Meeting Report Open Access

The Cellular and Metabolic Bases of Organ Fibrosis: UNIA Workshop 2023 in Baeza, Spain

Fibrosis. 2024, 2(1), 10001; https://doi.org/10.35534/fibrosis.2024.10001
1
Centro de Biología Molecular “Severo Ochoa”, Consejo Superior de Investigaciones Científicas (C.S.I.C.), Universidad Autónoma de Madrid (U.A.M.), 28049 Madrid, Spain
2
Renal, Electrolyte, and Hypertension Division, Department of Medicine/Institute for Diabetes, Obesity and Metabolism/Department of Genetics; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
*
Authors to whom correspondence should be addressed.

Received: 30 Jan 2024    Accepted: 18 Feb 2024    Published: 20 Feb 2024   

Abstract

Fibrosis is defined by scarring and tissue hardening caused by excess deposition of extracellular matrix components, mainly collagens. A fibrotic response can occur in any tissue of the body and is the final outcome of an unbalanced reaction to inflammation and wound healing induced by a variety of insults, including persistent infections, autoimmune reactions, allergic responses, chemical exposure, radiation, and tissue injury. The accumulation of extracellular matrix proteins replaces the living tissue and disrupts the architecture leading to organ malfunction. Fibrosis remains a major clinical and therapeutic challenge and has been estimated to account for 45% of deaths in the developed world. While major advances regarding mechanistic knowledge on the underlying cell biology alterations in fibrosis have helped to characterize the main phases and mediators involved, this knowledge has not yielded significant progress in treatment. Only recently, the metabolic features associated to fibrosis have begun to emerge. This information, likely representing only the tip of the iceberg, suggests that metabolic derangement is a key culprit in the pathophysiology of fibrogenesis. The Workshop on The Cellular and Metabolic Bases of Organ Fibrosis, International University of Andalusia, Baeza, Spain, October 8–11, 2023 aimed to discuss the current knowledge and novel perspectives on the mechanisms contributing to the development of fibrosis in different organs and tissues, with particular focus on new methodological developments in metabolomics and therapeutic strategies.

