1
|
Yamamoto T, Kase S, Shinkai A, Murata M, Kikuchi K, Wu D, Kageyama Y, Shinohara M, Sasase T, Ishida S. Phosphorylation of αB-Crystallin Involves Interleukin-1β-Mediated Intracellular Retention in Retinal Müller Cells: A New Mechanism Underlying Fibrovascular Membrane Formation. Invest Ophthalmol Vis Sci 2023; 64:20. [PMID: 37459063 PMCID: PMC10362920 DOI: 10.1167/iovs.64.10.20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023] Open
Abstract
Purpose Chronic inflammation plays a pivotal role in the pathology of proliferative diabetic retinopathy (PDR), in which biological alterations of retinal glial cells are one of the key elements. The phosphorylation of αB-crystallin/CRYAB modulates its molecular dynamics and chaperone activity, and attenuates αB-crystallin secretion via exosomes. In this study, we investigated the effect of phosphorylated αB-crystallin in retinal Müller cells on diabetic mimicking conditions, including interleukin (IL)-1β stimuli. Methods Human retinal Müller cells (MIO-M1) were used to examine gene and protein expressions with real-time quantitative PCR, enzyme linked immunosorbent assay (ELISA), and immunoblot analyses. Cell apoptosis was assessed by Caspase-3/7 assay and TdT-mediated dUTP nick-end labeling staining. Retinal tissues isolated from the Spontaneously Diabetic Torii (SDT) fatty rat, a type 2 diabetic animal model with obesity, and fibrovascular membranes from patients with PDR were examined by double-staining immunofluorescence. Results CRYAB mRNA was downregulated in MIO-M1 cells with the addition of 10 ng/mL IL-1β; however, intracellular αB-crystallin protein levels were maintained. The αB-crystallin serine 59 (Ser59) residue was phosphorylated with IL-1β application in MIO-M1 cells. Cell apoptosis in MIO-M1 cells was induced by CRYAB knockdown. Immunoreactivity for Ser59-phosphorylated αB-crystallin and glial fibrillary acidic protein was colocalized in glial cells of SDT fatty rats and fibrovascular membranes. Conclusions The Ser59 phosphorylation of αB-crystallin was modulated by IL-1β in Müller cells under diabetic mimicking inflammatory conditions, suggesting that αB-crystallin contributes to the pathogenesis of PDR through an anti-apoptotic effect.
Collapse
Affiliation(s)
- Taku Yamamoto
- Laboratory of Ocular Cell Biology and Visual Science, Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Satoru Kase
- Laboratory of Ocular Cell Biology and Visual Science, Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Akihiro Shinkai
- Laboratory of Ocular Cell Biology and Visual Science, Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Miyuki Murata
- Laboratory of Ocular Cell Biology and Visual Science, Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kasumi Kikuchi
- Laboratory of Ocular Cell Biology and Visual Science, Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Di Wu
- Eye Center of the Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | | | | | - Tomohiko Sasase
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Susumu Ishida
- Laboratory of Ocular Cell Biology and Visual Science, Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
2
|
Tanguy J, Boutanquoi PM, Burgy O, Dondaine L, Beltramo G, Uyanik B, Garrido C, Bonniaud P, Bellaye PS, Goirand F. HSPB5 Inhibition by NCI-41356 Reduces Experimental Lung Fibrosis by Blocking TGF-β1 Signaling. Pharmaceuticals (Basel) 2023; 16:177. [PMID: 37259327 PMCID: PMC9960643 DOI: 10.3390/ph16020177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/26/2024] Open
Abstract
Idiopathic pulmonary fibrosis is a chronic, progressive and lethal disease of unknown etiology that ranks among the most frequent interstitial lung diseases. Idiopathic pulmonary fibrosis is characterized by dysregulated healing mechanisms that lead to the accumulation of large amounts of collagen in the lung tissue that disrupts the alveolar architecture. The two currently available treatments, nintedanib and pirfenidone, are only able to slow down the disease without being curative. We demonstrated in the past that HSPB5, a low molecular weight heat shock protein, was involved in the development of fibrosis and therefore was a potential therapeutic target. Here, we have explored whether NCI-41356, a chemical inhibitor of HSPB5, can limit the development of pulmonary fibrosis. In vivo, we used a mouse model in which fibrosis was induced by intratracheal injection of bleomycin. Mice were treated with NaCl or NCI-41356 (six times intravenously or three times intratracheally). Fibrosis was evaluated by collagen quantification, immunofluorescence and TGF-β gene expression. In vitro, we studied the specific role of NCI-41356 on the chaperone function of HSPB5 and the inhibitory properties of NCI-41356 on HSPB5 interaction with its partner SMAD4 during fibrosis. TGF-β1 signaling was evaluated by immunofluorescence and Western Blot in epithelial cells treated with TGF-β1 with or without NCI-41356. In vivo, NCI-41356 reduced the accumulation of collagen, the expression of TGF-β1 and pro-fibrotic markers (PAI-1, α-SMA). In vitro, NCI-41356 decreased the interaction between HSPB5 and SMAD4 and thus modulated the SMAD4 canonical nuclear translocation involved in TGF-β1 signaling, which may explain NCI-41356 anti-fibrotic properties. In this study, we determined that inhibition of HSPB5 by NCI-41356 could limit pulmonary fibrosis in mice by limiting the synthesis of collagen and pro-fibrotic markers. At the molecular level, this outcome may be explained by the effect of NCI-41356 inhibiting HSPB5/SMAD4 interaction, thus modulating SMAD4 and TGF-β1 signaling. Further investigations are needed to determine whether these results can be transposed to humans.
