Next Article in Journal
Characterization of 3D Organotypic Culture of Mouse Adipose-Derived Stem Cells
Previous Article in Journal
Is There a Place for Lewy Bodies before and beyond Alpha-Synuclein Accumulation? Provocative Issues in Need of Solid Explanations
Previous Article in Special Issue
Inhibition of the NF-κB Signaling Pathway Alleviates Pyroptosis in Bladder Epithelial Cells and Neurogenic Bladder Fibrosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Cell Death in Acute Organ Injury and Fibrosis

1
Department of Immunology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
2
Department of Diabetes and Endocrinology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(7), 3930; https://doi.org/10.3390/ijms25073930
Submission received: 9 March 2024 / Accepted: 25 March 2024 / Published: 1 April 2024
(This article belongs to the Special Issue Cell Death in Acute Organ Injury and Fibrosis)
Tissue fibrosis is characterized by the excessive accumulation of extracellular matrix in various organs, including the lungs, liver, skin, kidneys, pancreas, and heart, ultimately leading to organ failure [1,2]. This fibrotic process may be triggered by tissue injury resulting from diverse mechanisms such as infection, trauma, metabolic disorders, wounds, acute or chronic inflammation, autoimmune disorders, cancer, or unknown mechanisms [1]. Following injury, an abnormal tissue repair mechanism ensues, marked by the enhanced accumulation of fibroblasts and/or myofibroblasts within the affected organ, which overproduce extracellular matrix proteins [3]. Various cell types, including parenchymal epithelial cells, vascular endothelial cells, and cells from the innate or acquired immune systems, participate in this fibrotic process by secreting factors that recruit and activate fibroblasts to produce extracellular matrix proteins [4].
Importantly, during tissue injury and fibrosis, parenchymal cells undergo cell death, leading to their replacement by collagen-producing cells, thereby exacerbating the fibrotic process [5,6,7,8]. Profibrotic cytokines such as transforming growth factor-β1 (TGF-β1) promote the production and secretion of extracellular matrix proteins and may also cause parenchymal cell apoptosis, further contributing to tissue sclerosis [9]. TGF-β1 has been reported to induce alveolar epithelial cell apoptosis in lung fibrosis and hepatocytes in liver cirrhosis [10]. Increased levels of TGF-β1 have also been implicated in the pathogenesis of fibrosis in several organs in patients with diabetes mellitus [11,12]. TGF-β1 may induce the apoptosis of insulin-producing β-cells, increase insulin resistance contributing to the acceleration of diabetes mellitus, or induce the apoptosis of podocytes and renal tubular epithelial cells contributing to the pathogenesis of diabetic nephropathy [9,11].
Cell death is indispensable for numerous physiological processes, including embryonic development, tissue homeostasis, and immune responses [13]. During embryogenesis, apoptosis eliminates superfluous cells, shapes developing tissues, and regulates organ morphogenesis [13,14]. In adult organisms, programmed cell death maintains tissue integrity by eliminating damaged or senescent cells, thereby preventing the accumulation of potentially harmful cellular debris. Moreover, cell death serves as a defense mechanism against pathogens, facilitating the clearance of infected cells and promoting immune surveillance [14]. Cell death can occur through several mechanisms including apoptosis, pyroptosis, necrosis, and autophagy [13].
The death of parenchymal cells has been implicated in the pathogenesis of organ fibrosis [5,15]. Dead cells are replaced by fibroblasts, which perpetuate fibrosis by producing and releasing extracellular matrix proteins [5]. Furthermore, the progression of fibrosis is often preceded by acute tissue injury crises, such as acute exacerbation of interstitial lung disease and acute kidney injury, which are associated with an increased death of parenchymal and vascular endothelial cells [16,17,18].
The amelioration of organ injury/fibrosis by inhibitors of apoptosis or pyroptosis further supports the implication of cell death in the pathogenesis of tissue injury/fibrosis [19,20]. For example, the inhibition of pyroptosis of bladder epithelial cells improves neurogenic bladder fibrosis, lung overexpression of matrix metalloproteinase-2 has been shown to mitigate lung fibrosis by blocking apoptosis of lung epithelial cells, and selective cannabinoid type II receptors, which are known to block apoptosis, protect against inflammatory response in endotoxin-induced acute lung injury and hepatic ischemic/reperfusion injury [21,22,23,24,25].
The resistance of fibroblasts or myofibroblasts to cell death has also been reported to contribute to the progression of fibrosis [26]. Some drugs undergoing clinical trials accelerate the apoptosis of myofibroblasts [27,28,29]. However, it is worth noting that the injury or apoptosis of fibroblasts/myofibroblasts may also be detrimental under certain environmental or pathological conditions. For example, excessive mechanical stress, aging, or hypoxia can cause sublethal injury or the apoptosis of anterior cruciate ligament fibroblasts, hindering fibroblast cell motility and ligament regeneration [30]. An improvement in fibroblast motility or survival has been shown to protect and accelerate the healing of the anterior cruciate ligament [31,32,33]. In addition, components of extracellular matrix proteins may also be important to protect some organ normal resident cells from injury and apoptosis. For example, a previous study has shown that the presence of elastin in the skin may be important to protect against the loss of melanocytes in vitiligo [34]. Apoptosis caused by immune cells is involved in the loss of melanocytes in vitiligo conditions [35].
In summary, tissue fibrosis, triggered by various insults, involves excessive extracellular matrix accumulation leading to organ dysfunction. Profibrotic cytokines such as TGF-β1 promote matrix production and the apoptosis of parenchymal cells, worsening fibrosis. Cell death, vital for development and immunity, contributes to fibrosis progression by replacing dead cells with collagen-producing fibroblasts. Strategies targeting cell death pathways offer potential in mitigating fibrosis, but caution is needed due to its dual role in tissue repair and pathology.

