriani G, Catania A, Star RA, Murphy MT, Lipton JM (1996) alpha-MSH production, receptors, and influence on neopterin in a human monocyte/macrophage cell line. J Leukoc Biol 59(2):248–253PubMedCrossRef Rajora N, Ceriani G, Catania A, Star RA, Murphy MT, Lipton JM (1996) alpha-MSH production, receptors, and influence on neopterin in a human monocyte/macrophage cell line. J Leukoc Biol 59(2):248–253PubMedCrossRef
47.
go back to reference Brzoska T, Kalden DH, Scholzen T, Luger TA (1999) Molecular basis of the alpha-MSH/IL-1 antagonism. Ann N Y Acad Sci 885:230–238PubMedCrossRef Brzoska T, Kalden DH, Scholzen T, Luger TA (1999) Molecular basis of the alpha-MSH/IL-1 antagonism. Ann N Y Acad Sci 885:230–238PubMedCrossRef
48.
go back to reference Star RA, Rajora N, Huang J, Stock RC, Catania A, Lipton JM (1995) Evidence of autocrine modulation of macrophage nitric oxide synthase by alpha-melanocyte-stimulating hormone. Proc Natl Acad Sci U S A 92(17):8016–8020PubMedPubMedCentralCrossRef Star RA, Rajora N, Huang J, Stock RC, Catania A, Lipton JM (1995) Evidence of autocrine modulation of macrophage nitric oxide synthase by alpha-melanocyte-stimulating hormone. Proc Natl Acad Sci U S A 92(17):8016–8020PubMedPubMedCentralCrossRef
49.
go back to reference Raap U, Brzoska T, Sohl S, Path G, Emmel J, Herz U et al (2003) Alpha-melanocyte-stimulating hormone inhibits allergic airway inflammation. J Immunol 171(1):353–359PubMedCrossRef Raap U, Brzoska T, Sohl S, Path G, Emmel J, Herz U et al (2003) Alpha-melanocyte-stimulating hormone inhibits allergic airway inflammation. J Immunol 171(1):353–359PubMedCrossRef
50.
go back to reference Rajora N, Boccoli G, Catania A, Lipton JM (1997) alpha-MSH modulates experimental inflammatory bowel disease. Peptides 18(3):381–385PubMedCrossRef Rajora N, Boccoli G, Catania A, Lipton JM (1997) alpha-MSH modulates experimental inflammatory bowel disease. Peptides 18(3):381–385PubMedCrossRef
52.
go back to reference Robbins LS, Nadeau JH, Johnson KR, Kelly MA, Roselli-Rehfuss L, Baack E et al (1993) Pigmentation phenotypes of variant extension locus alleles result from point mutations that alter MSH receptor function. Cell 72(6):827–834PubMedCrossRef Robbins LS, Nadeau JH, Johnson KR, Kelly MA, Roselli-Rehfuss L, Baack E et al (1993) Pigmentation phenotypes of variant extension locus alleles result from point mutations that alter MSH receptor function. Cell 72(6):827–834PubMedCrossRef
53.
go back to reference Bohm M, Luger TA, Tobin DJ, Garcia-Borron JC (2006) Melanocortin receptor ligands: new horizons for skin biology and clinical dermatology. J Invest Dermatol 126(9):1966–1975PubMedCrossRef Bohm M, Luger TA, Tobin DJ, Garcia-Borron JC (2006) Melanocortin receptor ligands: new horizons for skin biology and clinical dermatology. J Invest Dermatol 126(9):1966–1975PubMedCrossRef
54.
go back to reference Brzoska T, Luger TA, Maaser C, Abels C, Bohm M (2008) Alpha-melanocyte-stimulating hormone and related tripeptides: biochemistry, antiinflammatory and protective effects in vitro and in vivo, and future perspectives for the treatment of immune-mediated inflammatory diseases. Endocr Rev 29(5):581–602PubMedCrossRef Brzoska T, Luger TA, Maaser C, Abels C, Bohm M (2008) Alpha-melanocyte-stimulating hormone and related tripeptides: biochemistry, antiinflammatory and protective effects in vitro and in vivo, and future perspectives for the treatment of immune-mediated inflammatory diseases. Endocr Rev 29(5):581–602PubMedCrossRef
55.
go back to reference Catania A (2008) Neuroprotective actions of melanocortins: a therapeutic opportunity. Trends Neurosci 31(7):353–360PubMedCrossRef Catania A (2008) Neuroprotective actions of melanocortins: a therapeutic opportunity. Trends Neurosci 31(7):353–360PubMedCrossRef
56.
go back to reference Maaser C, Kannengiesser K, Specht C, Lugering A, Brzoska T, Luger TA et al (2006) Crucial role of the melanocortin receptor MC1R in experimental colitis. Gut 55(10):1415–1422PubMedPubMedCentralCrossRef Maaser C, Kannengiesser K, Specht C, Lugering A, Brzoska T, Luger TA et al (2006) Crucial role of the melanocortin receptor MC1R in experimental colitis. Gut 55(10):1415–1422PubMedPubMedCentralCrossRef
57.
go back to reference Carter D, Warsen A, Mandell K, Cuschieri J, Maier RV, Arbabi S (2014) Delayed topical p38 MAPK inhibition attenuates full-thickness burn wound inflammatory signaling. J Burn Care Res 35(2):e83-92PubMedCrossRef Carter D, Warsen A, Mandell K, Cuschieri J, Maier RV, Arbabi S (2014) Delayed topical p38 MAPK inhibition attenuates full-thickness burn wound inflammatory signaling. J Burn Care Res 35(2):e83-92PubMedCrossRef
58.
go back to reference Carter DW, Sood RF, Seaton ME, Muffley LA, Honari S, Hocking AM et al (2018) MC1R gene polymorphisms are associated with dysfunctional immune responses and wound infection after burn injury. J Surg Res 231:448–452PubMedPubMedCentralCrossRef Carter DW, Sood RF, Seaton ME, Muffley LA, Honari S, Hocking AM et al (2018) MC1R gene polymorphisms are associated with dysfunctional immune responses and wound infection after burn injury. J Surg Res 231:448–452PubMedPubMedCentralCrossRef
59.
go back to reference Sood RF, Hocking AM, Muffley LA, Ga M, Honari S, Reiner AP et al (2015) Race and melanocortin 1 receptor polymorphism R163Q are associated with post-burn hypertrophic scarring: a prospective cohort study. J Invest Dermatol 135(10):2394–2401PubMedPubMedCentralCrossRef Sood RF, Hocking AM, Muffley LA, Ga M, Honari S, Reiner AP et al (2015) Race and melanocortin 1 receptor polymorphism R163Q are associated with post-burn hypertrophic scarring: a prospective cohort study. J Invest Dermatol 135(10):2394–2401PubMedPubMedCentralCrossRef
60.
go back to reference Spana C, Taylor AW, Yee DG, Makhlina M, Yang W, Dodd J (2018) Probing the role of melanocortin type 1 receptor agonists in diverse immunological diseases. Front Pharmacol 9:1535PubMedCrossRef Spana C, Taylor AW, Yee DG, Makhlina M, Yang W, Dodd J (2018) Probing the role of melanocortin type 1 receptor agonists in diverse immunological diseases. Front Pharmacol 9:1535PubMedCrossRef
61.
go back to reference Dodd J, Jordan R, Makhlina M, Barnett K, Roffel A, Spana C et al (2023) A novel oral formulation of the melanocortin-1 receptor agonist PL8177 resolves inflammation in preclinical studies of inflammatory bowel disease and is gut restricted in rats, dogs, and humans. Front Immunol 14:1083333PubMedPubMedCentralCrossRef Dodd J, Jordan R, Makhlina M, Barnett K, Roffel A, Spana C et al (2023) A novel oral formulation of the melanocortin-1 receptor agonist PL8177 resolves inflammation in preclinical studies of inflammatory bowel disease and is gut restricted in rats, dogs, and humans. Front Immunol 14:1083333PubMedPubMedCentralCrossRef
62.
go back to reference Kanti V, Puder L, Jahnke I, Krabusch PM, Kottner J, Vogt A et al (2021) A melanocortin-4 receptor agonist induces skin and hair pigmentation in patients with monogenic mutations in the leptin-melanocortin pathway. Skin Pharmacol Physiol 34(6):307–316PubMedCrossRef Kanti V, Puder L, Jahnke I, Krabusch PM, Kottner J, Vogt A et al (2021) A melanocortin-4 receptor agonist induces skin and hair pigmentation in patients with monogenic mutations in the leptin-melanocortin pathway. Skin Pharmacol Physiol 34(6):307–316PubMedCrossRef
63.
go back to reference Clement K, Biebermann H, Farooqi IS, Van der Ploeg L, Wolters B, Poitou C et al (2018) MC4R agonism promotes durable weight loss in patients with leptin receptor deficiency. Nat Med 24(5):551–555PubMedCrossRef Clement K, Biebermann H, Farooqi IS, Van der Ploeg L, Wolters B, Poitou C et al (2018) MC4R agonism promotes durable weight loss in patients with leptin receptor deficiency. Nat Med 24(5):551–555PubMedCrossRef
64.
go back to reference Clement K, van den Akker E, Argente J, Bahm A, Chung WK, Connors H et al (2020) Efficacy and safety of setmelanotide, an MC4R agonist, in individuals with severe obesity due to LEPR or POMC deficiency: single-arm, open-label, multicentre, phase 3 trials. Lancet Diabetes Endocrinol 8(12):960–970PubMedCrossRef Clement K, van den Akker E, Argente J, Bahm A, Chung WK, Connors H et al (2020) Efficacy and safety of setmelanotide, an MC4R agonist, in individuals with severe obesity due to LEPR or POMC deficiency: single-arm, open-label, multicentre, phase 3 trials. Lancet Diabetes Endocrinol 8(12):960–970PubMedCrossRef
65.
go back to reference Kamermans A, Verhoeven T, van Het Hof B, Koning JJ, Borghuis L, Witte M et al (2019) Setmelanotide, a novel, selective melanocortin receptor-4 agonist exerts anti-inflammatory actions in astrocytes and promotes an anti-inflammatory macrophage phenotype. Front Immunol 10:2312PubMedPubMedCentralCrossRef Kamermans A, Verhoeven T, van Het Hof B, Koning JJ, Borghuis L, Witte M et al (2019) Setmelanotide, a novel, selective melanocortin receptor-4 agonist exerts anti-inflammatory actions in astrocytes and promotes an anti-inflammatory macrophage phenotype. Front Immunol 10:2312PubMedPubMedCentralCrossRef
66.
go back to reference Gregor MF, Hotamisligil GS (2011) Inflammatory mechanisms in obesity. Annu Rev Immunol 29:415–445PubMedCrossRef Gregor MF, Hotamisligil GS (2011) Inflammatory mechanisms in obesity. Annu Rev Immunol 29:415–445PubMedCrossRef
67.
68.
go back to reference Ron D, Walter P (2007) Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol 8(7):519–529PubMedCrossRef Ron D, Walter P (2007) Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol 8(7):519–529PubMedCrossRef
69.
go back to reference Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, Harding HP et al (2000) Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 287(5453):664–666PubMedCrossRef Urano F, Wang X, Bertolotti A, Zhang Y, Chung P, Harding HP et al (2000) Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 287(5453):664–666PubMedCrossRef
70.
go back to reference Deng J, Lu PD, Zhang Y, Scheuner D, Kaufman RJ, Sonenberg N et al (2004) Translational repression mediates activation of nuclear factor kappa B by phosphorylated translation initiation factor 2. Mol Cell Biol 24(23):10161–10168PubMedPubMedCentralCrossRef Deng J, Lu PD, Zhang Y, Scheuner D, Kaufman RJ, Sonenberg N et al (2004) Translational repression mediates activation of nuclear factor kappa B by phosphorylated translation initiation factor 2. Mol Cell Biol 24(23):10161–10168PubMedPubMedCentralCrossRef
71.
go back to reference Gregor MF, Hotamisligil GS (2007) Thematic review series: adipocyte biology. Adipocyte stress: the endoplasmic reticulum and metabolic disease. J Lipid Res. 48(9):1905–14PubMedCrossRef Gregor MF, Hotamisligil GS (2007) Thematic review series: adipocyte biology. Adipocyte stress: the endoplasmic reticulum and metabolic disease. J Lipid Res. 48(9):1905–14PubMedCrossRef
72.
73.
go back to reference Yudkin JS (2003) Adipose tissue, insulin action and vascular disease: inflammatory signals. Int J Obes Relat Metab Disord 27(Suppl 3):S25–S28PubMedCrossRef Yudkin JS (2003) Adipose tissue, insulin action and vascular disease: inflammatory signals. Int J Obes Relat Metab Disord 27(Suppl 3):S25–S28PubMedCrossRef
74.
go back to reference Yudkin JS, Kumari M, Humphries SE, Mohamed-Ali V (2000) Inflammation, obesity, stress and coronary heart disease: is interleukin-6 the link? Atherosclerosis 148(2):209–214PubMedCrossRef Yudkin JS, Kumari M, Humphries SE, Mohamed-Ali V (2000) Inflammation, obesity, stress and coronary heart disease: is interleukin-6 the link? Atherosclerosis 148(2):209–214PubMedCrossRef
75.
go back to reference Hosogai N, Fukuhara A, Oshima K, Miyata Y, Tanaka S, Segawa K et al (2007) Adipose tissue hypoxia in obesity and its impact on adipocytokine dysregulation. Diabetes 56(4):901–911PubMedCrossRef Hosogai N, Fukuhara A, Oshima K, Miyata Y, Tanaka S, Segawa K et al (2007) Adipose tissue hypoxia in obesity and its impact on adipocytokine dysregulation. Diabetes 56(4):901–911PubMedCrossRef
76.
go back to reference Ye J, Gao Z, Yin J, He Q (2007) Hypoxia is a potential risk factor for chronic inflammation and adiponectin reduction in adipose tissue of ob/ob and dietary obese mice. Am J Physiol Endocrinol Metab 293(4):E1118–E1128PubMedCrossRef Ye J, Gao Z, Yin J, He Q (2007) Hypoxia is a potential risk factor for chronic inflammation and adiponectin reduction in adipose tissue of ob/ob and dietary obese mice. Am J Physiol Endocrinol Metab 293(4):E1118–E1128PubMedCrossRef
77.
go back to reference Wenger RH (2002) Cellular adaptation to hypoxia: O2-sensing protein hydroxylases, hypoxia-inducible transcription factors, and O2-regulated gene expression. FASEB J 16(10):1151–1162PubMedCrossRef Wenger RH (2002) Cellular adaptation to hypoxia: O2-sensing protein hydroxylases, hypoxia-inducible transcription factors, and O2-regulated gene expression. FASEB J 16(10):1151–1162PubMedCrossRef