Collapse
Affiliation(s)
- Julie Tanguy
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, 21000 Dijon, France
- UFR des Sciences de Santé, University of Bourgogne-Franche-Comté, 21000 Dijon, France
- Reference Center for Rare Pulmonary Diseases, University Hospital, Bourgogne-Franche Comté, 21000 Dijon, France
- Réseau OrphaLung, Filière RespiFIl, Department of Pulmonary Medicine and Intensive Care Unit, University Hospital, Bourgogne-Franche Comté, 21000 Dijon, France
| | - Pierre-Marie Boutanquoi
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, 21000 Dijon, France
| | - Olivier Burgy
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, 21000 Dijon, France
- UFR des Sciences de Santé, University of Bourgogne-Franche-Comté, 21000 Dijon, France
- Reference Center for Rare Pulmonary Diseases, University Hospital, Bourgogne-Franche Comté, 21000 Dijon, France
- Réseau OrphaLung, Filière RespiFIl, Department of Pulmonary Medicine and Intensive Care Unit, University Hospital, Bourgogne-Franche Comté, 21000 Dijon, France
| | - Lucile Dondaine
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, 21000 Dijon, France
- Reference Center for Rare Pulmonary Diseases, University Hospital, Bourgogne-Franche Comté, 21000 Dijon, France
- Réseau OrphaLung, Filière RespiFIl, Department of Pulmonary Medicine and Intensive Care Unit, University Hospital, Bourgogne-Franche Comté, 21000 Dijon, France
| | - Guillaume Beltramo
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, 21000 Dijon, France
- UFR des Sciences de Santé, University of Bourgogne-Franche-Comté, 21000 Dijon, France
- Reference Center for Rare Pulmonary Diseases, University Hospital, Bourgogne-Franche Comté, 21000 Dijon, France
- Réseau OrphaLung, Filière RespiFIl, Department of Pulmonary Medicine and Intensive Care Unit, University Hospital, Bourgogne-Franche Comté, 21000 Dijon, France
| | - Burhan Uyanik
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, 21000 Dijon, France
| | - Carmen Garrido
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, 21000 Dijon, France
- Reference Center for Rare Pulmonary Diseases, University Hospital, Bourgogne-Franche Comté, 21000 Dijon, France
- Réseau OrphaLung, Filière RespiFIl, Department of Pulmonary Medicine and Intensive Care Unit, University Hospital, Bourgogne-Franche Comté, 21000 Dijon, France
| | - Philippe Bonniaud
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, 21000 Dijon, France
- UFR des Sciences de Santé, University of Bourgogne-Franche-Comté, 21000 Dijon, France
- Reference Center for Rare Pulmonary Diseases, University Hospital, Bourgogne-Franche Comté, 21000 Dijon, France
- Réseau OrphaLung, Filière RespiFIl, Department of Pulmonary Medicine and Intensive Care Unit, University Hospital, Bourgogne-Franche Comté, 21000 Dijon, France
| | - Pierre-Simon Bellaye
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, 21000 Dijon, France
- Reference Center for Rare Pulmonary Diseases, University Hospital, Bourgogne-Franche Comté, 21000 Dijon, France
- Réseau OrphaLung, Filière RespiFIl, Department of Pulmonary Medicine and Intensive Care Unit, University Hospital, Bourgogne-Franche Comté, 21000 Dijon, France
- Cancer Center George François Leclerc, 21000 Dijon, France
| | - Françoise Goirand
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, 21000 Dijon, France
- UFR des Sciences de Santé, University of Bourgogne-Franche-Comté, 21000 Dijon, France
- Reference Center for Rare Pulmonary Diseases, University Hospital, Bourgogne-Franche Comté, 21000 Dijon, France
- Réseau OrphaLung, Filière RespiFIl, Department of Pulmonary Medicine and Intensive Care Unit, University Hospital, Bourgogne-Franche Comté, 21000 Dijon, France
| |
Collapse
|
3
|
Limb-Bud and Heart (LBH) Upregulation in Cardiomyocytes under Hypoxia Promotes the Activation of Cardiac Fibroblasts via Exosome Secretion. Mediators Inflamm 2022; 2022:8939449. [PMID: 36110098 PMCID: PMC9470350 DOI: 10.1155/2022/8939449] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022] Open
Abstract
The activation of cardiac fibroblasts (CFs) after myocardial infarction (MI) is essential for post-MI infarct healing, during which the regulation of transforming growth factor beta1 (TGF-β1) signaling is predominant. We have demonstrated that TGF-β1-mediated upregulation of LBH contributes to post-MI CF activation via modulating αB-crystallin (CRYAB), after being upregulated by TGF-β1. In this study, the effect of LBH-CRYAB signaling on the cardiac microenvironment via exosome communication and the corresponding mechanisms were investigated. The upregulation of LBH and CRYAB was verified in both cardiomyocytes (CMs) and CFs in hypoxic, post-MI peri-infarct tissues. CM-derived exosomes were isolated and identified, and LBH distribution was elevated in exosomes derived from LBH-upregulated CMs under hypoxia. Treatment with LBH+ exosomes promoted cellular proliferation, differentiation, and epithelial-mesenchymal transition-like processes in CFs. Additionally, in primary LBHKO CFs, western blotting showed that LBH knockout partially inhibited TGF-β1-induced CF activation, while LBH-CRYAB signaling affected TGF-β1 expression and secretion through a positive feedback loop. The administration of a Smad3 phosphorylation inhibitor to LBHKO CFs under TGF-β1 stimulation indicated that Smad3 phosphorylation partially accounted for TGF-β1-induced LBH upregulation. In conclusion, LBH upregulation in CMs in post-MI peri-infarct areas correlated with a hypoxic cardiac microenvironment and led to elevated exosomal LBH levels, promoting the activation of recipient CFs, which brings new insights into the studies and therapeutic strategies of post-MI cardiac repair.