References

1.
Moretti L, Stalfort J, Barker TH, Abebayehu D. The interplay of fibroblasts, the extracellular matrix, and inflammation in scar formation. J. Biol. Chem. 2022, 298, 101530. [Google Scholar]
2.
Henderson NC, Rieder F, Wynn TA. Fibrosis: from mechanisms to medicines. Nature 2020, 587, 555–566. [Google Scholar]
3.
Rosenbloom J, Macarak E, Piera-Velazquez S, Jimenez SA. Human Fibrotic Diseases: Current Challenges in Fibrosis Research. In Fibrosis: Methods and Protocols; Springer: New York, NY, USA; 2017; pp 1–23.
4.
Distler JHW, Györfi A-H, Ramanujam M, Whitfield ML, Königshoff M, Lafyatis R. Shared and distinct mechanisms of fibrosis. Nat. Rev. Rheumatol. 2019, 15, 705–730. [Google Scholar]
5.
Senior M. Fighting fibrosis. Nat. Biotechnol. 2022, 40, 1169–1173. [Google Scholar]
6.
Wynn T. Cellular and molecular mechanisms of fibrosis. J. Pathol. 2008, 214, 199–210. [Google Scholar]
7.
Zhao X, Kwan JYY, Yip K, Liu PP, Liu FF. Targeting metabolic dysregulation for fibrosis therapy. Nat. Rev. Drug Discov. 2020, 19, 57–75. [Google Scholar]
8.
Shin H, Park S, Hong J, Baek A-R, Lee J, Kim D-J, et al. Overexpression of fatty acid synthase attenuates bleomycin induced lung fibrosis by restoring mitochondrial dysfunction in mice. Sci. Rep. 2023, 13, 9044. [Google Scholar]
9.
Hamanaka RB, Mutlu GM. Metabolic requirements of pulmonary fibrosis: role of fibroblast metabolism. FEBS J. 2021, 288, 6331–6352. [Google Scholar]
10.
Kay EJ, Koulouras G, Zanivan S. Regulation of Extracellular Matrix Production in Activated Fibroblasts: Roles of Amino Acid Metabolism in Collagen Synthesis. Front. Oncol. 2021, 11, 719922. [Google Scholar]
11.
Staab-Weijnitz CA. Fighting the Fiber: Targeting Collagen in Lung Fibrosis. Am. J. Respir. Cell Mol. Biol. 2022, 66, 363–381. [Google Scholar]
12.
Hamanaka RB, O’Leary EM, Witt LJ, Tian Y, Gökalp GA, Meliton AY, et al. Glutamine Metabolism Is Required for Collagen Protein Synthesis in Lung Fibroblasts. Am. J. Respir. Cell Mol. Biol. 2019, 61, 597–606. [Google Scholar]
13.
Schwörer S, Berisa M, Violante S, Qin W, Zhu J, Hendrickson RC, et al. Proline biosynthesis is a vent for TGFβ‐induced mitochondrial redox stress. EMBO J. 2020, 39, e103334. [Google Scholar]
14.
Conroy LR, Clarke HA, Allison DB, Valenca SS, Sun Q, Hawkinson TR, et al. Spatial metabolomics reveals glycogen as an actionable target for pulmonary fibrosis. Nat. Commun. 2023, 14, 2759. [Google Scholar]
15.
Huang R, Fu P, Ma L.  Kidney fibrosis: from mechanisms to therapeutic medicines. Signal Transduct. Target. Ther. 2023, 8, 129. [Google Scholar]
16.
Balzer MS, Rohacs T, Susztak K. How Many Cell Types Are in the Kidney and What Do They Do? Ann. Rev. Physiol. 2022, 84, 507–531. [Google Scholar]
17.
Li L, Fu H, Liu Y. The fibrogenic niche in kidney fibrosis: components and mechanisms. Nat. Rev. Nephrol. 2022, 18, 545–557. [Google Scholar]
18.
Abedini A, Ma Z, Frederick J, Dhillon P, Balzer MS, Shrestha R, et al. Spatially resolved human kidney multi-omics single cell atlas highlights the key role of the fibrotic microenvironment in kidney disease progression. bioRxiv 2022, 2022.10.24.513598.
19.
Doke T, Mukherjee S, Mukhi D, Dhillon P, Abedini A, Davis JG, et al. NAD+ precursor supplementation prevents mtRNA/RIG-I-dependent inflammation during kidney injury. Nat. Metabol. 2023, 5, 414–430. [Google Scholar]
20.
Mukhi D, Li L, Liu H, Doke T, Kolligundla LP, Ha E, et al. ACSS2 gene variants determine kidney disease risk by controlling de novo lipogenesis in kidney tubules. J. Clin. Invest. 2023, 134, e172963. [Google Scholar]
21.
Li H, Dixon EE, Wu,H, Humphreys BD. Comprehensive single-cell transcriptional profiling defines shared and unique epithelial injury responses during kidney fibrosis. Cell Metabol. 2022, 34, 1977–1998.e9.
22.
Miguel V, Tituaña J, Herrero JI, Herrero L, Serra D, Cuevas P, et al. Renal tubule Cpt1a overexpression protects from kidney fibrosis by restoring mitochondrial homeostasis. J. Clin. Invest. 2021, 131, e140695. [Google Scholar]
23.
Wang G, Heijs B, Kostidis S, Mahfouz A, Rietjens RGJ, Bijkerk R, et al. Analyzing cell-type-specific dynamics of metabolism in kidney repair. Nat. Metabol. 2022, 4, 1109–1118. [Google Scholar]
24.
Tiwari R, Kapitsinou PP. Role of Endothelial Prolyl-4-Hydroxylase Domain Protein/Hypoxia-Inducible Factor Axis in Acute Kidney Injury. Nephron 2021, 146, 243–248. [Google Scholar]
25.
Tiwari R, Bommi PV, Gao P, Schipma MJ, Zhou Y, Quaggin SE, et al. Chemical inhibition of oxygen-sensing prolyl hydroxylases impairs angiogenic competence of human vascular endothelium through metabolic reprogramming.  iScience 2022, 25, 105086. [Google Scholar]
26.
Rajendran G, Schonfeld MP, Tiwari R, Huang S, Torosyan R, Fields T, et al. Inhibition of Endothelial PHD2 Suppresses Post-Ischemic Kidney Inflammation through Hypoxia-Inducible Factor-1.  J. Am. Soc. Nephrol. 2020, 31, 501–516. [Google Scholar]
27.
Rosell-Garcia T, Rivas-Muñoz S, Kin K, Romero-Albillo V, Alcaraz S, Fernandez-Tornero C, et al. Multimerization of HIF enhances transcription of target genes containing the hypoxia ancillary sequence. Biochim. Biophys. Acta Gene Regul. Mech. 2023, 1866, 194963. [Google Scholar]
28.
Zhao JL, Qiao XH, Mao JH, Liu F, Fu HD. The interaction between cellular senescence and chronic kidney disease as a therapeutic opportunity. Front. Pharmacol. 2022, 13, 974361. [Google Scholar]
29.
Marquez-Exposito L, Tejedor-Santamaria L, Valentijn FA, Tejera-Muñoz A, Rayego-Mateos S, Marchant V, et al. Oxidative Stress and Cellular Senescence Are Involved in the Aging Kidney. Antioxidants 2022, 11, 301. [Google Scholar]
30.
Valentijn FA, Knoppert SN, Marquez-Exposito L, Rodrigues-Diez RR, Pissas G, Tang J, et al. Cellular communication network 2 (connective tissue growth factor) aggravates acute DNA damage and subsequent DNA damage response-senescence-fibrosis following kidney ischemia reperfusion injury. Kidney Int. 2022, 102, 1305–1319. [Google Scholar]
31.
Wynn TA. Integrating mechanisms of pulmonary fibrosis. J. Exp. Med. 2011, 208, 1339–1350. [Google Scholar]
32.
Yue X, Shan B, Lasky JA. TGF-β: Titan of Lung Fibrogenesis. Curr. Enzym. Inhib. 2010, 6, 67–77. [Google Scholar]
33.
Jin J, Togo S, Kadoya K, Tulafu M, Namba Y, Iwai M, et al. Pirfenidone attenuates lung fibrotic fibroblast responses to transforming growth factor-β1. Respir. Res. 2019, 20, 119. [Google Scholar]
34.
Rangarajan S, Bernard K, Thannickal VJ. Mitochondrial Dysfunction in Pulmonary Fibrosis. Ann. Am. Thorac. Soc. 2017, 14, s383–s388. [Google Scholar]
35.
Rangarajan S, Bone NB, Zmijewska AA, Jiang S, Park DW, Bernard K, et al. Metformin reverses established lung fibrosis in a bleomycin model. Nat. Med. 2018, 24, 1121–1127. [Google Scholar]
36.
Ogawa T, Shichino S, Ueha S, Matsushima K. Macrophages in lung fibrosis. Int. Immunol. 2021, 33, 665–671. [Google Scholar]
37.
Wculek SK, Heras-Murillo I, Mastrangelo A, Mañanes D, Galán M, Miguel V, et al. Oxidative phosphorylation selectively orchestrates tissue macrophage homeostasis. Immunity 2023, 56, 516–530. [Google Scholar]
38.
Chandran RR, Xie Y, Gallardo-Vara E, Adams T, Garcia-Milian R, Kabir I, et al. Distinct roles of KLF4 in mesenchymal cell subtypes during lung fibrogenesis. Nat. Commun. 2021, 12, 7179. [Google Scholar]
39.
Marnell CS, Bick A, Natarajan P. Clonal hematopoiesis of indeterminate potential (CHIP): Linking somatic mutations, hematopoiesis, chronic inflammation and cardiovascular disease. J. Mol. Cell. Cardiol. 2021, 161, 98–105. [Google Scholar]
40.
Basil MC, Cardenas-Diaz FL, Kathiriya JJ, Morley MP, Carl J, Brumwell AN, et al. Human distal airways contain a multipotent secretory cell that can regenerate alveoli. Nature 2022, 604, 120–126. [Google Scholar]
41.
Roehlen N, Crouchet E, Baumert TF. Liver Fibrosis: Mechanistic Concepts and Therapeutic Perspectives. Cells 2020, 9, 875. [Google Scholar]
42.
Lazcanoiturburu N, García-Sáez J, González-Corralejo C, Roncero C, Sanz J, Martín-Rodríguez C, et al. Lack of EGFR catalytic activity in hepatocytes improves liver regeneration following DDC-induced cholestatic injury by promoting a pro-restorative inflammatory response. J. Pathol. 2022, 258, 312–324. [Google Scholar]
43.
López-Luque J, Caballero-Díaz D, Martinez-Palacián A, Roncero C, Moreno-Càceres J, García-Bravo M, et al. Dissecting the role of epidermal growth factor receptor catalytic activity during liver regeneration and hepatocarcinogenesis. Hepatology 2016, 63, 604–619. [Google Scholar]
44.
Loneker AE, Alisafaei F, Kant A, Li D, Janmey PA, Shenoy VB, et al. Lipid droplets are intracellular mechanical stressors that impair hepatocyte function. Proc. Nat. Acad. Sci. 2023, 120, e2216811120. [Google Scholar]
45.
Marín-Sedeño E, de Morentin XM, Pérez-Pomares JM, Gómez-Cabrero D, Ruiz-Villalba A.  Understanding the Adult Mammalian Heart at Single-Cell RNA-Seq Resolution. Front. Cell Dev. Biol. 2021, 9, 645276. [Google Scholar]
46.
Ruiz-Villalba A, Romero JP, Hernández SC, Vilas-Zornoza A, Fortelny N, Castro-Labrador L, 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 2020, 142, 1831–1847. [Google Scholar]
Creative Commons

© 2024 by the authors; licensee SCIEPublish, SCISCAN co. Ltd. This article is an open access article distributed under the CC BY license (https://creativecommons.org/licenses/by/4.0/).