78.
80.
go back to reference Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS (2006) TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest 116(11):3015–3025PubMedPubMedCentralCrossRef Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS (2006) TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest 116(11):3015–3025PubMedPubMedCentralCrossRef
81.
go back to reference Suganami T, Nishida J, Ogawa Y (2005) A paracrine loop between adipocytes and macrophages aggravates inflammatory changes: role of free fatty acids and tumor necrosis factor alpha. Arterioscler Thromb Vasc Biol 25(10):2062–2068PubMedCrossRef Suganami T, Nishida J, Ogawa Y (2005) A paracrine loop between adipocytes and macrophages aggravates inflammatory changes: role of free fatty acids and tumor necrosis factor alpha. Arterioscler Thromb Vasc Biol 25(10):2062–2068PubMedCrossRef
83.
go back to reference Harman-Boehm I, Bluher M, Redel H, Sion-Vardy N, Ovadia S, Avinoach E et al (2007) Macrophage infiltration into omental versus subcutaneous fat across different populations: effect of regional adiposity and the comorbidities of obesity. J Clin Endocrinol Metab 92(6):2240–2247PubMedCrossRef Harman-Boehm I, Bluher M, Redel H, Sion-Vardy N, Ovadia S, Avinoach E et al (2007) Macrophage infiltration into omental versus subcutaneous fat across different populations: effect of regional adiposity and the comorbidities of obesity. J Clin Endocrinol Metab 92(6):2240–2247PubMedCrossRef
84.
85.
go back to reference Hopkins TA, Ouchi N, Shibata R, Walsh K (2007) Adiponectin actions in the cardiovascular system. Cardiovasc Res 74(1):11–18PubMedCrossRef Hopkins TA, Ouchi N, Shibata R, Walsh K (2007) Adiponectin actions in the cardiovascular system. Cardiovasc Res 74(1):11–18PubMedCrossRef
86.
go back to reference Qiao L, Zou C, van der Westhuyzen DR, Shao J (2008) Adiponectin reduces plasma triglyceride by increasing VLDL triglyceride catabolism. Diabetes 57(7):1824–1833PubMedPubMedCentralCrossRef Qiao L, Zou C, van der Westhuyzen DR, Shao J (2008) Adiponectin reduces plasma triglyceride by increasing VLDL triglyceride catabolism. Diabetes 57(7):1824–1833PubMedPubMedCentralCrossRef
87.
go back to reference Aldhahi W, Hamdy O (2003) Adipokines, inflammation, and the endothelium in diabetes. Curr Diab Rep 3(4):293–298PubMedCrossRef Aldhahi W, Hamdy O (2003) Adipokines, inflammation, and the endothelium in diabetes. Curr Diab Rep 3(4):293–298PubMedCrossRef
88.
go back to reference Chen H, Montagnani M, Funahashi T, Shimomura I, Quon MJ (2003) Adiponectin stimulates production of nitric oxide in vascular endothelial cells. J Biol Chem 278(45):45021–45026PubMedCrossRef Chen H, Montagnani M, Funahashi T, Shimomura I, Quon MJ (2003) Adiponectin stimulates production of nitric oxide in vascular endothelial cells. J Biol Chem 278(45):45021–45026PubMedCrossRef
89.
go back to reference Twarda-Clapa A, Olczak A, Bialkowska AM, Koziolkiewicz M (2022) Advanced Glycation End-Products (AGEs): formation, chemistry, classification, receptors, and diseases related to AGEs. Cells 11(8):1312PubMedPubMedCentralCrossRef Twarda-Clapa A, Olczak A, Bialkowska AM, Koziolkiewicz M (2022) Advanced Glycation End-Products (AGEs): formation, chemistry, classification, receptors, and diseases related to AGEs. Cells 11(8):1312PubMedPubMedCentralCrossRef
91.
go back to reference Psaila AM, Vohralik EJ, Quinlan KGR (2022) Shades of white: new insights into tissue-resident leukocyte heterogeneity. FEBS J 289(2):308–318PubMedCrossRef Psaila AM, Vohralik EJ, Quinlan KGR (2022) Shades of white: new insights into tissue-resident leukocyte heterogeneity. FEBS J 289(2):308–318PubMedCrossRef
92.
go back to reference Weinstock A, Brown EJ, Garabedian ML, Pena S, Sharma M, Lafaille J et al (2019) 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 1:e190008PubMedPubMedCentral Weinstock A, Brown EJ, Garabedian ML, Pena S, Sharma M, Lafaille J et al (2019) 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 1:e190008PubMedPubMedCentral
94.
go back to reference Elgazar-Carmon V, Rudich A, Hadad N, Levy R (2008) Neutrophils transiently infiltrate intra-abdominal fat early in the course of high-fat feeding. J Lipid Res 49(9):1894–1903PubMedCrossRef Elgazar-Carmon V, Rudich A, Hadad N, Levy R (2008) Neutrophils transiently infiltrate intra-abdominal fat early in the course of high-fat feeding. J Lipid Res 49(9):1894–1903PubMedCrossRef
95.
go back to reference Gordon S, Martinez FO (2010) Alternative activation of macrophages: mechanism and functions. Immunity 32(5):593–604PubMedCrossRef Gordon S, Martinez FO (2010) Alternative activation of macrophages: mechanism and functions. Immunity 32(5):593–604PubMedCrossRef
97.
go back to reference Wensveen FM, Jelencic V, Valentic S, Sestan M, Wensveen TT, Theurich S et al (2015) NK cells link obesity-induced adipose stress to inflammation and insulin resistance. Nat Immunol 16(4):376–385PubMedCrossRef Wensveen FM, Jelencic V, Valentic S, Sestan M, Wensveen TT, Theurich S et al (2015) NK cells link obesity-induced adipose stress to inflammation and insulin resistance. Nat Immunol 16(4):376–385PubMedCrossRef
98.
go back to reference Dungan LS, McGuinness NC, Boon L, Lynch MA, Mills KH (2014) Innate IFN-gamma promotes development of experimental autoimmune encephalomyelitis: a role for NK cells and M1 macrophages. Eur J Immunol 44(10):2903–2917PubMedCrossRef Dungan LS, McGuinness NC, Boon L, Lynch MA, Mills KH (2014) Innate IFN-gamma promotes development of experimental autoimmune encephalomyelitis: a role for NK cells and M1 macrophages. Eur J Immunol 44(10):2903–2917PubMedCrossRef
99.
go back to reference DeFuria J, Belkina AC, Jagannathan-Bogdan M, Snyder-Cappione J, Carr JD, Nersesova YR et al (2013) B cells promote inflammation in obesity and type 2 diabetes through regulation of T-cell function and an inflammatory cytokine profile. Proc Natl Acad Sci U S A 110(13):5133–5138PubMedPubMedCentralCrossRef DeFuria J, Belkina AC, Jagannathan-Bogdan M, Snyder-Cappione J, Carr JD, Nersesova YR et al (2013) B cells promote inflammation in obesity and type 2 diabetes through regulation of T-cell function and an inflammatory cytokine profile. Proc Natl Acad Sci U S A 110(13):5133–5138PubMedPubMedCentralCrossRef
100.
go back to reference Desai HR, Sivasubramaniyam T, Revelo XS, Schroer SA, Luk CT, Rikkala PR et al (2017) Macrophage JAK2 deficiency protects against high-fat diet-induced inflammation. Sci Rep 7(1):7653PubMedPubMedCentralCrossRef Desai HR, Sivasubramaniyam T, Revelo XS, Schroer SA, Luk CT, Rikkala PR et al (2017) Macrophage JAK2 deficiency protects against high-fat diet-induced inflammation. Sci Rep 7(1):7653PubMedPubMedCentralCrossRef
101.
go back to reference Dror E, Dalmas E, Meier DT, Wueest S, Thevenet J, Thienel C et al (2017) Postprandial macrophage-derived IL-1beta stimulates insulin, and both synergistically promote glucose disposal and inflammation. Nat Immunol 18(3):283–292PubMedCrossRef Dror E, Dalmas E, Meier DT, Wueest S, Thevenet J, Thienel C et al (2017) Postprandial macrophage-derived IL-1beta stimulates insulin, and both synergistically promote glucose disposal and inflammation. Nat Immunol 18(3):283–292PubMedCrossRef
103.
go back to reference Stienstra R, van Diepen JA, Tack CJ, Zaki MH, van de Veerdonk FL, Perera D et al (2011) Inflammasome is a central player in the induction of obesity and insulin resistance. Proc Natl Acad Sci U S A 108(37):15324–15329PubMedPubMedCentralCrossRef Stienstra R, van Diepen JA, Tack CJ, Zaki MH, van de Veerdonk FL, Perera D et al (2011) Inflammasome is a central player in the induction of obesity and insulin resistance. Proc Natl Acad Sci U S A 108(37):15324–15329PubMedPubMedCentralCrossRef
104.
go back to reference Stienstra R, Joosten LA, Koenen T, van Tits B, van Diepen JA, van den Berg SA et al (2010) The inflammasome-mediated caspase-1 activation controls adipocyte differentiation and insulin sensitivity. Cell Metab 12(6):593–605PubMedPubMedCentralCrossRef Stienstra R, Joosten LA, Koenen T, van Tits B, van Diepen JA, van den Berg SA et al (2010) The inflammasome-mediated caspase-1 activation controls adipocyte differentiation and insulin sensitivity. Cell Metab 12(6):593–605PubMedPubMedCentralCrossRef
105.
go back to reference Vandanmagsar B, Youm YH, Ravussin A, Galgani JE, Stadler K, Mynatt RL et al (2011) The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med 17(2):179–188PubMedPubMedCentralCrossRef Vandanmagsar B, Youm YH, Ravussin A, Galgani JE, Stadler K, Mynatt RL et al (2011) The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med 17(2):179–188PubMedPubMedCentralCrossRef
106.
go back to reference Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG et al (2010) NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 464(7293):1357–1361PubMedPubMedCentralCrossRef Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG et al (2010) NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 464(7293):1357–1361PubMedPubMedCentralCrossRef
107.
go back to reference Wen H, Gris D, Lei Y, Jha S, Zhang L, Huang MT et al (2011) Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat Immunol 12(5):408–415PubMedPubMedCentralCrossRef Wen H, Gris D, Lei Y, Jha S, Zhang L, Huang MT et al (2011) Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat Immunol 12(5):408–415PubMedPubMedCentralCrossRef
108.
go back to reference Ruscitti P, Masedu F, Alvaro S, Airo P, Battafarano N, Cantarini L et al (2019) Anti-interleukin-1 treatment in patients with rheumatoid arthritis and type 2 diabetes (TRACK): a multicentre, open-label, randomised controlled trial. PLoS Med 16(9):e1002901PubMedPubMedCentralCrossRef Ruscitti P, Masedu F, Alvaro S, Airo P, Battafarano N, Cantarini L et al (2019) Anti-interleukin-1 treatment in patients with rheumatoid arthritis and type 2 diabetes (TRACK): a multicentre, open-label, randomised controlled trial. PLoS Med 16(9):e1002901PubMedPubMedCentralCrossRef
109.
go back to reference Antohe JL, Bili A, Sartorius JA, Kirchner HL, Morris SJ, Dancea S et al (2012) Diabetes mellitus risk in rheumatoid arthritis: reduced incidence with anti-tumor necrosis factor alpha therapy. Arthritis Care Res (Hoboken) 64(2):215–221PubMedCrossRef Antohe JL, Bili A, Sartorius JA, Kirchner HL, Morris SJ, Dancea S et al (2012) Diabetes mellitus risk in rheumatoid arthritis: reduced incidence with anti-tumor necrosis factor alpha therapy. Arthritis Care Res (Hoboken) 64(2):215–221PubMedCrossRef
110.
go back to reference Solomon DH, Massarotti E, Garg R, Liu J, Canning C, Schneeweiss S (2011) Association between disease-modifying antirheumatic drugs and diabetes risk in patients with rheumatoid arthritis and psoriasis. JAMA 305(24):2525–2531PubMedCrossRef Solomon DH, Massarotti E, Garg R, Liu J, Canning C, Schneeweiss S (2011) Association between disease-modifying antirheumatic drugs and diabetes risk in patients with rheumatoid arthritis and psoriasis. JAMA 305(24):2525–2531PubMedCrossRef
111.
go back to reference Gonzalez-Gay MA, De Matias JM, Gonzalez-Juanatey C, Garcia-Porrua C, Sanchez-Andrade A, Martin J et al (2006) Anti-tumor necrosis factor-alpha blockade improves insulin resistance in patients with rheumatoid arthritis. Clin Exp Rheumatol 24(1):83–86PubMed Gonzalez-Gay MA, De Matias JM, Gonzalez-Juanatey C, Garcia-Porrua C, Sanchez-Andrade A, Martin J et al (2006) Anti-tumor necrosis factor-alpha blockade improves insulin resistance in patients with rheumatoid arthritis. Clin Exp Rheumatol 24(1):83–86PubMed
112.
go back to reference Huvers FC, Popa C, Netea MG, van den Hoogen FH, Tack CJ (2007) Improved insulin sensitivity by anti-TNFalpha antibody treatment in patients with rheumatic diseases. Ann Rheum Dis 66(4):558–559PubMedPubMedCentralCrossRef Huvers FC, Popa C, Netea MG, van den Hoogen FH, Tack CJ (2007) Improved insulin sensitivity by anti-TNFalpha antibody treatment in patients with rheumatic diseases. Ann Rheum Dis 66(4):558–559PubMedPubMedCentralCrossRef
113.
go back to reference Kiortsis DN, Mavridis AK, Vasakos S, Nikas SN, Drosos AA (2005) Effects of infliximab treatment on insulin resistance in patients with rheumatoid arthritis and ankylosing spondylitis. Ann Rheum Dis 64(5):765–766PubMedCrossRef Kiortsis DN, Mavridis AK, Vasakos S, Nikas SN, Drosos AA (2005) Effects of infliximab treatment on insulin resistance in patients with rheumatoid arthritis and ankylosing spondylitis. Ann Rheum Dis 64(5):765–766PubMedCrossRef
114.
go back to reference Marra M, Campanati A, Testa R, Sirolla C, Bonfigli AR, Franceschi C et al (2007) Effect of etanercept on insulin sensitivity in nine patients with psoriasis. Int J Immunopathol Pharmacol 20(4):731–736PubMedCrossRef Marra M, Campanati A, Testa R, Sirolla C, Bonfigli AR, Franceschi C et al (2007) Effect of etanercept on insulin sensitivity in nine patients with psoriasis. Int J Immunopathol Pharmacol 20(4):731–736PubMedCrossRef