Collapse
|
4
|
Tucker TA, Idell S. Update on Novel Targeted Therapy for Pleural Organization and Fibrosis. Int J Mol Sci 2022; 23:ijms23031587. [PMID: 35163509 PMCID: PMC8835949 DOI: 10.3390/ijms23031587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 11/22/2022] Open
Abstract
Pleural injury and subsequent loculation is characterized by acute injury, sustained inflammation and, when severe, pathologic tissue reorganization. While fibrin deposition is a normal part of the injury response, disordered fibrin turnover can promote pleural loculation and, when unresolved, fibrosis of the affected area. Within this review, we present a brief discussion of the current IPFT therapies, including scuPA, for the treatment of pathologic fibrin deposition and empyema. We also discuss endogenously expressed PAI-1 and how it may affect the efficacy of IPFT therapies. We further delineate the role of pleural mesothelial cells in the progression of pleural injury and subsequent pleural remodeling resulting from matrix deposition. We also describe how pleural mesothelial cells promote pleural fibrosis as myofibroblasts via mesomesenchymal transition. Finally, we discuss novel therapeutic targets which focus on blocking and/or reversing the myofibroblast differentiation of pleural mesothelial cells for the treatment of pleural fibrosis.
Collapse
|
5
|
Herzog R, Sacnun JM, González-Mateo G, Bartosova M, Bialas K, Wagner A, Unterwurzacher M, Sobieszek IJ, Daniel-Fischer L, Rusai K, Pascual-Antón L, Kaczirek K, Vychytil A, Schmitt CP, López-Cabrera M, Alper SL, Aufricht C, Kratochwill K. Lithium preserves peritoneal membrane integrity by suppressing mesothelial cell αB-crystallin. Sci Transl Med 2021; 13:13/608/eaaz9705. [PMID: 34433641 DOI: 10.1126/scitranslmed.aaz9705] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 04/30/2021] [Accepted: 08/04/2021] [Indexed: 01/18/2023]
Abstract
Life-saving renal replacement therapy by peritoneal dialysis (PD) is limited in use and duration by progressive impairment of peritoneal membrane integrity and homeostasis. Preservation of peritoneal membrane integrity during chronic PD remains an urgent but long unmet medical need. PD therapy failure results from peritoneal fibrosis and angiogenesis caused by hypertonic PD fluid (PDF)-induced mesothelial cytotoxicity. However, the pathophysiological mechanisms involved are incompletely understood, limiting identification of therapeutic targets. We report that addition of lithium chloride (LiCl) to PDF is a translatable intervention to counteract PDF-induced mesothelial cell death, peritoneal membrane fibrosis, and angiogenesis. LiCl improved mesothelial cell survival in a dose-dependent manner. Combined transcriptomic and proteomic characterization of icodextrin-based PDF-induced mesothelial cell injury identified αB-crystallin as the mesothelial cell protein most consistently counter-regulated by LiCl. In vitro and in vivo overexpression of αB-crystallin triggered a fibrotic phenotype and PDF-like up-regulation of vascular endothelial growth factor (VEGF), CD31-positive cells, and TGF-β-independent activation of TGF-β-regulated targets. In contrast, αB-crystallin knockdown decreased VEGF expression and early mesothelial-to-mesenchymal transition. LiCl reduced VEGF release and counteracted fibrosis- and angiogenesis-associated processes. αB-crystallin in patient-derived mesothelial cells was specifically up-regulated in response to PDF and increased in peritoneal mesothelial cells from biopsies from pediatric patients undergoing PD, correlating with markers of angiogenesis and fibrosis. LiCl-supplemented PDF promoted morphological preservation of mesothelial cells and the submesothelial zone in a mouse model of chronic PD. Thus, repurposing LiCl as a cytoprotective PDF additive may offer a translatable therapeutic strategy to combat peritoneal membrane deterioration during PD therapy.
