Resolution to Inflammation: Its Role in Reducing Fibrosis and Tissue Repair

Anna Meiliana, Andi Wijaya

Abstract


BACKGROUND: Many chronic disorders, including vascular diseases, metabolic syndrome, and neurological diseases, are known to share a common factor of excessive inflammation. It takes a lot of energy to heal damaged tissue, which is a complicated process. The outcome is often suboptimal, with some degree of fibrosis, depending on the tissue's ability to regenerate and the strength of the inflammatory response. We may get new insights into disease causation and therapeutic strategies by better understanding endogenous regulatory points within the inflammatory response.

CONTENT: Despite of the benefit in raising an inflammatory response, it also have unfavourable effects. Unresolved inflammation can over accumulate collagenous connective tissue and induce fibrosis, promote tissue dysfunction, and finally organ failure. Currently, the resolution of inflammation was described in terms of contemporary molecules as a different mechanisms from anti-inflammatory, since in resolution, the pathogen and apoptotic cells crumbs will be cleared and the macrophages will set back the tissue homeostasis. An active transition in the mediators that predominate in exudates occurs in conjunction with the remission of inflammation. These groups of inborn pro-resolution named resolvins, maresins, and protectins work to reduce inflammation by triggering certain pathways that support homeostasis rather than by suppressing the immune system.

SUMMARY: The resolution of inflammation, once believed to be a passive process, is now understood to entail active biochemical programs that allow inflamed tissues to regain equilibrium. In this review, we spotlight the resolution to inflammation as a strategy to prevent tissue fibrosis and hinder the organ damage.

KEYWORDS: inflammation, resolution, fibrosis, wound healing, specialized pro-resolving mediators


Full Text:

PDF

References


Alfaddagh A, Martin SS, Leucker TM, Michos ED, Blaha MJ, Lowenstein CJ, et al. Inflammation and cardiovascular disease: From mechanisms to therapeutics. Am J Prev Cardiol. 2020; 4: 100130, CrossRef.

Degan D, Ornello R, Tiseo C, Carolei A, Sacco S, Pistoia F. The role of inflammation in neurological disorders. Curr Pharm Des. 2018; 24(14): 1485–501, CrossRef.

Fullerton JN, Gilroy DW. Resolution of inflammation: a new therapeutic frontier. Nat Rev Drug Discov. 2016; 15(8): 551–67, CrossRef.

Wynn TA. Fibrotic disease and the T(H)1/T(H)2 paradigm. Nat Rev Immunol. 2004; 4(8): 583–94, CrossRef.

Wynn T. Cellular and molecular mechanisms of fibrosis. J Pathol. 2008; 214(2): 199–210, CrossRef.

Nanthakumar CB, Hatley RJD, Lemma S, Gauldie J, Marshall RP, Macdonald SJF. Dissecting fibrosis: therapeutic insights from the small-molecule toolbox. Nat Rev Drug Discov. 2015; 14(10): 693–720, CrossRef.

Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature. 2013; 493(7434): 674–8, CrossRef.

Wynn TA, Vannella KM. Macrophages in tissue repair, regeneration, and fibrosis. Immunity. 2016; 44(3): 450–62, CrossRef.

Murray PJ. Macrophage Polarization. Annu Rev Physiol. 2017; 79(1): 541–66, CrossRef.

Ho KJ, Spite M, Owens CD, Lancero H, Kroemer AHK, Pande R, et al. Aspirin-triggered lipoxin and resolvin E1 modulate vascular smooth muscle phenotype and correlate with peripheral atherosclerosis. Am J Pathol. 2010; 177(4): 2116–23, CrossRef.

Dalli J, Colas RA, Quintana C, Barragan-Bradford D, Hurwitz S, Levy BD, et al. Human sepsis eicosanoid and proresolving lipid mediator temporal profiles: Correlations with survival and clinical outcomes. Crit Care Med. 2017; 45(1): 58–68, CrossRef.

Serhan CN, Chiang N, Dalli J. New pro-resolving n-3 mediators bridge resolution of infectious inflammation to tissue regeneration. Mol Aspects Med. 2018; 64: 1–17, CrossRef.

Calder PC. Marine omega-3 fatty acids and inflammatory processes: Effects, mechanisms and clinical relevance. Biochim Biophys Acta. 2015; 1851(4): 469–84, CrossRef.

Abbas AK, Aster JC, Kumar V. Robbins and Cotran Pathologic Basis of Disease. 4th Edition. Philadelphia: Elsevier Saunders; 2015, NLMID.

Libby P. Atherosclerosis: the new view. Sci Am. 2002; 286(5): 46–55, CrossRef.

Hansson GK, Libby P. The immune response in atherosclerosis: a double-edged sword. Nat Rev Immunol. 2006; 6(7): 508–19, CrossRef.

Serhan CN, Clish CB, Brannon J, Colgan SP, Chiang N, Gronert K. Novel functional sets of lipid-derived mediators with antiinflammatory actions generated from omega-3 fatty acids via cyclooxygenase 2-nonsteroidal antiinflammatory drugs and transcellular processing. J Exp Med. 2000; 192(8): 1197–204, CrossRef.

Levy BD, Clish CB, Schmidt B, Gronert K, Serhan CN. Lipid mediator class switching during acute inflammation: signals in resolution. Nat Immunol. 2001; 2(7): 612–9, CrossRef.

Lawrence T, Willoughby DA, Gilroy DW. Anti-inflammatory lipid mediators and insights into the resolution of inflammation. Nat Rev Immunol. 2002; 2(10): 787–95, CrossRef.