115.
go back to reference Stanley TL, Zanni MV, Johnsen S, Rasheed S, Makimura H, Lee H et al (2011) TNF-alpha antagonism with etanercept decreases glucose and increases the proportion of high molecular weight adiponectin in obese subjects with features of the metabolic syndrome. J Clin Endocrinol Metab 96(1):E146–E150PubMedCrossRef Stanley TL, Zanni MV, Johnsen S, Rasheed S, Makimura H, Lee H et al (2011) TNF-alpha antagonism with etanercept decreases glucose and increases the proportion of high molecular weight adiponectin in obese subjects with features of the metabolic syndrome. J Clin Endocrinol Metab 96(1):E146–E150PubMedCrossRef
116.
117.
go back to reference Yazdani-Biuki B, Mueller T, Brezinschek HP, Hermann J, Graninger W, Wascher TC (2006) Relapse of diabetes after interruption of chronic administration of anti-tumor necrosis factor-alpha antibody infliximab: a case observation. Diabetes Care 29(7):1712–1713PubMedCrossRef Yazdani-Biuki B, Mueller T, Brezinschek HP, Hermann J, Graninger W, Wascher TC (2006) Relapse of diabetes after interruption of chronic administration of anti-tumor necrosis factor-alpha antibody infliximab: a case observation. Diabetes Care 29(7):1712–1713PubMedCrossRef
118.
go back to reference Yazdani-Biuki B, Stelzl H, Brezinschek HP, Hermann J, Mueller T, Krippl P et al (2004) Improvement of insulin sensitivity in insulin resistant subjects during prolonged treatment with the anti-TNF-alpha antibody infliximab. Eur J Clin Invest 34(9):641–642PubMedCrossRef Yazdani-Biuki B, Stelzl H, Brezinschek HP, Hermann J, Mueller T, Krippl P et al (2004) Improvement of insulin sensitivity in insulin resistant subjects during prolonged treatment with the anti-TNF-alpha antibody infliximab. Eur J Clin Invest 34(9):641–642PubMedCrossRef
119.
go back to reference Larsen CM, Faulenbach M, Vaag A, Volund A, Ehses JA, Seifert B et al (2007) Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med 356(15):1517–1526PubMedCrossRef Larsen CM, Faulenbach M, Vaag A, Volund A, Ehses JA, Seifert B et al (2007) Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med 356(15):1517–1526PubMedCrossRef
120.
go back to reference Cavelti-Weder C, Furrer R, Keller C, Babians-Brunner A, Solinger AM, Gast H et al (2011) Inhibition of IL-1beta improves fatigue in type 2 diabetes. Diabetes Care 34(10):e158PubMedPubMedCentralCrossRef Cavelti-Weder C, Furrer R, Keller C, Babians-Brunner A, Solinger AM, Gast H et al (2011) Inhibition of IL-1beta improves fatigue in type 2 diabetes. Diabetes Care 34(10):e158PubMedPubMedCentralCrossRef
121.
go back to reference Rissanen A, Howard CP, Botha J, Thuren T, Global I (2012) Effect of anti-IL-1beta antibody (canakinumab) on insulin secretion rates in impaired glucose tolerance or type 2 diabetes: results of a randomized, placebo-controlled trial. Diabetes Obes Metab 14(12):1088–1096PubMedCrossRef Rissanen A, Howard CP, Botha J, Thuren T, Global I (2012) Effect of anti-IL-1beta antibody (canakinumab) on insulin secretion rates in impaired glucose tolerance or type 2 diabetes: results of a randomized, placebo-controlled trial. Diabetes Obes Metab 14(12):1088–1096PubMedCrossRef
122.
go back to reference Sloan-Lancaster J, Abu-Raddad E, Polzer J, Miller JW, Scherer JC, De Gaetano A et al (2013) Double-blind, randomized study evaluating the glycemic and anti-inflammatory effects of subcutaneous LY2189102, a neutralizing IL-1beta antibody, in patients with type 2 diabetes. Diabetes Care 36(8):2239–2246PubMedPubMedCentralCrossRef Sloan-Lancaster J, Abu-Raddad E, Polzer J, Miller JW, Scherer JC, De Gaetano A et al (2013) Double-blind, randomized study evaluating the glycemic and anti-inflammatory effects of subcutaneous LY2189102, a neutralizing IL-1beta antibody, in patients with type 2 diabetes. Diabetes Care 36(8):2239–2246PubMedPubMedCentralCrossRef
123.
go back to reference Furuhashi M, Ura N, Higashiura K, Murakami H, Tanaka M, Moniwa N et al (2003) Blockade of the renin-angiotensin system increases adiponectin concentrations in patients with essential hypertension. Hypertension 42(1):76–81PubMedCrossRef Furuhashi M, Ura N, Higashiura K, Murakami H, Tanaka M, Moniwa N et al (2003) Blockade of the renin-angiotensin system increases adiponectin concentrations in patients with essential hypertension. Hypertension 42(1):76–81PubMedCrossRef
124.
go back to reference Decochez K, Rippley RK, Miller JL, De Smet M, Yan KX, Matthijs Z et al (2006) A dual PPAR alpha/gamma agonist increases adiponectin and improves plasma lipid profiles in healthy subjects. Drugs R D 7(2):99–110PubMedCrossRef Decochez K, Rippley RK, Miller JL, De Smet M, Yan KX, Matthijs Z et al (2006) A dual PPAR alpha/gamma agonist increases adiponectin and improves plasma lipid profiles in healthy subjects. Drugs R D 7(2):99–110PubMedCrossRef
125.
go back to reference Li M, Kim DH, Tsenovoy PL, Peterson SJ, Rezzani R, Rodella LF et al (2008) Treatment of obese diabetic mice with a heme oxygenase inducer reduces visceral and subcutaneous adiposity, increases adiponectin levels, and improves insulin sensitivity and glucose tolerance. Diabetes 57(6):1526–1535PubMedCrossRef Li M, Kim DH, Tsenovoy PL, Peterson SJ, Rezzani R, Rodella LF et al (2008) Treatment of obese diabetic mice with a heme oxygenase inducer reduces visceral and subcutaneous adiposity, increases adiponectin levels, and improves insulin sensitivity and glucose tolerance. Diabetes 57(6):1526–1535PubMedCrossRef
126.
go back to reference Phillips SA, Ciaraldi TP, Kong AP, Bandukwala R, Aroda V, Carter L et al (2003) Modulation of circulating and adipose tissue adiponectin levels by antidiabetic therapy. Diabetes 52(3):667–674PubMedCrossRef Phillips SA, Ciaraldi TP, Kong AP, Bandukwala R, Aroda V, Carter L et al (2003) Modulation of circulating and adipose tissue adiponectin levels by antidiabetic therapy. Diabetes 52(3):667–674PubMedCrossRef
127.
go back to reference Yamauchi T, Kamon J, Waki H, Terauchi Y, Kubota N, Hara K et al (2001) The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat Med 7(8):941–946PubMedCrossRef Yamauchi T, Kamon J, Waki H, Terauchi Y, Kubota N, Hara K et al (2001) The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat Med 7(8):941–946PubMedCrossRef
128.
go back to reference Matsuda M, Shimomura I, Sata M, Arita Y, Nishida M, Maeda N et al (2002) Role of adiponectin in preventing vascular stenosis. The missing link of adipo-vascular axis. J Biol Chem. 277(40):37487–91PubMedCrossRef Matsuda M, Shimomura I, Sata M, Arita Y, Nishida M, Maeda N et al (2002) Role of adiponectin in preventing vascular stenosis. The missing link of adipo-vascular axis. J Biol Chem. 277(40):37487–91PubMedCrossRef
129.
go back to reference Bastard JP, Maachi M, Lagathu C, Kim MJ, Caron M, Vidal H et al (2006) Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur Cytokine Netw 17(1):4–12PubMed Bastard JP, Maachi M, Lagathu C, Kim MJ, Caron M, Vidal H et al (2006) Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur Cytokine Netw 17(1):4–12PubMed
130.
go back to reference Goldstein BJ, Scalia R (2004) Adiponectin: a novel adipokine linking adipocytes and vascular function. J Clin Endocrinol Metab 89(6):2563–2568PubMedCrossRef Goldstein BJ, Scalia R (2004) Adiponectin: a novel adipokine linking adipocytes and vascular function. J Clin Endocrinol Metab 89(6):2563–2568PubMedCrossRef
131.
go back to reference Nakamura Y, Shimada K, Fukuda D, Shimada Y, Ehara S, Hirose M et al (2004) Implications of plasma concentrations of adiponectin in patients with coronary artery disease. Heart 90(5):528–533PubMedPubMedCentralCrossRef Nakamura Y, Shimada K, Fukuda D, Shimada Y, Ehara S, Hirose M et al (2004) Implications of plasma concentrations of adiponectin in patients with coronary artery disease. Heart 90(5):528–533PubMedPubMedCentralCrossRef
132.
go back to reference Ouedraogo R, Wu X, Xu SQ, Fuchsel L, Motoshima H, Mahadev K et al (2006) Adiponectin suppression of high-glucose-induced reactive oxygen species in vascular endothelial cells: evidence for involvement of a cAMP signaling pathway. Diabetes 55(6):1840–1846PubMedCrossRef Ouedraogo R, Wu X, Xu SQ, Fuchsel L, Motoshima H, Mahadev K et al (2006) Adiponectin suppression of high-glucose-induced reactive oxygen species in vascular endothelial cells: evidence for involvement of a cAMP signaling pathway. Diabetes 55(6):1840–1846PubMedCrossRef
133.
go back to reference Harmancey R, Wilson CR, Taegtmeyer H (2008) Adaptation and maladaptation of the heart in obesity. Hypertension 52(2):181–187PubMedCrossRef Harmancey R, Wilson CR, Taegtmeyer H (2008) Adaptation and maladaptation of the heart in obesity. Hypertension 52(2):181–187PubMedCrossRef
134.
go back to reference De Rosa A, Monaco ML, Capasso M, Forestieri P, Pilone V, Nardelli C et al (2013) Adiponectin oligomers as potential indicators of adipose tissue improvement in obese subjects. Eur J Endocrinol 169(1):37–43PubMedCrossRef De Rosa A, Monaco ML, Capasso M, Forestieri P, Pilone V, Nardelli C et al (2013) Adiponectin oligomers as potential indicators of adipose tissue improvement in obese subjects. Eur J Endocrinol 169(1):37–43PubMedCrossRef
135.
go back to reference Papatriantafyllou M (2011) Mucosal immunology: inflammasome shapes the microbiota. Nat Rev Immunol 11(7):439PubMedCrossRef Papatriantafyllou M (2011) Mucosal immunology: inflammasome shapes the microbiota. Nat Rev Immunol 11(7):439PubMedCrossRef
136.
go back to reference Cancello R, Henegar C, Viguerie N, Taleb S, Poitou C, Rouault C et al (2005) Reduction of macrophage infiltration and chemoattractant gene expression changes in white adipose tissue of morbidly obese subjects after surgery-induced weight loss. Diabetes 54(8):2277–2286PubMedCrossRef Cancello R, Henegar C, Viguerie N, Taleb S, Poitou C, Rouault C et al (2005) Reduction of macrophage infiltration and chemoattractant gene expression changes in white adipose tissue of morbidly obese subjects after surgery-induced weight loss. Diabetes 54(8):2277–2286PubMedCrossRef
137.
go back to reference Kim KA, Gu W, Lee IA, Joh EH, Kim DH (2012) High fat diet-induced gut microbiota exacerbates inflammation and obesity in mice via the TLR4 signaling pathway. PLoS One 7(10):e47713PubMedPubMedCentralCrossRef Kim KA, Gu W, Lee IA, Joh EH, Kim DH (2012) High fat diet-induced gut microbiota exacerbates inflammation and obesity in mice via the TLR4 signaling pathway. PLoS One 7(10):e47713PubMedPubMedCentralCrossRef
138.
go back to reference Tilg H, Zmora N, Adolph TE, Elinav E (2020) The intestinal microbiota fuelling metabolic inflammation. Nat Rev Immunol 20(1):40–54PubMedCrossRef Tilg H, Zmora N, Adolph TE, Elinav E (2020) The intestinal microbiota fuelling metabolic inflammation. Nat Rev Immunol 20(1):40–54PubMedCrossRef
139.
go back to reference Kreutzer C, Peters S, Schulte DM, Fangmann D, Turk K, Wolff S et al (2017) Hypothalamic inflammation in human obesity is mediated by environmental and genetic factors. Diabetes 66(9):2407–2415PubMedCrossRef Kreutzer C, Peters S, Schulte DM, Fangmann D, Turk K, Wolff S et al (2017) Hypothalamic inflammation in human obesity is mediated by environmental and genetic factors. Diabetes 66(9):2407–2415PubMedCrossRef
140.
go back to reference Fouesnard M, Zoppi J, Petera M, Le Gleau L, Migne C, Devime F et al (2021) Dietary switch to Western diet induces hypothalamic adaptation associated with gut microbiota dysbiosis in rats. Int J Obes (Lond) 45(6):1271–1283PubMedCrossRef Fouesnard M, Zoppi J, Petera M, Le Gleau L, Migne C, Devime F et al (2021) Dietary switch to Western diet induces hypothalamic adaptation associated with gut microbiota dysbiosis in rats. Int J Obes (Lond) 45(6):1271–1283PubMedCrossRef
141.
142.
go back to reference Kim DW, Glendining KA, Grattan DR, Jasoni CL (2016) Maternal obesity in the mouse compromises the blood-brain barrier in the arcuate nucleus of offspring. Endocrinology 157(6):2229–2242PubMedCrossRef Kim DW, Glendining KA, Grattan DR, Jasoni CL (2016) Maternal obesity in the mouse compromises the blood-brain barrier in the arcuate nucleus of offspring. Endocrinology 157(6):2229–2242PubMedCrossRef
144.
go back to reference Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Toth M et al (2014) The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med. 6(263):263ra158PubMedPubMedCentralCrossRef Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Toth M et al (2014) The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med. 6(263):263ra158PubMedPubMedCentralCrossRef
145.
146.