Collapse
Affiliation(s)
- Rebecca Herzog
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Juan Manuel Sacnun
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria.,Zytoprotec GmbH, 1090 Vienna, Austria
| | - Guadalupe González-Mateo
- Tissue and Organ Homeostasis, Molecular Biology Centre Severo Ochoa, CSIC-UAM, 28049 Madrid, Spain
| | - Maria Bartosova
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University of Heidelberg, 69120 Heidelberg, Germany
| | - Katarzyna Bialas
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria.,Zytoprotec GmbH, 1090 Vienna, Austria
| | - Anja Wagner
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Markus Unterwurzacher
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Isabel J Sobieszek
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Lisa Daniel-Fischer
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria.,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Krisztina Rusai
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Lucía Pascual-Antón
- Tissue and Organ Homeostasis, Molecular Biology Centre Severo Ochoa, CSIC-UAM, 28049 Madrid, Spain
| | - Klaus Kaczirek
- Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Andreas Vychytil
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, 1090 Vienna, Austria
| | - Claus Peter Schmitt
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University of Heidelberg, 69120 Heidelberg, Germany
| | - Manuel López-Cabrera
- Tissue and Organ Homeostasis, Molecular Biology Centre Severo Ochoa, CSIC-UAM, 28049 Madrid, Spain
| | - Seth L Alper
- Division of Nephrology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Christoph Aufricht
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Klaus Kratochwill
- Christian Doppler Laboratory for Molecular Stress Research in Peritoneal Dialysis, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, 1090 Vienna, Austria. .,Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
6
|
Limb-bud and Heart (LBH) mediates proliferation, fibroblast-to-myofibroblast transition and EMT-like processes in cardiac fibroblasts. Mol Cell Biochem 2021; 476:2685-2701. [PMID: 33666830 DOI: 10.1007/s11010-021-04111-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/12/2021] [Indexed: 10/22/2022]
Abstract
Cardiac fibrosis is an important pathological change after myocardial infarction (MI). Its progression is essential for post-MI infarct healing, during which transforming growth factor beta1 (TGF-β1) plays a critical role. Limb-bud and Heart (LBH), a newly discovered target gene of TGF-β1, was shown to promote normal cardiogenesis. αB-crystallin (CRYAB), an LBH-interacting protein, was demonstrated to be involved in TGF-β1-induced fibrosis. The roles and molecular mechanisms of LBH and CRYAB during cardiac fibrosis remain largely unexplored. In this study, we investigated the alterations of LBH and CRYAB expression in mouse cardiac tissue after MI. LBH and CRYAB were upregulated in activated cardiac fibroblasts (CFs), while in vitro TGF-β1 stimulation induced the upregulation of LBH, CRYAB, and fibrogenic genes in primary CFs of neonatal rats. The results of the ectopic expression of LBH proved that LBH accelerated CF proliferation under hypoxia, mediated the expression of CRYAB and fibrogenic genes, and promoted epithelial-mesenchymal transition (EMT)-like processes in rat CFs, while subsequent CRYAB silencing reversed the effects induced by elevated LBH expression. We also verified the protein-protein interaction (PPI) between LBH and CRYAB in fibroblasts. In summary, our work demonstrated that LBH promotes the proliferation of CFs, mediates TGF-β1-induced fibroblast-to-myofibroblast transition and EMT-like processes through CRYAB upregulation, jointly functioning in post-MI infarct healing. These findings suggest that LBH could be a promising potential target for the study of cardiac repair and cardiac fibrosis.
Collapse
|
7
|
Chen X, Xu H, Hou J, Wang H, Zheng Y, Li H, Cai H, Han X, Dai J. Epithelial cell senescence induces pulmonary fibrosis through Nanog-mediated fibroblast activation. Aging (Albany NY) 2019; 12:242-259. [PMID: 31891567 PMCID: PMC6977687 DOI: 10.18632/aging.102613] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive lung disease tightly correlated with aging. The pathological features of IPF include epithelial cell senescence and abundant foci of highly activated pulmonary fibroblasts. However, the underlying mechanism between epithelial cell senescence and pulmonary fibroblast activation remain to be elucidated. In our study, we demonstrated that Nanog, as a pluripotency gene, played an essential role in the activation of pulmonary fibroblasts. In the progression of IPF, senescent epithelial cells could contribute to the activation of pulmonary fibroblasts via increasing the expression of senescence-associated secretory phenotype (SASP). In addition, we found activated pulmonary fibroblasts exhibited aberrant activation of Wnt/β-catenin signalling and elevated expression of Nanog. Further study revealed that the activation of Wnt/β-catenin signalling was responsible for senescent epithelial cell-induced Nanog phenotype in pulmonary fibroblasts. β-catenin was observed to bind to the promoter of Nanog during the activation of pulmonary fibroblasts. Targeted inhibition of epithelial cell senescence or Nanog could effectively suppress the activation of pulmonary fibroblasts and impair the development of pulmonary fibrosis, indicating a potential for the exploration of novel anti-fibrotic strategies.
