Abstract
Fibrosis is a pathophysiological mechanism involved in chronic and progressive diseases that results in excessive tissue scarring. Diseases associated with fibrosis include metabolic dysfunction-associated steatohepatitis (MASH), inflammatory bowel diseases (IBDs), chronic kidney disease (CKD), idiopathic pulmonary fibrosis (IPF) and systemic sclerosis (SSc), which are collectively responsible for substantial morbidity and mortality. Although a few drugs with direct antifibrotic activity are approved for pulmonary fibrosis and considerable progress has been made in the understanding of mechanisms of fibrosis, translation of this knowledge into effective therapies continues to be limited and challenging. With the aim of assisting developers of novel antifibrotic drugs, this Review integrates viewpoints of biologists and physician-scientists on core pathways involved in fibrosis across organs, as well as on specific characteristics and approaches to assess therapeutic interventions for fibrotic diseases of the lung, gut, kidney, skin and liver. This discussion is used as a basis to propose strategies to improve the translation of potential antifibrotic therapies.
This is a preview of subscription content, access via your institution
Access options
Access through your institution
Change institution
Buy or subscribe
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Learn more
Subscribe to this journal
Receive 12 print issues and online access
$209.00 per year
only $17.42 per issue
Learn more
Buy this article
Purchase on SpringerLink
Instant access to full article PDF
Buy now
Prices may be subject to local taxes which are calculated during checkout
Additional access options:
Log in
Learn about institutional subscriptions
Read our FAQs
Contact customer support
Fig. 1: Initiation of fibrosis.
Fig. 2: Myofibroblast activation states and pathways.
Fig. 3: Key mechanisms involved in the development of pulmonary fibrosis.
Fig. 4: Core mechanisms of intestinal fibrogenesis.
Fig. 5: Key pathways of chronic kidney disease and fibrosis.
Fig. 6: Mechanisms of fibrotic tissue remodelling in systemic sclerosis.
Fig. 7: Key pathways of liver fibrosis progression and regression.
References
Hinz, B. et al. Recent developments in myofibroblast biology: paradigms for connective tissue remodeling. Am. J. Pathol. 180, 1340–1355 (2012).
CASPubMedGoogle Scholar
Wynn, T. A. & Ramalingam, T. R. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat. Med. 18, 1028–1040 (2012).
CASPubMedGoogle Scholar
Henderson, N. C., Rieder, F. & Wynn, T. A. Fibrosis: from mechanisms to medicines. Nature 587, 555–566 (2020).
CASPubMedGoogle Scholar
Szaszi, K. & Amoozadeh, Y. New insights into functions, regulation, and pathological roles of tight junctions in kidney tubular epithelium. Int. Rev. Cell Mol. Biol. 308, 205–271 (2014).
CASPubMedGoogle Scholar
Selman, M. & Pardo, A. The leading role of epithelial cells in the pathogenesis of idiopathic pulmonary fibrosis. Cell Signal. 66, 109482 (2020).
CASPubMedGoogle Scholar
Yamashita, N. & Kramann, R. Mechanisms of kidney fibrosis and routes towards therapy. Trends Endocrinol. Metab. 35, 31–48 (2024).
CASPubMedGoogle Scholar
Prunotto, M. et al. From acute injury to chronic disease: pathophysiological hypothesis of an epithelial/mesenchymal crosstalk alteration in CKD. Nephrol. Dial. Transpl. 27, iii43–iii50 (2012).
Google Scholar
Hinz, B. & Lagares, D. Evasion of apoptosis by myofibroblasts: a hallmark of fibrotic diseases. Nat. Rev. Rheumatol. 16, 11–31 (2020).
CASPubMedGoogle Scholar
Pakshir, P. & Hinz, B. The big five in fibrosis: macrophages, myofibroblasts, matrix, mechanics, and miscommunication. Matrix Biol. 68-69, 81–93 (2018).
CASPubMedGoogle Scholar
Bonventre, J. V. & Yang, L. Cellular pathophysiology of ischemic acute kidney injury. J. Clin. Invest. 121, 4210–4221 (2011).
CASPubMedGoogle Scholar
Venkatachalam, M. A., Weinberg, J. M., Kriz, W. & Bidani, A. K. Failed tubule recovery, AKI-CKD transition, and kidney disease progression. J. Am. Soc. Nephrol. 26, 1765–1776 (2015).
CASPubMedGoogle Scholar
Yang, L., Besschetnova, T. Y., Brooks, C. R., Shah, J. V. & Bonventre, J. V. Epithelial cell cycle arrest in G2/M mediates kidney fibrosis after injury. Nat. Med. 16, 535–543 (2010).
CASPubMedGoogle Scholar
Koesters, R. et al. Tubular overexpression of transforming growth factor-beta1 induces autophagy and fibrosis but not mesenchymal transition of renal epithelial cells. Am. J. Pathol. 177, 632–643 (2010).
CASPubMedGoogle Scholar
Grgic, I. et al. Targeted proximal tubule injury triggers interstitial fibrosis and glomerulosclerosis. Kidney Int. 82, 172–183 (2012).
CASPubMedGoogle Scholar
Takaori, K. et al. Severity and frequency of proximal tubule injury determines renal prognosis. J. Am. Soc. Nephrol. 27, 2393–2406 (2016).
PubMedGoogle Scholar
Liu, B. C., Tang, T. T., Lv, L. L. & Lan, H. Y. Renal tubule injury: a driving force toward chronic kidney disease. Kidney Int. 93, 568–579 (2018).
CASPubMedGoogle Scholar
Liu, J. et al. Accelerated senescence of renal tubular epithelial cells is associated with disease progression of patients with immunoglobulin A (IgA) nephropathy. Transl. Res. 159, 454–463 (2012).
CASPubMedGoogle Scholar
Seibold, M. A. et al. A common MUC5B promoter polymorphism and pulmonary fibrosis. N. Engl. J. Med. 364, 1503–1512 (2011).
CASPubMedGoogle Scholar
Fingerlin, T. E. et al. Genome-wide association study identifies multiple susceptibility loci for pulmonary fibrosis. Nat. Genet. 45, 613–620 (2013).
CASPubMedGoogle Scholar
Noth, I. et al. Genetic variants associated with idiopathic pulmonary fibrosis susceptibility and mortality: a genome-wide association study. Lancet Respir. Med. 1, 309–317 (2013).
CASPubMedGoogle Scholar
Allen, R. J. et al. Genetic variants associated with susceptibility to idiopathic pulmonary fibrosis in people of European ancestry: a genome-wide association study. Lancet Respir. Med. 5, 869–880 (2017).
CASPubMedGoogle Scholar
Dressen, A. et al. Analysis of protein-altering variants in telomerase genes and their association with MUC5B common variant status in patients with idiopathic pulmonary fibrosis: a candidate gene sequencing study. Lancet Respir. Med. 6, 603–614 (2018).
CASPubMedGoogle Scholar
Garcia, O. et al. Targeted type 2 alveolar cell depletion. a dynamic functional model for lung injury repair. Am. J. Respir. Cell Mol. Biol. 54, 319–330 (2016).
CASPubMedGoogle Scholar
Maher, T. M. et al. Diminished prostaglandin E2 contributes to the apoptosis paradox in idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 182, 73–82 (2010).
CASPubMedGoogle Scholar
Naikawadi, R. P. et al. Telomere dysfunction in alveolar epithelial cells causes lung remodeling and fibrosis. JCI Insight 1, e86704 (2016).
PubMedGoogle Scholar
Nureki, S. I. et al. Expression of mutant Sftpc in murine alveolar epithelia drives spontaneous lung fibrosis. J. Clin. Invest. 128, 4008–4024 (2018).
PubMedGoogle Scholar
Gabele, E. et al. Role of TLR9 in hepatic stellate cells and experimental liver fibrosis. Biochem. Biophys. Res. Commun. 376, 271–276 (2008).
PubMedGoogle Scholar
Watanabe, A. et al. Apoptotic hepatocyte DNA inhibits hepatic stellate cell chemotaxis via toll-like receptor 9. Hepatology 46, 1509–1518 (2007).
CASPubMedGoogle Scholar
Chen, Y. et al. Study on the relationship between hepatic fibrosis and epithelial-mesenchymal transition in intrahepatic cells. Biomed. Pharmacother. 129, 110413 (2020).
CASPubMedGoogle Scholar
Taura, K. et al. Hepatocytes do not undergo epithelial-mesenchymal transition in liver fibrosis in mice. Hepatology 51, 1027–1036 (2010).
PubMedGoogle Scholar
Borthwick, L. A. The IL-1 cytokine family and its role in inflammation and fibrosis in the lung. Semin. Immunopathol. 38, 517–534 (2016).
CASPubMedGoogle Scholar
Scarpa, M. et al. The epithelial danger signal IL-1alpha is a potent activator of fibroblasts and reactivator of intestinal inflammation. Am. J. Pathol. 185, 1624–1637 (2015).
CASPubMedGoogle Scholar
Bersudsky, M. et al. Non-redundant properties of IL-1alpha and IL-1beta during acute colon inflammation in mice. Gut 63, 598–609 (2014).
CASPubMedGoogle Scholar
Lopetuso, L. R., Scaldaferri, F. & Pizarro, T. T. Emerging role of the interleukin (IL)-33/ST2 axis in gut mucosal wound healing and fibrosis. Fibrogenesis Tissue Repair 5, 18 (2012).
CASPubMedGoogle Scholar
Beltran, C. J. et al. Characterization of the novel ST2/IL-33 system in patients with inflammatory bowel disease. Inflamm. Bowel Dis. 16, 1097–1107 (2010).
PubMedGoogle Scholar
Kobori, A. et al. Interleukin-33 expression is specifically enhanced in inflamed mucosa of ulcerative colitis. J. Gastroenterol. 45, 999–1007 (2010).
CASPubMedGoogle Scholar
Flier, S. N. et al. Identification of epithelial to mesenchymal transition as a novel source of fibroblasts in intestinal fibrosis. J. Biol. Chem. 285, 20202–20212 (2010).
CASPubMedGoogle Scholar
Mostmans, Y. et al. The role of endothelial cells in the vasculopathy of systemic sclerosis: a systematic review. Autoimmun. Rev. 16, 774–786 (2017).
CASPubMedGoogle Scholar
Sgonc, R. et al. Endothelial cell apoptosis is a primary pathogenetic event underlying skin lesions in avian and human scleroderma. J. Clin. Invest. 98, 785–792 (1996).
CASPubMedGoogle Scholar
Distler, J. H. W. et al. Shared and distinct mechanisms of fibrosis. Nat. Rev. Rheumatol. 15, 705–730 (2019).
CASPubMedGoogle Scholar
Maurer, B. et al. Vascular endothelial growth factor aggravates fibrosis and vasculopathy in experimental models of systemic sclerosis. Ann. Rheum. Dis. 73, 1880–1887 (2014).
PubMedGoogle Scholar
Di Benedetto, P. et al. Endothelial-to-mesenchymal transition in systemic sclerosis. Clin. Exp. Immunol. 205, 12–27 (2021).
PubMedGoogle Scholar
Dees, C. et al. Platelet-derived serotonin links vascular disease and tissue fibrosis. J. Exp. Med. 208, 961–972 (2011).
CASPubMedGoogle Scholar
Distler, J. H. et al. Hypoxia-induced increase in the production of extracellular matrix proteins in systemic sclerosis. Arthritis Rheum. 56, 4203–4215 (2007).
CASPubMedGoogle Scholar
Aden, N. et al. Epithelial cells promote fibroblast activation via IL-1alpha in systemic sclerosis. J. Invest. Dermatol. 130, 2191–2200 (2010).