go back to reference Chen KE, Lainez NM, Nair MG, Coss D (2021) Visceral adipose tissue imparts peripheral macrophage influx into the hypothalamus. J Neuroinflammation 18(1):140PubMedPubMedCentralCrossRef Chen KE, Lainez NM, Nair MG, Coss D (2021) Visceral adipose tissue imparts peripheral macrophage influx into the hypothalamus. J Neuroinflammation 18(1):140PubMedPubMedCentralCrossRef
147.
go back to reference Valdearcos M, Douglass JD, Robblee MM, Dorfman MD, Stifler DR, Bennett ML et al (2017) Microglial inflammatory signaling orchestrates the hypothalamic immune response to dietary excess and mediates obesity susceptibility. Cell Metab. 26(1):185–97.e3PubMedPubMedCentralCrossRef Valdearcos M, Douglass JD, Robblee MM, Dorfman MD, Stifler DR, Bennett ML et al (2017) Microglial inflammatory signaling orchestrates the hypothalamic immune response to dietary excess and mediates obesity susceptibility. Cell Metab. 26(1):185–97.e3PubMedPubMedCentralCrossRef
148.
go back to reference Andre C, Guzman-Quevedo O, Rey C, Remus-Borel J, Clark S, Castellanos-Jankiewicz A et al (2017) Inhibiting microglia expansion prevents diet-induced hypothalamic and peripheral inflammation. Diabetes 66(4):908–919PubMedCrossRef Andre C, Guzman-Quevedo O, Rey C, Remus-Borel J, Clark S, Castellanos-Jankiewicz A et al (2017) Inhibiting microglia expansion prevents diet-induced hypothalamic and peripheral inflammation. Diabetes 66(4):908–919PubMedCrossRef
151.
go back to reference Valdearcos M, Robblee MM, Benjamin DI, Nomura DK, Xu AW, Koliwad SK (2014) Microglia dictate the impact of saturated fat consumption on hypothalamic inflammation and neuronal function. Cell Rep 9(6):2124–2138PubMedPubMedCentralCrossRef Valdearcos M, Robblee MM, Benjamin DI, Nomura DK, Xu AW, Koliwad SK (2014) Microglia dictate the impact of saturated fat consumption on hypothalamic inflammation and neuronal function. Cell Rep 9(6):2124–2138PubMedPubMedCentralCrossRef
152.
go back to reference Thaler JP, Yi CX, Schur EA, Guyenet SJ, Hwang BH, Dietrich MO et al (2012) Obesity is associated with hypothalamic injury in rodents and humans. J Clin Invest 122(1):153–162PubMedCrossRef Thaler JP, Yi CX, Schur EA, Guyenet SJ, Hwang BH, Dietrich MO et al (2012) Obesity is associated with hypothalamic injury in rodents and humans. J Clin Invest 122(1):153–162PubMedCrossRef
153.
154.
go back to reference De Simone R, Ajmone-Cat MA, Pandolfi M, Bernardo A, De Nuccio C, Minghetti L et al (2015) The mitochondrial uncoupling protein-2 is a master regulator of both M1 and M2 microglial responses. J Neurochem 135(1):147–156PubMedCrossRef De Simone R, Ajmone-Cat MA, Pandolfi M, Bernardo A, De Nuccio C, Minghetti L et al (2015) The mitochondrial uncoupling protein-2 is a master regulator of both M1 and M2 microglial responses. J Neurochem 135(1):147–156PubMedCrossRef
155.
go back to reference Arsenijevic D, Clavel S, Sanchis D, Plamondon J, Huang Q, Ricquier D et al (2007) Induction of Ucp2 expression in brain phagocytes and neurons following murine toxoplasmosis: an essential role of IFN-gamma and an association with negative energy balance. J Neuroimmunol 186(1–2):121–132PubMedCrossRef Arsenijevic D, Clavel S, Sanchis D, Plamondon J, Huang Q, Ricquier D et al (2007) Induction of Ucp2 expression in brain phagocytes and neurons following murine toxoplasmosis: an essential role of IFN-gamma and an association with negative energy balance. J Neuroimmunol 186(1–2):121–132PubMedCrossRef
156.
go back to reference Parton LE, Ye CP, Coppari R, Enriori PJ, Choi B, Zhang CY et al (2007) Glucose sensing by POMC neurons regulates glucose homeostasis and is impaired in obesity. Nature 449(7159):228–232PubMedCrossRef Parton LE, Ye CP, Coppari R, Enriori PJ, Choi B, Zhang CY et al (2007) Glucose sensing by POMC neurons regulates glucose homeostasis and is impaired in obesity. Nature 449(7159):228–232PubMedCrossRef
157.
go back to reference Diano S, Horvath TL (2012) Mitochondrial uncoupling protein 2 (UCP2) in glucose and lipid metabolism. Trends Mol Med 18(1):52–58PubMedCrossRef Diano S, Horvath TL (2012) Mitochondrial uncoupling protein 2 (UCP2) in glucose and lipid metabolism. Trends Mol Med 18(1):52–58PubMedCrossRef
158.
go back to reference Affourtit C, Crichton PG, Parker N, Brand MD (2007) Novel uncoupling proteins. Novartis Found Symp. 287:70–80. Discussion -91PubMedCrossRef Affourtit C, Crichton PG, Parker N, Brand MD (2007) Novel uncoupling proteins. Novartis Found Symp. 287:70–80. Discussion -91PubMedCrossRef
159.
go back to reference Echtay KS (2007) Mitochondrial uncoupling proteins–what is their physiological role? Free Radic Biol Med 43(10):1351–1371PubMedCrossRef Echtay KS (2007) Mitochondrial uncoupling proteins–what is their physiological role? Free Radic Biol Med 43(10):1351–1371PubMedCrossRef
160.
go back to reference Krauss S, Zhang CY, Lowell BB (2005) The mitochondrial uncoupling-protein homologues. Nat Rev Mol Cell Biol 6(3):248–261PubMedCrossRef Krauss S, Zhang CY, Lowell BB (2005) The mitochondrial uncoupling-protein homologues. Nat Rev Mol Cell Biol 6(3):248–261PubMedCrossRef
161.
go back to reference Alan L, Smolkova K, Kronusova E, Santorova J, Jezek P (2009) Absolute levels of transcripts for mitochondrial uncoupling proteins UCP2, UCP3, UCP4, and UCP5 show different patterns in rat and mice tissues. J Bioenerg Biomembr 41(1):71–78PubMedCrossRef Alan L, Smolkova K, Kronusova E, Santorova J, Jezek P (2009) Absolute levels of transcripts for mitochondrial uncoupling proteins UCP2, UCP3, UCP4, and UCP5 show different patterns in rat and mice tissues. J Bioenerg Biomembr 41(1):71–78PubMedCrossRef
162.
go back to reference Andrews ZB, Diano S, Horvath TL (2005) Mitochondrial uncoupling proteins in the CNS: in support of function and survival. Nat Rev Neurosci 6(11):829–840PubMedCrossRef Andrews ZB, Diano S, Horvath TL (2005) Mitochondrial uncoupling proteins in the CNS: in support of function and survival. Nat Rev Neurosci 6(11):829–840PubMedCrossRef
163.
go back to reference Le Fur S, Le Stunff C, Dos Santos C, Bougneres P (2004) The common -866 G/A polymorphism in the promoter of uncoupling protein 2 is associated with increased carbohydrate and decreased lipid oxidation in juvenile obesity. Diabetes 53(1):235–239PubMedCrossRef Le Fur S, Le Stunff C, Dos Santos C, Bougneres P (2004) The common -866 G/A polymorphism in the promoter of uncoupling protein 2 is associated with increased carbohydrate and decreased lipid oxidation in juvenile obesity. Diabetes 53(1):235–239PubMedCrossRef
164.
go back to reference Bulotta A, Ludovico O, Coco A, Di Paola R, Quattrone A, Carella M et al (2005) The common -866G/A polymorphism in the promoter region of the UCP-2 gene is associated with reduced risk of type 2 diabetes in Caucasians from Italy. J Clin Endocrinol Metab 90(2):1176–1180PubMedCrossRef Bulotta A, Ludovico O, Coco A, Di Paola R, Quattrone A, Carella M et al (2005) The common -866G/A polymorphism in the promoter region of the UCP-2 gene is associated with reduced risk of type 2 diabetes in Caucasians from Italy. J Clin Endocrinol Metab 90(2):1176–1180PubMedCrossRef
165.
go back to reference Su M, Chen X, Chen Y, Wang C, Li S, Ying X et al (2018) UCP2 and UCP3 variants and gene-environment interaction associated with prediabetes and T2DM in a rural population: a case control study in China. BMC Med Genet 19(1):43PubMedPubMedCentralCrossRef Su M, Chen X, Chen Y, Wang C, Li S, Ying X et al (2018) UCP2 and UCP3 variants and gene-environment interaction associated with prediabetes and T2DM in a rural population: a case control study in China. BMC Med Genet 19(1):43PubMedPubMedCentralCrossRef
166.
go back to reference Mehta SL, Li PA (2009) Neuroprotective role of mitochondrial uncoupling protein 2 in cerebral stroke. J Cereb Blood Flow Metab 29(6):1069–1078PubMedCrossRef Mehta SL, Li PA (2009) Neuroprotective role of mitochondrial uncoupling protein 2 in cerebral stroke. J Cereb Blood Flow Metab 29(6):1069–1078PubMedCrossRef
167.
go back to reference De Souza CT, Araujo EP, Bordin S, Ashimine R, Zollner RL, Boschero AC et al (2005) Consumption of a fat-rich diet activates a proinflammatory response and induces insulin resistance in the hypothalamus. Endocrinology 146(10):4192–4199PubMedCrossRef De Souza CT, Araujo EP, Bordin S, Ashimine R, Zollner RL, Boschero AC et al (2005) Consumption of a fat-rich diet activates a proinflammatory response and induces insulin resistance in the hypothalamus. Endocrinology 146(10):4192–4199PubMedCrossRef
169.
go back to reference Ohta S, Misawa A, Lefebvre V, Okano H, Kawakami Y, Toda M (2013) Sox6 up-regulation by macrophage migration inhibitory factor promotes survival and maintenance of mouse neural stem/progenitor cells. PLoS One 8(9):e74315PubMedPubMedCentralCrossRef Ohta S, Misawa A, Lefebvre V, Okano H, Kawakami Y, Toda M (2013) Sox6 up-regulation by macrophage migration inhibitory factor promotes survival and maintenance of mouse neural stem/progenitor cells. PLoS One 8(9):e74315PubMedPubMedCentralCrossRef
170.
go back to reference Wegner M, Stolt CC (2005) From stem cells to neurons and glia: a Soxist’s view of neural development. Trends Neurosci 28(11):583–588PubMedCrossRef Wegner M, Stolt CC (2005) From stem cells to neurons and glia: a Soxist’s view of neural development. Trends Neurosci 28(11):583–588PubMedCrossRef
171.
go back to reference Zhang J, Jiao J (2015) Molecular biomarkers for embryonic and adult neural stem cell and neurogenesis. Biomed Res Int 2015:727542PubMedPubMedCentral Zhang J, Jiao J (2015) Molecular biomarkers for embryonic and adult neural stem cell and neurogenesis. Biomed Res Int 2015:727542PubMedPubMedCentral
172.
go back to reference Baier PC, May U, Scheller J, Rose-John S, Schiffelholz T (2009) Impaired hippocampus-dependent and -independent learning in IL-6 deficient mice. Behav Brain Res 200(1):192–196PubMedCrossRef Baier PC, May U, Scheller J, Rose-John S, Schiffelholz T (2009) Impaired hippocampus-dependent and -independent learning in IL-6 deficient mice. Behav Brain Res 200(1):192–196PubMedCrossRef
173.
go back to reference Gomes da Silva S, Simoes PS, Mortara RA, Scorza FA, Cavalheiro EA, da Graca Naffah-Mazzacoratti M et al (2013) Exercise-induced hippocampal anti-inflammatory response in aged rats. J Neuroinflammation. 10:61PubMedPubMedCentralCrossRef Gomes da Silva S, Simoes PS, Mortara RA, Scorza FA, Cavalheiro EA, da Graca Naffah-Mazzacoratti M et al (2013) Exercise-induced hippocampal anti-inflammatory response in aged rats. J Neuroinflammation. 10:61PubMedPubMedCentralCrossRef
174.
go back to reference Ropelle ER, Flores MB, Cintra DE, Rocha GZ, Pauli JR, Morari J et al (2010) IL-6 and IL-10 anti-inflammatory activity links exercise to hypothalamic insulin and leptin sensitivity through IKKbeta and ER stress inhibition. PLoS Biol 8(8):e100046CrossRef Ropelle ER, Flores MB, Cintra DE, Rocha GZ, Pauli JR, Morari J et al (2010) IL-6 and IL-10 anti-inflammatory activity links exercise to hypothalamic insulin and leptin sensitivity through IKKbeta and ER stress inhibition. PLoS Biol 8(8):e100046CrossRef
175.
go back to reference Silva VRR, Micheletti TO, Katashima CK, Lenhare L, Morari J, Moura-Assis A et al (2018) Exercise activates the hypothalamic S1PR1-STAT3 axis through the central action of interleukin 6 in mice. J Cell Physiol 233(12):9426–9436PubMedCrossRef Silva VRR, Micheletti TO, Katashima CK, Lenhare L, Morari J, Moura-Assis A et al (2018) Exercise activates the hypothalamic S1PR1-STAT3 axis through the central action of interleukin 6 in mice. J Cell Physiol 233(12):9426–9436PubMedCrossRef
176.
go back to reference Bobbo VC, Engel DF, Jara CP, Mendes NF, Haddad-Tovolli R, Prado TP et al (2021) Interleukin-6 actions in the hypothalamus protects against obesity and is involved in the regulation of neurogenesis. J Neuroinflammation 18(1):192PubMedPubMedCentralCrossRef Bobbo VC, Engel DF, Jara CP, Mendes NF, Haddad-Tovolli R, Prado TP et al (2021) Interleukin-6 actions in the hypothalamus protects against obesity and is involved in the regulation of neurogenesis. J Neuroinflammation 18(1):192PubMedPubMedCentralCrossRef
177.
go back to reference Bjorbaek C, El-Haschimi K, Frantz JD, Flier JS (1999) The role of SOCS-3 in leptin signaling and leptin resistance. J Biol Chem 274(42):30059–30065PubMedCrossRef Bjorbaek C, El-Haschimi K, Frantz JD, Flier JS (1999) The role of SOCS-3 in leptin signaling and leptin resistance. J Biol Chem 274(42):30059–30065PubMedCrossRef
178.
go back to reference Bjorbaek C, Elmquist JK, Frantz JD, Shoelson SE, Flier JS (1998) Identification of SOCS-3 as a potential mediator of central leptin resistance. Mol Cell 1(4):619–625PubMedCrossRef Bjorbaek C, Elmquist JK, Frantz JD, Shoelson SE, Flier JS (1998) Identification of SOCS-3 as a potential mediator of central leptin resistance. Mol Cell 1(4):619–625PubMedCrossRef
179.