Collapse
Affiliation(s)
- Xiang Chen
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China.,Immunology and Reproduction Biology Laboratory and State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Hongyang Xu
- Department of Critical Care Medicine, The Affiliated WuXi People's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Jiwei Hou
- Immunology and Reproduction Biology Laboratory and State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Hui Wang
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yi Zheng
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Hui Li
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Hourong Cai
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Xiaodong Han
- Immunology and Reproduction Biology Laboratory and State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Jinghong Dai
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| |
Collapse
|
8
|
The pleural mesothelium and transforming growth factor-β1 pathways in restrictive allograft syndrome: A pre-clinical investigation. J Heart Lung Transplant 2019; 38:570-579. [DOI: 10.1016/j.healun.2019.02.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 12/21/2022] Open
|
9
|
A monoclonal antibody targeted to the functional peptide of αB-crystallin inhibits the chaperone and anti-apoptotic activities. J Immunol Methods 2019; 467:37-47. [PMID: 30738041 DOI: 10.1016/j.jim.2019.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/24/2019] [Accepted: 02/05/2019] [Indexed: 01/18/2023]
Abstract
αB-Crystallin is a member of the small heat shock protein family. It is a molecular chaperone and an anti-apoptotic protein. Previous studies have shown that the peptide (73DRFSVNLDVKHFSPEELKVKV93, hereafter referred to as peptain-1) from the core domain of αB-crystallin exhibits both chaperone and anti-apoptotic properties similar to the parent protein. We developed a mouse monoclonal antibody against peptain-1 with the aim of blocking the functions of αB-crystallin. The antibody reacted with peptain-1, it did not react with the chaperone peptide of αA-crystallin. The antibody strongly reacted with human recombinant αB-crystallin but weakly with Hsp20; it did not react with αA-crystallin or Hsp27. The antibody specifically reacted with αB-crystallin in human and mouse lens proteins but not with αA-crystallin. The antibody reacted with αB-crystallin in human lens epithelial cells, human retinal endothelial cells, and with peptain-1 in peptain-1-transduced cells. Unlike the commercial antibodies against αB-crystallin, the antibody against peptain-1 inhibited the chaperone and anti-apoptotic activities of peptain-1. The antibody might find use in inhibiting αB-crystallin's chaperone and anti-apoptotic activities in diseases where αB-crystallin is a causative or contributing factor.
Collapse
|
10
|
Matrix-bound AGEs enhance TGFβ2-mediated mesenchymal transition of lens epithelial cells via the noncanonical pathway: implications for secondary cataract formation. Biochem J 2018; 475:1427-1440. [PMID: 29588342 DOI: 10.1042/bcj20170856] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/19/2018] [Accepted: 03/27/2018] [Indexed: 12/28/2022]
Abstract
Advanced glycation end products (AGEs) are post-translational modifications formed from the reaction of reactive carbonyl compounds with amino groups in proteins. Our laboratory has previously shown that AGEs in extracellular matrix (ECM) proteins promote TGFβ2 (transforming growth factor-beta 2)-mediated epithelial-to-mesenchymal transition (EMT) of lens epithelial cells (LECs), which could play a role in fibrosis associated with posterior capsule opacification. We have also shown that αB-crystallin plays an important role in TGFβ2-mediated EMT of LECs. Here, we investigated the signaling mechanisms by which ECM-AGEs enhance TGFβ2-mediated EMT in LECs. We found that in LECs cultured on AGE-modified basement protein extract (AGE-BME), TGFβ2 treatment up-regulated the mesenchymal markers α-SMA (α-smooth muscle actin) and αB-crystallin and down-regulated the epithelial marker E-cadherin more than LECs cultured on unmodified BME and treated with TGFβ2. Using a Multiplex Assay, we found that AGE-BME significantly up-regulated the noncanonical pathway by promoting phosphorylation of ERK (extracellular signal-regulated kinases), AKT, and p38 MAPK (mitogen-activated protein kinases) during TGFβ2-mediated EMT. This EMT response was strongly suppressed by inhibition of AKT and p38 MAPK phosphorylation. The AKT inhibitor LY294002 also suppressed TGFβ2-induced up-regulation of nuclear Snail and reduced phosphorylation of GSK3β. Inhibition of Snail expression suppressed TGFβ2-mediated α-SMA expression. αB-Crystallin was up-regulated in an AKT-dependent manner during AGE-BME/TGFβ2-mediated EMT in LECs. The absence of αB-crystallin in LECs suppressed TGFβ2-induced GSK3β phosphorylation, resulting in lower Snail levels. Taken together, these results show that ECM-AGEs enhance the TGFβ2-mediated EMT response through activation of the AKT/Snail pathway, in which αB-crystallin plays an important role as a linker between the TGFβ2 and AGE-mediated signaling pathways.
Collapse
|
11
|
Besnard V, Dagher R, Madjer T, Joannes A, Jaillet M, Kolb M, Bonniaud P, Murray LA, Sleeman MA, Crestani B. Identification of periplakin as a major regulator of lung injury and repair in mice. JCI Insight 2018. [PMID: 29515024 DOI: 10.1172/jci.insight.90163] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Periplakin is a component of the desmosomes that acts as a cytolinker between intermediate filament scaffolding and the desmosomal plaque. Periplakin is strongly expressed by epithelial cells in the lung and is a target antigen for autoimmunity in idiopathic pulmonary fibrosis. The aim of this study was to determine the role of periplakin during lung injury and remodeling in a mouse model of lung fibrosis induced by bleomycin. We found that periplakin expression was downregulated in the whole lung and in alveolar epithelial cells following bleomycin-induced injury. Deletion of the Ppl gene in mice improved survival and reduced lung fibrosis development after bleomycin-induced injury. Notably, Ppl deletion promoted an antiinflammatory alveolar environment linked to profound changes in type 2 alveolar epithelial cells, including overexpression of antiinflammatory cytokines, decreased expression of profibrotic mediators, and altered cell signaling with a reduced response to TGF-β1. These results identify periplakin as a previously unidentified regulator of the response to injury in the lung.