Serhan CN, Hong S, Gronert K, Colgan SP, Devchand PR, Mirick G, et al. Resolvins: a family of bioactive products of omega-3 fatty acid transformation circuits initiated by aspirin treatment that counter proinflammation signals. J Exp Med. 2002; 196(8): 1025–37, CrossRef.

Serhan CN, Petasis NA. Resolvins and protectins in inflammation resolution. Chem Rev. 2011; 111(10): 5922–43, CrossRef.

Schuppan D, Ruehl M, Somasundaram R, Hahn EG. Matrix as a modulator of hepatic fibrogenesis. Semin Liver Dis. 2001; 21(3): 351–72, CrossRef.

Wynn TA. Common and unique mechanisms regulate fibrosis in various fibroproliferative diseases. J Clin Invest. 2007; 117(3): 524–9, CrossRef.

Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat Rev Mol Cell Biol. 2002; 3(5): 349–63, CrossRef.

Friedman SL. Mechanisms of disease: Mechanisms of hepatic fibrosis and therapeutic implications. Nat Clin Pract Gastroenterol Hepatol. 2004; 1(2): 98–105, CrossRef.

Serhan CN. Pro-resolving lipid mediators are leads for resolution physiology. Nature. 2014; 510(7503): 92–101, CrossRef.

Ward P. Acute and chronic inflammation. In: Serhan C, Ward P, Gilroy D, editors. Fundamentals of Inflammation. New York: Cambridge University Press; 2010. p. 1–16, NLMID.

Chiang N, Serhan CN. Structural elucidation and physiologic functions of specialized pro-resolving mediators and their receptors. Mol Aspects Med. 2017; 58: 114–29, CrossRef.

Bandeira-Melo C, Serra MF, Diaz BL, Cordeiro RS, Silva PM, Lenzi HL, et al. Cyclooxygenase-2-derived prostaglandin E2 and lipoxin A4 accelerate resolution of allergic edema in Angiostrongylus costaricensis-infected rats: relationship with concurrent eosinophilia. J Immunol. 2000; 164(2): 1029–36, CrossRef.

Serhan CN. The resolution of inflammation: the devil in the flask and in the details. FASEB J. 2011; 25(5): 1441–8, CrossRef.

Burdge GC, Calder PC. Introduction to fatty acids and lipids. In: Calder PC, Waitzberg DL, Koletzko B, editors. World Review of Nutrition and Dietetics. Basel: Karger AG; 2014. p. 1–16, CrossRef.

Lewis NE, Schramm G, Bordbar A, Schellenberger J, Andersen MP, Cheng JK, et al. Large-scale in silico modeling of metabolic interactions between cell types in the human brain. Nat Biotechnol. 2010; 28(12): 1279–85, CrossRef.

Tilley SL, Coffman TM, Koller BH. Mixed messages: modulation of inflammation and immune responses by prostaglandins and thromboxanes. J Clin Invest. 2001; 108(1): 15–23, CrossRef.

Kalinski P. Regulation of immune responses by prostaglandin E 2. J Immunol. 2012; 188(1): 21–8, CrossRef.

Calder PC. Omega-3 fatty acids and inflammatory processes: from molecules to man. Biochem Soc Trans. 2017; 45(5): 1105–15, CrossRef.

Flower RJ. Prostaglandins, bioassay and inflammation. Br J Pharmacol. 2006; 147(S1): S182–92, CrossRef.

Samuelsson B. Leukotrienes: mediators of immediate hypersensitivity reactions and inflammation. Science. 1983; 220(4597): 568–75, CrossRef.

Serhan CN. A search for endogenous mechanisms of anti-inflammation uncovers novel chemical mediators: missing links to resolution. Histochem Cell Biol. 2004; 122(4): 305–21, CrossRef.

Serhan CN, Chiang N, Van Dyke TE. Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators. Nat Rev Immunol. 2008; 8(5): 349–61, CrossRef.

Leslie CC. Cytosolic phospholipase A₂: physiological function and role in disease. J Lipid Res. 2015; 56(8): 1386–402, CrossRef.

Rådmark O, Werz O, Steinhilber D, Samuelsson B. 5-Lipoxygenase, a key enzyme for leukotriene biosynthesis in health and disease. Biochim Biophys Acta. 2015; 1851(4): 331–9, CrossRef.

Haeggström JZ. Leukotriene biosynthetic enzymes as therapeutic targets. J Clin Invest. 2018; 128(7): 2680–90, CrossRef.

Gordon S. Phagocytosis: An immunobiologic process. Immunity. 2016; 44(3): 463–75, CrossRef.

Godson C, Mitchell S, Harvey K, Petasis NA, Hogg N, Brady HR. Cutting edge: lipoxins rapidly stimulate nonphlogistic phagocytosis of apoptotic neutrophils by monocyte-derived macrophages. J Immunol. 2000; 164(4): 1663–7, CrossRef.

Serhan CN. Lipoxins and novel aspirin-triggered 15-epi-lipoxins (ATL): a jungle of cell-cell interactions or a therapeutic opportunity? Prostaglandins. 1997; 53(2): 107–37, CrossRef.

Bannenberg GL, Chiang N, Ariel A, Arita M, Tjonahen E, Gotlinger KH, et al. Molecular circuits of resolution: formation and actions of resolvins and protectins. J Immunol. 2005; 174(7): 4345–55, CrossRef.

Hong S, Gronert K, Devchand PR, Moussignac RL, Serhan CN. Novel docosatrienes and 17S-resolvins generated from docosahexaenoic acid in murine brain, human blood, and glial cells. Autacoids in anti-inflammation. J Biol Chem. 2003; 278(17): 14677–87, CrossRef.