go back to reference Wunderlich CM, Hovelmeyer N, Wunderlich FT (2013) Mechanisms of chronic JAK-STAT3-SOCS3 signaling in obesity. JAKSTAT 2(2):e23878PubMedPubMedCentral Wunderlich CM, Hovelmeyer N, Wunderlich FT (2013) Mechanisms of chronic JAK-STAT3-SOCS3 signaling in obesity. JAKSTAT 2(2):e23878PubMedPubMedCentral
180.
go back to reference Yang Z, Hulver M, McMillan RP, Cai L, Kershaw EE, Yu L et al (2012) Regulation of insulin and leptin signaling by muscle suppressor of cytokine signaling 3 (SOCS3). PLoS One 7(10):e47493PubMedPubMedCentralCrossRef Yang Z, Hulver M, McMillan RP, Cai L, Kershaw EE, Yu L et al (2012) Regulation of insulin and leptin signaling by muscle suppressor of cytokine signaling 3 (SOCS3). PLoS One 7(10):e47493PubMedPubMedCentralCrossRef
181.
go back to reference Pedroso JA, Buonfiglio DC, Cardinali LI, Furigo IC, Ramos-Lobo AM, Tirapegui J et al (2014) Inactivation of SOCS3 in leptin receptor-expressing cells protects mice from diet-induced insulin resistance but does not prevent obesity. Mol Metab 3(6):608–618PubMedPubMedCentralCrossRef Pedroso JA, Buonfiglio DC, Cardinali LI, Furigo IC, Ramos-Lobo AM, Tirapegui J et al (2014) Inactivation of SOCS3 in leptin receptor-expressing cells protects mice from diet-induced insulin resistance but does not prevent obesity. Mol Metab 3(6):608–618PubMedPubMedCentralCrossRef
182.
go back to reference Bernardi M, Moreau R, Angeli P, Schnabl B, Arroyo V (2015) Mechanisms of decompensation and organ failure in cirrhosis: From peripheral arterial vasodilation to systemic inflammation hypothesis. J Hepatol 63(5):1272–1284PubMedCrossRef Bernardi M, Moreau R, Angeli P, Schnabl B, Arroyo V (2015) Mechanisms of decompensation and organ failure in cirrhosis: From peripheral arterial vasodilation to systemic inflammation hypothesis. J Hepatol 63(5):1272–1284PubMedCrossRef
183.
go back to reference Seidman JS, Troutman TD, Sakai M, Gola A, Spann NJ, Bennett H et al (2020) Niche-Specific Reprogramming of Epigenetic Landscapes Drives Myeloid Cell Diversity in Nonalcoholic Steatohepatitis. Immunity. 52(6):1057–74.e7PubMedPubMedCentralCrossRef Seidman JS, Troutman TD, Sakai M, Gola A, Spann NJ, Bennett H et al (2020) Niche-Specific Reprogramming of Epigenetic Landscapes Drives Myeloid Cell Diversity in Nonalcoholic Steatohepatitis. Immunity. 52(6):1057–74.e7PubMedPubMedCentralCrossRef
184.
go back to reference Loomba R, Gindin Y, Jiang Z, Lawitz E, Caldwell S, Djedjos CS et al (2018) DNA methylation signatures reflect aging in patients with nonalcoholic steatohepatitis. JCI Insight. 3(2):e96685PubMedPubMedCentralCrossRef Loomba R, Gindin Y, Jiang Z, Lawitz E, Caldwell S, Djedjos CS et al (2018) DNA methylation signatures reflect aging in patients with nonalcoholic steatohepatitis. JCI Insight. 3(2):e96685PubMedPubMedCentralCrossRef
185.
go back to reference Loomba R, Friedman SL, Shulman GI (2021) Mechanisms and disease consequences of nonalcoholic fatty liver disease. Cell 184(10):2537–2564PubMedCrossRef Loomba R, Friedman SL, Shulman GI (2021) Mechanisms and disease consequences of nonalcoholic fatty liver disease. Cell 184(10):2537–2564PubMedCrossRef
186.
go back to reference Feldstein AE, Canbay A, Angulo P, Taniai M, Burgart LJ, Lindor KD et al (2003) Hepatocyte apoptosis and fas expression are prominent features of human nonalcoholic steatohepatitis. Gastroenterology 125(2):437–443PubMedCrossRef Feldstein AE, Canbay A, Angulo P, Taniai M, Burgart LJ, Lindor KD et al (2003) Hepatocyte apoptosis and fas expression are prominent features of human nonalcoholic steatohepatitis. Gastroenterology 125(2):437–443PubMedCrossRef
187.
go back to reference Buzzetti E, Pinzani M, Tsochatzis EA (2016) The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism 65(8):1038–1048PubMedCrossRef Buzzetti E, Pinzani M, Tsochatzis EA (2016) The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism 65(8):1038–1048PubMedCrossRef
188.
go back to reference Brunt EM, Wong VW, Nobili V, Day CP, Sookoian S, Maher JJ et al (2015) Nonalcoholic fatty liver disease. Nat Rev Dis Primers 1:15080PubMedCrossRef Brunt EM, Wong VW, Nobili V, Day CP, Sookoian S, Maher JJ et al (2015) Nonalcoholic fatty liver disease. Nat Rev Dis Primers 1:15080PubMedCrossRef
189.
go back to reference Begriche K, Massart J, Robin MA, Bonnet F, Fromenty B (2013) Mitochondrial adaptations and dysfunctions in nonalcoholic fatty liver disease. Hepatology 58(4):1497–1507PubMedCrossRef Begriche K, Massart J, Robin MA, Bonnet F, Fromenty B (2013) Mitochondrial adaptations and dysfunctions in nonalcoholic fatty liver disease. Hepatology 58(4):1497–1507PubMedCrossRef
190.
go back to reference Cusi K (2012) Role of obesity and lipotoxicity in the development of nonalcoholic steatohepatitis: pathophysiology and clinical implications. Gastroenterology. 142(4):711–25.e6PubMedCrossRef Cusi K (2012) Role of obesity and lipotoxicity in the development of nonalcoholic steatohepatitis: pathophysiology and clinical implications. Gastroenterology. 142(4):711–25.e6PubMedCrossRef
191.
go back to reference Schnabl B, Brenner DA (2014) Interactions between the intestinal microbiome and liver diseases. Gastroenterology 146(6):1513–1524PubMedCrossRef Schnabl B, Brenner DA (2014) Interactions between the intestinal microbiome and liver diseases. Gastroenterology 146(6):1513–1524PubMedCrossRef
192.
go back to reference Potter JJ, Rennie-Tankesley L, Mezey E (2003) Influence of leptin in the development of hepatic fibrosis produced in mice by Schistosoma mansoni infection and by chronic carbon tetrachloride administration. J Hepatol 38(3):281–288PubMedCrossRef Potter JJ, Rennie-Tankesley L, Mezey E (2003) Influence of leptin in the development of hepatic fibrosis produced in mice by Schistosoma mansoni infection and by chronic carbon tetrachloride administration. J Hepatol 38(3):281–288PubMedCrossRef
193.
go back to reference Wang J, Leclercq I, Brymora JM, Xu N, Ramezani-Moghadam M, London RM et al (2009) Kupffer cells mediate leptin-induced liver fibrosis. Gastroenterology 137(2):713–723PubMedCrossRef Wang J, Leclercq I, Brymora JM, Xu N, Ramezani-Moghadam M, London RM et al (2009) Kupffer cells mediate leptin-induced liver fibrosis. Gastroenterology 137(2):713–723PubMedCrossRef
194.
go back to reference Ikejima K, Takei Y, Honda H, Hirose M, Yoshikawa M, Zhang YJ et al (2002) Leptin receptor-mediated signaling regulates hepatic fibrogenesis and remodeling of extracellular matrix in the rat. Gastroenterology 122(5):1399–1410PubMedCrossRef Ikejima K, Takei Y, Honda H, Hirose M, Yoshikawa M, Zhang YJ et al (2002) Leptin receptor-mediated signaling regulates hepatic fibrogenesis and remodeling of extracellular matrix in the rat. Gastroenterology 122(5):1399–1410PubMedCrossRef
195.
go back to reference De Minicis S, Seki E, Oesterreicher C, Schnabl B, Schwabe RF, Brenner DA (2008) Reduced nicotinamide adenine dinucleotide phosphate oxidase mediates fibrotic and inflammatory effects of leptin on hepatic stellate cells. Hepatology 48(6):2016–2026PubMedCrossRef De Minicis S, Seki E, Oesterreicher C, Schnabl B, Schwabe RF, Brenner DA (2008) Reduced nicotinamide adenine dinucleotide phosphate oxidase mediates fibrotic and inflammatory effects of leptin on hepatic stellate cells. Hepatology 48(6):2016–2026PubMedCrossRef
196.
go back to reference Adachi M, Brenner DA (2008) High molecular weight adiponectin inhibits proliferation of hepatic stellate cells via activation of adenosine monophosphate-activated protein kinase. Hepatology 47(2):677–685PubMedCrossRef Adachi M, Brenner DA (2008) High molecular weight adiponectin inhibits proliferation of hepatic stellate cells via activation of adenosine monophosphate-activated protein kinase. Hepatology 47(2):677–685PubMedCrossRef
197.
go back to reference Xu A, Wang Y, Keshaw H, Xu LY, Lam KS, Cooper GJ (2003) The fat-derived hormone adiponectin alleviates alcoholic and nonalcoholic fatty liver diseases in mice. J Clin Invest 112(1):91–100PubMedPubMedCentralCrossRef Xu A, Wang Y, Keshaw H, Xu LY, Lam KS, Cooper GJ (2003) The fat-derived hormone adiponectin alleviates alcoholic and nonalcoholic fatty liver diseases in mice. J Clin Invest 112(1):91–100PubMedPubMedCentralCrossRef
198.
go back to reference Xu H, Zhao Q, Song N, Yan Z, Lin R, Wu S et al (2020) AdipoR1/AdipoR2 dual agonist recovers nonalcoholic steatohepatitis and related fibrosis via endoplasmic reticulum-mitochondria axis. Nat Commun 11(1):5807PubMedPubMedCentralCrossRef Xu H, Zhao Q, Song N, Yan Z, Lin R, Wu S et al (2020) AdipoR1/AdipoR2 dual agonist recovers nonalcoholic steatohepatitis and related fibrosis via endoplasmic reticulum-mitochondria axis. Nat Commun 11(1):5807PubMedPubMedCentralCrossRef
199.
go back to reference Hueber W, Patel DD, Dryja T, Wright AM, Koroleva I, Bruin G et al (2010) Effects of AIN457, a fully human antibody to interleukin-17A, on psoriasis, rheumatoid arthritis, and uveitis. Sci Transl Med. 2(52):52ra72PubMedCrossRef Hueber W, Patel DD, Dryja T, Wright AM, Koroleva I, Bruin G et al (2010) Effects of AIN457, a fully human antibody to interleukin-17A, on psoriasis, rheumatoid arthritis, and uveitis. Sci Transl Med. 2(52):52ra72PubMedCrossRef
200.
go back to reference Friedman S, Sanyal A, Goodman Z, Lefebvre E, Gottwald M, Fischer L et al (2016) Efficacy and safety study of cenicriviroc for the treatment of non-alcoholic steatohepatitis in adult subjects with liver fibrosis: CENTAUR Phase 2b study design. Contemp Clin Trials 47:356–365PubMedCrossRef Friedman S, Sanyal A, Goodman Z, Lefebvre E, Gottwald M, Fischer L et al (2016) Efficacy and safety study of cenicriviroc for the treatment of non-alcoholic steatohepatitis in adult subjects with liver fibrosis: CENTAUR Phase 2b study design. Contemp Clin Trials 47:356–365PubMedCrossRef
201.
go back to reference Seki E, De Minicis S, Gwak GY, Kluwe J, Inokuchi S, Bursill CA et al (2009) CCR1 and CCR5 promote hepatic fibrosis in mice. J Clin Invest 119(7):1858–1870PubMedPubMedCentral Seki E, De Minicis S, Gwak GY, Kluwe J, Inokuchi S, Bursill CA et al (2009) CCR1 and CCR5 promote hepatic fibrosis in mice. J Clin Invest 119(7):1858–1870PubMedPubMedCentral
202.
go back to reference Seki E, de Minicis S, Inokuchi S, Taura K, Miyai K, van Rooijen N et al (2009) CCR2 promotes hepatic fibrosis in mice. Hepatology 50(1):185–197PubMedCrossRef Seki E, de Minicis S, Inokuchi S, Taura K, Miyai K, van Rooijen N et al (2009) CCR2 promotes hepatic fibrosis in mice. Hepatology 50(1):185–197PubMedCrossRef
203.
go back to reference Mitchell C, Couton D, Couty JP, Anson M, Crain AM, Bizet V et al (2009) Dual role of CCR2 in the constitution and the resolution of liver fibrosis in mice. Am J Pathol 174(5):1766–1775PubMedPubMedCentralCrossRef Mitchell C, Couton D, Couty JP, Anson M, Crain AM, Bizet V et al (2009) Dual role of CCR2 in the constitution and the resolution of liver fibrosis in mice. Am J Pathol 174(5):1766–1775PubMedPubMedCentralCrossRef
204.
go back to reference Miura K, Yang L, van Rooijen N, Ohnishi H, Seki E (2012) Hepatic recruitment of macrophages promotes nonalcoholic steatohepatitis through CCR2. Am J Physiol Gastrointest Liver Physiol 302(11):G1310–G1321PubMedPubMedCentralCrossRef Miura K, Yang L, van Rooijen N, Ohnishi H, Seki E (2012) Hepatic recruitment of macrophages promotes nonalcoholic steatohepatitis through CCR2. Am J Physiol Gastrointest Liver Physiol 302(11):G1310–G1321PubMedPubMedCentralCrossRef
205.
go back to reference Berres ML, Koenen RR, Rueland A, Zaldivar MM, Heinrichs D, Sahin H et al (2010) Antagonism of the chemokine Ccl5 ameliorates experimental liver fibrosis in mice. J Clin Invest 120(11):4129–4140PubMedPubMedCentralCrossRef Berres ML, Koenen RR, Rueland A, Zaldivar MM, Heinrichs D, Sahin H et al (2010) Antagonism of the chemokine Ccl5 ameliorates experimental liver fibrosis in mice. J Clin Invest 120(11):4129–4140PubMedPubMedCentralCrossRef
206.
go back to reference Ling H, Roux E, Hempel D, Tao J, Smith M, Lonning S et al (2013) Transforming growth factor beta neutralization ameliorates pre-existing hepatic fibrosis and reduces cholangiocarcinoma in thioacetamide-treated rats. PLoS One 8(1):e54499PubMedPubMedCentralCrossRef Ling H, Roux E, Hempel D, Tao J, Smith M, Lonning S et al (2013) Transforming growth factor beta neutralization ameliorates pre-existing hepatic fibrosis and reduces cholangiocarcinoma in thioacetamide-treated rats. PLoS One 8(1):e54499PubMedPubMedCentralCrossRef
207.
go back to reference Lacouture ME, Morris JC, Lawrence DP, Tan AR, Olencki TE, Shapiro GI et al (2015) Cutaneous keratoacanthomas/squamous cell carcinomas associated with neutralization of transforming growth factor beta by the monoclonal antibody fresolimumab (GC1008). Cancer Immunol Immunother 64(4):437–446PubMedPubMedCentralCrossRef Lacouture ME, Morris JC, Lawrence DP, Tan AR, Olencki TE, Shapiro GI et al (2015) Cutaneous keratoacanthomas/squamous cell carcinomas associated with neutralization of transforming growth factor beta by the monoclonal antibody fresolimumab (GC1008). Cancer Immunol Immunother 64(4):437–446PubMedPubMedCentralCrossRef