CASPubMedGoogle Scholar
Nikitorowicz-Buniak, J., Shiwen, X., Denton, C. P., Abraham, D. & Stratton, R. Abnormally differentiating keratinocytes in the epidermis of systemic sclerosis patients show enhanced secretion of CCN2 and S100A9. J. Invest. Dermatol. 134, 2693–2702 (2014).
CASPubMedGoogle Scholar
McCoy, S. S. et al. Scleroderma keratinocytes promote fibroblast activation independent of transforming growth factor beta. Rheumatology 56, 1970–1981 (2017).
CASPubMedGoogle Scholar
Distler, J. H. et al. Expression of interleukin-21 receptor in epidermis from patients with systemic sclerosis. Arthritis Rheum. 52, 856–864 (2005).
CASPubMedGoogle Scholar
Russo, B. et al. Dysfunctional keratinocytes increase dermal inflammation in systemic sclerosis: results from studies using tissue-engineered scleroderma epidermis. Arthritis Rheumatol. 73, 1311–1317 (2021).
CASPubMedGoogle Scholar
Soliman, H. et al. Multipotent stromal cells: one name, multiple identities. Cell Stem Cell 28, 1690–1707 (2021).
CASPubMedGoogle Scholar
Rognoni, E. et al. Fibroblast state switching orchestrates dermal maturation and wound healing. Mol. Syst. Biol. 14, e8174 (2018).
PubMedGoogle Scholar
Mascharak, S., desJardins-Park, H. E. & Longaker, M. T. Fibroblast heterogeneity in wound healing: hurdles to clinical translation. Trends Mol. Med. 26, 1101–1106 (2020).
CASPubMedGoogle Scholar
Ligresti, G. et al. Mesenchymal cells in the lung: evolving concepts and their role in fibrosis. Gene 859, 147142 (2023).
CASPubMedGoogle Scholar
Kuppe, C. et al. Spatial multi-omic map of human myocardial infarction. Nature 608, 766–777 (2022).
CASPubMedGoogle Scholar
Schreibing, F., Anslinger, T. M. & Kramann, R. Fibrosis in pathology of heart and kidney: from deep RNA-sequencing to novel molecular targets. Circ. Res. 132, 1013–1033 (2023).
CASPubMedGoogle Scholar
Filliol, A. et al. Opposing roles of hepatic stellate cell subpopulations in hepatocarcinogenesis. Nature 610, 356–365 (2022).
CASPubMedGoogle Scholar
Ushakumary, M. G., Riccetti, M. & Perl, A. T. Resident interstitial lung fibroblasts and their role in alveolar stem cell niche development, homeostasis, injury, and regeneration. Stem Cell Transl. Med. 10, 1021–1032 (2021).
CASGoogle Scholar
Tsukui, T. et al. Collagen-producing lung cell atlas identifies multiple subsets with distinct localization and relevance to fibrosis. Nat. Commun. 11, 1920 (2020).
CASPubMedGoogle Scholar
Kadur Lakshminarasimha Murthy, P. et al. Human distal lung maps and lineage hierarchies reveal a bipotent progenitor. Nature 604, 111–119 (2022).
CASPubMedGoogle Scholar
Ascension, A. M., Fuertes-Alvarez, S., Ibanez-Sole, O., Izeta, A. & Arauzo-Bravo, M. J. Human dermal fibroblast subpopulations are conserved across single-cell RNA sequencing studies. J. Invest. Dermatol. 141, 1735–1744 e1735 (2021).
CASPubMedGoogle Scholar
Kisseleva, T. et al. Myofibroblasts revert to an inactive phenotype during regression of liver fibrosis. Proc. Natl Acad. Sci. USA 109, 9448–9453 (2012).
CASPubMedGoogle Scholar
Yao, L. et al. Temporal control of PDGFRalpha regulates the fibroblast-to-myofibroblast transition in wound healing. Cell Rep. 40, 111192 (2022).
CASPubMedGoogle Scholar
Buechler, M. B. et al. Cross-tissue organization of the fibroblast lineage. Nature 593, 575–579 (2021).
CASPubMedGoogle Scholar
Hortells, L. et al. A specialized population of periostin-expressing cardiac fibroblasts contributes to postnatal cardiomyocyte maturation and innervation. Proc. Natl Acad. Sci. USA 117, 21469–21479 (2020).
CASPubMedGoogle Scholar
Younesi, F. S., Son, D. O., Firmino, J. & Hinz, B. Myofibroblast markers and microscopy detection methods in cell culture and histology. Methods Mol. Biol. 2299, 17–47 (2021).
CASPubMedGoogle Scholar
Kramann, R. et al. Perivascular Gli1+ progenitors are key contributors to injury-induced organ fibrosis. Cell Stem Cell 16, 51–66 (2015).
CASPubMedGoogle Scholar
Scott, R. W., Arostegui, M., Schweitzer, R., Rossi, F. M. V. & Underhill, T. M. Hic1 defines quiescent mesenchymal progenitor subpopulations with distinct functions and fates in skeletal muscle regeneration. Cell Stem Cell 25, 797–813 e799 (2019).
CASPubMedGoogle Scholar
Hagood, J. S. et al. Loss of fibroblast Thy-1 expression correlates with lung fibrogenesis. Am. J. Pathol. 167, 365–379 (2005).
CASPubMedGoogle Scholar
Fendt, B. M. et al. Protein atlas of fibroblast specific protein 1 (FSP1)/S100A4. Histol. Histopathol. 38, 1391–14001 (2023).
CASPubMedGoogle Scholar
Schuster, R., Rockel, J. S., Kapoor, M. & Hinz, B. The inflammatory speech of fibroblasts. Immunol. Rev. 302, 126–146 (2021).
CASPubMedGoogle Scholar
Sahai, E. et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer 20, 174–186 (2020).
CASPubMedGoogle Scholar
MacCarthy-Morrogh, L. & Martin, P. The hallmarks of cancer are also the hallmarks of wound healing. Sci. Signal. 13, eaay8690 (2020).
CASPubMedGoogle Scholar
Ruiz-Villalba, A. et al. Single-cell RNA sequencing analysis reveals a crucial role for CTHRC1 (collagen triple helix repeat containing 1) cardiac fibroblasts after myocardial infarction. Circulation 142, 1831–1847 (2020).
CASPubMedGoogle Scholar
Pakshir, P. et al. The myofibroblast at a glance. J. Cell Sci. 133, jcs227900 (2020).
CASPubMedGoogle Scholar
Gabbiani, G., Ryan, G. B. & Majno, G. Presence of modified fibroblasts in granulation tissue and their possible role in wound contraction. Experientia 27, 549–550 (1971).
CASPubMedGoogle Scholar
Brugger, M. D. & Basler, K. The diverse nature of intestinal fibroblasts in development, homeostasis, and disease. Trends Cell Biol. 33, 834–849 (2023).
PubMedGoogle Scholar
Younesi, F. S., Miller, A. E., Barker, T. H., Rossi, F. M. V. & Hinz, B. Fibroblast and myofibroblast activation in normal tissue repair and fibrosis. Nat. Rev. Mol. Cell Biol. 25, 617–638 (2024).
CASPubMedGoogle Scholar
de Oliveira Camargo, R., Abual’anaz, B., Rattan, S. G., Filomeno, K. L. & Dixon, I. M. C. Novel factors that activate and deactivate cardiac fibroblasts: a new perspective for treatment of cardiac fibrosis. Wound Repair Regen. 29, 667–677 (2021).
PubMedGoogle Scholar
Tacke, F., Puengel, T., Loomba, R. & Friedman, S. L. An integrated view of anti-inflammatory and antifibrotic targets for the treatment of NASH. J. Hepatol. 79, 552–566 (2023).
CASPubMedGoogle Scholar
Wang, J. et al. Novel mechanisms and clinical trial endpoints in intestinal fibrosis. Immunol. Rev. 302, 211–227 (2021).
CASPubMedGoogle Scholar
Martinez, F. J. et al. Idiopathic pulmonary fibrosis. Nat. Rev. Dis. Primers 3, 17074 (2017).
PubMedGoogle Scholar
Schuster, R., Younesi, F., Ezzo, M. & Hinz, B. The role of myofibroblasts in physiological and pathological tissue repair. Cold Spring Harb. Perspect. Biol. 15, a041231 (2023).
CASPubMedGoogle Scholar
Volkmann, E. R. & Varga, J. Emerging targets of disease-modifying therapy for systemic sclerosis. Nat. Rev. Rheumatol. 15, 208–224 (2019).
PubMedGoogle Scholar
Talbott, H. E., Mascharak, S., Griffin, M., Wan, D. C. & Longaker, M. T. Wound healing, fibroblast heterogeneity, and fibrosis. Cell Stem Cell 29, 1161–1180 (2022).
CASPubMedGoogle Scholar
Younesi, F. S. & Hinz, B. The myofibroblast fate of therapeutic mesenchymal stromal cells: regeneration, repair, or despair? Int. J. Mol. Sci. 25, 8712 (2024).
CASPubMedGoogle Scholar
Kojima, Y. et al. Autocrine TGF-beta and stromal cell-derived factor-1 (SDF-1) signaling drives the evolution of tumor-promoting mammary stromal myofibroblasts. Proc. Natl Acad. Sci. USA 107, 20009–20014 (2010).
CASPubMedGoogle Scholar
Tschumperlin, D. J. & Lagares, D. Mechano-therapeutics: targeting mechanical signaling in fibrosis and tumor stroma. Pharmacol. Ther. 212, 107575 (2020).
CASPubMedGoogle Scholar
Venugopal, H., Hanna, A., Humeres, C. & Frangogiannis, N. G. Properties and functions of fibroblasts and myofibroblasts in myocardial infarction. Cells 11, 1386 (2022).
CASPubMedGoogle Scholar
Nho, R. S., Ballinger, M. N., Rojas, M. M., Ghadiali, S. N. & Horowitz, J. C. Biomechanical force and cellular stiffness in lung fibrosis. Am. J. Pathol. 192, 750–761 (2022).
CASPubMedGoogle Scholar
Chen, K. et al. Disrupting mechanotransduction decreases fibrosis and contracture in split-thickness skin grafting. Sci. Transl. Med. 14, eabj9152 (2022).
CASPubMedGoogle Scholar
Ezzo, M. & Hinz, B. Novel approaches to target fibroblast mechanotransduction in fibroproliferative diseases. Pharmacol. Ther. 250, 108528 (2023).
CASPubMedGoogle Scholar
Lee, M., Du, H., Winer, D. A., Clemente-Casares, X. & Tsai, S. Mechanosensing in macrophages and dendritic cells in steady-state and disease. Front. Cell Dev. Biol. 10, 1044729 (2022).
PubMedGoogle Scholar
Noskovicova, N., Hinz, B. & Pakshir, P. Implant fibrosis and the underappreciated role of myofibroblasts in the foreign body reaction. Cells 10, 1794 (2021).
CASPubMedGoogle Scholar
Mao, Y. & Wickstrom, S. A. Mechanical state transitions in the regulation of tissue form and function. Nat. Rev. Mol. Cell Biol. 25, 654–670 (2024).
CASPubMedGoogle Scholar
Levine, D. et al. Expression of the integrin alpha8beta1 during pulmonary and hepatic fibrosis. Am. J. Pathol. 156, 1927–1935 (2000).
CASPubMedGoogle Scholar
Primac, I. et al. Stromal integrin alpha11 regulates PDGFR-beta signaling and promotes breast cancer progression. J. Clin. Invest. 129, 4609–4628 (2019).