Collapse
Affiliation(s)
| | | | | | | | | | - Martin Kolb
- Department of Medecine, Firestone Institute for respiratory Health, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, Canada
| | | | - Lynne A Murray
- MedImmune Ltd, Granta Park, Cambridgeshire, United Kingdom.,Respiratory, Inflammation, Autoimmunity (RIA) IMED Biotech unit, AstraZeneca, Gothenburg, Sweden
| | | | - Bruno Crestani
- INSERM U1152, Paris, France.,Université Paris Diderot, LABEX INFLAMEX, Paris, France.,Assistance Publique-Hôpitaux de Paris, DHU FIRE, Hôpital Bichat, Paris, France
| |
Collapse
|
12
|
Bonniaud P, Burgy O, Garrido C. Heat shock protein-90 toward theranostics: a breath of fresh air in idiopathic pulmonary fibrosis. Eur Respir J 2018; 51:13993003.02612-2017. [PMID: 29437951 DOI: 10.1183/13993003.02612-2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 12/21/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Philippe Bonniaud
- Service de Pneumologie et Soins Intensifs Respiratoires, Centre Hospitalo-Universitaire de Dijon-Bourgogne, Dijon, France .,Faculté de Médecine et Pharmacie, Université de Bourgogne-Franche Comté, Dijon, France.,INSERM UMR 1231, Dijon, France
| | - Olivier Burgy
- Faculté de Médecine et Pharmacie, Université de Bourgogne-Franche Comté, Dijon, France.,INSERM UMR 1231, Dijon, France.,Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Carmen Garrido
- Faculté de Médecine et Pharmacie, Université de Bourgogne-Franche Comté, Dijon, France.,INSERM UMR 1231, Dijon, France
| |
Collapse
|
13
|
Mümmler C, Burgy O, Hermann S, Mutze K, Günther A, Königshoff M. Cell-specific expression of runt-related transcription factor 2 contributes to pulmonary fibrosis. FASEB J 2018; 32:703-716. [PMID: 28986417 DOI: 10.1096/fj.201700482r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease with limited therapeutic options and unknown etiology. IPF is characterized by epithelial cell injury, impaired cellular crosstalk between epithelial cells and fibroblasts, and the formation of fibroblast foci with increased extracellular matrix deposition (ECM). We investigated the role of runt-related transcription factor 2 (RUNX2), a master regulator of bone development that has been linked to profibrotic signaling. RUNX2 expression was up-regulated in lung homogenates from patients with IPF and in experimental bleomycin-induced lung fibrosis. The RUNX2 level correlated with disease severity as measured by decreased diffusing capacity and increased levels of the IPF biomarker, matrix metalloproteinase 7. Nuclear RUNX2 was observed in prosurfactant protein C-positive hyperplastic epithelial cells and was rarely found in myofibroblasts. We discovered an up-regulation of RUNX2 in fibrotic alveolar epithelial type II (ATII) cells as well as an increase of RUNX2-negative fibroblasts in experimental and human pulmonary fibrosis. Functionally, small interfering RNA-mediated RUNX2 knockdown decreased profibrotic ATII cell function, such as proliferation and migration, whereas fibroblasts displayed activation markers and increased ECM expression after RUNX2 knockdown. This study reveals that RUNX2 is differentially expressed in ATII cells vs. fibroblasts in lung fibrosis, which contributes to profibrotic cell function. Cell-specific targeting of RUNX2 pathways may represent a therapeutic approach for IPF.-Mümmler, C., Burgy, O., Hermann, S., Mutze, K., Günther, A., Königshoff, M. Cell-specific expression of runt-related transcription factor 2 contributes to pulmonary fibrosis.
Collapse
Affiliation(s)
- Carlo Mümmler
- Comprehensive Pneumology Center, Helmholtz Center Munich, University Hospital Grosshadern, Ludwig Maximilians University München, Munich, Germany
| | - Olivier Burgy
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Denver, Colorado, USA
| | - Sarah Hermann
- Comprehensive Pneumology Center, Helmholtz Center Munich, University Hospital Grosshadern, Ludwig Maximilians University München, Munich, Germany
| | - Kathrin Mutze
- Comprehensive Pneumology Center, Helmholtz Center Munich, University Hospital Grosshadern, Ludwig Maximilians University München, Munich, Germany
| | - Andreas Günther
- Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany
| | - Melanie Königshoff
- Comprehensive Pneumology Center, Helmholtz Center Munich, University Hospital Grosshadern, Ludwig Maximilians University München, Munich, Germany.,Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Denver, Colorado, USA
| |
Collapse
|
14
|
Burgy O, Bellaye PS, Causse S, Beltramo G, Wettstein G, Boutanquoi PM, Goirand F, Garrido C, Bonniaud P. Pleural inhibition of the caspase-1/IL-1β pathway diminishes profibrotic lung toxicity of bleomycin. Respir Res 2016; 17:162. [PMID: 27894300 PMCID: PMC5127006 DOI: 10.1186/s12931-016-0475-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/22/2016] [Indexed: 01/21/2023] Open
Abstract