208.
go back to reference Morris JC, Tan AR, Olencki TE, Shapiro GI, Dezube BJ, Reiss M et al (2014) Phase I study of GC1008 (fresolimumab): a human anti-transforming growth factor-beta (TGFbeta) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma. PLoS One 9(3):e90353PubMedPubMedCentralCrossRef Morris JC, Tan AR, Olencki TE, Shapiro GI, Dezube BJ, Reiss M et al (2014) Phase I study of GC1008 (fresolimumab): a human anti-transforming growth factor-beta (TGFbeta) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma. PLoS One 9(3):e90353PubMedPubMedCentralCrossRef
209.
go back to reference Trachtman H, Fervenza FC, Gipson DS, Heering P, Jayne DR, Peters H et al (2011) A phase 1, single-dose study of fresolimumab, an anti-TGF-beta antibody, in treatment-resistant primary focal segmental glomerulosclerosis. Kidney Int 79(11):1236–1243PubMedPubMedCentralCrossRef Trachtman H, Fervenza FC, Gipson DS, Heering P, Jayne DR, Peters H et al (2011) A phase 1, single-dose study of fresolimumab, an anti-TGF-beta antibody, in treatment-resistant primary focal segmental glomerulosclerosis. Kidney Int 79(11):1236–1243PubMedPubMedCentralCrossRef
210.
go back to reference Svegliati-Baroni G, Ridolfi F, Di Sario A, Casini A, Marucci L, Gaggiotti G et al (1999) Insulin and insulin-like growth factor-1 stimulate proliferation and type I collagen accumulation by human hepatic stellate cells: differential effects on signal transduction pathways. Hepatology 29(6):1743–1751PubMedCrossRef Svegliati-Baroni G, Ridolfi F, Di Sario A, Casini A, Marucci L, Gaggiotti G et al (1999) Insulin and insulin-like growth factor-1 stimulate proliferation and type I collagen accumulation by human hepatic stellate cells: differential effects on signal transduction pathways. Hepatology 29(6):1743–1751PubMedCrossRef
211.
go back to reference Ota T, Takamura T, Kurita S, Matsuzawa N, Kita Y, Uno M et al (2007) Insulin resistance accelerates a dietary rat model of nonalcoholic steatohepatitis. Gastroenterology 132(1):282–293PubMedCrossRef Ota T, Takamura T, Kurita S, Matsuzawa N, Kita Y, Uno M et al (2007) Insulin resistance accelerates a dietary rat model of nonalcoholic steatohepatitis. Gastroenterology 132(1):282–293PubMedCrossRef
212.
go back to reference Liang T, Zhang Q, Sun W, Xin Y, Zhang Z, Tan Y et al (2015) Zinc treatment prevents type 1 diabetes-induced hepatic oxidative damage, endoplasmic reticulum stress, and cell death, and even prevents possible steatohepatitis in the OVE26 mouse model: Important role of metallothionein. Toxicol Lett 233(2):114–124PubMedCrossRef Liang T, Zhang Q, Sun W, Xin Y, Zhang Z, Tan Y et al (2015) Zinc treatment prevents type 1 diabetes-induced hepatic oxidative damage, endoplasmic reticulum stress, and cell death, and even prevents possible steatohepatitis in the OVE26 mouse model: Important role of metallothionein. Toxicol Lett 233(2):114–124PubMedCrossRef
213.
go back to reference Masarone M, Rosato V, Aglitti A, Bucci T, Caruso R, Salvatore T et al (2017) Liver biopsy in type 2 diabetes mellitus: Steatohepatitis represents the sole feature of liver damage. PLoS One 12(6):e0178473PubMedPubMedCentralCrossRef Masarone M, Rosato V, Aglitti A, Bucci T, Caruso R, Salvatore T et al (2017) Liver biopsy in type 2 diabetes mellitus: Steatohepatitis represents the sole feature of liver damage. PLoS One 12(6):e0178473PubMedPubMedCentralCrossRef
214.
go back to reference Londos C, Honnor RC, Dhillon GS (1985) cAMP-dependent protein kinase and lipolysis in rat adipocytes. III. Multiple modes of insulin regulation of lipolysis and regulation of insulin responses by adenylate cyclase regulators. J Biol Chem 260(28):15139–45PubMedCrossRef Londos C, Honnor RC, Dhillon GS (1985) cAMP-dependent protein kinase and lipolysis in rat adipocytes. III. Multiple modes of insulin regulation of lipolysis and regulation of insulin responses by adenylate cyclase regulators. J Biol Chem 260(28):15139–45PubMedCrossRef
215.
go back to reference Boden G, She P, Mozzoli M, Cheung P, Gumireddy K, Reddy P et al (2005) Free fatty acids produce insulin resistance and activate the proinflammatory nuclear factor-kappaB pathway in rat liver. Diabetes 54(12):3458–3465PubMedCrossRef Boden G, She P, Mozzoli M, Cheung P, Gumireddy K, Reddy P et al (2005) Free fatty acids produce insulin resistance and activate the proinflammatory nuclear factor-kappaB pathway in rat liver. Diabetes 54(12):3458–3465PubMedCrossRef
216.
go back to reference Zhang J, Zhao Y, Xu C, Hong Y, Lu H, Wu J et al (2014) Association between serum free fatty acid levels and nonalcoholic fatty liver disease: a cross-sectional study. Sci Rep 4:5832PubMedPubMedCentralCrossRef Zhang J, Zhao Y, Xu C, Hong Y, Lu H, Wu J et al (2014) Association between serum free fatty acid levels and nonalcoholic fatty liver disease: a cross-sectional study. Sci Rep 4:5832PubMedPubMedCentralCrossRef
217.
go back to reference Crespo J, Cayon A, Fernandez-Gil P, Hernandez-Guerra M, Mayorga M, Dominguez-Diez A et al (2001) Gene expression of tumor necrosis factor alpha and TNF-receptors, p55 and p75, in nonalcoholic steatohepatitis patients. Hepatology 34(6):1158–1163PubMedCrossRef Crespo J, Cayon A, Fernandez-Gil P, Hernandez-Guerra M, Mayorga M, Dominguez-Diez A et al (2001) Gene expression of tumor necrosis factor alpha and TNF-receptors, p55 and p75, in nonalcoholic steatohepatitis patients. Hepatology 34(6):1158–1163PubMedCrossRef
218.
go back to reference Coulon S, Francque S, Colle I, Verrijken A, Blomme B, Heindryckx F et al (2012) Evaluation of inflammatory and angiogenic factors in patients with non-alcoholic fatty liver disease. Cytokine 59(2):442–449PubMedCrossRef Coulon S, Francque S, Colle I, Verrijken A, Blomme B, Heindryckx F et al (2012) Evaluation of inflammatory and angiogenic factors in patients with non-alcoholic fatty liver disease. Cytokine 59(2):442–449PubMedCrossRef
219.
go back to reference Sabio G, Das M, Mora A, Zhang Z, Jun JY, Ko HJ et al (2008) A stress signaling pathway in adipose tissue regulates hepatic insulin resistance. Science 322(5907):1539–1543PubMedPubMedCentralCrossRef Sabio G, Das M, Mora A, Zhang Z, Jun JY, Ko HJ et al (2008) A stress signaling pathway in adipose tissue regulates hepatic insulin resistance. Science 322(5907):1539–1543PubMedPubMedCentralCrossRef
221.
go back to reference Nadeau KJ, Klingensmith G, Zeitler P (2005) Type 2 diabetes in children is frequently associated with elevated alanine aminotransferase. J Pediatr Gastroenterol Nutr 41(1):94–98PubMedCrossRef Nadeau KJ, Klingensmith G, Zeitler P (2005) Type 2 diabetes in children is frequently associated with elevated alanine aminotransferase. J Pediatr Gastroenterol Nutr 41(1):94–98PubMedCrossRef
222.
go back to reference Burgert TS, Taksali SE, Dziura J, Goodman TR, Yeckel CW, Papademetris X et al (2006) Alanine aminotransferase levels and fatty liver in childhood obesity: associations with insulin resistance, adiponectin, and visceral fat. J Clin Endocrinol Metab 91(11):4287–4294PubMedCrossRef Burgert TS, Taksali SE, Dziura J, Goodman TR, Yeckel CW, Papademetris X et al (2006) Alanine aminotransferase levels and fatty liver in childhood obesity: associations with insulin resistance, adiponectin, and visceral fat. J Clin Endocrinol Metab 91(11):4287–4294PubMedCrossRef
223.
go back to reference Sheldon RD, Kanosky KM, Wells KD, Miles L, Perfield JW 2nd, Xanthakos S et al (2016) Transcriptomic differences in intra-abdominal adipose tissue in extremely obese adolescents with different stages of NAFLD. Physiol Genomics 48(12):897–911PubMedPubMedCentralCrossRef Sheldon RD, Kanosky KM, Wells KD, Miles L, Perfield JW 2nd, Xanthakos S et al (2016) Transcriptomic differences in intra-abdominal adipose tissue in extremely obese adolescents with different stages of NAFLD. Physiol Genomics 48(12):897–911PubMedPubMedCentralCrossRef
224.
go back to reference Joo E, Harada N, Yamane S, Fukushima T, Taura D, Iwasaki K et al (2017) Inhibition of gastric inhibitory polypeptide receptor signaling in adipose tissue reduces insulin resistance and hepatic steatosis in high-fat diet-fed mice. Diabetes 66(4):868–879PubMedCrossRef Joo E, Harada N, Yamane S, Fukushima T, Taura D, Iwasaki K et al (2017) Inhibition of gastric inhibitory polypeptide receptor signaling in adipose tissue reduces insulin resistance and hepatic steatosis in high-fat diet-fed mice. Diabetes 66(4):868–879PubMedCrossRef
225.
go back to reference Nie Y, Ma RC, Chan JC, Xu H, Xu G (2012) Glucose-dependent insulinotropic peptide impairs insulin signaling via inducing adipocyte inflammation in glucose-dependent insulinotropic peptide receptor-overexpressing adipocytes. FASEB J 26(6):2383–2393PubMedCrossRef Nie Y, Ma RC, Chan JC, Xu H, Xu G (2012) Glucose-dependent insulinotropic peptide impairs insulin signaling via inducing adipocyte inflammation in glucose-dependent insulinotropic peptide receptor-overexpressing adipocytes. FASEB J 26(6):2383–2393PubMedCrossRef
226.
go back to reference Timper K, Grisouard J, Sauter NS, Herzog-Radimerski T, Dembinski K, Peterli R et al (2013) Glucose-dependent insulinotropic polypeptide induces cytokine expression, lipolysis, and insulin resistance in human adipocytes. Am J Physiol Endocrinol Metab 304(1):E1-13PubMedCrossRef Timper K, Grisouard J, Sauter NS, Herzog-Radimerski T, Dembinski K, Peterli R et al (2013) Glucose-dependent insulinotropic polypeptide induces cytokine expression, lipolysis, and insulin resistance in human adipocytes. Am J Physiol Endocrinol Metab 304(1):E1-13PubMedCrossRef
227.
go back to reference Chen S, Okahara F, Osaki N, Shimotoyodome A (2015) Increased GIP signaling induces adipose inflammation via a HIF-1alpha-dependent pathway and impairs insulin sensitivity in mice. Am J Physiol Endocrinol Metab 308(5):E414–E425PubMedCrossRef Chen S, Okahara F, Osaki N, Shimotoyodome A (2015) Increased GIP signaling induces adipose inflammation via a HIF-1alpha-dependent pathway and impairs insulin sensitivity in mice. Am J Physiol Endocrinol Metab 308(5):E414–E425PubMedCrossRef
228.
go back to reference Gogebakan O, Osterhoff MA, Schuler R, Pivovarova O, Kruse M, Seltmann AC et al (2015) GIP increases adipose tissue expression and blood levels of MCP-1 in humans and links high energy diets to inflammation: a randomised trial. Diabetologia 58(8):1759–1768PubMedCrossRef Gogebakan O, Osterhoff MA, Schuler R, Pivovarova O, Kruse M, Seltmann AC et al (2015) GIP increases adipose tissue expression and blood levels of MCP-1 in humans and links high energy diets to inflammation: a randomised trial. Diabetologia 58(8):1759–1768PubMedCrossRef
229.
go back to reference Beaudry JL, Kaur KD, Varin EM, Baggio LL, Cao X, Mulvihill EE et al (2019) Physiological roles of the GIP receptor in murine brown adipose tissue. Mol Metab 28:14–25PubMedPubMedCentralCrossRef Beaudry JL, Kaur KD, Varin EM, Baggio LL, Cao X, Mulvihill EE et al (2019) Physiological roles of the GIP receptor in murine brown adipose tissue. Mol Metab 28:14–25PubMedPubMedCentralCrossRef
230.
go back to reference Fu Y, Kaneko K, Lin HY, Mo Q, Xu Y, Suganami T et al (2020) Gut hormone GIP induces inflammation and insulin resistance in the hypothalamus. Endocrinology. 161(9):bqaa102PubMedPubMedCentralCrossRef Fu Y, Kaneko K, Lin HY, Mo Q, Xu Y, Suganami T et al (2020) Gut hormone GIP induces inflammation and insulin resistance in the hypothalamus. Endocrinology. 161(9):bqaa102PubMedPubMedCentralCrossRef
231.
go back to reference Kaneko K, Fu Y, Lin HY, Cordonier EL, Mo Q, Gao Y et al (2019) Gut-derived GIP activates central Rap1 to impair neural leptin sensitivity during overnutrition. J Clin Invest 129(9):3786–3791PubMedPubMedCentralCrossRef Kaneko K, Fu Y, Lin HY, Cordonier EL, Mo Q, Gao Y et al (2019) Gut-derived GIP activates central Rap1 to impair neural leptin sensitivity during overnutrition. J Clin Invest 129(9):3786–3791PubMedPubMedCentralCrossRef
232.
go back to reference Campbell JE, Drucker DJ (2013) Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab 17(6):819–837PubMedCrossRef Campbell JE, Drucker DJ (2013) Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab 17(6):819–837PubMedCrossRef
233.
go back to reference Finan B, Muller TD, Clemmensen C, Perez-Tilve D, DiMarchi RD, Tschop MH (2016) Reappraisal of GIP pharmacology for metabolic diseases. Trends Mol Med 22(5):359–376PubMedCrossRef Finan B, Muller TD, Clemmensen C, Perez-Tilve D, DiMarchi RD, Tschop MH (2016) Reappraisal of GIP pharmacology for metabolic diseases. Trends Mol Med 22(5):359–376PubMedCrossRef
234.