Author Contributions

Conceptualization, E.C.G.; writing—original draft preparation, E.C.G. and T.Y.; writing—review and editing, E.C.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Lurje, I.; Gaisa, N.T.; Weiskirchen, R.; Tacke, F. Mechanisms of organ fibrosis: Emerging concepts and implications for novel treatment strategies. Mol. Asp. Med. 2023, 92, 101191. [Google Scholar] [CrossRef] [PubMed]
  2. Singh, V.; Ulasov, I.; Gupta, S.; Singh, A.; Roy, V.K.; Kharwar, R.K. Idiopathic Pulmonary Fibrosis: Where do We Stand and How Far to Go? Discov. Med. 2024, 36, 22–47. [Google Scholar] [CrossRef] [PubMed]
  3. Miao, H.; Wu, X.Q.; Zhang, D.D.; Wang, Y.N.; Guo, Y.; Li, P.; Xiong, Q.; Zhao, Y.Y. Deciphering the cellular mechanisms underlying fibrosis-associated diseases and therapeutic avenues. Pharmacol. Res. 2021, 163, 105316. [Google Scholar] [CrossRef] [PubMed]
  4. Caligiuri, A.; Gentilini, A.; Pastore, M.; Gitto, S.; Marra, F. Cellular and Molecular Mechanisms Underlying Liver Fibrosis Regression. Cells 2021, 10, 2759. [Google Scholar] [CrossRef] [PubMed]
  5. Gao, R.; Tang, H.; Mao, J. Programmed Cell Death in Liver Fibrosis. J. Inflamm. Res. 2023, 16, 3897–3910. [Google Scholar] [CrossRef] [PubMed]
  6. Picca, A.; Calvani, R.; Coelho-Junior, H.J.; Marzetti, E. Cell Death and Inflammation: The Role of Mitochondria in Health and Disease. Cells 2021, 10, 537. [Google Scholar] [CrossRef] [PubMed]
  7. Piek, A.; de Boer, R.A.; Sillje, H.H. The fibrosis-cell death axis in heart failure. Heart Fail. Rev. 2016, 21, 199–211. [Google Scholar] [CrossRef] [PubMed]
  8. Song, Z.; Gong, Q.; Guo, J. Pyroptosis: Mechanisms and Links with Fibrosis. Cells 2021, 10, 3509. [Google Scholar] [CrossRef] [PubMed]
  9. D’Alessandro, V.F.; Takeshita, A.; Yasuma, T.; Toda, M.; D’Alessandro-Gabazza, C.N.; Okano, Y.; Tharavecharak, S.; Inoue, C.; Nishihama, K.; Fujimoto, H.; et al. Transforming Growth Factorbeta1 Overexpression Is Associated with Insulin Resistance and Rapidly Progressive Kidney Fibrosis under Diabetic Conditions. Int. J. Mol. Sci. 2022, 23, 14265. [Google Scholar] [CrossRef]
  10. Chung, J.Y.; Chan, M.K.; Li, J.S.; Chan, A.S.; Tang, P.C.; Leung, K.T.; To, K.F.; Lan, H.Y.; Tang, P.M. TGF-beta Signaling: From Tissue Fibrosis to Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 7575. [Google Scholar] [CrossRef]
  11. Wang, L.; Wang, H.L.; Liu, T.T.; Lan, H.Y. TGF-Beta as a Master Regulator of Diabetic Nephropathy. Int. J. Mol. Sci. 2021, 22, 7881. [Google Scholar] [CrossRef]
  12. Yue, Y.; Meng, K.; Pu, Y.; Zhang, X. Transforming growth factor beta (TGF-beta) mediates cardiac fibrosis and induces diabetic cardiomyopathy. Diabetes Res. Clin. Pract. 2017, 133, 124–130. [Google Scholar] [CrossRef] [PubMed]
  13. Newton, K.; Strasser, A.; Kayagaki, N.; Dixit, V.M. Cell death. Cell 2024, 187, 235–256. [Google Scholar] [CrossRef]