Background Idiopathic and toxic pulmonary fibrosis are severe diseases starting classically in the subpleural area of the lung. It has recently been suggested that pleural mesothelial cells acquire a myofibroblast phenotype under fibrotic conditions induced by TGF-β1 or bleomycin. The importance and role of inflammation in fibrogenesis are still controversial. In this work, we explored the role of IL-1β/caspase-1 signaling in bleomycin lung toxicity and in pleural mesothelial cell transformation. Methods C57BL/6 mice were intravenously injected with either bleomycin or nigericin or NaCl as control. In vitro, the Met5A cell line was used as a model of human pleural mesothelial cells. Results Intravenous injections of bleomycin induced lung fibrosis with histologically-proven peripheral distribution, collagen accumulation in the pleural and subpleural area, and overexpression of markers of myofibroblast transformation of pleural cells which migrated into the lung. These events were associated with an inflammatory process with an increase in neutrophil recruitment in pleural lavage fluid and increased caspase-1 activity. TGF-β1 was also overexpressed in pleural lavage fluid and was produced by pleural cells following intravenous bleomycin. In this model, local pleural inhibition of IL-1β with the IL-1β inhibitor anakinra diminished TGF-β1 and collagen accumulation. In vitro, caspase-1 inhibition interfered with Met5A cell transformation into the myofibroblast-like phenotype induced by bleomycin or TGF-β1. Moreover, nigericin, a caspase-1 activator, triggered transformation of Met5A cells and its intra-pleural delivery induced fibrogenesis in mice. Conclusions We demonstrated, after intravenous bleomycin injection in mice, the role of the pleura and highlighted the key role of IL-1β/caspase-1 axis in this fibrogenesis process. Electronic supplementary material The online version of this article (doi:10.1186/s12931-016-0475-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Olivier Burgy
- INSERM, LNC UMR866, LipSTIC LabEx team, Université Bourgogne Franche-Comté, 21000, Dijon, France
| | - Pierre-Simon Bellaye
- INSERM, LNC UMR866, LipSTIC LabEx team, Université Bourgogne Franche-Comté, 21000, Dijon, France
| | - Sebastien Causse
- INSERM, LNC UMR866, LipSTIC LabEx team, Université Bourgogne Franche-Comté, 21000, Dijon, France
| | - Guillaume Beltramo
- INSERM, LNC UMR866, LipSTIC LabEx team, Université Bourgogne Franche-Comté, 21000, Dijon, France.,Service de Pneumologie et Soins Intensifs Respiratoires, Centre Hospitalo-Universitaire de Bourgogne, 21000, Dijon, France
| | - Guillaume Wettstein
- INSERM, LNC UMR866, LipSTIC LabEx team, Université Bourgogne Franche-Comté, 21000, Dijon, France
| | - Pierre-Marie Boutanquoi
- INSERM, LNC UMR866, LipSTIC LabEx team, Université Bourgogne Franche-Comté, 21000, Dijon, France
| | - Françoise Goirand
- INSERM, LNC UMR866, LipSTIC LabEx team, Université Bourgogne Franche-Comté, 21000, Dijon, France
| | - Carmen Garrido
- INSERM, LNC UMR866, LipSTIC LabEx team, Université Bourgogne Franche-Comté, 21000, Dijon, France.,Anticancer Centre Georges François Leclerc, CGFL, 21000, Dijon, France
| | - Philippe Bonniaud
- INSERM, LNC UMR866, LipSTIC LabEx team, Université Bourgogne Franche-Comté, 21000, Dijon, France. .,Service de Pneumologie et Soins Intensifs Respiratoires, Centre Hospitalo-Universitaire de Bourgogne, 21000, Dijon, France.
| |
Collapse
|
15
|
Boehme SA, Franz-Bacon K, DiTirro DN, Ly TW, Bacon KB. MAP3K19 Is a Novel Regulator of TGF-β Signaling That Impacts Bleomycin-Induced Lung Injury and Pulmonary Fibrosis. PLoS One 2016; 11:e0154874. [PMID: 27144281 PMCID: PMC4856290 DOI: 10.1371/journal.pone.0154874] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 04/20/2016] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, debilitating disease for which two medications, pirfenidone and nintedanib, have only recently been approved for treatment. The cytokine TGF-β has been shown to be a central mediator in the disease process. We investigated the role of a novel kinase, MAP3K19, upregulated in IPF tissue, in TGF-β-induced signal transduction and in bleomycin-induced pulmonary fibrosis. MAP3K19 has a very limited tissue expression, restricted primarily to the lungs and trachea. In pulmonary tissue, expression was predominantly localized to alveolar and interstitial macrophages, bronchial epithelial cells and type II pneumocytes of the epithelium. MAP3K19 was also found to be overexpressed in bronchoalveolar lavage macrophages from IPF patients compared to normal patients. Treatment of A549 or THP-1 cells with either MAP3K19 siRNA or a highly potent and specific inhibitor reduced phospho-Smad2 & 3 nuclear translocation following TGF-β stimulation. TGF-β-induced gene transcription was also strongly inhibited by both the MAP3K19 inhibitor and nintedanib, whereas pirfenidone had a much less pronounced effect. In combination, the MAP3K19 inhibitor appeared to act synergistically with either pirfenidone or nintedanib, at the level of target gene transcription or protein production. Finally, in an animal model of IPF, inhibition of MAP3K19 strongly attenuated bleomycin-induced pulmonary fibrosis when administered either prophylactically ortherapeutically. In summary, these results strongly suggest that inhibition of MAP3K19 may have a beneficial therapeutic effect in the treatment of IPF and represents a novel strategy to target this disease.