Dalli J, Chiang N, Serhan CN. Elucidation of novel 13-series resolvins that increase with atorvastatin and clear infections. Nat Med. 2015; 21(9): 1071–5, CrossRef.

Dalli J, Colas RA, Serhan CN. Novel n-3 immunoresolvents: structures and actions. Sci Rep. 2013; 3: 1940, CrossRef.

Levy BD, Clish CB, Schmidt B, Gronert K, Serhan CN. Lipid mediator class switching during acute inflammation: signals in resolution. Nat Immunol. 2001; 2(7): 612–9, CrossRef.

Basil MC, Levy BD. Specialized pro-resolving mediators: endogenous regulators of infection and inflammation. Nat Rev Immunol. 2016; 16(1): 51–67, CrossRef.

Serhan CN, Chiang N, Dalli J. The resolution code of acute inflammation: Novel pro-resolving lipid mediators in resolution. Semin Immunol. 2015; 27(3): 200–15, CrossRef.

Duffney PF, Falsetta ML, Rackow AR, Thatcher TH, Phipps RP, Sime PJ. Key roles for lipid mediators in the adaptive immune response. J Clin Invest. 2018; 128(7): 2724–31, CrossRef.

Behn A, Ur E. The obesity epidemic and its cardiovascular consequences. Curr Opin Cardiol. 2006; 21(4): 353–60, CrossRef.

Lusis AJ. Atherosclerosis. Nature. 2000; 407(6801): 233–41, CrossRef.

Tanaka H, Sukhova GK, Swanson SJ, Clinton SK, Ganz P, Cybulsky MI, et al. Sustained activation of vascular cells and leukocytes in the rabbit aorta after balloon injury. Circulation. 1993; 88(4 Pt 1): 1788–803, CrossRef.

Kornowski R, Hong MK, Tio FO, Bramwell O, Wu H, Leon MB. In-stent restenosis: contributions of inflammatory responses and arterial injury to neointimal hyperplasia. J Am Coll Cardiol. 1998; 31(1): 224–30, CrossRef.

Shah PK. Inflammation, neointimal hyperplasia, and restenosis: as the leukocytes roll, the arteries thicken. Circulation. 2003; 107(17): 2175–7, CrossRef.

de Vries MR, Quax PHA. Inflammation in Vein Graft Disease. Front Cardiovasc Med. 2018; 5: 3, CrossRef.

Conte MS, Desai TA, Wu B, Schaller M, Werlin E. Pro-resolving lipid mediators in vascular disease. J Clin Investig. 2018; 128(9): 3727–35, CrossRef.

Geering B, Stoeckle C, Conus S, Simon HU. Living and dying for inflammation: neutrophils, eosinophils, basophils. Trends Immunol. 2013; 34(8): 398–409, CrossRef.

Merched AJ, Ko K, Gotlinger KH, Serhan CN, Chan L. Atherosclerosis: evidence for impairment of resolution of vascular inflammation governed by specific lipid mediators. FASEB J. 2008; 22(10): 3595–606, CrossRef.

White PJ, Arita M, Taguchi R, Kang JX, Marette A. Transgenic restoration of long-chain n-3 fatty acids in insulin target tissues improves resolution capacity and alleviates obesity-linked inflammation and insulin resistance in high-fat-fed mice. Diabetes. 2010; 59(12): 3066–73, CrossRef.

Titos E, Rius B, González-Périz A, López-Vicario C, Morán-Salvador E, Martínez-Clemente M, et al. Resolvin D1 and its precursor docosahexaenoic acid promote resolution of adipose tissue inflammation by eliciting macrophage polarization toward an M2-like phenotype. J Immunol. 2011; 187(10): 5408–18, CrossRef.

Medeiros R, Kitazawa M, Passos GF, Baglietto-Vargas D, Cheng D, Cribbs DH, et al. Aspirin-triggered lipoxin A4 stimulates alternative activation of microglia and reduces Alzheimer disease-like pathology in mice. Am J Pathol. 2013; 182(5): 1780–9, CrossRef.

Planagumà A, Kazani S, Marigowda G, Haworth O, Mariani TJ, Israel E, et al. Airway lipoxin A4 generation and lipoxin A4 receptor expression are decreased in severe asthma. Am J Respir Crit Care Med. 2008; 178(6): 574–82, CrossRef.

Serhan CN. Novel lipid mediators and resolution mechanisms in acute inflammation: to resolve or not? Am J Pathol. 2010; 177(4): 1576–91, CrossRef.

Serhan CN. Resolution phase of inflammation: novel endogenous anti-inflammatory and proresolving lipid mediators and pathways. Annu Rev Immunol. 2007; 25: 101–37, CrossRef.

Clària J, Serhan CN. Aspirin triggers previously undescribed bioactive eicosanoids by human endothelial cell-leukocyte interactions. Proc Natl Acad Sci USA. 1995; 92(21): 9475–9, CrossRef.

Tabas I. Macrophage death and defective inflammation resolution in atherosclerosis. Nat Rev Immunol. 2010; 10(1): 36–46, CrossRef.

Fredman G, Hellmann J, Proto JD, Kuriakose G, Colas RA, Dorweiler B, et al. An imbalance between specialized pro-resolving lipid mediators and pro-inflammatory leukotrienes promotes instability of atherosclerotic plaques. Nat Commun. 2016; 7: 12859, CrossRef.