CASPubMedGoogle Scholar
Nuchel, J. et al. TGFB1 is secreted through an unconventional pathway dependent on the autophagic machinery and cytoskeletal regulators. Autophagy 14, 465–486 (2018).
PubMedGoogle Scholar
Noll, N. A. et al. Loss of talin in cardiac fibroblasts results in augmented ventricular cardiomyocyte hypertrophy in response to pressure overload. Am. J. Physiol. Heart Circ. Physiol 322, H857–H866 (2022).
CASPubMedGoogle Scholar
Godbout, E. et al. Kindlin-2 mediates mechanical activation of cardiac myofibroblasts. Cells 9, 2702 (2020).
CASPubMedGoogle Scholar
Zhang, P. et al. Kindlin-2 acts as a key mediator of lung fibroblast activation and pulmonary fibrosis progression. Am. J. Respir. Cell Mol. Biol. 65, 54–69 (2021).
CASPubMedGoogle Scholar
Razinia, Z. et al. Stiffness-dependent motility and proliferation uncoupled by deletion of CD44. Sci. Rep. 7, 16499 (2017).
PubMedGoogle Scholar
Guerrot, D. et al. Discoidin domain receptor 1 is a major mediator of inflammation and fibrosis in obstructive nephropathy. Am. J. Pathol. 179, 83–91 (2011).
CASPubMedGoogle Scholar
Moll, S. et al. DDR1 role in fibrosis and its pharmacological targeting. Biochim. Biophys. Acta Mol. Cell Res. 1886, 118474 (2019).
Google Scholar
Grove, L. M. et al. Translocation of TRPV4-PI3Kgamma complexes to the plasma membrane drives myofibroblast transdifferentiation. Sci. Signal. 12, eaau1533 (2019).
CASPubMedGoogle Scholar
Sharma, S. et al. TRPV4 ion channel is a novel regulator of dermal myofibroblast differentiation. Am. J. Physiol. Cell Physiol. 312, C562–C572 (2017).
PubMedGoogle Scholar
Fu, Y. et al. Targeting mechanosensitive Piezo1 alleviated renal fibrosis through p38MAPK-YAP pathway. Front. Cell Dev. Biol. 9, 741060 (2021).
PubMedGoogle Scholar
Emig, R. et al. Piezo1 channels contribute to the regulation of human atrial fibroblast mechanical properties and matrix stiffness sensing. Cells 10, 663 (2021).
CASPubMedGoogle Scholar
Rahaman, S. O. et al. TRPV4 mediates myofibroblast differentiation and pulmonary fibrosis in mice. J. Clin. Invest. 124, 5225–5238 (2014).
PubMedGoogle Scholar
Zhang, H., Ren, L. & Shivnaraine, R. V. Targeting GPCRs to treat cardiac fibrosis. Front. Cardiovasc. Med. 9, 1011176 (2022).
CASPubMedGoogle Scholar
Zhou, Y. et al. Inhibition of mechanosensitive signaling in myofibroblasts ameliorates experimental pulmonary fibrosis. J. Clin. Invest. 123, 1096–1108 (2013).
CASPubMedGoogle Scholar
Jenkins, R. G. et al. Ligation of protease-activated receptor 1 enhances alpha(v)beta6 integrin-dependent TGF-beta activation and promotes acute lung injury. J. Clin. Invest. 116, 1606–1614 (2006).
CASPubMedGoogle Scholar
Tager, A. M. et al. The lysophosphatidic acid receptor LPA1 links pulmonary fibrosis to lung injury by mediating fibroblast recruitment and vascular leak. Nat. Med. 14, 45–54 (2008).
CASPubMedGoogle Scholar
Haak, A. J. et al. Selective YAP/TAZ inhibition in fibroblasts via dopamine receptor D1 agonism reverses fibrosis. Sci. Transl. Med. 11, eaau6296 (2019).
CASPubMedGoogle Scholar
Dooling, L. J. & Discher, D. E. Inhibiting tumor fibrosis and actomyosin through GPCR activation. Trends Cancer 5, 197–199 (2019).
CASPubMedGoogle Scholar
Piccolo, S., Panciera, T., Contessotto, P. & Cordenonsi, M. YAP/TAZ as master regulators in cancer: modulation, function and therapeutic approaches. Nat. Cancer 4, 9–26 (2023).
CASPubMedGoogle Scholar
He, X. et al. Myofibroblast YAP/TAZ activation is a key step in organ fibrogenesis. JCI Insight 7, e146243 (2022).
PubMedGoogle Scholar
Kofler, M. et al. Mediated nuclear import and export of TAZ and the underlying molecular requirements. Nat. Commun. 9, 4966 (2018).
PubMedGoogle Scholar
Masszi, A. et al. Fate-determining mechanisms in epithelial-myofibroblast transition: major inhibitory role for Smad3. J. Cell Biol. 188, 383–399 (2010).
CASPubMedGoogle Scholar
Yu-Wai-Man, C. et al. Local delivery of novel MRTF/SRF inhibitors prevents scar tissue formation in a preclinical model of fibrosis. Sci. Rep. 7, 518 (2017).
Google Scholar
Leal, A. S., Misek, S. A., Lisabeth, E. M., Neubig, R. R. & Liby, K. T. The Rho/MRTF pathway inhibitor CCG-222740 reduces stellate cell activation and modulates immune cell populations in Kras(G12D); Pdx1-Cre (KC) mice. Sci. Rep. 9, 7072 (2019).
PubMedGoogle Scholar
Johnson, L. A. et al. Novel Rho/MRTF/SRF inhibitors block matrix-stiffness and TGF-beta-induced fibrogenesis in human colonic myofibroblasts. Inflamm. Bowel Dis. 20, 154–165 (2014).
PubMedGoogle Scholar
Link, P. A. et al. Combined control of the fibroblast contractile program by YAP and TAZ. Am. J. Physiol. Lung Cell. Mol. Physiol. 322, L23–L32 (2022).
CASPubMedGoogle Scholar
Reggiani, F., Gobbi, G., Ciarrocchi, A. & Sancisi, V. YAP and TAZ are not identical twins. Trends Biochem. Sci. 46, 154–168 (2021).
CASPubMedGoogle Scholar
Szeto, S. G. et al. YAP/TAZ are mechanoregulators of TGF-beta-smad signaling and renal fibrogenesis. J. Am. Soc. Nephrol. 27, 3117–3128 (2016).
CASPubMedGoogle Scholar
Piersma, B. et al. YAP1 is a driver of myofibroblast differentiation in normal and diseased fibroblasts. Am. J. Pathol. 185, 3326–3337 (2015).
CASPubMedGoogle Scholar
Santos, D. M. et al. Screening for YAP inhibitors identifies statins as modulators of fibrosis. Am. J. Respir. Cell Mol. Biol. 62, 479–492 (2020).
CASPubMedGoogle Scholar
Shi, M. et al. Latent TGF-beta structure and activation. Nature 474, 343–349 (2011).
CASPubMedGoogle Scholar
Buscemi, L. et al. The single-molecule mechanics of the latent TGF-beta1 complex. Curr. Biol. 21, 2046–2054 (2011).
CASPubMedGoogle Scholar
Wipff, P. J., Rifkin, D. B., Meister, J. J. & Hinz, B. Myofibroblast contraction activates latent TGF-beta1 from the extracellular matrix. J. Cell Biol. 179, 1311–1323 (2007).
CASPubMedGoogle Scholar
Henderson, N. C. et al. Targeting of αv integrin identifies a core molecular pathway that regulates fibrosis in several organs. Nat. Med. 19, 1617–1624 (2013).
CASPubMedGoogle Scholar
Annes, J. P., Chen, Y., Munger, J. S. & Rifkin, D. B. Integrin αVβ6-mediated activation of latent TGF-β requires the latent TGF-β binding protein-1. J. Cell Biol. 165, 723–734 (2004).
CASPubMedGoogle Scholar
Lodyga, M. & Hinz, B. TGF-beta1 - a truly transforming growth factor in fibrosis and immunity. Semin. Cell Dev. Biol. 101, 123–139 (2020).
CASPubMedGoogle Scholar
Klingberg, F. et al. Prestress in the extracellular matrix sensitizes latent TGF-beta1 for activation. J. Cell Biol. 207, 283–297 (2014).
CASPubMedGoogle Scholar
Massague, J. & Sheppard, D. TGF-beta signaling in health and disease. Cell 186, 4007–4037 (2023).
CASPubMedGoogle Scholar
Reed, N. I. et al. The αvβ1 integrin plays a critical in vivo role in tissue fibrosis. Sci. Transl. Med. 7, 288ra279 (2015).
Google Scholar
Lancaster, L. H. PLN-74809 shows favorable safety and tolerability and indicates antifibrotic activity in a phase 2a study for the treatment of idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 207, A2777 (2023).
Google Scholar
Wirth, M. et al. A multicenter phase 1 study of EMD 525797 (DI17E6), a novel humanized monoclonal antibody targeting alphav integrins, in progressive castration-resistant prostate cancer with bone metastases after chemotherapy. Eur. Urol. 65, 897–904 (2014).
CASPubMedGoogle Scholar
Hussain, M. et al. Differential effect on bone lesions of targeting integrins: randomized phase ii trial of abituzumab in patients with metastatic castration-resistant prostate cancer. Clin. Cancer Res. 22, 3192–3200 (2016).
CASPubMedGoogle Scholar
Distler, O. et al. Nintedanib for systemic sclerosis-associated interstitial lung disease. N. Engl. J. Med. 380, 2518–2528 (2019).
CASPubMedGoogle Scholar
Richeldi, L. et al. Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N. Engl. J. Med. 370, 2071–2082 (2014).
PubMedGoogle Scholar
Sun, Y. W. et al. Pirfenidone prevents radiation-induced intestinal fibrosis in rats by inhibiting fibroblast proliferation and differentiation and suppressing the TGF-beta1/Smad/CTGF signaling pathway. Eur. J. Pharmacol. 822, 199–206 (2018).
CASPubMedGoogle Scholar
King, T. E. Jr. et al. A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N. Engl. J. Med. 370, 2083–2092 (2014).
PubMedGoogle Scholar
Bhattacharyya, S. et al. Pharmacological inhibition of toll-like receptor-4 signaling by TAK242 prevents and induces regression of experimental organ fibrosis. Front. Immunol. 9, 2434 (2018).
PubMedGoogle Scholar
Schuppan, D., Ashfaq-Khan, M., Yang, A. T. & Kim, Y. O. Liver fibrosis: direct antifibrotic agents and targeted therapies. Matrix Biol. 68-69, 435–451 (2018).
CASPubMedGoogle Scholar
Sato, S., Yanagihara, T. & Kolb, M. R. J. Therapeutic targets and early stage clinical trials for pulmonary fibrosis. Expert Opin. Invest. Drugs 28, 19–28 (2019).
CASGoogle Scholar
O’Reilly, S. Interleukin-11 and its eminent role in tissue fibrosis: a possible therapeutic target. Clin. Exp. Immunol. 214, 154–161 (2023).
PubMedGoogle Scholar
Dolivo, D. M., Larson, S. A. & Dominko, T. Fibroblast growth factor 2 as an antifibrotic: antagonism of myofibroblast differentiation and suppression of pro-fibrotic gene expression. Cytokine Growth Factor Rev. 38, 49–58 (2017).
CASPubMedGoogle Scholar
Ornitz, D. M. & Itoh, N. New developments in the biology of fibroblast growth factors. WIREs Mech. Dis. 14, e1549 (2022).