Collapse
Affiliation(s)
- Stefen A. Boehme
- AxikinPharmaceuticals, Inc., San Diego, California, United States of America
| | - Karin Franz-Bacon
- DNA Consulting, Inc., San Diego, California, United States of America
| | - Danielle N. DiTirro
- AxikinPharmaceuticals, Inc., San Diego, California, United States of America
| | - Tai Wei Ly
- AxikinPharmaceuticals, Inc., San Diego, California, United States of America
| | - Kevin B. Bacon
- AxikinPharmaceuticals, Inc., San Diego, California, United States of America
| |
Collapse
|
16
|
Ishikawa K, Sreekumar PG, Spee C, Nazari H, Zhu D, Kannan R, Hinton DR. αB-Crystallin Regulates Subretinal Fibrosis by Modulation of Epithelial-Mesenchymal Transition. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:859-73. [PMID: 26878210 PMCID: PMC4822331 DOI: 10.1016/j.ajpath.2015.11.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 10/20/2015] [Accepted: 11/19/2015] [Indexed: 01/18/2023]
Abstract
Subretinal fibrosis is an end stage of neovascular age-related macular degeneration, characterized by fibrous membrane formation after choroidal neovascularization. An initial step of the pathogenesis is an epithelial-mesenchymal transition (EMT) of retinal pigment epithelium cells. αB-crystallin plays multiple roles in age-related macular degeneration, including cytoprotection and angiogenesis. However, the role of αB-crystallin in subretinal EMT and fibrosis is unknown. Herein, we showed attenuation of subretinal fibrosis after regression of laser-induced choroidal neovascularization and a decrease in mesenchymal retinal pigment epithelium cells in αB-crystallin knockout mice compared with wild-type mice. αB-crystallin was prominently expressed in subretinal fibrotic lesions in mice. In vitro, overexpression of αB-crystallin induced EMT, whereas suppression of αB-crystallin induced a mesenchymal-epithelial transition. Transforming growth factor-β2-induced EMT was further enhanced by overexpression of αB-crystallin but was inhibited by suppression of αB-crystallin. Silencing of αB-crystallin inhibited multiple fibrotic processes, including cell proliferation, migration, and fibronectin production. Bone morphogenetic protein 4 up-regulated αB-crystallin, and its EMT induction was inhibited by knockdown of αB-crystallin. Furthermore, inhibition of αB-crystallin enhanced monotetraubiquitination of SMAD4, which can impair its nuclear localization. Overexpression of αB-crystallin enhanced nuclear translocation and accumulation of SMAD4 and SMAD5. Thus, αB-crystallin is an important regulator of EMT, acting as a molecular chaperone for SMAD4 and as its potential therapeutic target for preventing subretinal fibrosis development in neovascular age-related macular degeneration.
Collapse
Affiliation(s)
- Keijiro Ishikawa
- Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, California; Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | | | - Christine Spee
- Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Hossein Nazari
- Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Danhong Zhu
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Ram Kannan
- Arnold and Mabel Beckman Macular Research Center, Doheny Eye Institute, Los Angeles, California
| | - David R Hinton
- Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California; Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California.
| |
Collapse
|
17
|
Nagaraj RH, Nahomi RB, Mueller NH, Raghavan CT, Ammar DA, Petrash JM. Therapeutic potential of α-crystallin. Biochim Biophys Acta Gen Subj 2015; 1860:252-7. [PMID: 25840354 DOI: 10.1016/j.bbagen.2015.03.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 03/26/2015] [Indexed: 01/18/2023]
Abstract
BACKGROUND The findings that α-crystallins are multi-functional proteins with diverse biological functions have generated considerable interest in understanding their role in health and disease. Recent studies have shown that chaperone peptides of α-crystallin could be delivered into cultured cells and in experimental animals with beneficial effects against protein aggregation, oxidation, inflammation and apoptosis. SCOPE OF REVIEW In this review, we will summarize the latest developments on the therapeutic potential of α-crystallins and their functional peptides. MAJOR CONCLUSIONS α-Crystallins and their functional peptides have shown significant favorable effects against several diseases. Their targeted delivery to tissues would be of great therapeutic benefit. However, α-crystallins can also function as disease-causing proteins. These seemingly contradictory functions must be carefully considered prior to their therapeutic use. GENERAL SIGNIFICANCE αA and αB-Crystallin are members of the small heat shock protein family. These proteins exhibit molecular chaperone and anti-apoptotic activities. The core crystallin domain within these proteins is largely responsible for these prosperities. Recent studies have identified peptides within the crystallin domain of both α- and αB-crystallins with remarkable chaperone and anti-apoptotic activities. Administration of α-crystallin or their functional peptides has shown substantial inhibition of pathologies in several diseases. However, α-crystallins have been shown to promote disease-causing pathways. These two sides of the proteins are discussed in this review. This article is part of a Special Issue entitled Crystallin Biochemistry in Health and Disease.
Collapse
Affiliation(s)
- Ram H Nagaraj
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | - Rooban B Nahomi
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Niklaus H Mueller
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Cibin T Raghavan
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - David A Ammar
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - J Mark Petrash
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|