Viola JR, Lemnitzer P, Jansen Y, Csaba G, Winter C, Neideck C, et al. Resolving lipid mediators maresin 1 and resolvin D2 prevent atheroprogression in mice. Circ Res. 2016; 119(9): 1030–8, CrossRef.

Bacon PA, Townend JN. Nails in the coffin: increasing evidence for the role of rheumatic disease in the cardiovascular mortality of rheumatoid arthritis. Arthritis Rheum. 2001; 44(12): 2707–10, CrossRef.

Buckley CD, Gilroy DW, Serhan CN. Proresolving lipid mediators and mechanisms in the resolution of acute inflammation. Immunity. 2014; 40(3): 315–27, CrossRef.

Raza K, Buckley CE, Salmon M, Buckley CD. Treating very early rheumatoid arthritis. Best Pract Res Clin Rheumatol. 2006; 20(5): 849–63, CrossRef.

Feldmann M, Maini RN. Perspectives from masters in rheumatology and autoimmunity: Can we get closer to a cure for rheumatoid arthritis? Arthritis Rheumatol. 2015; 67(9): 2283–91, CrossRef.

Klarenbeek NB, Güler-Yüksel M, van der Kooij SM, Han KH, Ronday HK, Kerstens PJSM, et al. The impact of four dynamic, goal-steered treatment strategies on the 5-year outcomes of rheumatoid arthritis patients in the BeSt study. Ann Rheum Dis. 2011; 70(6): 1039–46, CrossRef.

Siebert S, Tsoukas A, Robertson J, McInnes I. Cytokines as therapeutic targets in rheumatoid arthritis and other inflammatory diseases. Pharmacol Rev. 2015; 67(2): 280–309, CrossRef.

Norling LV, Headland SE, Dalli J, Arnardottir HH, Haworth O, Jones HR, et al. Proresolving and cartilage-protective actions of resolvin D1 in inflammatory arthritis. JCI Insight. 2016; 1(5): e85922, CrossRef.

Neuman MG. Immune dysfunction in inflammatory bowel disease. Transl Res. 2007; 149(4): 173–86, CrossRef.

Strober W, Fuss I, Mannon P. The fundamental basis of inflammatory bowel disease. J Clin Invest. 2007; 117(3): 514–21, CrossRef.

Sands BE. Inflammatory bowel disease: past, present, and future. J Gastroenterol. 2007; 42(1): 16–25, CrossRef.

Harris RE. Cyclooxygenase-2 (cox-2) blockade in the chemoprevention of cancers of the colon, breast, prostate, and lung. Inflammopharmacology. 2009; 17(2): 55–67, CrossRef.

Algra AM, Rothwell PM. Effects of regular aspirin on long-term cancer incidence and metastasis: a systematic comparison of evidence from observational studies versus randomised trials. Lancet Oncol. 2012; 13(5): 518–27, CrossRef.

Rothwell PM, Price JF, Fowkes FGR, Zanchetti A, Roncaglioni MC, Tognoni G, et al. Short-term effects of daily aspirin on cancer incidence, mortality, and non-vascular death: analysis of the time course of risks and benefits in 51 randomised controlled trials. Lancet. 2012; 379(9826): 1602–12, CrossRef.

Rothwell PM, Wilson M, Price JF, Belch JFF, Meade TW, Mehta Z. Effect of daily aspirin on risk of cancer metastasis: a study of incident cancers during randomised controlled trials. Lancet. 2012; 379(9826): 1591–601, CrossRef.

Wang D, DuBois RN. The role of anti-inflammatory drugs in colorectal cancer. Annu Rev Med. 2013; 64: 131–44, CrossRef.

Drew DA, Cao Y, Chan AT. Aspirin and colorectal cancer: the promise of precision chemoprevention. Nat Rev Cancer. 2016; 16(3): 173–86, CrossRef.

Kong P, Wu R, Liu X, Liu J, Chen S, Ye M, et al. The effects of anti-inflammatory drug treatment in gastric cancer prevention: An update of a meta-analysis. J Cancer. 2016; 7(15): 2247–57, CrossRef.

Eberhart CE, Coffey RJ, Radhika A, Giardiello FM, Ferrenbach S, DuBois RN. Up-regulation of cyclooxygenase 2 gene expression in human colorectal adenomas and adenocarcinomas. Gastroenterology. 1994; 107(4): 1183–8, CrossRef.

Gupta RA, Dubois RN. Colorectal cancer prevention and treatment by inhibition of cyclooxygenase-2. Nat Rev Cancer. 2001; 1(1): 11–21, CrossRef.

Marnett LJ, DuBois RN. COX-2: a target for colon cancer prevention. Annu Rev Pharmacol Toxicol. 2002; 42: 55–80, CrossRef.

Mayers JR, Wu C, Clish CB, Kraft P, Torrence ME, Fiske BP, et al. Elevation of circulating branched-chain amino acids is an early event in human pancreatic adenocarcinoma development. Nat Med. 2014; 20(10): 1193–8, CrossRef.

Hu Z, Yang Y, Zhao Y, Huang Y. The prognostic value of cyclooxygenase-2 expression in patients with esophageal cancer: evidence from a meta-analysis. Onco Targets Ther. 2017; 10: 2893–901, CrossRef.

Cheng J, Fan XM. Role of cyclooxygenase-2 in gastric cancer development and progression. World J Gastroenterol. 2013; 19(42): 7361–8, CrossRef.

Scher JU, Pillinger MH. The anti-inflammatory effects of prostaglandins. J Investig Med. 2009; 57(6): 703–8, CrossRef.