CASPubMedGoogle Scholar
Akhmetshina, A. et al. The cannabinoid receptor CB2 exerts antifibrotic effects in experimental dermal fibrosis. Arthritis Rheum. 60, 1129–1136 (2009).
PubMedGoogle Scholar
Balistreri, E. et al. The cannabinoid WIN55, 212-2 abrogates dermal fibrosis in scleroderma bleomycin model. Ann. Rheum. Dis. 70, 695–699 (2011).
CASPubMedGoogle Scholar
Spiera, R. et al. Safety and efficacy of lenabasum in a phase II, randomized, placebo-controlled trial in adults with systemic sclerosis. Arthritis Rheumatol. 72, 1350–1360 (2020).
CASPubMedGoogle Scholar
Widjaja, A. A. et al. Molecular dissection of pro-fibrotic IL11 signaling in cardiac and pulmonary fibroblasts. Front. Mol. Biosci. 8, 740650 (2021).
CASPubMedGoogle Scholar
Corden, B., Adami, E., Sweeney, M., Schafer, S. & Cook, S. A. IL-11 in cardiac and renal fibrosis: late to the party but a central player. Br. J. Pharmacol. 177, 1695–1708 (2020).
CASPubMedGoogle Scholar
Schafer, S. et al. IL-11 is a crucial determinant of cardiovascular fibrosis. Nature 552, 110–115 (2017).
CASPubMedGoogle Scholar
Gieseck, R. L. 3rd, Wilson, M. S. & Wynn, T. A. Type 2 immunity in tissue repair and fibrosis. Nat. Rev. Immunol. 18, 62–76 (2018).
CASPubMedGoogle Scholar
Lee, C. G. et al. Interleukin-13 induces tissue fibrosis by selectively stimulating and activating transforming growth factor beta(1). J. Exp. Med. 194, 809–821 (2001).
CASPubMedGoogle Scholar
Gieseck, R. L. 3rd et al. Interleukin-13 activates distinct cellular pathways leading to ductular reaction, steatosis, and fibrosis. Immunity 45, 145–158 (2016).
CASPubMedGoogle Scholar
Hart, K. M. et al. Type 2 immunity is protective in metabolic disease but exacerbates NAFLD collaboratively with TGF-beta. Sci. Transl. Med. 9, eaal3694 (2017).
PubMedGoogle Scholar
Xue, J. et al. Alternatively activated macrophages promote pancreatic fibrosis in chronic pancreatitis. Nat. Commun. 6, 7158 (2015).
CASPubMedGoogle Scholar
Adler, M. et al. Principles of cell circuits for tissue repair and fibrosis. iScience 23, 100841 (2020).
CASPubMedGoogle Scholar
Buechler, M. B., Fu, W. & Turley, S. J. Fibroblast-macrophage reciprocal interactions in health, fibrosis, and cancer. Immunity 54, 903–915 (2021).
CASPubMedGoogle Scholar
Wynn, T. A. & Vannella, K. M. Macrophages in tissue repair, regeneration, and fibrosis. Immunity 44, 450–462 (2016).
CASPubMedGoogle Scholar
Eming, S. A., Wynn, T. A. & Martin, P. Inflammation and metabolism in tissue repair and regeneration. Science 356, 1026–1030 (2017).
CASPubMedGoogle Scholar
Minutti, C. M., Knipper, J. A., Allen, J. E. & Zaiss, D. M. Tissue-specific contribution of macrophages to wound healing. Semin. Cell Dev. Biol. 61, 3–11 (2017).
CASPubMedGoogle Scholar
Shook, B. A. et al. Myofibroblast proliferation and heterogeneity are supported by macrophages during skin repair. Science 362, eaar2971 (2018).
PubMedGoogle Scholar
Mirza, R., DiPietro, L. A. & Koh, T. J. Selective and specific macrophage ablation is detrimental to wound healing in mice. Am. J. Pathol. 175, 2454–2462 (2009).
CASPubMedGoogle Scholar
MacLeod, A. S. & Mansbridge, J. N. The innate immune system in acute and chronic wounds. Adv. Wound Care 5, 65–78 (2016).
Google Scholar
Lavin, Y. et al. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 159, 1312–1326 (2014).
CASPubMedGoogle Scholar
Braga, T. T., Agudelo, J. S. & Camara, N. O. Macrophages during the fibrotic process: M2 as friend and foe. Front. Immunol. 6, 602 (2015).
PubMedGoogle Scholar
Misharin, A. V. et al. Monocyte-derived alveolar macrophages drive lung fibrosis and persist in the lung over the life span. J. Exp. Med. 214, 2387–2404 (2017).
CASPubMedGoogle Scholar
Forbes, S. J. & Rosenthal, N. Preparing the ground for tissue regeneration: from mechanism to therapy. Nat. Med. 20, 857–869 (2014).
CASPubMedGoogle Scholar
Vannella, K. M. & Wynn, T. A. Mechanisms of organ injury and repair by macrophages. Annu. Rev. Physiol. 79, 593–617 (2017).
CASPubMedGoogle Scholar
Fritz, J. M. et al. Depletion of tumor-associated macrophages slows the growth of chemically induced mouse lung adenocarcinomas. Front. Immunol. 5, 587 (2014).
PubMedGoogle Scholar
Duffield, J. S., Lupher, M., Thannickal, V. J. & Wynn, T. A. Host responses in tissue repair and fibrosis. Annu. Rev. Pathol. 8, 241–276 (2013).
CASPubMedGoogle Scholar
Morse, C. et al. Proliferating SPP1/MERTK-expressing macrophages in idiopathic pulmonary fibrosis. Eur. Respir. J. 54, 1802441 (2019).
CASPubMedGoogle Scholar
Bhattacharya, M. & Ramachandran, P. Immunology of human fibrosis. Nat. Immunol. 24, 1423–1433 (2023).
CASPubMedGoogle Scholar
Ramachandran, P. et al. Resolving the fibrotic niche of human liver cirrhosis at single-cell level. Nature 575, 512–518 (2019).
CASPubMedGoogle Scholar
Hoeft, K. et al. Platelet-instructed SPP1+ macrophages drive myofibroblast activation in fibrosis in a CXCL4-dependent manner. Cell Rep. 42, 112131 (2023).
CASPubMedGoogle Scholar
Fabre, T. et al. Identification of a broadly fibrogenic macrophage subset induced by type 3 inflammation. Sci. Immunol. 8, eadd8945 (2023).
CASPubMedGoogle Scholar
Pakshir, P. et al. Dynamic fibroblast contractions attract remote macrophages in fibrillar collagen matrix. Nat. Commun. 10, 1850 (2019).
PubMedGoogle Scholar
Lodyga, M. et al. Cadherin-11-mediated adhesion of macrophages to myofibroblasts establishes a profibrotic niche of active TGF-beta. Sci. Signal. 12, eaao3469 (2019).
CASPubMedGoogle Scholar
Tang, P. M., Nikolic-Paterson, D. J. & Lan, H. Y. Macrophages: versatile players in renal inflammation and fibrosis. Nat. Rev. Nephrol. 15, 144–158 (2019).
PubMedGoogle Scholar
Guillot, A. & Tacke, F. Liver macrophages revisited: the expanding universe of versatile responses in a spatiotemporal context. Hepatol. Commun. 8, e0491 (2024).
PubMedGoogle Scholar
Long, H. et al. Macrophages and fibrosis: how resident and infiltrating mononuclear phagocytes account for organ injury, regeneration or atrophy. Front. Immunol. 14, 1194988 (2023).
CASPubMedGoogle Scholar
Kinchen, J. et al. Structural remodeling of the human colonic mesenchyme in inflammatory bowel disease. Cell 175, 372–386.e317 (2018).
CASPubMedGoogle Scholar
Smolgovsky, S., Theall, B., Wagner, N. & Alcaide, P. Fibroblasts and immune cells: at the crossroad of organ inflammation and fibrosis. Am. J. Physiol. Heart Circ. Physiol 326, H303–H316 (2024).
CASPubMedGoogle Scholar
Lopez, B. et al. Diffuse myocardial fibrosis: mechanisms, diagnosis and therapeutic approaches. Nat. Rev. Cardiol. 18, 479–498 (2021).
PubMedGoogle Scholar
Ravassa, S. et al. Cardiac fibrosis in heart failure: focus on non-invasive diagnosis and emerging therapeutic strategies. Mol. Asp. Med. 93, 101194 (2023).
CASGoogle Scholar
Travers, J. G., Tharp, C. A., Rubino, M. & McKinsey, T. A. Therapeutic targets for cardiac fibrosis: from old school to next-gen. J. Clin. Invest. 132, e148554 (2022).
CASPubMedGoogle Scholar
Wijsenbeek, M., Suzuki, A. & Maher, T. M. Interstitial lung diseases. Lancet 400, 769–786 (2022).
PubMedGoogle Scholar
Kaul, B. et al. Epidemiology of idiopathic pulmonary fibrosis among U.S. Veterans, 2010-2019. Ann. Am. Thorac. Soc. 19, 196–203 (2022).
PubMedGoogle Scholar
Arimura-Omori, M. et al. Association between telomere-related polymorphisms and the risk of IPF and COPD as a precursor lesion of lung cancer: findings from the fukuoka tobacco-related lung disease (FOLD) registry. Asian Pac. J. Cancer Prev. 21, 667–673 (2020).
CASPubMedGoogle Scholar
Korthagen, N. M. et al. Association between variations in cell cycle genes and idiopathic pulmonary fibrosis. PLoS ONE 7, e30442 (2012).
CASPubMedGoogle Scholar
Juge, P. A. et al. MUC5B promoter variant and rheumatoid arthritis with interstitial lung disease. N. Engl. J. Med. 379, 2209–2219 (2018).
CASPubMedGoogle Scholar
Furusawa, H. et al. Common idiopathic pulmonary fibrosis risk variants are associated with hypersensitivity pneumonitis. Thorax 77, 508–510 (2022).
PubMedGoogle Scholar
Pauchet, A. et al. Idiopathic pulmonary fibrosis: what do we know about the role of occupational and environmental determinants? A systematic literature review and meta-analysis. J. Toxicol. Env. Health B Crit. Rev. 25, 372–392 (2022).
CASGoogle Scholar
Lee, J. S. et al. Molecular markers of telomere dysfunction and senescence are common findings in the usual interstitial pneumonia pattern of lung fibrosis. Histopathology 79, 67–76 (2021).
PubMedGoogle Scholar
Allden, S. J. et al. The transferrin receptor CD71 delineates functionally distinct airway macrophage subsets during idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 200, 209–219 (2019).
CASPubMedGoogle Scholar
Asano, S. et al. Matrix stiffness regulates migration of human lung fibroblasts. Physiol. Rep. 5, e13281 (2017).
PubMedGoogle Scholar
Valenzi, E. et al. Disparate interferon signaling and shared aberrant basaloid cells in single-cell profiling of idiopathic pulmonary fibrosis and systemic sclerosis-associated interstitial lung disease. Front. Immunol. 12, 595811 (2021).
CASPubMedGoogle Scholar
Khanna, D. et al. Etiology, risk factors, and biomarkers in systemic sclerosis with interstitial lung disease. Am. J. Respir. Crit. Care Med. 201, 650–660 (2020).
CASPubMedGoogle Scholar
Renzoni, E. A. et al. Interstitial vascularity in fibrosing alveolitis. Am. J. Respir. Crit. Care Med. 167, 438–443 (2003).
PubMedGoogle Scholar
Yanagihara, T. et al. Vascular-parenchymal cross-talk promotes lung fibrosis through BMPR2 signaling. Am. J. Respir. Crit. Care Med. 207, 1498–1514 (2023).
CASPubMedGoogle Scholar
Fliesser, E. et al. Lung fibrosis is linked to increased endothelial cell activation and dysfunctional vascular barrier integrity. Am. J. Respir. Cell Mol. Biol. 71, 318–331 (2024).
CASPubMedGoogle Scholar
Waxman, A. et al. Inhaled treprostinil in pulmonary hypertension due to interstitial lung disease. N. Engl. J. Med. 384, 325–334 (2021).
CASPubMedGoogle Scholar
Ask, K. et al. Comparison between conventional and “clinical” assessment of experimental lung fibrosis. J. Transl. Med. 6, 16 (2008).
PubMedGoogle Scholar
Maher, T. M. Interstitial lung disease: a review. JAMA 331, 1655–1665 (2024).
CASPubMedGoogle Scholar
Xu, F. et al. The transition from normal lung anatomy to minimal and established fibrosis in idiopathic pulmonary fibrosis (IPF). EBioMedicine 66, 103325 (2021).
CASPubMedGoogle Scholar
Raghu, G. et al. Idiopathic pulmonary fibrosis (an Update) and progressive pulmonary fibrosis in adults: an official ATS/ERS/JRS/ALAT clinical practice guideline. Am. J. Respir. Crit. Care Med. 205, e18–e47 (2022).
PubMedGoogle Scholar
Walsh, S. L. F., Calandriello, L., Silva, M. & Sverzellati, N. Deep learning for classifying fibrotic lung disease on high-resolution computed tomography: a case-cohort study. Lancet Respir. Med. 6, 837–845 (2018).
PubMedGoogle Scholar
Wells, A. U. et al. Idiopathic pulmonary fibrosis: a composite physiologic index derived from disease extent observed by computed tomography. Am. J. Respir. Crit Care Med. 167, 962–969 (2003).
PubMedGoogle Scholar
Zappala, C. J. et al. Marginal decline in forced vital capacity is associated with a poor outcome in idiopathic pulmonary fibrosis. Eur. Respir. J. 35, 830–836 (2010).
CASPubMedGoogle Scholar
Karimi-Shah, B. A. & Chowdhury, B. A. Forced vital capacity in idiopathic pulmonary fibrosis-FDA review of pirfenidone and nintedanib. N. Engl. J. Med. 372, 1189–1191 (2015).
PubMedGoogle Scholar
Raghu, G. et al. Meaningful endpoints for idiopathic pulmonary fibrosis (IPF) clinical trials: emphasis on ‘feels, functions, survives’. Am. J. Respir. Crit. Care Med. 209, 647–669 (2024).
PubMedGoogle Scholar
Wu, X. et al. Computed tomographic biomarkers in idiopathic pulmonary fibrosis. the future of quantitative analysis. Am. J. Respir. Crit. Care Med. 199, 12–21 (2019).
PubMedGoogle Scholar
Jenkins, R. G. et al. Longitudinal change in collagen degradation biomarkers in idiopathic pulmonary fibrosis: an analysis from the prospective, multicentre PROFILE study. Lancet Respir. Med. 3, 462–472 (2015).
CASPubMedGoogle Scholar
Maher, T. M. et al. An epithelial biomarker signature for idiopathic pulmonary fibrosis: an analysis from the multicentre PROFILE cohort study. Lancet Respir. Med. 5, 946–955 (2017).
CASPubMedGoogle Scholar
Organ, L. A. et al. Biomarkers of collagen synthesis predict progression in the PROFILE idiopathic pulmonary fibrosis cohort. Respir. Res. 20, 148 (2019).
PubMedGoogle Scholar
Rosas, I. O. et al. MMP1 and MMP7 as potential peripheral blood biomarkers in idiopathic pulmonary fibrosis. PLoS Med. 5, e93 (2008).
PubMedGoogle Scholar
Prasse, A. et al. Serum CC-chemokine ligand 18 concentration predicts outcome in idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 179, 717–723 (2009).
CASPubMedGoogle Scholar
Maher, T. M. et al. Biomarkers of extracellular matrix turnover in patients with idiopathic pulmonary fibrosis given nintedanib (INMARK study): a randomised, placebo-controlled study. Lancet Respir. Med. 7, 771–779 (2019).
CASPubMedGoogle Scholar
Ma, H. Y. et al. Inhibition of MRTF activation as a clinically achievable anti-fibrotic mechanism for pirfenidone. Eur. Respir. J. 61, 2200604 (2023).
CASPubMedGoogle Scholar
Wollin, L. et al. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur. Respir. J. 45, 1434–1445 (2015).
CASPubMedGoogle Scholar
Maher, T. M. et al. Phase 2 trial to assess lebrikizumab in patients with idiopathic pulmonary fibrosis. Eur. Respir. J. 57, 1902442 (2021).
CASPubMedGoogle Scholar
Maher, T. M. et al. Ziritaxestat, a novel autotaxin inhibitor, and lung function in idiopathic pulmonary fibrosis: the ISABELA 1 and 2 randomized clinical trials. JAMA 329, 1567–1578 (2023).
CASPubMedGoogle Scholar
Richeldi, L. et al. Trial of a preferential phosphodiesterase 4B inhibitor for idiopathic pulmonary fibrosis. N. Engl. J. Med. 386, 2178–2187 (2022).
CASPubMedGoogle Scholar
Lancaster, L. et al. Bexotegrast in patients with idiopathic pulmonary fibrosis: the INTEGRIS-IPF clinical trial. Am. J. Respir. Crit. Care Med. 210, 424–434 (2024).
CASPubMedGoogle Scholar
Corte, T. J. et al. Efficacy and safety of admilparant, an LPA(1) antagonist in pulmonary fibrosis: a phase 2 randomized clinical trial. Am. J. Respir. Crit. Care Med. 211, 230–238 (2024).
Google Scholar
Herrmann, F. E., Hesslinger, C., Wollin, L. & Nickolaus, P. BI 1015550 is a PDE4B inhibitor and a clinical drug candidate for the oral treatment of idiopathic pulmonary fibrosis. Front. Pharmacol. 13, 838449 (2022).
CASPubMedGoogle Scholar
Kolb, M. et al. The antifibrotic effects of inhaled treprostinil: an emerging option for ILD. Adv. Ther. 39, 3881–3895 (2022).
CASPubMedGoogle Scholar
Rieder, F., Mukherjee, P. K., Massey, W. J., Wang, Y. & Fiocchi, C. Fibrosis in IBD: from pathogenesis to therapeutic targets. Gut 73, 854–866 (2024).
CASGoogle Scholar
Gordon, I. O. et al. Fibrosis in ulcerative colitis is directly linked to severity and chronicity of mucosal inflammation. Aliment. Pharmacol. Ther. 47, 922–939 (2018).
CASPubMedGoogle Scholar
Jarmakiewicz-Czaja, S., Zielinska, M., Sokal, A. & Filip, R. Genetic and epigenetic etiology of inflammatory bowel disease: an update. Genes 13, 2388 (2022).
CASPubMedGoogle Scholar
Steiner, C. A. et al. Biomarkers for the prediction and diagnosis of fibrostenosing Crohn’s disease: a systematic review. Clin. Gastroenterol. Hepatol. 20, 817–846.e10 (2021).
PubMedGoogle Scholar
Adler, J. et al. Computed tomography enterography findings correlate with tissue inflammation, not fibrosis in resected small bowel Crohn’s disease. Inflamm. Bowel Dis. 18, 849–856 (2011).
PubMedGoogle Scholar
Farmer, R. G., Whelan, G. & Fazio, V. W. Long-term follow-up of patients with Crohn’s disease. Relationship between the clinical pattern and prognosis. Gastroenterology 88, 1818–1825 (1985).
CASPubMedGoogle Scholar
Smillie, C. S. et al. Intra- and inter-cellular rewiring of the human colon during ulcerative colitis. Cell 178, 714–730.e722 (2019).
CASPubMedGoogle Scholar
Lim, W. W. et al. Transgenic interleukin 11 expression causes cross-tissue fibro-inflammation and an inflammatory bowel phenotype in mice. PLoS ONE 15, e0227505 (2020).
CASPubMedGoogle Scholar
Imai, J. et al. Flagellin-mediated activation of IL-33-ST2 signaling by a pathobiont promotes intestinal fibrosis. Mucosal Immunol. 12, 632–643 (2019).
CASPubMedGoogle Scholar
Franze, E. et al. Interleukin-34 stimulates gut fibroblasts to produce collagen synthesis. J. Crohns Colitis 14, 1436–1445 (2020).
PubMedGoogle Scholar
Scheibe, K. et al. Inhibiting interleukin 36 receptor signaling reduces fibrosis in mice with chronic intestinal inflammation. Gastroenterology 156, 1082–1097.e1011 (2019).
CASPubMedGoogle Scholar
Shih, D. Q. et al. Constitutive TL1A (TNFSF15) expression on lymphoid or myeloid cells leads to mild intestinal inflammation and fibrosis. PLoS ONE 6, e16090 (2011).
CASPubMedGoogle Scholar
Jacob, N. et al. Inflammation-independent TL1A-mediated intestinal fibrosis is dependent on the gut microbiome. Mucosal Immunol. 11, 1466–1476 (2018).
CASPubMedGoogle Scholar
Wang, J. et al. Preventing fibrosis in IBD: update on immune pathways and clinical strategies. Expert Rev. Clin. Immunol. 20, 727–734 (2024).
CASPubMedGoogle Scholar
Zhao, S. et al. Selective deletion of MyD88 signaling in alpha-SMA positive cells ameliorates experimental intestinal fibrosis via post-transcriptional regulation. Mucosal Immunol. 13, 665–678 (2020).
CASPubMedGoogle Scholar
Mao, R. et al. Activated intestinal muscle cells promote preadipocyte migration: a novel mechanism for creeping fat formation in Crohn’s disease. Gut 71, 55–67 (2022).
CASPubMedGoogle Scholar
Mao, R. et al. The mesenteric fat and intestinal muscle interface: creeping fat influencing stricture formation in Crohn’s disease. Inflamm. Bowel Dis. 25, 421–426 (2019).
PubMedGoogle Scholar
Chen, W. et al. Smooth muscle hyperplasia/hypertrophy is the most prominent histological change in Crohn’s fibrostenosing bowel strictures: a semiquantitative analysis by using a novel histological grading scheme. J. Crohns Colitis 11, 92–104 (2017).
PubMedGoogle Scholar
Mukherjee, P. K. et al. Stricturing Crohn’s disease single-cell RNA sequencing reveals fibroblast heterogeneity and intercellular interactions. Gastroenterology 165, 1180–1196 (2023).
CASPubMedGoogle Scholar
Rieder, F. et al. Inflammation-induced endothelial-to-mesenchymal transition: a novel mechanism of intestinal fibrosis. Am. J. Pathol. 179, 2660–2673 (2012).
Google Scholar
Yamamoto, T., Fazio, V. W. & Tekkis, P. P. Safety and efficacy of strictureplasty for Crohn’s disease: a systematic review and meta-analysis. Dis. Colon Rectum 50, 1968–1986 (2007).
PubMedGoogle Scholar
Fazio, V. W. et al. Long-term follow-up of strictureplasty in Crohn’s disease. Dis. Colon Rectum 36, 355–361 (1993).
CASPubMedGoogle Scholar
Maconi, G. et al. Preoperative characteristics and postoperative behavior of bowel wall on risk of recurrence after conservative surgery in Crohn’s disease: a prospective study. Ann. Surg. 233, 345–352 (2001).
CASPubMedGoogle Scholar
Bettenworth, D. & Rieder, F. Reversibility of stricturing Crohn’s disease — fact or fiction? Inflamm. Bowel Dis. 22, 241–247 (2016).
PubMedGoogle Scholar
Rieder, F. et al. An expert consensus to standardise definitions, diagnosis and treatment targets for anti-fibrotic stricture therapies in Crohn’s disease. Aliment. Pharmacol. Ther. 48, 347–357 (2018).
CASPubMedGoogle Scholar
Bettenworth, D. et al. Assessment of Crohn’s disease-associated small bowel strictures and fibrosis on cross-sectional imaging: a systematic review. Gut 68, 1115–1126 (2019).
CASPubMedGoogle Scholar
Rieder, F. et al. Reliability of MR enterography features for describing fibrostenosing Crohn disease. Radiology 312, e233039 (2024).
PubMedGoogle Scholar
Rieder, F. et al. Reliability of CT enterography for describing fibrostenosing Crohn disease. Radiology 312, e233038 (2024).
PubMedGoogle Scholar
Rimola, J. et al. Characterization of inflammation and fibrosis in Crohn’s disease lesions by magnetic resonance imaging. Am. J. Gastroenterol. 110, 432–440 (2015).
PubMedGoogle Scholar
Dillman, J. R. et al. US elastography-derived shear wave velocity helps distinguish acutely inflamed from fibrotic bowel in a Crohn disease animal model. Radiology 267, 757–766 (2013).
PubMedGoogle Scholar
Pazahr, S. et al. Magnetization transfer for the assessment of bowel fibrosis in patients with Crohn’s disease: initial experience. MAGMA 26, 291–301 (2013).
CASPubMedGoogle Scholar
Li, X. H. et al. Characterization of degree of intestinal fibrosis in patients with Crohn disease by using magnetization transfer MR imaging. Radiology 287, 494–503 (2018).
PubMedGoogle Scholar
Chiorean, M. V. et al. Correlation of CT enteroclysis with surgical pathology in Crohn’s disease. Am. J. Gastroenterol. 102, 2541–2550 (2007).
PubMedGoogle Scholar
Jensen, M. D., Kjeldsen, J., Rafaelsen, S. R. & Nathan, T. Diagnostic accuracies of MR enterography and CT enterography in symptomatic Crohn’s disease. Scand. J. Gastroenterol. 46, 1449–1457 (2011).
PubMedGoogle Scholar
Guyatt, G. H., Kirshner, B. & Jaeschke, R. Measuring health status: what are the necessary measurement properties? J. Clin. Epidemiol. 45, 1341–1345 (1992).
CASPubMedGoogle Scholar
Gordon, I. O. et al. Histopathology scoring systems of stenosis associated with small bowel Crohn’s disease: a systematic review. Gastroenterology 158, 137–150.e131 (2020).
PubMedGoogle Scholar
Li, P. et al. Histopathologic correlates of kidney function: insights from nephrectomy specimens. Am. J. Kidney Dis. 77, 336–345 (2021).
PubMedGoogle Scholar
Tziastoudi, M. et al. Key genetic components of fibrosis in diabetic nephropathy: an updated systematic review and meta-analysis. Int. J. Mol. Sci. 23, 15331 (2022).
CASPubMedGoogle Scholar
Corredor, Z. et al. Genetic variants associated with chronic kidney disease in a Spanish population. Sci. Rep. 10, 144 (2020).
CASPubMedGoogle Scholar
Fountoglou, A., Deltas, C., Siomou, E. & Dounousi, E. Genome-wide association studies reconstructing chronic kidney disease. Nephrol. Dial. Transpl. 39, 395–402 (2024).
CASGoogle Scholar
Kuppe, C. et al. Decoding myofibroblast origins in human kidney fibrosis. Nature 589, 281–286 (2021).
CASPubMedGoogle Scholar
Kida, Y., Ieronimakis, N., Schrimpf, C., Reyes, M. & Duffield, J. S. EphrinB2 reverse signaling protects against capillary rarefaction and fibrosis after kidney injury. J. Am. Soc. Nephrol. 24, 559–572 (2013).
CASPubMedGoogle Scholar
Duffield, J. S. Cellular and molecular mechanisms in kidney fibrosis. J. Clin. Invest. 124, 2299–2306 (2014).
CASPubMedGoogle Scholar
Kramann, R., Tanaka, M. & Humphreys, B. D. Fluorescence microangiography for quantitative assessment of peritubular capillary changes after AKI in mice. J. Am. Soc. Nephrol. 25, 1924–1931 (2014).
PubMedGoogle Scholar
Kramann, R., Wongboonsin, J., Chang-Panesso, M., Machado, F. G. & Humphreys, B. D. Gli1+ pericyte loss induces capillary rarefaction and proximal tubular injury. J. Am. Soc. Nephrol. 28, 776–784 (2017).
CASPubMedGoogle Scholar
Babickova, J. et al. Regardless of etiology, progressive renal disease causes ultrastructural and functional alterations of peritubular capillaries. Kidney Int. 91, 70–85 (2017).
CASPubMedGoogle Scholar
Fioretto, P., Steffes, M. W., Sutherland, D. E., Goetz, F. C. & Mauer, M. Reversal of lesions of diabetic nephropathy after pancreas transplantation. N. Engl. J. Med. 339, 69–75 (1998).
CASPubMedGoogle Scholar
Adamczak, M. et al. Reversal of glomerulosclerosis after high-dose enalapril treatment in subtotally nephrectomized rats. J. Am. Soc. Nephrol. 14, 2833–2842 (2003).
CASPubMedGoogle Scholar
Ferguson, C. M. et al. Renal fibrosis detected by diffusion-weighted magnetic resonance imaging remains unchanged despite treatment in subjects with renovascular disease. Sci. Rep. 10, 16300 (2020).
CASPubMedGoogle Scholar
Cho, M. E., Smith, D. C., Branton, M. H., Penzak, S. R. & Kopp, J. B. Pirfenidone slows renal function decline in patients with focal segmental glomerulosclerosis. Clin. J. Am. Soc. Nephrol. 2, 906–913 (2007).
CASPubMedGoogle Scholar
Sharma, K. et al. Pirfenidone for diabetic nephropathy. J. Am. Soc. Nephrol. 22, 1144–1151 (2011).
CASPubMedGoogle Scholar
Vincenti, F. et al. A phase 2, double-blind, placebo-controlled, randomized study of fresolimumab in patients with steroid-resistant primary focal segmental glomerulosclerosis. Kidney Int. Rep. 2, 800–810 (2017).
PubMedGoogle Scholar
Voelker, J. et al. Anti-TGF-beta1 antibody therapy in patients with diabetic nephropathy. J. Am. Soc. Nephrol. 28, 953–962 (2017).
CASPubMedGoogle Scholar
Sun, Q. et al. Elastin imaging enables noninvasive staging and treatment monitoring of kidney fibrosis. Sci. Transl. Med. 11, eaat4865 (2019).
CASPubMedGoogle Scholar
Denton, C. P. & Khanna, D. Systemic sclerosis. Lancet 390, 1685–1699 (2017).
PubMedGoogle Scholar
Jeschke, M. G. et al. Scars. Nat. Rev. Dis. Primers 9, 64 (2023).
PubMedGoogle Scholar
Finnerty, C. C. et al. Hypertrophic scarring: the greatest unmet challenge after burn injury. Lancet 388, 1427–1436 (2016).
PubMedGoogle Scholar
Tyndall, A. J. 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. 69, 1809–1815 (2010).
PubMedGoogle Scholar
Ishikawa, Y. & Terao, C. Genetics of systemic sclerosis. J. Scleroderma Relat. Disord. 5, 192–201 (2020).
PubMedGoogle Scholar
Ortiz-Fernandez, L., Martin, J. & Alarcon-Riquelme, M. E. A summary on the genetics of systemic lupus erythematosus, rheumatoid arthritis, systemic sclerosis, and Sjogren’s syndrome. Clin. Rev. Allergy Immunol. 64, 392–411 (2023).
CASPubMedGoogle Scholar
Asano, Y. The pathogenesis of systemic sclerosis: an understanding based on a common pathologic cascade across multiple organs and additional organ-specific pathologies. J. Clin. Med. 9, 2687 (2020).
PubMedGoogle Scholar
Dumoitier, N. et al. Scleroderma peripheral B lymphocytes secrete interleukin-6 and transforming growth factor beta and activate fibroblasts. Arthritis Rheumatol. 69, 1078–1089 (2017).
CASPubMedGoogle Scholar
Berger, M. & Steen, V. D. Role of anti-receptor autoantibodies in pathophysiology of scleroderma. Autoimmun. Rev. 16, 1029–1035 (2017).
CASPubMedGoogle Scholar
Baroni, S. S. et al. Stimulatory autoantibodies to the PDGF receptor in systemic sclerosis. N. Engl. J. Med. 354, 2667–2676 (2006).
CASPubMedGoogle Scholar
Riemekasten, G. et al. Involvement of functional autoantibodies against vascular receptors in systemic sclerosis. Ann. Rheum. Dis. 70, 530–536 (2011).
CASPubMedGoogle Scholar
Marangoni, R. G. et al. Myofibroblasts in murine cutaneous fibrosis originate from adiponectin-positive intradermal progenitors. Arthritis Rheumatol. 67, 1062–1073 (2015).
CASPubMedGoogle Scholar
Bergmann, C. & Distler, J. H. Epigenetic factors as drivers of fibrosis in systemic sclerosis. Epigenomics 9, 463–477 (2017).
CASPubMedGoogle Scholar
Rius Rigau, A., Luber, M. & Distler, J. H. W. Mouse models of skin fibrosis. Methods Mol. Biol. 2299, 371–383 (2021).
CASPubMedGoogle Scholar
Beyer, C., Schett, G., Distler, O. & Distler, J. H. Animal models of systemic sclerosis: prospects and limitations. Arthritis Rheum. 62, 2831–2844 (2010).
CASPubMedGoogle Scholar
Steen, V. D., Medsger, T. A. Jr. & Rodnan, G. P. d-Penicillamine therapy in progressive systemic sclerosis (scleroderma): a retrospective analysis. Ann. Intern. Med. 97, 652–659 (1982).
CASPubMedGoogle Scholar
Tashkin, D. P. et al. Mycophenolate mofetil versus oral cyclophosphamide in scleroderma-related interstitial lung disease (SLS II): a randomised controlled, double-blind, parallel group trial. Lancet Respir. Med. 4, 708–719 (2016).
CASPubMedGoogle Scholar
Tashkin, D. P. et al. Cyclophosphamide versus placebo in scleroderma lung disease. N. Engl. J. Med. 354, 2655–2666 (2006).
CASPubMedGoogle Scholar
Khanna, D. et al. Tocilizumab in systemic sclerosis: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir. Med. 8, 963–974 (2020).
CASPubMedGoogle Scholar
Khanna, D. et al. Safety and efficacy of subcutaneous tocilizumab in adults with systemic sclerosis (faSScinate): a phase 2, randomised, controlled trial. Lancet 387, 2630–2640 (2016).
CASPubMedGoogle Scholar
Highland, K. B. et al. Efficacy and safety of nintedanib in patients with systemic sclerosis-associated interstitial lung disease treated with mycophenolate: a subgroup analysis of the SENSCIS trial. Lancet Respir. Med. 9, 96–106 (2021).
CASPubMedGoogle Scholar
Dobrota, R. et al. Circulating collagen neo-epitopes and their role in the prediction of fibrosis in patients with systemic sclerosis: a multicentre cohort study. Lancet Rheumatol. 3, e175–e184 (2021).
CASPubMedGoogle Scholar
Bergmann, C. et al. 68Ga-FAPI-04 PET-CT for molecular assessment of fibroblast activation and risk evaluation in systemic sclerosis-associated interstitial lung disease: a single-centre, pilot study. Lancet Rheumatol. 3, e185–e194 (2021).
CASPubMedGoogle Scholar
Treutlein, C. et al. Assessment of myocardial fibrosis in patients with systemic sclerosis using [68Ga]Ga-FAPI-04-PET-CT. Eur. J. Nucl. Med. Mol. Imaging 50, 1629–1635 (2023).
CASPubMedGoogle Scholar
Rinella, M. E. et al. A multisociety Delphi consensus statement on new fatty liver disease nomenclature. Hepatology 78, 1966–1986 (2023).
PubMedGoogle Scholar
Estes, C. et al. Modeling NAFLD disease burden in China, France, Germany, Italy, Japan, Spain, United Kingdom, and United States for the period 2016–2030. J. Hepatol. 69, 896–904 (2018).
PubMedGoogle Scholar
Linden, D. & Romeo, S. Therapeutic opportunities for the treatment of NASH with genetically validated targets. J. Hepatol. 79, 1056–1064 (2023).
CASPubMedGoogle Scholar
Caon, E. et al. Exploring the impact of the PNPLA3 I148M variant on primary human hepatic stellate cells using 3D extracellular matrix models. J. Hepatol. 80, 941–956 (2024).
CASPubMedGoogle Scholar
Rady, B. et al. PNPLA3 downregulation exacerbates the fibrotic response in human hepatic stellate cells. PLoS ONE 16, e0260721 (2021).
CASPubMedGoogle Scholar
Dwi Astarini, F., Ratnasari, N. & Wasityastuti, W. Update on non-alcoholic fatty liver disease-associated single nucleotide polymorphisms and their involvement in liver steatosis, inflammation, and fibrosis: a narrative review. Iran. Biomed. J. 26, 252–268 (2022).
PubMedGoogle Scholar
Wu, X. et al. Recent advances in understanding of pathogenesis of alcohol-associated liver disease. Annu. Rev. Pathol. 18, 411–438 (2023).
CASPubMedGoogle Scholar
Iwakiri, Y., Shah, V. & Rockey, D. C. Vascular pathobiology in chronic liver disease and cirrhosis – current status and future directions. J. Hepatol. 61, 912–924 (2014).
CASPubMedGoogle Scholar
Torres Rojas, A. M. & Lorente, S. Liver fibrosis emulation: impact of the vascular fibrotic alterations on hemodynamics. Comput. Biol. Med. 166, 107563 (2023).
CASPubMedGoogle Scholar
Lee, Y. S. & Seki, E. In vivo and in vitro models to study liver fibrosis: mechanisms and limitations. Cell Mol. Gastroenterol. Hepatol. 16, 355–367 (2023).
CASPubMedGoogle Scholar
Ravichandra, A. & Schwabe, R. F. Mouse models of liver fibrosis. Methods Mol. Biol. 2299, 339–356 (2021).
CASPubMedGoogle Scholar
Creeden, J. F. et al. Hepatic kinome atlas: an in-depth identification of kinase pathways in liver fibrosis of humans and rodents. Hepatology 76, 1376–1388 (2022).
CASPubMedGoogle Scholar
Benegiamo, G. et al. The genetic background shapes the susceptibility to mitochondrial dysfunction and NASH progression. J. Exp. Med. 220, e20221738 (2023).
CASPubMedGoogle Scholar
Delire, B., Starkel, P. & Leclercq, I. Animal models for fibrotic liver diseases: what we have, what we need, and what is under development. J. Clin. Transl. Hepatol. 3, 53–66 (2015).
PubMedGoogle Scholar
Park, J. et al. IL-6/STAT3 axis dictates the PNPLA3-mediated susceptibility to non-alcoholic fatty liver disease. J. Hepatol. 78, 45–56 (2023).
CASPubMedGoogle Scholar
Rockey, D. C. & Friedman, S. L. Fibrosis regression after eradication of hepatitis C virus: from bench to bedside. Gastroenterology 160, 1502–1520.e1501 (2021).
CASPubMedGoogle Scholar
Koda, Y., Nakamoto, N. & Kanai, T. Regulation of progression and resolution of liver fibrosis by immune cells. Semin. Liver Dis. 42, 475–488 (2022).
CASPubMedGoogle Scholar
Sanyal, A. J., Castera, L. & Wong, V. W. Noninvasive assessment of liver fibrosis in NAFLD. Clin. Gastroenterol. Hepatol. 21, 2026–2039 (2023).
CASPubMedGoogle Scholar
Natarajan, Y. & Loomba, R. Magnetic resonance elastography for the clinical risk assessment of fibrosis, cirrhosis, and portal hypertension in patients with NAFLD. J. Clin. Exp. Hepatol. 12, 174–179 (2022).
CASPubMedGoogle Scholar
Sanyal, A. J. et al. Cirrhosis regression is associated with improved clinical outcomes in patients with nonalcoholic steatohepatitis. Hepatology 75, 1235–1246 (2022).
CASPubMedGoogle Scholar
Niu, L. et al. Noninvasive proteomic biomarkers for alcohol-related liver disease. Nat. Med. 28, 1277–1287 (2022).
CASPubMedGoogle Scholar
Loomba, R. et al. Liver stiffness thresholds to predict disease progression and clinical outcomes in bridging fibrosis and cirrhosis. Gut 72, 581–589 (2023).
CASPubMedGoogle Scholar
Harrison, S. A., Loomba, R., Dubourg, J., Ratziu, V. & Noureddin, M. Clinical trial landscape in NASH. Clin. Gastroenterol. Hepatol. 21, 2001–2014 (2023).
CASPubMedGoogle Scholar
Dufour, J. F. et al. Current therapies and new developments in NASH. Gut 71, 2123–2134 (2022).
CASPubMedGoogle Scholar
Harrison, S. A. et al. A phase 3, randomized, controlled trial of resmetirom in NASH with liver fibrosis. N. Engl. J. Med. 390, 497–509 (2024).
PubMedGoogle Scholar
Rogliani, P., Calzetta, L., Cavalli, F., Matera, M. G. & Cazzola, M. Pirfenidone, nintedanib and N-acetylcysteine for the treatment of idiopathic pulmonary fibrosis: a systematic review and meta-analysis. Pulm. Pharmacol. Ther. 40, 95–103 (2016).
CASPubMedGoogle Scholar
Naqvi, M. et al. Antifibrotic therapy in progressive pulmonary fibrosis: a review of recent advances. Expert Rev. Respir. Med. 18, 397–407 (2024).
CASPubMedGoogle Scholar
Akasaka, E., Kleiser, S., Sengle, G., Bruckner-Tuderman, L. & Nystrom, A. Diversity of mechanisms underlying latent TGF-beta activation in recessive dystrophic epidermolysis bullosa. J. Invest. Dermatol. 141, 1450–1460 e1459 (2021).
CASPubMedGoogle Scholar
Leoncini, G. et al. Blood pressure reduction and RAAS inhibition in diabetic kidney disease: therapeutic potentials and limitations. J. Nephrol. 33, 949–963 (2020).
CASPubMedGoogle Scholar
van der Aart-van der Beek, A. B., de Boer, R. A. & Heerspink, H. J. L. Kidney and heart failure outcomes associated with SGLT2 inhibitor use. Nat. Rev. Nephrol. 18, 294–306 (2022).
PubMedGoogle Scholar
Perkovic, V. et al. Effects of semaglutide on chronic kidney disease in patients with type 2 diabetes. N. Engl. J. Med. 391, 109–121 (2024).
CASPubMedGoogle Scholar
Ratziu, V. & Friedman, S. L. Why do so many nonalcoholic steatohepatitis trials fail? Gastroenterology 165, 5–10 (2023).
PubMedGoogle Scholar
Choi, S. E., Fogo, A. B. & Lim, B. J. Histologic evaluation of activity and chronicity of lupus nephritis and its clinical significance. Kidney Res. Clin. Pract. 42, 166–173 (2023).
PubMedGoogle Scholar
Torok, N. J., Dranoff, J. A., Schuppan, D. & Friedman, S. L. Strategies and endpoints of antifibrotic drug trials: summary and recommendations from the AASLD Emerging Trends Conference, Chicago, June 2014. Hepatology 62, 627–634 (2015).
PubMedGoogle Scholar
Rea, G. et al. Beyond visual interpretation: quantitative analysis and artificial intelligence in interstitial lung disease diagnosis “Expanding Horizons in Radiology”. Diagnostics 13, 2333 (2023).
PubMedGoogle Scholar
Walker, P. D. et al. Practice guidelines for the renal biopsy. Mod. Pathol. 17, 1555–1563 (2004).
PubMedGoogle Scholar
Showalter, K. & Gordon, J. K. Skin histology in systemic sclerosis: a relevant clinical biomarker. Curr. Rheumatol. Rep. 23, 3 (2020).
PubMedGoogle Scholar
Santiago, T. et al. Ultrasound and elastography in the assessment of skin involvement in systemic sclerosis: a systematic literature review focusing on validation and standardization - WSF Skin Ultrasound Group. Semin. Arthritis Rheum. 52, 151954 (2022).
PubMedGoogle Scholar
Furst, D. E. et al. The modified Rodnan skin score is an accurate reflection of skin biopsy thickness in systemic sclerosis. J. Rheumatol. 25, 84–88 (1998).
CASPubMedGoogle Scholar
Sanyal, A. J. et al. Endpoints and clinical trial design for nonalcoholic steatohepatitis. Hepatology 54, 344–353 (2011).
PubMedGoogle Scholar
Maher, T. M. et al. Rituximab versus intravenous cyclophosphamide in patients with connective tissue disease-associated interstitial lung disease in the UK (RECITAL): a double-blind, double-dummy, randomised, controlled, phase 2b trial. Lancet Respir. Med. 11, 45–54 (2023).
CASPubMedGoogle Scholar
Visca, D. et al. Effect of ambulatory oxygen on quality of life for patients with fibrotic lung disease (AmbOx): a prospective, open-label, mixed-method, crossover randomised controlled trial. Lancet Respir. Med. 6, 759–770 (2018).
PubMedGoogle Scholar
Meima-van Praag, E. M., Buskens, C. J., Hompes, R. & Bemelman, W. A. Surgical management of Crohn’s disease: a state of the art review. Int. J. Colorectal Dis. 36, 1133–1145 (2021).
PubMedGoogle Scholar
Yamamoto, H. et al. Guidelines for endoscopic balloon dilation in treating Crohn’s disease-associated small intestinal strictures (supplement to the Clinical Practice Guidelines for Enteroscopy). Dig. Endosc. 34, 1278–1296 (2022).
PubMedGoogle Scholar
Levine, M. J. Empagliflozin for type 2 diabetes mellitus: an overview of phase 3 clinical trials. Curr. Diabetes Rev. 13, 405–423 (2017).
CASPubMedGoogle Scholar
Chertow, G. M. et al. Effects of dapagliflozin in chronic kidney disease, with and without other cardiovascular medications: DAPA-CKD trial. J. Am. Heart Assoc. 12, e028739 (2023).
CASPubMedGoogle Scholar
Bakris, G. L. et al. Effect of finerenone on chronic kidney disease outcomes in type 2 diabetes. N. Engl. J. Med. 383, 2219–2229 (2020).
CASPubMedGoogle Scholar
Brenner, B. M. et al. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N. Engl. J. Med. 345, 861–869 (2001).
CASPubMedGoogle Scholar
Heerspink, H. J. L. et al. Zibotentan in combination with dapagliflozin compared with dapagliflozin in patients with chronic kidney disease (ZENITH-CKD): a multicentre, randomised, active-controlled, phase 2b, clinical trial. Lancet 402, 2004–2017 (2023).
CASPubMedGoogle Scholar
Heerspink, H. J. L. et al. Atrasentan and renal events in patients with type 2 diabetes and chronic kidney disease (SONAR): a double-blind, randomised, placebo-controlled trial. Lancet 393, 1937–1947 (2019).
CASPubMedGoogle Scholar
Hoyles, R. K. et al. A multicenter, prospective, randomized, double-blind, placebo-controlled trial of corticosteroids and intravenous cyclophosphamide followed by oral azathioprine for the treatment of pulmonary fibrosis in scleroderma. Arthritis Rheum. 54, 3962–3970 (2006).
CASPubMedGoogle Scholar
Macrea, M. et al. Rituximab in patients with systemic sclerosis-associated interstitial lung disease: a systematic review and meta-analysis. Ann. Am. Thorac. Soc. 21, 317–327 (2024).
PubMedGoogle Scholar
Kuzumi, A. et al. Long-term outcomes after rituximab treatment for patients with systemic sclerosis: follow-up of the DESIRES trial with a focus on serum immunoglobulin levels. JAMA Dermatol. 159, 374–383 (2023).
PubMedGoogle Scholar
Ebata, S. et al. Safety and efficacy of rituximab in systemic sclerosis (DESIRES): open-label extension of a double-blind, investigators-initiated, randomised, placebo-controlled trial. Lancet Rheumatol. 4, e546–e555 (2022).
CASPubMedGoogle Scholar
Sullivan, K. M. et al. Myeloablative autologous stem-cell transplantation for severe scleroderma. N. Engl. J. Med. 378, 35–47 (2018).
PubMedGoogle Scholar
van Laar, J. M. et al. Autologous hematopoietic stem cell transplantation vs intravenous pulse cyclophosphamide in diffuse cutaneous systemic sclerosis: a randomized clinical trial. JAMA 311, 2490–2498 (2014).
PubMedGoogle Scholar
Bergmann, C. et al. Treatment of a patient with severe systemic sclerosis (SSc) using CD19-targeted CAR T cells. Ann. Rheum. Dis. 82, 1117–1120 (2023).
PubMedGoogle Scholar
Muller, F. et al. CD19 CAR T-cell therapy in autoimmune disease - a case series with follow-up. N. Engl. J. Med. 390, 687–700 (2024).
PubMedGoogle Scholar
Schett, G., Mackensen, A. & Mougiakakos, D. CAR T-cell therapy in autoimmune diseases. Lancet 402, 2034–2044 (2023).
CASPubMedGoogle Scholar
Merkt, W. et al. Third-generation CD19.CAR-T cell-containing combination therapy in Scl70+ systemic sclerosis. Ann. Rheum. Dis. 83, 543–546 (2024).
PubMedGoogle Scholar
Zhang, C. Y., Liu, S. & Yang, M. Treatment of liver fibrosis: past, current, and future. World J. Hepatol. 15, 755–774 (2023).
PubMedGoogle Scholar
Cao, J. et al. Joint profiling of chromatin accessibility and gene expression in thousands of single cells. Science 361, 1380–1385 (2018).
CASPubMedGoogle Scholar
Peterson, V. M. et al. Multiplexed quantification of proteins and transcripts in single cells. Nat. Biotechnol. 35, 936–939 (2017).
CASPubMedGoogle Scholar
Stoeckius, M. et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods 14, 865–868 (2017).
CASPubMedGoogle Scholar
Muhl, L. et al. Single-cell analysis uncovers fibroblast heterogeneity and criteria for fibroblast and mural cell identification and discrimination. Nat. Commun. 11, 3953 (2020).
CASPubMedGoogle Scholar
Rodriques, S. G. et al. Slide-seq: a scalable technology for measuring genome-wide expression at high spatial resolution. Science 363, 1463–1467 (2019).
CASPubMedGoogle Scholar
Vickovic, S. et al. High-definition spatial transcriptomics for in situ tissue profiling. Nat. Methods 16, 987–990 (2019).
CASPubMedGoogle Scholar
Eng, C. L. et al. Transcriptome-scale super-resolved imaging in tissues by RNA seqFISH. Nature 568, 235–239 (2019).
CASPubMedGoogle Scholar
Sommerfeld, S. D. et al. Interleukin-36gamma-producing macrophages drive IL-17-mediated fibrosis. Sci. Immunol. 4, eaax4783 (2019).
CASPubMedGoogle Scholar
Xu, Y. et al. A transient wave of Bhlhe41+ resident macrophages enables remodeling of the developing infarcted myocardium. Cell Rep. 42, 113174 (2023).
CASPubMedGoogle Scholar
Wu, H., Kirita, Y., Donnelly, E. L. & Humphreys, B. D. Advantages of single-nucleus over single-cell RNA sequencing of adult kidney: rare cell types and novel cell states revealed in fibrosis. J. Am. Soc. Nephrol. 30, 23–32 (2019).
CASPubMedGoogle Scholar
Dudley, J. T. et al. Computational repositioning of the anticonvulsant topiramate for inflammatory bowel disease. Sci. Transl. Med. 3, 96ra76 (2011).
CASPubMedGoogle Scholar
Download references
Acknowledgements
The authors thank the Helmsley Charitable Trust for supporting the construction of a pathway to test antifibrotic therapies in IBD. The research of L.E.N. is funded in part by grants from NIH P50AA024333, R01AA027456, U01AA026398 and R01AA030699. B.H.’s research is supported by grants from the Canadian Institutes of Health Research CIHR (#375597, #190081) and joint support from the Canada Foundation for Innovation (CFI) and the Ontario Research Fund (ORF) (#36050, #38861, #36349). T.M.M. is supported by an NIHR Clinician Scientist Fellowship (NIHR Ref: CS-2013-13-017) British Lung Foundation Chair in Respiratory Research (C17-3).
Author information
Author notes
These authors contributed equally: Florian Rieder, Marco Prunotto.
Authors and Affiliations
Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
Florian Rieder
Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, USA
Florian Rieder & Laura E. Nagy
Program for Global Translational Inflammatory Bowel Diseases (GRID), Chicago, IL, USA
Florian Rieder
Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, USA
Laura E. Nagy
Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
Toby M. Maher
National Heart and Lung Institute, Imperial College, London, UK
Toby M. Maher
Department of Rheumatology, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
Jörg H. W. Distler
Hiller Research Center, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
Jörg H. W. Distler
Department of Nephrology and Clinical Immunology, RWTH Aachen; Medical Faculty, Aachen, Germany
Rafael Kramann
Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
Rafael Kramann
Keenan Research Institute for Biomedical Science of the St Michael’s Hospital, Toronto, Ontario, Canada
Boris Hinz
Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
Boris Hinz
Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
Marco Prunotto
Authors
Florian Rieder
View author publications
You can also search for this author inPubMedGoogle Scholar
2. Laura E. Nagy
View author publications
You can also search for this author inPubMedGoogle Scholar
3. Toby M. Maher
View author publications
You can also search for this author inPubMedGoogle Scholar
4. Jörg H. W. Distler
View author publications
You can also search for this author inPubMedGoogle Scholar
5. Rafael Kramann
View author publications
You can also search for this author inPubMedGoogle Scholar
6. Boris Hinz
View author publications
You can also search for this author inPubMedGoogle Scholar
7. Marco Prunotto
View author publications
You can also search for this author inPubMedGoogle Scholar
Contributions
All authors contributed to discussion of the content and wrote sections of the article. All authors reviewed and/or edited the manuscript before submission.
Corresponding authors
Correspondence to Florian Rieder or Marco Prunotto.
Ethics declarations
Competing interests
F.R. is on the advisory board of or consultant to Adiso, Adnovate, Agomab, Allergan, AbbVie, Arena, AstraZeneca, Bausch & Lomb, Boehringer-Ingelheim, Celgene/BMS, Celltrion, CDISC, Celsius, Cowen, Eugit, Ferring, Galapagos, Galmed, Genentech, Gilead, Gossamer, Granite, Guidepoint, Helmsley, Horizon Therapeutics, Image Analysis Limited, Index Pharma, Landos, Janssen, Koutif, Mestag, Metacrine, Mirum, Mopac, Morphic, Myka Labs, Organovo, Origo, Palisade, Pfizer, Pliant, Prometheus Biosciences, Receptos, RedX, Roche, Samsung, Sanofi, Surmodics, Surrozen, Takeda, Techlab, Teva, Theravance, Thetis, Trix Bio, UCB, Ysios and 89Bio. J.H.W.D. has consultancy relationships with and/or is part of the speaker or advisory board of AbbVie, Active Biotech, Anamar, ARXX, AstraZeneca, Bayer Pharma, Boehringer-Ingelheim, Calliditas Therapeutics, Celgene, Galapagos, Genentech, GSK, Inventiva, Janssen, Novartis, Pfizer, Roche and UCB. J.H.W.D. has received research funding from Anamar, Argenx, ARXX, BMS, Bayer Pharma, Boehringer-Ingelheim, Cantargia, Celgene, CSL Behring, Galapagos, GSK, Inventiva, Kiniksa, Lassen, Sanofi-Aventis, RedX and UCB. J.H.W.D. is CEO of 4D Science and Scientific Lead of FibroCure. R.K. is founder and shareholder of Sequantrix GmbH and has grants from Travere Therapeutics, Galapagos, Chugai, AskBio and Novo Nordisk and is a consultant for Bayer, Roche, Chugai, Pfizer, Novo Nordisk, Hybridize Therapeutics and Gruenenthal. T.M.M., via his institution, has received industry-academic funding from AstraZeneca and GlaxoSmithKline R&D; and consultancy or speaker fees from AstraZeneca, Bayer, Boehringer-Ingelheim, BMS, CSL Behring, Fibrogen, Galapagos, Galecto, GlaxoSmithKline, IQVIA, Merck, Pliant, Pfizer, Qureight, Roche, Sanofi-Aventis, Structure Therapeutics, Trevi and Veracyte. M.P. is an employee of Medicxi Ventures. L.E.N. and B.H. declare no competing interests.
Peer review
Peer review information
Nature Reviews Drug Discovery thanks Prakash Ramachandran, Frank Tacke and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Related links
Pliant Therapeutics provides update on BEACON-IPF, a phase 2b/3 trial in patients with idiopathic pulmonary fibrosis: https://ir.pliantrx.com/news-releases/news-release-details/pliant-therapeutics-provides-update-beacon-ipf-phase-2b3-trial-0
ClinicalTrials.gov: https://clinicaltrials.gov
Supplementary information
Supporting information
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
Reprints and permissions
About this article
Check for updates. Verify currency and authenticity via CrossMark
Cite this article
Rieder, F., Nagy, L.E., Maher, T.M. et al. Fibrosis: cross-organ biology and pathways to development of innovative drugs. Nat Rev Drug Discov (2025). https://doi.org/10.1038/s41573-025-01158-9
Download citation
Accepted:10 February 2025
Published:18 March 2025
DOI:https://doi.org/10.1038/s41573-025-01158-9
Share this article
Anyone you share the following link with will be able to read this content:
Get shareable link
Sorry, a shareable link is not currently available for this article.
Copy to clipboard
Provided by the Springer Nature SharedIt content-sharing initiative