go back to reference Kim SJ, Nian C, Karunakaran S, Clee SM, Isales CM, McIntosh CH (2012) GIP-overexpressing mice demonstrate reduced diet-induced obesity and steatosis, and improved glucose homeostasis. PLoS One 7(7):e40156PubMedPubMedCentralCrossRef Kim SJ, Nian C, Karunakaran S, Clee SM, Isales CM, McIntosh CH (2012) GIP-overexpressing mice demonstrate reduced diet-induced obesity and steatosis, and improved glucose homeostasis. PLoS One 7(7):e40156PubMedPubMedCentralCrossRef
235.
go back to reference Miyawaki K, Yamada Y, Ban N, Ihara Y, Tsukiyama K, Zhou H et al (2002) Inhibition of gastric inhibitory polypeptide signaling prevents obesity. Nat Med 8(7):738–742PubMedCrossRef Miyawaki K, Yamada Y, Ban N, Ihara Y, Tsukiyama K, Zhou H et al (2002) Inhibition of gastric inhibitory polypeptide signaling prevents obesity. Nat Med 8(7):738–742PubMedCrossRef
236.
go back to reference Bates HE, Campbell JE, Ussher JR, Baggio LL, Maida A, Seino Y et al (2012) Gipr is essential for adrenocortical steroidogenesis; however, corticosterone deficiency does not mediate the favorable metabolic phenotype of Gipr(-/-) mice. Diabetes 61(1):40–48PubMedCrossRef Bates HE, Campbell JE, Ussher JR, Baggio LL, Maida A, Seino Y et al (2012) Gipr is essential for adrenocortical steroidogenesis; however, corticosterone deficiency does not mediate the favorable metabolic phenotype of Gipr(-/-) mice. Diabetes 61(1):40–48PubMedCrossRef
237.
go back to reference Hansotia T, Maida A, Flock G, Yamada Y, Tsukiyama K, Seino Y et al (2007) Extrapancreatic incretin receptors modulate glucose homeostasis, body weight, and energy expenditure. J Clin Invest 117(1):143–152PubMedCrossRef Hansotia T, Maida A, Flock G, Yamada Y, Tsukiyama K, Seino Y et al (2007) Extrapancreatic incretin receptors modulate glucose homeostasis, body weight, and energy expenditure. J Clin Invest 117(1):143–152PubMedCrossRef
238.
go back to reference Nasteska D, Harada N, Suzuki K, Yamane S, Hamasaki A, Joo E et al (2014) Chronic reduction of GIP secretion alleviates obesity and insulin resistance under high-fat diet conditions. Diabetes 63(7):2332–2343PubMedCrossRef Nasteska D, Harada N, Suzuki K, Yamane S, Hamasaki A, Joo E et al (2014) Chronic reduction of GIP secretion alleviates obesity and insulin resistance under high-fat diet conditions. Diabetes 63(7):2332–2343PubMedCrossRef
239.
go back to reference Campbell JE, Ussher JR, Mulvihill EE, Kolic J, Baggio LL, Cao X et al (2016) TCF1 links GIPR signaling to the control of beta cell function and survival. Nat Med 22(1):84–90PubMedCrossRef Campbell JE, Ussher JR, Mulvihill EE, Kolic J, Baggio LL, Cao X et al (2016) TCF1 links GIPR signaling to the control of beta cell function and survival. Nat Med 22(1):84–90PubMedCrossRef
240.
go back to reference Althage MC, Ford EL, Wang S, Tso P, Polonsky KS, Wice BM (2008) Targeted ablation of glucose-dependent insulinotropic polypeptide-producing cells in transgenic mice reduces obesity and insulin resistance induced by a high fat diet. J Biol Chem 283(26):18365–18376PubMedPubMedCentralCrossRef Althage MC, Ford EL, Wang S, Tso P, Polonsky KS, Wice BM (2008) Targeted ablation of glucose-dependent insulinotropic polypeptide-producing cells in transgenic mice reduces obesity and insulin resistance induced by a high fat diet. J Biol Chem 283(26):18365–18376PubMedPubMedCentralCrossRef
241.
go back to reference Killion EA, Wang J, Yie J, Shi SD, Bates D, Min X et al (2018) Anti-obesity effects of GIPR antagonists alone and in combination with GLP-1R agonists in preclinical models. Sci Transl Med. 10(472):eaat3392PubMedCrossRef Killion EA, Wang J, Yie J, Shi SD, Bates D, Min X et al (2018) Anti-obesity effects of GIPR antagonists alone and in combination with GLP-1R agonists in preclinical models. Sci Transl Med. 10(472):eaat3392PubMedCrossRef
242.
go back to reference Mantelmacher FD, Fishman S, Cohen K, Pasmanik Chor M, Yamada Y, Zvibel I et al (2017) Glucose-dependent insulinotropic polypeptide receptor deficiency leads to impaired bone marrow hematopoiesis. J Immunol 198(8):3089–3098PubMedCrossRef Mantelmacher FD, Fishman S, Cohen K, Pasmanik Chor M, Yamada Y, Zvibel I et al (2017) Glucose-dependent insulinotropic polypeptide receptor deficiency leads to impaired bone marrow hematopoiesis. J Immunol 198(8):3089–3098PubMedCrossRef
243.
go back to reference Pujadas G, Varin EM, Baggio LL, Mulvihill EE, Bang KWA, Koehler JA et al (2020) The gut hormone receptor GIPR links energy availability to the control of hematopoiesis. Mol Metab 39:101008PubMedPubMedCentralCrossRef Pujadas G, Varin EM, Baggio LL, Mulvihill EE, Bang KWA, Koehler JA et al (2020) The gut hormone receptor GIPR links energy availability to the control of hematopoiesis. Mol Metab 39:101008PubMedPubMedCentralCrossRef
244.
go back to reference Ravn P, Madhurantakam C, Kunze S, Matthews E, Priest C, O’Brien S et al (2013) Structural and pharmacological characterization of novel potent and selective monoclonal antibody antagonists of glucose-dependent insulinotropic polypeptide receptor. J Biol Chem 288(27):19760–19772PubMedPubMedCentralCrossRef Ravn P, Madhurantakam C, Kunze S, Matthews E, Priest C, O’Brien S et al (2013) Structural and pharmacological characterization of novel potent and selective monoclonal antibody antagonists of glucose-dependent insulinotropic polypeptide receptor. J Biol Chem 288(27):19760–19772PubMedPubMedCentralCrossRef
245.
go back to reference Miyawaki K, Yamada Y, Yano H, Niwa H, Ban N, Ihara Y et al (1999) Glucose intolerance caused by a defect in the entero-insular axis: a study in gastric inhibitory polypeptide receptor knockout mice. Proc Natl Acad Sci U S A 96(26):14843–14847PubMedPubMedCentralCrossRef Miyawaki K, Yamada Y, Yano H, Niwa H, Ban N, Ihara Y et al (1999) Glucose intolerance caused by a defect in the entero-insular axis: a study in gastric inhibitory polypeptide receptor knockout mice. Proc Natl Acad Sci U S A 96(26):14843–14847PubMedPubMedCentralCrossRef
246.
go back to reference Goralska J, Razny U, Polus A, Stancel-Mozwillo J, Chojnacka M, Gruca A et al (2018) Pro-inflammatory gene expression profile in obese adults with high plasma GIP levels. Int J Obes (Lond) 42(4):826–834PubMedCrossRef Goralska J, Razny U, Polus A, Stancel-Mozwillo J, Chojnacka M, Gruca A et al (2018) Pro-inflammatory gene expression profile in obese adults with high plasma GIP levels. Int J Obes (Lond) 42(4):826–834PubMedCrossRef
247.
go back to reference Jorsal T, Rhee NA, Pedersen J, Wahlgren CD, Mortensen B, Jepsen SL et al (2018) Enteroendocrine K and L cells in healthy and type 2 diabetic individuals. Diabetologia 61(2):284–294PubMedCrossRef Jorsal T, Rhee NA, Pedersen J, Wahlgren CD, Mortensen B, Jepsen SL et al (2018) Enteroendocrine K and L cells in healthy and type 2 diabetic individuals. Diabetologia 61(2):284–294PubMedCrossRef
248.
go back to reference Hammoud R, Drucker DJ (2023) Beyond the pancreas: contrasting cardiometabolic actions of GIP and GLP1. Nat Rev Endocrinol 19(4):201–216PubMedCrossRef Hammoud R, Drucker DJ (2023) Beyond the pancreas: contrasting cardiometabolic actions of GIP and GLP1. Nat Rev Endocrinol 19(4):201–216PubMedCrossRef
249.
go back to reference Drucker DJ, Philippe J, Mojsov S, Chick WL, Habener JF (1987) Glucagon-like peptide I stimulates insulin gene expression and increases cyclic AMP levels in a rat islet cell line. Proc Natl Acad Sci U S A 84(10):3434–3438PubMedPubMedCentralCrossRef Drucker DJ, Philippe J, Mojsov S, Chick WL, Habener JF (1987) Glucagon-like peptide I stimulates insulin gene expression and increases cyclic AMP levels in a rat islet cell line. Proc Natl Acad Sci U S A 84(10):3434–3438PubMedPubMedCentralCrossRef
250.
go back to reference Arakawa M, Ebato C, Mita T, Hirose T, Kawamori R, Fujitani Y et al (2009) Effects of exendin-4 on glucose tolerance, insulin secretion, and beta-cell proliferation depend on treatment dose, treatment duration and meal contents. Biochem Biophys Res Commun 390(3):809–814PubMedCrossRef Arakawa M, Ebato C, Mita T, Hirose T, Kawamori R, Fujitani Y et al (2009) Effects of exendin-4 on glucose tolerance, insulin secretion, and beta-cell proliferation depend on treatment dose, treatment duration and meal contents. Biochem Biophys Res Commun 390(3):809–814PubMedCrossRef
251.
go back to reference Kawamori D, Shirakawa J, Liew CW, Hu J, Morioka T, Duttaroy A et al (2017) GLP-1 signalling compensates for impaired insulin signalling in regulating beta cell proliferation in betaIRKO mice. Diabetologia 60(8):1442–1453PubMedPubMedCentralCrossRef Kawamori D, Shirakawa J, Liew CW, Hu J, Morioka T, Duttaroy A et al (2017) GLP-1 signalling compensates for impaired insulin signalling in regulating beta cell proliferation in betaIRKO mice. Diabetologia 60(8):1442–1453PubMedPubMedCentralCrossRef
252.
go back to reference Park S, Dong X, Fisher TL, Dunn S, Omer AK, Weir G et al (2006) Exendin-4 uses Irs2 signaling to mediate pancreatic beta cell growth and function. J Biol Chem 281(2):1159–1168PubMedCrossRef Park S, Dong X, Fisher TL, Dunn S, Omer AK, Weir G et al (2006) Exendin-4 uses Irs2 signaling to mediate pancreatic beta cell growth and function. J Biol Chem 281(2):1159–1168PubMedCrossRef
253.
go back to reference Buteau J, El-Assaad W, Rhodes CJ, Rosenberg L, Joly E, Prentki M (2004) Glucagon-like peptide-1 prevents beta cell glucolipotoxicity. Diabetologia 47(5):806–815PubMedCrossRef Buteau J, El-Assaad W, Rhodes CJ, Rosenberg L, Joly E, Prentki M (2004) Glucagon-like peptide-1 prevents beta cell glucolipotoxicity. Diabetologia 47(5):806–815PubMedCrossRef
254.
go back to reference Ellingsgaard H, Hauselmann I, Schuler B, Habib AM, Baggio LL, Meier DT et al (2011) Interleukin-6 enhances insulin secretion by increasing glucagon-like peptide-1 secretion from L cells and alpha cells. Nat Med 17(11):1481–1489PubMedPubMedCentralCrossRef Ellingsgaard H, Hauselmann I, Schuler B, Habib AM, Baggio LL, Meier DT et al (2011) Interleukin-6 enhances insulin secretion by increasing glucagon-like peptide-1 secretion from L cells and alpha cells. Nat Med 17(11):1481–1489PubMedPubMedCentralCrossRef
255.
go back to reference Lebrun LJ, Lenaerts K, Kiers D, Pais de Barros JP, Le Guern N, Plesnik J et al (2017) Enteroendocrine L Cells Sense LPS after Gut Barrier Injury to Enhance GLP-1 Secretion. Cell Rep. 21(5):1160–8PubMedCrossRef Lebrun LJ, Lenaerts K, Kiers D, Pais de Barros JP, Le Guern N, Plesnik J et al (2017) Enteroendocrine L Cells Sense LPS after Gut Barrier Injury to Enhance GLP-1 Secretion. Cell Rep. 21(5):1160–8PubMedCrossRef
256.
go back to reference Chimerel C, Emery E, Summers DK, Keyser U, Gribble FM, Reimann F (2014) Bacterial metabolite indole modulates incretin secretion from intestinal enteroendocrine L cells. Cell Rep 9(4):1202–1208PubMedPubMedCentralCrossRef Chimerel C, Emery E, Summers DK, Keyser U, Gribble FM, Reimann F (2014) Bacterial metabolite indole modulates incretin secretion from intestinal enteroendocrine L cells. Cell Rep 9(4):1202–1208PubMedPubMedCentralCrossRef
257.
go back to reference Breton J, Tennoune N, Lucas N, Francois M, Legrand R, Jacquemot J et al (2016) Gut commensal E. coli proteins activate host satiety pathways following nutrient-induced bacterial growth. Cell Metab. 23(2):324–34PubMedCrossRef Breton J, Tennoune N, Lucas N, Francois M, Legrand R, Jacquemot J et al (2016) Gut commensal E. coli proteins activate host satiety pathways following nutrient-induced bacterial growth. Cell Metab. 23(2):324–34PubMedCrossRef
258.
go back to reference Brakenridge SC, Moore FA, Mercier NR, Cox M, Wu Q, Moldawer LL et al (2019) Persistently elesvated glucagon-like peptide-1 levels among critically Ill surgical patients after sepsis and development of chronic critical illness and dismal long-term outcomes. J Am Coll Surg. 229(1):58-67.e1PubMedPubMedCentralCrossRef Brakenridge SC, Moore FA, Mercier NR, Cox M, Wu Q, Moldawer LL et al (2019) Persistently elesvated glucagon-like peptide-1 levels among critically Ill surgical patients after sepsis and development of chronic critical illness and dismal long-term outcomes. J Am Coll Surg. 229(1):58-67.e1PubMedPubMedCentralCrossRef
259.
go back to reference Lebherz C, Schlieper G, Mollmann J, Kahles F, Schwarz M, Brunsing J et al (2017) GLP-1 levels predict mortality in patients with critical illness as well as end-stage renal disease. Am J Med. 130(7):833–41.e3PubMedCrossRef Lebherz C, Schlieper G, Mollmann J, Kahles F, Schwarz M, Brunsing J et al (2017) GLP-1 levels predict mortality in patients with critical illness as well as end-stage renal disease. Am J Med. 130(7):833–41.e3PubMedCrossRef
260.
go back to reference Wong CK, Yusta B, Koehler JA, Baggio LL, McLean BA, Matthews D et al (2022) Divergent roles for the gut intraepithelial lymphocyte GLP-1R in control of metabolism, microbiota, and T cell-induced inflammation. Cell Metab. 34(10):1514–31.e7PubMedCrossRef Wong CK, Yusta B, Koehler JA, Baggio LL, McLean BA, Matthews D et al (2022) Divergent roles for the gut intraepithelial lymphocyte GLP-1R in control of metabolism, microbiota, and T cell-induced inflammation. Cell Metab. 34(10):1514–31.e7PubMedCrossRef
261.
go back to reference Noyan-Ashraf MH, Shikatani EA, Schuiki I, Mukovozov I, Wu J, Li RK et al (2013) A glucagon-like peptide-1 analog reverses the molecular pathology and cardiac dysfunction of a mouse model of obesity. Circulation 127(1):74–85PubMedCrossRef Noyan-Ashraf MH, Shikatani EA, Schuiki I, Mukovozov I, Wu J, Li RK et al (2013) A glucagon-like peptide-1 analog reverses the molecular pathology and cardiac dysfunction of a mouse model of obesity. Circulation 127(1):74–85PubMedCrossRef
262.
go back to reference Drucker DJ (2018) Mechanisms of action and therapeutic application of glucagon-like peptide-1. Cell Metab 27(4):740–756PubMedCrossRef Drucker DJ (2018) Mechanisms of action and therapeutic application of glucagon-like peptide-1. Cell Metab 27(4):740–756PubMedCrossRef
263.
go back to reference Trevaskis JL, Griffin PS, Wittmer C, Neuschwander-Tetri BA, Brunt EM, Dolman CS et al (2012) Glucagon-like peptide-1 receptor agonism improves metabolic, biochemical, and histopathological indices of nonalcoholic steatohepatitis in mice. Am J Physiol Gastrointest Liver Physiol 302(8):G762–G772PubMedCrossRef Trevaskis JL, Griffin PS, Wittmer C, Neuschwander-Tetri BA, Brunt EM, Dolman CS et al (2012) Glucagon-like peptide-1 receptor agonism improves metabolic, biochemical, and histopathological indices of nonalcoholic steatohepatitis in mice. Am J Physiol Gastrointest Liver Physiol 302(8):G762–G772PubMedCrossRef
264.
go back to reference Somm E, Montandon SA, Loizides-Mangold U, Gaia N, Lazarevic V, De Vito C et al (2021) The GLP-1R agonist liraglutide limits hepatic lipotoxicity and inflammatory response in mice fed a methionine-choline deficient diet. Transl Res 227:75–88PubMedCrossRef Somm E, Montandon SA, Loizides-Mangold U, Gaia N, Lazarevic V, De Vito C et al (2021) The GLP-1R agonist liraglutide limits hepatic lipotoxicity and inflammatory response in mice fed a methionine-choline deficient diet. Transl Res 227:75–88PubMedCrossRef
265.
go back to reference McLean BA, Wong CK, Kaur KD, Seeley RJ, Drucker DJ (2021) Differential importance of endothelial and hematopoietic cell GLP-1Rs for cardiometabolic versus hepatic actions of semaglutide. JCI Insight 6(22):e153732PubMedPubMedCentralCrossRef McLean BA, Wong CK, Kaur KD, Seeley RJ, Drucker DJ (2021) Differential importance of endothelial and hematopoietic cell GLP-1Rs for cardiometabolic versus hepatic actions of semaglutide. JCI Insight 6(22):e153732PubMedPubMedCentralCrossRef
266.
go back to reference Newsome PN, Buchholtz K, Cusi K, Linder M, Okanoue T, Ratziu V et al (2021) A placebo-controlled trial of subcutaneous semaglutide in nonalcoholic steatohepatitis. N Engl J Med 384(12):1113–1124PubMedCrossRef Newsome PN, Buchholtz K, Cusi K, Linder M, Okanoue T, Ratziu V et al (2021) A placebo-controlled trial of subcutaneous semaglutide in nonalcoholic steatohepatitis. N Engl J Med 384(12):1113–1124PubMedCrossRef
267.
go back to reference Armstrong MJ, Gaunt P, Aithal GP, Barton D, Hull D, Parker R et al (2016) Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet 387(10019):679–690PubMedCrossRef Armstrong MJ, Gaunt P, Aithal GP, Barton D, Hull D, Parker R et al (2016) Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet 387(10019):679–690PubMedCrossRef
268.
go back to reference Nystrom T, Gutniak MK, Zhang Q, Zhang F, Holst JJ, Ahren B et al (2004) Effects of glucagon-like peptide-1 on endothelial function in type 2 diabetes patients with stable coronary artery disease. Am J Physiol Endocrinol Metab 287(6):E1209–E1215PubMedCrossRef Nystrom T, Gutniak MK, Zhang Q, Zhang F, Holst JJ, Ahren B et al (2004) Effects of glucagon-like peptide-1 on endothelial function in type 2 diabetes patients with stable coronary artery disease. Am J Physiol Endocrinol Metab 287(6):E1209–E1215PubMedCrossRef
269.
go back to reference Wei Y, Mojsov S (1995) Tissue-specific expression of the human receptor for glucagon-like peptide-I: brain, heart and pancreatic forms have the same deduced amino acid sequences. FEBS Lett 358(3):219–224PubMedCrossRef Wei Y, Mojsov S (1995) Tissue-specific expression of the human receptor for glucagon-like peptide-I: brain, heart and pancreatic forms have the same deduced amino acid sequences. FEBS Lett 358(3):219–224PubMedCrossRef
270.
go back to reference Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA et al (2016) Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med 375(4):311–322PubMedPubMedCentralCrossRef Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA et al (2016) Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med 375(4):311–322PubMedPubMedCentralCrossRef
271.
go back to reference Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jodar E, Leiter LA et al (2016) Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 375(19):1834–1844PubMedCrossRef Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jodar E, Leiter LA et al (2016) Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 375(19):1834–1844PubMedCrossRef
272.
go back to reference Ammar RA, Mohamed AF, Kamal MM, Safar MM, Abdelkader NF (2022) Neuroprotective effect of liraglutide in an experimental mouse model of multiple sclerosis: role of AMPK/SIRT1 signaling and NLRP3 inflammasome. Inflammopharmacology 30(3):919–934PubMedPubMedCentralCrossRef Ammar RA, Mohamed AF, Kamal MM, Safar MM, Abdelkader NF (2022) Neuroprotective effect of liraglutide in an experimental mouse model of multiple sclerosis: role of AMPK/SIRT1 signaling and NLRP3 inflammasome. Inflammopharmacology 30(3):919–934PubMedPubMedCentralCrossRef
273.
go back to reference Song S, Guo R, Mehmood A, Zhang L, Yin B, Yuan C et al (2022) Liraglutide attenuate central nervous inflammation and demyelination through AMPK and pyroptosis-related NLRP3 pathway. CNS Neurosci Ther 28(3):422–434PubMedPubMedCentralCrossRef Song S, Guo R, Mehmood A, Zhang L, Yin B, Yuan C et al (2022) Liraglutide attenuate central nervous inflammation and demyelination through AMPK and pyroptosis-related NLRP3 pathway. CNS Neurosci Ther 28(3):422–434PubMedPubMedCentralCrossRef
274.
go back to reference Svendsen B, Capozzi ME, Nui J, Hannou SA, Finan B, Naylor J et al (2020) Pharmacological antagonism of the incretin system protects against diet-induced obesity. Mol Metab 32:44–55PubMedCrossRef Svendsen B, Capozzi ME, Nui J, Hannou SA, Finan B, Naylor J et al (2020) Pharmacological antagonism of the incretin system protects against diet-induced obesity. Mol Metab 32:44–55PubMedCrossRef
275.
go back to reference Frias JP, Nauck MA, Van J, Kutner ME, Cui X, Benson C et al (2018) Efficacy and safety of LY3298176, a novel dual GIP and GLP-1 receptor agonist, in patients with type 2 diabetes: a randomised, placebo-controlled and active comparator-controlled phase 2 trial. Lancet 392(10160):2180–2193PubMedCrossRef Frias JP, Nauck MA, Van J, Kutner ME, Cui X, Benson C et al (2018) Efficacy and safety of LY3298176, a novel dual GIP and GLP-1 receptor agonist, in patients with type 2 diabetes: a randomised, placebo-controlled and active comparator-controlled phase 2 trial. Lancet 392(10160):2180–2193PubMedCrossRef
276.
go back to reference Finan B, Ma T, Ottaway N, Muller TD, Habegger KM, Heppner KM et al (2013) Unimolecular dual incretins maximize metabolic benefits in rodents, monkeys, and humans. Sci Transl Med. 5(209):209ra151PubMedCrossRef Finan B, Ma T, Ottaway N, Muller TD, Habegger KM, Heppner KM et al (2013) Unimolecular dual incretins maximize metabolic benefits in rodents, monkeys, and humans. Sci Transl Med. 5(209):209ra151PubMedCrossRef
277.
go back to reference Coskun T, Sloop KW, Loghin C, Alsina-Fernandez J, Urva S, Bokvist KB et al (2018) LY3298176, a novel dual GIP and GLP-1 receptor agonist for the treatment of type 2 diabetes mellitus: from discovery to clinical proof of concept. Mol Metab 18:3–14PubMedPubMedCentralCrossRef Coskun T, Sloop KW, Loghin C, Alsina-Fernandez J, Urva S, Bokvist KB et al (2018) LY3298176, a novel dual GIP and GLP-1 receptor agonist for the treatment of type 2 diabetes mellitus: from discovery to clinical proof of concept. Mol Metab 18:3–14PubMedPubMedCentralCrossRef
278.
go back to reference Mroz PA, Finan B, Gelfanov V, Yang B, Tschop MH, DiMarchi RD et al (2019) Optimized GIP analogs promote body weight lowering in mice through GIPR agonism not antagonism. Mol Metab 20:51–62PubMedCrossRef Mroz PA, Finan B, Gelfanov V, Yang B, Tschop MH, DiMarchi RD et al (2019) Optimized GIP analogs promote body weight lowering in mice through GIPR agonism not antagonism. Mol Metab 20:51–62PubMedCrossRef
279.
go back to reference Jastreboff AM, Aronne LJ, Ahmad NN, Wharton S, Connery L, Alves B et al (2022) Tirzepatide once weekly for the treatment of obesity. N Engl J Med 387(3):205–216PubMedCrossRef Jastreboff AM, Aronne LJ, Ahmad NN, Wharton S, Connery L, Alves B et al (2022) Tirzepatide once weekly for the treatment of obesity. N Engl J Med 387(3):205–216PubMedCrossRef
280.
go back to reference Gastaldelli A, Cusi K, Fernandez Lando L, Bray R, Brouwers B, Rodriguez A (2022) Effect of tirzepatide versus insulin degludec on liver fat content and abdominal adipose tissue in people with type 2 diabetes (SURPASS-3 MRI): a substudy of the randomised, open-label, parallel-group, phase 3 SURPASS-3 trial. Lancet Diabetes Endocrinol 10(6):393–406PubMedCrossRef Gastaldelli A, Cusi K, Fernandez Lando L, Bray R, Brouwers B, Rodriguez A (2022) Effect of tirzepatide versus insulin degludec on liver fat content and abdominal adipose tissue in people with type 2 diabetes (SURPASS-3 MRI): a substudy of the randomised, open-label, parallel-group, phase 3 SURPASS-3 trial. Lancet Diabetes Endocrinol 10(6):393–406PubMedCrossRef
281.
go back to reference Li T, Jiao JJ, Su Q, Holscher C, Zhang J, Yan XD et al (2020) A GLP-1/GIP/Gcg receptor triagonist improves memory behavior, as well as synaptic transmission, neuronal excitability and Ca(2+) homeostasis in 3xTg-AD mice. Neuropharmacology 170:108042PubMedCrossRef Li T, Jiao JJ, Su Q, Holscher C, Zhang J, Yan XD et al (2020) A GLP-1/GIP/Gcg receptor triagonist improves memory behavior, as well as synaptic transmission, neuronal excitability and Ca(2+) homeostasis in 3xTg-AD mice. Neuropharmacology 170:108042PubMedCrossRef
282.
go back to reference Kleber M, Lass N, Papcke S, Wabitsch M, Reinehr T (2010) One-year follow-up of untreated obese white children and adolescents with impaired glucose tolerance: high conversion rate to normal glucose tolerance. Diabet Med 27(5):516–521PubMedCrossRef Kleber M, Lass N, Papcke S, Wabitsch M, Reinehr T (2010) One-year follow-up of untreated obese white children and adolescents with impaired glucose tolerance: high conversion rate to normal glucose tolerance. Diabet Med 27(5):516–521PubMedCrossRef
283.
go back to reference Feldstein AE, Charatcharoenwitthaya P, Treeprasertsuk S, Benson JT, Enders FB, Angulo P (2009) The natural history of non-alcoholic fatty liver disease in children: a follow-up study for up to 20 years. Gut 58(11):1538–1544PubMedCrossRef Feldstein AE, Charatcharoenwitthaya P, Treeprasertsuk S, Benson JT, Enders FB, Angulo P (2009) The natural history of non-alcoholic fatty liver disease in children: a follow-up study for up to 20 years. Gut 58(11):1538–1544PubMedCrossRef
Metadata
Title
Relevance and consequence of chronic inflammation for obesity development
Authors
Lisa Ruck
Susanna Wiegand
Peter Kühnen
Publication date
14-11-2023
Publisher
Springer International Publishing
Published in
Molecular and Cellular Pediatrics / Issue 1/2023
Electronic ISSN: 2194-7791
DOI
https://doi.org/10.1186/s40348-023-00170-6

Other articles of this Issue 1/2023

Molecular and Cellular Pediatrics 1/2023 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

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

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

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

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

At a glance: The STEP trials

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

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

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

Year in Review: Pulmonary vascular disease

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

Year in Review: Valvular heart disease

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

Year in Review: Heart failure and cardiomyopathies

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