Straus DS, Pascual G, Li M, Welch JS, Ricote M, Hsiang CH, et al. 15-deoxy-delta 12,14-prostaglandin J2 inhibits multiple steps in the NF-kappa B signaling pathway. Proc Natl Acad Sci USA. 2000; 97(9): 4844–9, CrossRef.

Wang D, Dubois RN. Eicosanoids and cancer. Nat Rev Cancer. 2010; 10(3): 181–93, CrossRef.

Wang D, Mann JR, DuBois RN. The role of prostaglandins and other eicosanoids in the gastrointestinal tract. Gastroenterology. 2005; 128(5): 1445–61, CrossRef.

Narumiya S. Prostanoids and inflammation: a new concept arising from receptor knockout mice. J Mol Med. 2009; 87(10): 1015–22, CrossRef.

Wang D, DuBois RN. Role of prostanoids in gastrointestinal cancer. J Clin Invest. 2018; 128(7): 2732–42, CrossRef.

Weiskirchen R, Weiskirchen S, Tacke F. Organ and tissue fibrosis: Molecular signals, cellular mechanisms and translational implications. Mol Aspects Med. 2019; 65: 2–15, CrossRef.

Pakshir P, Hinz B. The big five in fibrosis: Macrophages, myofibroblasts, matrix, mechanics, and miscommunication. Matrix Biology. 2018; 68–69: 81–93, CrossRef.

Rosenbloom J, Castro SV, Jimenez SA. Narrative review: Fibrotic diseases: Cellular and molecular mechanisms and novel therapies. Ann Intern Med. 2010; 152(3): 159–66, CrossRef.

Rockey DC, Bell PD, Hill JA. Fibrosis--a common pathway to organ injury and failure. N Engl J Med. 2015; 372(12): 1138–49, CrossRef.

Gabbiani G. The myofibroblast: a key cell for wound healing and fibrocontractive diseases. Prog Clin Biol Res. 1981; 54: 183–94, PMID.

Desmoulière A. Factors influencing myofibroblast differentiation during wound healing and fibrosis. Cell Biol Int. 1995; 19(5): 471–6, CrossRef.

McAnulty RJ. Fibroblasts and myofibroblasts: their source, function and role in disease. Int J Biochem Cell Biol. 2007; 39(4): 666–71, CrossRef.

Hinz B, Phan SH, Thannickal VJ, Galli A, Bochaton-Piallat ML, Gabbiani G. The myofibroblast: one function, multiple origins. Am J Pathol. 2007; 170(6): 1807–16, CrossRef.

Hinz B, Phan SH, Thannickal VJ, Prunotto M, Desmoulière A, Varga J, et al. Recent developments in myofibroblast biology: paradigms for connective tissue remodeling. Am J Pathol. 2012; 180(4): 1340–55, CrossRef.

Hu B, Phan SH. Myofibroblasts. Curr Opin Rheumatol. 2013; 25(1): 71–7, CrossRef.

Kirk TZ, Mark ME, Chua CC, Chua BH, Mayes MD. Myofibroblasts from scleroderma skin synthesize elevated levels of collagen and tissue inhibitor of metalloproteinase (TIMP-1) with two forms of TIMP-1. J Biol Chem. 1995; 270(7): 3423–8, CrossRef.

Abraham DJ, Eckes B, Rajkumar V, Krieg T. New developments in fibroblast and myofibroblast biology: implications for fibrosis and scleroderma. Curr Rheumatol Rep. 2007; 9(2): 136–43, CrossRef.

Gilbane AJ, Denton CP, Holmes AM. Scleroderma pathogenesis: a pivotal role for fibroblasts as effector cells. Arthritis Res Ther. 2013; 15(3): 215, CrossRef.

Kendall RT, Feghali-Bostwick CA. Fibroblasts in fibrosis: novel roles and mediators. Front Pharmacol. 2014; 5: 123, CrossRef.

Laurent GJ, Chambers RC, Hill MR, McAnulty RJ. Regulation of matrix turnover: fibroblasts, forces, factors and fibrosis. Biochem Soc Trans. 2007; 35(Pt 4): 647–51, CrossRef.

Wells RG, Discher DE. Matrix elasticity, cytoskeletal tension, and TGF-beta: the insoluble and soluble meet. Sci Signal. 2008; 1(10): pe13, CrossRef.

Hinz B. Tissue stiffness, latent TGF-β1 activation, and mechanical signal transduction: Implications for the pathogenesis and treatment of fibrosis. Curr Rheumatol Rep. 2009; 11(2): 120–6, CrossRef.

Parker MW, Rossi D, Peterson M, Smith K, Sikström K, White ES, et al. Fibrotic extracellular matrix activates a profibrotic positive feedback loop. J Clin Invest. 2014; 124(4): 1622–35, CrossRef.

Rittié L. Another dimension to the importance of the extracellular matrix in fibrosis. J Cell Commun Signal. 2015; 9(1): 99–100, CrossRef.

Postlethwaite AE, Shigemitsu H, Kanangat S. Cellular origins of fibroblasts: possible implications for organ fibrosis in systemic sclerosis. Curr Opin Rheumatol. 2004; 16(6): 733–8, CrossRef.

Rosenbloom J, Macarak E, Piera-Velazquez S, Jimenez SA. Human fibrotic diseases: Current challenges in fibrosis research. In: Rittié L, editor. Fibrosis. New York: Springer New York; 2017. p. 1–23, CrossRef.

Thannickal VJ, Zhou Y, Gaggar A, Duncan SR. Fibrosis: ultimate and proximate causes. J Clin Invest. 2014; 124(11): 4673–7, CrossRef.