  14. Kayagaki, N.; Webster, J.D.; Newton, K. Control of Cell Death in Health and Disease. Annu. Rev. Pathol. 2024, 19, 157–180. [Google Scholar] [CrossRef]
  15. D’Alessandro-Gabazza, C.N.; Kobayashi, T.; Yasuma, T.; Toda, M.; Kim, H.; Fujimoto, H.; Hataji, O.; Takeshita, A.; Nishihama, K.; Okano, T.; et al. A Staphylococcus pro-apoptotic peptide induces acute exacerbation of pulmonary fibrosis. Nat. Commun. 2020, 11, 1539. [Google Scholar] [CrossRef] [PubMed]
  16. Enomoto, N. Pathological Roles of Pulmonary Cells in Acute Lung Injury: Lessons from Clinical Practice. Int. J. Mol. Sci. 2022, 23, 15027. [Google Scholar] [CrossRef] [PubMed]
  17. Havasi, A.; Borkan, S.C. Apoptosis and acute kidney injury. Kidney Int. 2011, 80, 29–40. [Google Scholar] [CrossRef]
  18. Power, C.; Fanning, N.; Redmond, H.P. Cellular apoptosis and organ injury in sepsis: A review. Shock 2002, 18, 197–211. [Google Scholar] [CrossRef]
  19. D’Alessandro-Gabazza, C.N.; Yasuma, T.; Kobayashi, T.; Toda, M.; Abdel-Hamid, A.M.; Fujimoto, H.; Hataji, O.; Nakahara, H.; Takeshita, A.; Nishihama, K.; et al. Inhibition of lung microbiota-derived proapoptotic peptides ameliorates acute exacerbation of pulmonary fibrosis. Nat. Commun. 2022, 13, 1558. [Google Scholar] [CrossRef]
  20. Fridman D’Alessandro, V.; D’Alessandro-Gabazza, C.N.; Yasuma, T.; Toda, M.; Takeshita, A.; Tomaru, A.; Tharavecharak, S.; Lasisi, I.O.; Hess, R.Y.; Nishihama, K.; et al. Inhibition of a Microbiota-Derived Peptide Ameliorates Established Acute Lung Injury. Am. J. Pathol. 2023, 193, 740–754. [Google Scholar] [CrossRef]
  21. Chen, J.; Li, Q.; Hong, Y.; Zhou, X.; Yu, C.; Tian, X.; Zhao, J.; Long, C.; Shen, L.; Wu, S.; et al. Inhibition of the NF-kappaB Signaling Pathway Alleviates Pyroptosis in Bladder Epithelial Cells and Neurogenic Bladder Fibrosis. Int. J. Mol. Sci. 2023, 24, 1160. [Google Scholar] [CrossRef]
  22. Hall, S.; Faridi, S.; Trivedi, P.; Sultana, S.; Ray, B.; Myers, T.; Euodia, I.; Vlatten, D.; Castonguay, M.; Zhou, J.; et al. Selective CB(2) Receptor Agonist, HU-308, Reduces Systemic Inflammation in Endotoxin Model of Pneumonia-Induced Acute Lung Injury. Int. J. Mol. Sci. 2022, 23, 15857. [Google Scholar] [CrossRef] [PubMed]
  23. Inoue, R.; Yasuma, T.; Fridman D’Alessandro, V.; Toda, M.; Ito, T.; Tomaru, A.; D’Alessandro-Gabazza, C.N.; Tsuruga, T.; Okano, T.; Takeshita, A.; et al. Amelioration of Pulmonary Fibrosis by Matrix Metalloproteinase-2 Overexpression. Int. J. Mol. Sci. 2023, 24, 6695. [Google Scholar] [CrossRef] [PubMed]
  24. Mukhopadhyay, P.; Rajesh, M.; Pan, H.; Patel, V.; Mukhopadhyay, B.; Batkai, S.; Gao, B.; Hasko, G.; Pacher, P. Cannabinoid-2 receptor limits inflammation, oxidative/nitrosative stress, and cell death in nephropathy. Free Radic. Biol. Med. 2010, 48, 457–467. [Google Scholar] [CrossRef] [PubMed]