Micallef L, Vedrenne N, Billet F, Coulomb B, Darby IA, Desmoulière A. The myofibroblast, multiple origins for major roles in normal and pathological tissue repair. Fibrogenesis Tissue Repair. 2012; 5(Suppl 1): S5, CrossRef.

Distler JHW, Györfi AH, Ramanujam M, Whitfield ML, Königshoff M, Lafyatis R. Shared and distinct mechanisms of fibrosis. Nat Rev Rheumatol. 2019; 15(12): 705–30, CrossRef.

Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med. 2012; 18(7): 1028–40, CrossRef.

Eming SA, Krieg T, Davidson JM. Inflammation in wound repair: molecular and cellular mechanisms. J Invest Dermatol. 2007; 127(3): 514–25, CrossRef.

Shook B, Xiao E, Kumamoto Y, Iwasaki A, Horsley V. CD301b+ macrophages are rssential for effective skin wound healing. J Invest Dermatol. 2016; 136(9): 1885–91, CrossRef.

Karlmark KR, Weiskirchen R, Zimmermann HW, Gassler N, Ginhoux F, Weber C, et al. Hepatic recruitment of the inflammatory Gr1+ monocyte subset upon liver injury promotes hepatic fibrosis. Hepatology. 2009; 50(1): 261–74, CrossRef.

Weng SY, Wang X, Vijayan S, Tang Y, Kim YO, Padberg K, et al. IL-4 receptor alpha signaling through macrophages differentially regulates liver fibrosis progression and reversal. EBioMedicine. 2018; 29: 92–103, CrossRef.

Belperio JA, Dy M, Burdick MD, Xue YY, Li K, Elias JA, et al. Interaction of IL-13 and C10 in the pathogenesis of bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol. 2002; 27(4): 419–27, CrossRef.

Lee CG, Homer RJ, Zhu Z, Lanone S, Wang X, Koteliansky V, et al. Interleukin-13 induces tissue fibrosis by selectively stimulating and activating transforming growth factor beta(1). J Exp Med. 2001; 194(6): 809–21, CrossRef.

Reyfman PA, Walter JM, Joshi N, Anekalla KR, McQuattie-Pimentel AC, Chiu S, et al. Single-cell transcriptomic analysis of human lung provides insights into the pathobiology of pulmonary fibrosis. Am J Respir Crit Care Med. 2019; 199(12): 1517–36, CrossRef.

Morse C, Tabib T, Sembrat J, Buschur KL, Bittar HT, Valenzi E, et al. Proliferating SPP1/MERTK-expressing macrophages in idiopathic pulmonary fibrosis. Eur Respir J. 2019; 54(2): 1802441, CrossRef.

Kazlauskas A. PDGFs and their receptors. Gene. 2017; 614: 1–7, CrossRef.

Borkham-Kamphorst E, Weiskirchen R. The PDGF system and its antagonists in liver fibrosis. Cytokine Growth Factor Rev. 2016; 28: 53–61, CrossRef.

Duffield JS, Hong S, Vaidya VS, Lu Y, Fredman G, Serhan CN, et al. Resolvin D series and protectin D1 mitigate acute kidney injury. J Immunol. 2006; 177(9): 5902–11, CrossRef.

Poss KD, Wilson LG, Keating MT. Heart regeneration in zebrafish. Science. 2002; 298(5601): 2188–90, CrossRef.

Varga J, Trojanowska M, Kuwana M. Pathogenesis of systemic sclerosis: recent insights of molecular and cellular mechanisms and therapeutic opportunities. J Scleroderma Relat Disord. 2017; 2(3): 137–52, CrossRef.

Brown M, O’Reilly S. The immunopathogenesis of fibrosis in systemic sclerosis. Clin Exp Immunol. 2019; 195(3): 310–21, CrossRef.

Hamaguchi Y, Takehara K. Anti-nuclear autoantibodies in systemic sclerosis : News and perspectives. J Scleroderma Relat Disord. 2018; 3(3): 201–13, CrossRef.

Tyndall AJ, Bannert B, Vonk M, Airo P, Cozzi F, Carreira PE, et al. Causes and risk factors for death in systemic sclerosis: a study from the EULAR Scleroderma Trials and Research (EUSTAR) database. Ann Rheum Dis. 2010; 69(10): 1809–15, CrossRef.

Volkmann ER, Varga J. Emerging targets of disease-modifying therapy for systemic sclerosis. Nat Rev Rheumatol. 2019; 15(4): 208–24, CrossRef.

Avanoǧlu Güler A, Rossi FW, Bellando-Randone S, Prevete N, Tufan A, Manetti M, et al. The role of endogenous eicosapentaenoic acid and docosahexaenoic acid-derived resolvins in systemic sclerosis. Front Immunol. 2020; 11: 1249, CrossRef.

Cutolo M, Soldano S, Smith V. Pathophysiology of systemic sclerosis: current understanding and new insights. Expert Rev Clin Immunol. 2019; 15(7): 753–64, CrossRef.

Huang J, Maier C, Zhang Y, Soare A, Dees C, Beyer C, et al. Nintedanib inhibits macrophage activation and ameliorates vascular and fibrotic manifestations in the Fra2 mouse model of systemic sclerosis. Ann Rheum Dis. 2017; 76(11): 1941–8, CrossRef.

Martinez FJ, Collard HR, Pardo A, Raghu G, Richeldi L, Selman M, et al. Idiopathic pulmonary fibrosis. Nat Rev Dis Primers. 2017; 3(1): 17074, CrossRef.

Kim HJ, Perlman D, Tomic R. Natural history of idiopathic pulmonary fibrosis. Respir Med. 2015; 109(6): 661–70, CrossRef.

Ley B, Collard HR. Epidemiology of idiopathic pulmonary fibrosis. Clin Epidemiol. 2013; 5: 483–92, CrossRef.

Wolters PJ, Collard HR, Jones KD. Pathogenesis of idiopathic pulmonary fibrosis. Annu Rev Pathol. 2014; 9(1): 157–79, CrossRef.

Selman M, Pardo A. Revealing the pathogenic and aging-related mechanisms of the enigmatic idiopathic pulmonary fibrosis. An integral model. Am J Respir Crit Care Med. 2014; 189(10): 1161–72, CrossRef.

Selman M, López-Otín C, Pardo A. Age-driven developmental drift in the pathogenesis of idiopathic pulmonary fibrosis. Eur Respir J. 2016; 48(2): 538–52, CrossRef.

Mokdad AA, Lopez AD, Shahraz S, Lozano R, Mokdad AH, Stanaway J, et al. Liver cirrhosis mortality in 187 countries between 1980 and 2010: a systematic analysis. BMC Med. 2014; 12: 145, CrossRef.

Sayiner M, Koenig A, Henry L, Younossi ZM. Epidemiology of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis in the United States and the rest of the world. Clin Liver Dis. 2016; 20(2): 205–14, CrossRef.

Satapathy SK, Sanyal AJ. Epidemiology and natural history of nonalcoholic fatty liver disease. Semin Liver Dis. 2015; 35(3): 221–35, CrossRef.

Koyama Y, Brenner DA. Liver inflammation and fibrosis. J Clin Invest. 2017; 127(1): 55–64, CrossRef.

Kisseleva T, Cong M, Paik Y, Scholten D, Jiang C, Benner C, et al. Myofibroblasts revert to an inactive phenotype during regression of liver fibrosis. Proc Natl Acad Sci USA. 2012; 109(24): 9448–53, CrossRef.

Blaner WS, O’Byrne SM, Wongsiriroj N, Kluwe J, D’Ambrosio DM, Jiang H, et al. Hepatic stellate cell lipid droplets: a specialized lipid droplet for retinoid storage. Biochim Biophys Acta. 2009; 1791(6): 467–73, CrossRef.

Marra F, Efsen E, Romanelli RG, Caligiuri A, Pastacaldi S, Batignani G, et al. Ligands of peroxisome proliferator-activated receptor gamma modulate profibrogenic and proinflammatory actions in hepatic stellate cells. Gastroenterology. 2000; 119(2): 466–78, CrossRef.

Bataller R, Brenner DA. Liver fibrosis. J Clin Invest. 2005; 115(2): 209–18, CrossRef.

Hellerbrand C, Stefanovic B, Giordano F, Burchardt ER, Brenner DA. The role of TGFbeta1 in initiating hepatic stellate cell activation in vivo. J Hepatol. 1999; 30(1): 77–87, CrossRef.

Pradere JP, Kluwe J, De Minicis S, Jiao JJ, Gwak GY, Dapito DH, et al. Hepatic macrophages but not dendritic cells contribute to liver fibrosis by promoting the survival of activated hepatic stellate cells in mice. Hepatology. 2013; 58(4): 1461–73, CrossRef.

Serhan CN, Brain SD, Buckley CD, Gilroy DW, Haslett C, O’Neill LAJ, et al. Resolution of inflammation: state of the art, definitions and terms. FASEB J. 2007; 21(2): 325–32, CrossRef.

Mi S, Li Z, Yang HZ, Liu H, Wang JP, Ma YG, et al. Blocking IL-17A promotes the resolution of pulmonary inflammation and fibrosis via TGF-beta1-dependent and -independent mechanisms. J Immunol. 2011; 187(6): 3003–14, CrossRef.

Reddy NM, Potteti HR, Mariani TJ, Biswal S, Reddy SP. Conditional deletion of Nrf2 in airway epithelium exacerbates acute lung injury and impairs the resolution of inflammation. Am J Respir Cell Mol Biol. 2011; 45(6): 1161–8, CrossRef.

Bynoe MS, Waickman AT, Mahamed DA, Mueller C, Mills JH, Czopik A. CD73 is critical for the resolution of murine colonic inflammation. J Biomed Biotechnol. 2012; 2012: 260983, CrossRef.

Mavers M, Cuda CM, Misharin AV, Gierut AK, Agrawal H, Weber E, et al. Cyclin-dependent kinase inhibitor p21, via its C-terminal domain, is essential for resolution of murine inflammatory arthritis. Arthritis Rheum. 2012; 64(1): 141–52, CrossRef.

Chiang N, Fredman G, Bäckhed F, Oh SF, Vickery T, Schmidt BA, et al. Infection regulates pro-resolving mediators that lower antibiotic requirements. Nature. 2012; 484(7395): 524–8, CrossRef.

Serhan CN, Yang R, Martinod K, Kasuga K, Pillai PS, Porter TF, et al. Maresins: novel macrophage mediators with potent antiinflammatory and proresolving actions. J Exp Med. 2009; 206(1): 15–23, CrossRef.

Fetterman JW, Zdanowicz MM. Therapeutic potential of n-3 polyunsaturated fatty acids in disease. Am J Health Syst Pharm. 2009; 66(13): 1169–79, CrossRef.

Reátegui E, Jalali F, Khankhel AH, Wong E, Cho H, Lee J, et al. Microscale arrays for the profiling of start and stop signals coordinating human-neutrophil swarming. Nat Biomed Eng. 2017; 1: 0094, CrossRef.

Werz O, Gerstmeier J, Libreros S, De la Rosa X, Werner M, Norris PC, et al. Human macrophages differentially produce specific resolvin or leukotriene signals that depend on bacterial pathogenicity. Nat Commun. 2018; 9: 59, CrossRef.

Motwani MP, Colas RA, George MJ, Flint JD, Dalli J, Richard-Loendt A, et al. Pro-resolving mediators promote resolution in a human skin model of UV-killed Escherichia coli-driven acute inflammation. JCI Insight. 2018; 3(6): e94463, CrossRef.

Dalli J, Serhan CN. Pro-resolving mediators in regulating and conferring macrophage function. Front Immunol. 2017; 8: 1400, CrossRef.

Serhan CN, Krishnamoorthy S, Recchiuti A, Chiang N. Novel anti-inflammatory--pro-resolving mediators and their receptors. Curr Top Med Chem. 2011; 11(6): 629–47, CrossRef.

Dalli J, Zhu M, Vlasenko NA, Deng B, Haeggström JZ, Petasis NA, et al. The novel 13S,14S-epoxy-maresin is converted by human macrophages to maresin 1 (MaR1), inhibits leukotriene A4 hydrolase (LTA4H), and shifts macrophage phenotype. FASEB J. 2013; 27(7): 2573–83, CrossRef.

Mitchell S, Thomas G, Harvey K, Cottell D, Reville K, Berlasconi G, et al. Lipoxins, aspirin-triggered epi-lipoxins, lipoxin stable analogues, and the resolution of inflammation: Stimulation of macrophage phagocytosis of apoptotic neutrophils in vivo. J Am Soc Nephrol. 2002; 13(10): 2497–507, CrossRef.

Moore KJ, Tabas I. Macrophages in the pathogenesis of atherosclerosis. Cell. 2011; 145(3): 341–55, CrossRef.

Schrijvers DM, De Meyer GRY, Kockx MM, Herman AG, Martinet W. Phagocytosis of apoptotic cells by macrophages is impaired in atherosclerosis. Arterioscler Thromb Vasc Biol. 2005; 25(6): 1256–61, CrossRef.

Linton MF, Babaev VR, Huang J, Linton EF, Tao H, Yancey PG. Macrophage apoptosis and efferocytosis in the pathogenesis of atherosclerosis. Circ J. 2016; 80(11): 2259–68, CrossRef.

Thorp E, Tabas I. Mechanisms and consequences of efferocytosis in advanced atherosclerosis. J Leukoc Biol. 2009; 86(5): 1089–95, CrossRef.

Schwab JM, Chiang N, Arita M, Serhan CN. Resolvin E1 and protectin D1 activate inflammation-resolution programmes. Nature. 2007; 447(7146): 869–74, CrossRef.

Dalli J, Serhan CN. Specific lipid mediator signatures of human phagocytes: microparticles stimulate macrophage efferocytosis and pro-resolving mediators. Blood. 2012; 120(15): e60–72, CrossRef.

Ohira T, Arita M, Omori K, Recchiuti A, Van Dyke TE, Serhan CN. Resolvin E1 receptor activation signals phosphorylation and phagocytosis. J Biol Chem. 2010; 285(5): 3451–61, CrossRef.

Maddox JF, Serhan CN. Lipoxin A4 and B4 are potent stimuli for human monocyte migration and adhesion: selective inactivation by dehydrogenation and reduction. J Exp Med. 1996; 183(1): 137–46, CrossRef.

Maddox JF, Hachicha M, Takano T, Petasis NA, Fokin VV, Serhan CN. Lipoxin A4 stable analogs are potent mimetics that stimulate human monocytes and THP-1 cells via a G-protein-linked lipoxin A4 receptor. J Biol Chem. 1997; 272(11): 6972–8, CrossRef.

Kasikara C, Doran AC, Cai B, Tabas I. The role of non-resolving inflammation in atherosclerosis. J Clin Invest. 2018; 128(7): 2713–23, CrossRef.

Qiu W, Guo K, Yi L, Gong Y, Huang L, Zhong W. Resolvin E1 reduces hepatic fibrosis in mice with Schistosoma japonicum infection. Exp Ther Med. 2014; 7(6): 1481–5, CrossRef.

Qu X, Zhang X, Yao J, Song J, Nikolic-Paterson DJ, Li J. Resolvins E1 and D1 inhibit interstitial fibrosis in the obstructed kidney via inhibition of local fibroblast proliferation: Resolvins prevent fibrosis via inhibition of fibroblast proliferation. J Pathol. 2012; 228(4): 506–19, CrossRef.

Yatomi M, Hisada T, Ishizuka T, Koga Y, Ono A, Kamide Y, et al. 17(R)-resolvin D1 ameliorates bleomycin-induced pulmonary fibrosis in mice. Physiol Rep. 2015; 3(12): e12628, CrossRef.

Kain V, Ingle KA, Colas RA, Dalli J, Prabhu SD, Serhan CN, et al. Resolvin D1 activates the inflammation resolving response at splenic and ventricular site following myocardial infarction leading to improved ventricular function. J Mol Cell Cardiol. 2015; 84: 24–35, CrossRef.




DOI: https://doi.org/10.18585/inabj.v15i2.2181

Copyright (c) 2023 The Prodia Education and Research Institute

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

 

Indexed by:

                  

               

                

 

 

The Prodia Education and Research Institute