  25. Rajesh, M.; Pan, H.; Mukhopadhyay, P.; Batkai, S.; Osei-Hyiaman, D.; Hasko, G.; Liaudet, L.; Gao, B.; Pacher, P. Cannabinoid-2 receptor agonist HU-308 protects against hepatic ischemia/reperfusion injury by attenuating oxidative stress, inflammatory response, and apoptosis. J. Leukoc. Biol. 2007, 82, 1382–1389. [Google Scholar] [CrossRef]
  26. Hinz, B.; Lagares, D. Evasion of apoptosis by myofibroblasts: A hallmark of fibrotic diseases. Nat. Rev. Rheumatol. 2020, 16, 11–31. [Google Scholar] [CrossRef] [PubMed]
  27. Siani, A. Pharmacological treatment of fibrosis: A systematic review of clinical trials. SN Compr. Clin. Med. 2020, 2, 531–550. [Google Scholar] [CrossRef]
  28. Juillerat-Jeanneret, L.; Aubert, J.D.; Mikulic, J.; Golshayan, D. Fibrogenic Disorders in Human Diseases: From Inflammation to Organ Dysfunction. J. Med. Chem. 2018, 61, 9811–9840. [Google Scholar] [CrossRef]
  29. Henderson, N.C.; Rieder, F.; Wynn, T.A. Fibrosis: From mechanisms to medicines. Nature 2020, 587, 555–566. [Google Scholar] [CrossRef]
  30. Leite, C.B.G.; Merkely, G.; Charles, J.F.; Lattermann, C. From Inflammation to Resolution: Specialized Pro-resolving Mediators in Posttraumatic Osteoarthritis. Curr. Osteoporos. Rep. 2023, 21, 758–770. [Google Scholar] [CrossRef]
  31. Cheng, M.; Johnson, V.M.; Murray, M.M. Effects of age and platelet-rich plasma on ACL cell viability and collagen gene expression. J. Orthop. Res. 2012, 30, 79–85. [Google Scholar] [CrossRef] [PubMed]
  32. Sha, Y.; Yang, L.; Lv, Y. MGF E peptide improves anterior cruciate ligament repair by inhibiting hypoxia-induced cell apoptosis and accelerating angiogenesis. J. Cell Physiol. 2019, 234, 8846–8861. [Google Scholar] [CrossRef] [PubMed]
  33. Sha, Y.; Zhang, B.; Chen, L.; Hong, H.; Chi, Q. Mechano Growth Factor Accelerates ACL Repair and Improves Cell Mobility of Mechanically Injured Human ACL Fibroblasts by Targeting Rac1-PAK1/2 and RhoA-ROCK1 Pathways. Int. J. Mol. Sci. 2022, 23, 4331. [Google Scholar] [CrossRef] [PubMed]
  34. Hirobe, T.; Enami, H. Reduced Elastin Fibers and Melanocyte Loss in Vitiliginous Skin Are Restored after Repigmentation by Phototherapy and/or Autologous Minigraft Transplantation. Int. J. Mol. Sci. 2022, 23, 15361. [Google Scholar] [CrossRef] [PubMed]
  35. Riding, R.L.; Harris, J.E. The Role of Memory CD8(+) T Cells in Vitiligo. J. Immunol. 2019, 203, 11–19. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yasuma, T.; Gabazza, E.C. Cell Death in Acute Organ Injury and Fibrosis. Int. J. Mol. Sci. 2024, 25, 3930. https://doi.org/10.3390/ijms25073930

AMA Style

Yasuma T, Gabazza EC. Cell Death in Acute Organ Injury and Fibrosis. International Journal of Molecular Sciences. 2024; 25(7):3930. https://doi.org/10.3390/ijms25073930

Chicago/Turabian Style

Yasuma, Taro, and Esteban C. Gabazza. 2024. "Cell Death in Acute Organ Injury and Fibrosis" International Journal of Molecular Sciences 25, no. 7: 3930. https://doi.org/10.3390/ijms25073930

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop