1
|
Abbad L, Esteve E, Chatziantoniou C. Advances and challenges in kidney fibrosis therapeutics. Nat Rev Nephrol 2025; 21:314-329. [PMID: 39934355 DOI: 10.1038/s41581-025-00934-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 02/13/2025]
Abstract
Chronic kidney disease (CKD) is a major global health burden that affects more than 10% of the adult population. Current treatments, including dialysis and transplantation, are costly and not curative. Kidney fibrosis, defined as an abnormal accumulation of extracellular matrix in the kidney parenchyma, is a common outcome in CKD, regardless of disease aetiology, and is a major cause of loss of kidney function and kidney failure. For this reason, research efforts have focused on identifying mediators of kidney fibrosis to inform the development of effective anti-fibrotic treatments. Given the prominent role of the transforming growth factor-β (TGFβ) family in fibrosis, efforts have focused on inhibiting TGFβ signalling. Despite hopes raised by the efficacy of this approach in preclinical models, translation into clinical practice has not met expectations. Antihypertensive and antidiabetic drugs slow the decline in kidney function and could slow fibrosis but, owing to the lack of technologies for in vivo renal imaging, their anti-fibrotic effect cannot be truly assessed at present. The emergence of new drugs targeting pro-fibrotic signalling, or enabling cell repair and cell metabolic reprogramming, combined with better stratification of people with CKD and the arrival of nanotechnologies for kidney-specific drug delivery, open up new perspectives for the treatment of this major public health challenge.
Collapse
Affiliation(s)
- Lilia Abbad
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France
| | - Emmanuel Esteve
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France
| | - Christos Chatziantoniou
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France.
| |
Collapse
|
2
|
Zhao T, Su Y. Mechanisms and Therapeutic Potential of Myofibroblast Transformation in Pulmonary Fibrosis. JOURNAL OF RESPIRATORY BIOLOGY AND TRANSLATIONAL MEDICINE 2025; 2:10001. [PMID: 40190620 PMCID: PMC11970920 DOI: 10.70322/jrbtm.2025.10001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, irreversible, and fatal disease with an increasing incidence and limited therapeutic options. It is characterized by the formation and deposition of excess extracellular matrix proteins resulting in the gradual replacement of normal lung architecture by fibrous tissue. The cellular and molecular mechanism of IPF has not been fully understood. A hallmark in IPF is pulmonary fibroblast to myofibroblast transformation (FMT). During excessive lung repair upon exposure to harmful stimuli, lung fibroblasts transform into myofibroblasts under stimulation of cytokines, chemokines, and vesicles from various cells. These mediators interact with lung fibroblasts, initiating multiple signaling cascades, such as TGFβ1, MAPK, Wnt/β-catenin, NF-κB, AMPK, endoplasmic reticulum stress, and autophagy, contributing to lung FMT. Furthermore, single-cell transcriptomic analysis has revealed significant heterogeneity among lung myofibroblasts, which arise from various cell types and are adapted to the altered microenvironment during pathological lung repair. This review provides an overview of recent research on the origins of lung myofibroblasts and the molecular pathways driving their formation, with a focus on the interactions between lung fibroblasts and epithelial cells, endothelial cells, and macrophages in the context of lung fibrosis. Based on these molecular insights, targeting the lung FMT could offer promising avenues for the treatment of IPF.
Collapse
Affiliation(s)
- Tianming Zhao
- Department of Pharmacology & Toxicology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Yunchao Su
- Department of Pharmacology & Toxicology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30912, USA
| |
Collapse
|
3
|
Jung W, Yang MJ, Kang MS, Lim J, Choi H, Lee JA, Yoon KS, Kim JB, Park EJ. Didecyldimethylammonium chloride-induced lung fibrosis may be associated with phospholipidosis. Toxicol Appl Pharmacol 2025; 495:117211. [PMID: 39710153 DOI: 10.1016/j.taap.2024.117211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
In the current study, we dosed didecyldimethylammonium chloride (DDAC) in mice by pharyngeal aspiration for 28 days or 90 days (weekly) and tried to elucidate the relationship between lamellar body formation and the lesions. When exposed for 28 days (0, 5, 10, 50, and 100 μg/head), all the mice in the 50 and 100 μg/head groups died since Day 2 after the third dosing (Day 16 after the first dosing). Edema, necrosis of bronchiolar and alveolar epithelium, and fibrinous exudate were observed in the lungs of all the dead mice, and chronic inflammatory lesions were observed in the lung tissues of alive mice. When dosed with DDAC of 0, 1, 4, and 8 μg/head for 13 weeks, the total number of pulmonary cells and the pulmonary levels of pro- and anti-inflammatory cytokines significantly increased, and chronic inflammatory lesions were detected with the production of collagen, collagen fibers, and lamellar body-like structures. Swelling of the nuclear envelope and nucleoplasmic components and generation of lipid droplets were also notably observed in the lung tissues of DDAC (8 μg/head)-treated mice. Furthermore, transcriptomic analysis performed using human bronchial epithelial cells showed that DDAC affected the expression of DNA damage, ER stress, lipid metabolism, and transcription regulation-related genes at 6 h after treatment, as it did 24 h treatment and that early growth response factor 1 gene was added to a list of the most up-regulated genes. Meanwhile, cytokines that are associated with the pathology of chronic lung diseases (IL-11, IL-24, and TGF-β) were slightly increased in the lung of DDAC-treated mice, and only the pulmonary level of CCL-2, but not CXCL-1 and CCL-3, increased in both sexes of mice. More importantly, the GM-CSF level increased dose-dependently in the lungs of both sexes of mice exposed to DDAC. Considering that the wound-healing process can take several weeks to complete, we suggest that DDAC-induced pulmonary fibrosis may be attributable to disruption of the wound-healing process due to continuous exposure to DDAC. We also hypothesize that the formation of lamellar bodies may be attributable to lysosomal accumulation of phospholipids separated from the destroyed lung tissue membrane.
Collapse
Affiliation(s)
- Wonkyun Jung
- College of Medicine, Graduate School, Kyung Hee University, 02447, Republic of Korea
| | - Mi-Jin Yang
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongup 56212, Republic of Korea
| | - Min-Sung Kang
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongup 56212, Republic of Korea
| | - Jiyun Lim
- College of Medicine, Graduate School, Kyung Hee University, 02447, Republic of Korea
| | - Hyosun Choi
- National Instrumentation Center for Environmental management, Seoul National University
| | - Ji Ae Lee
- College of Medicine, Graduate School, Kyung Hee University, 02447, Republic of Korea
| | - Kyung-Sik Yoon
- College of Medicine, Graduate School, Kyung Hee University, 02447, Republic of Korea; Human Health and Environmental Toxins Research Center, Kyung Hee University, 02447, Republic of Korea
| | - Jin-Bae Kim
- Human Health and Environmental Toxins Research Center, Kyung Hee University, 02447, Republic of Korea; Division of Cardiology, Department of Internal Medicine, Kyung-Hee University Hospital, Kyung Hee University, 02447, Republic of Korea
| | - Eun-Jung Park
- College of Medicine, Graduate School, Kyung Hee University, 02447, Republic of Korea; Human Health and Environmental Toxins Research Center, Kyung Hee University, 02447, Republic of Korea.
| |
Collapse
|
4
|
Liu L, Wang M, Yu S. Identification of Common Angiogenesis Marker Genes in Chronic Lung Diseases and Their Relationship with Immune Infiltration Based on Bioinformatics Approaches. Biomedicines 2025; 13:331. [PMID: 40002743 PMCID: PMC11852874 DOI: 10.3390/biomedicines13020331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
Objective: This study aims to explore the role of angiogenesis-related genes in chronic lung diseases (ILD and COPD) using bioinformatics methods, with the goal of identifying novel therapeutic targets to slow disease progression and prevent its deterioration into fibrosis or pulmonary artery hypertension. Methods: The research methods encompassed differential analysis, WGCNA (Weighted Gene Co-expression Network Analysis), and multiple machine learning approaches to screen for key genes. Gene Set Enrichment Analysis (GSEA), Gene Ontology (GO), and the Kyoto Encyclopedia of Genes and Genomes (KEGG) were utilized to assess related biological functions and pathways. Additionally, immune cell infiltration was analyzed to evaluate the immune status of the disease and the correlation between genes and immunity. Results: COPD and ILD are closely associated with pathways related to angiogenesis, immune responses, and others, with differential genes in both groups linked to inflammation-related signaling pathways. The study established a chronic lung disease-related gene set comprising 171 genes and further screened out 21 genes related to angiogenesis. Ultimately, four key genes-COL10A1, EDN1, MMP1, and RRAS-were identified through machine learning methods. These four genes are closely related to angiogenesis and immune processes, and clustering analysis based on them can reflect different disease states and variations in immune cell infiltration. Conclusions:COL10A1, EDN1, MMP1, and RRAS represent potential therapeutic targets for slowing the progression of chronic lung diseases and preventing their deterioration. Furthermore, monocytes exhibited consistent infiltration patterns across disease and control groups, as well as among different subgroups, suggesting their potential significant role in the development of chronic lung diseases.
Collapse
Affiliation(s)
| | | | - Shihuan Yu
- Department of Respiratory Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, China; (L.L.); (M.W.)
| |
Collapse
|
5
|
Lalmanach G, Rigoux B, David A, Tahri-Joutey M, Lecaille F, Marchand-Adam S, Saidi A. Human cystatin C in fibrotic diseases. Clin Chim Acta 2025; 565:120016. [PMID: 39461496 DOI: 10.1016/j.cca.2024.120016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
Human cystatin C (hCC), which has a pervasive distribution within body fluids and is ubiquitously expressed by numerous cells and tissues, is a highly potent extracellular inhibitor of cysteine proteases. Besides measurement of serum creatinine, which is the most widely used technique for appraising glomerular filtration rate (GFR), hCC has emerged as a relevant GFR biomarker, because its quantification in serum is less sensitive to interferences with factors such as age, muscle mass or diet. Moreover, there are growing body of evidence that hCC overexpression and/or oversecretion, which is primarily driven by TGF-β1, occur during fibrogenesis (cardiac, liver, oral, and lung fibrosis). Even though molecular mechanisms and signaling pathways governing the regulation of hCC remain to be deciphered more acutely, current data sustain that hCC expression relates to myofibrogenesis and that hCC could be a specific and valuable biomarker of fibrotic disease.
Collapse
Affiliation(s)
- Gilles Lalmanach
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France.
| | - Baptiste Rigoux
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France
| | - Alexis David
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France
| | - Mounia Tahri-Joutey
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France
| | - Fabien Lecaille
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France
| | - Sylvain Marchand-Adam
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France; The University Hospital Center of Tours (CHRU Tours), Pulmonology Department, Tours, France
| | - Ahlame Saidi
- University of Tours, Tours, France; INSERM, UMR1100, Research Center for Respiratory Diseases (CEPR), Team "Proteolytic Enzymes and Their Pharmacological Targeting in Lung Diseases", Tours, France
| |
Collapse
|
6
|
Kim TY, Yeo H, Jung E, Shin SY. EGR1 interacts with p-SMAD at the endothelin-1 gene promoter to regulate gene expression in TGFβ1-stimulated IMR-90 fibroblasts. Biochem Biophys Res Commun 2024; 739:150567. [PMID: 39186868 DOI: 10.1016/j.bbrc.2024.150567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/12/2024] [Accepted: 08/17/2024] [Indexed: 08/28/2024]
Abstract
Pulmonary fibrosis is a severe and progressive lung disease characterized by lung tissue scarring. Transforming growth factor beta 1 (TGFβ1) is crucial in causing pulmonary fibrosis by promoting the activation of fibroblasts and their differentiation into myofibroblasts, which are responsible for excessive extracellular matrix deposition. This study aimed to identify genes activated by TGFβ1 that promote fibrosis and to understand the regulatory pathway controlling myofibroblast. Endothelin-1 (ET-1) was identified as the top-ranking gene in the fibrosis-related gene set using quantitative PCR array analysis. TGFβ1 upregulated EGR1 expression through the ERK1/2 and JNK1/2 MAPK pathways. EGR1 and p-SMAD2 proteins interacted with the ET-1 gene promoter region to regulate TGFβ1-induced ET-1 expression in IMR-90 pulmonary fibroblasts. Mice lacking the Egr1 gene showed reduced ET-1 levels in a model of pulmonary fibrosis induced by intratracheal administration of bleomycin. These findings suggest that targeting EGR1 is a promising approach for treating pulmonary fibrosis, especially idiopathic pulmonary fibrosis, by affecting ET-1 expression and profibrotic reactions.
Collapse
Affiliation(s)
- Tae Yoon Kim
- Department of Biological Sciences, Sanghuh College of Life Sciences, Konkuk University, Seoul, 05029, Republic of Korea
| | - Hyunjin Yeo
- Department of Biological Sciences, Sanghuh College of Life Sciences, Konkuk University, Seoul, 05029, Republic of Korea
| | - Euitaek Jung
- Department of Biological Sciences, Sanghuh College of Life Sciences, Konkuk University, Seoul, 05029, Republic of Korea
| | - Soon Young Shin
- Department of Biological Sciences, Sanghuh College of Life Sciences, Konkuk University, Seoul, 05029, Republic of Korea; Cancer and Metabolism Institute, Konkuk University, Seoul, 05029, Republic of Korea.
| |
Collapse
|
7
|
Huang W, Zheng J, Wang M, Du LY, Bai L, Tang H. The potential therapeutic role of melatonin in organ fibrosis: a comprehensive review. Front Med (Lausanne) 2024; 11:1502368. [PMID: 39735699 PMCID: PMC11681627 DOI: 10.3389/fmed.2024.1502368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/30/2024] [Indexed: 12/31/2024] Open
Abstract
Organ fibrosis is a pathological process characterized by the inability of normal tissue cells to regenerate sufficiently to meet the dynamic repair demands of chronic injury, resulting in excessive extracellular matrix deposition and ultimately leading to organ dysfunction. Despite the increasing depth of research in the field of organ fibrosis and a more comprehensive understanding of its pathogenesis, effective treatments for fibrosis-related diseases are still lacking. Melatonin, a neuroendocrine hormone synthesized by the pineal gland, plays a crucial role in regulating biological rhythms, sleep, and antioxidant defenses. Recent studies have shown that melatonin may have potential in inhibiting organ fibrosis, possibly due to its functions in anti-oxidative stress, anti-inflammation, remodeling the extracellular matrix (ECM), inhibiting epithelial-mesenchymal transition (EMT), and regulating apoptosis, thereby alleviating fibrosis. This review aims to explore the therapeutic potential of melatonin in fibrosis-related human diseases using findings from various in vivo and in vitro studies. These discoveries should provide important insights for the further development of new drugs to treat fibrosis.
Collapse
Affiliation(s)
- Wei Huang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Juan Zheng
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Ming Wang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Ling-Yao Du
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Lang Bai
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Jeong JY, Bafor AE, Freeman BH, Chen PR, Park ES, Kim E. Pathophysiology in Brain Arteriovenous Malformations: Focus on Endothelial Dysfunctions and Endothelial-to-Mesenchymal Transition. Biomedicines 2024; 12:1795. [PMID: 39200259 PMCID: PMC11351371 DOI: 10.3390/biomedicines12081795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
Brain arteriovenous malformations (bAVMs) substantially increase the risk for intracerebral hemorrhage (ICH), which is associated with significant morbidity and mortality. However, the treatment options for bAVMs are severely limited, primarily relying on invasive methods that carry their own risks for intraoperative hemorrhage or even death. Currently, there are no pharmaceutical agents shown to treat this condition, primarily due to a poor understanding of bAVM pathophysiology. For the last decade, bAVM research has made significant advances, including the identification of novel genetic mutations and relevant signaling in bAVM development. However, bAVM pathophysiology is still largely unclear. Further investigation is required to understand the detailed cellular and molecular mechanisms involved, which will enable the development of safer and more effective treatment options. Endothelial cells (ECs), the cells that line the vascular lumen, are integral to the pathogenesis of bAVMs. Understanding the fundamental role of ECs in pathological conditions is crucial to unraveling bAVM pathophysiology. This review focuses on the current knowledge of bAVM-relevant signaling pathways and dysfunctions in ECs, particularly the endothelial-to-mesenchymal transition (EndMT).
Collapse
Affiliation(s)
| | | | | | | | | | - Eunhee Kim
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.Y.J.); (A.E.B.); (B.H.F.); (P.R.C.); (E.S.P.)
| |
Collapse
|
9
|
Mago E, Zhao X, Zhang W, Shao Q, Li P, Huang S, Ding X, Liu H, Sun T, He F, Weng D. RIP1 kinase inactivation protects against LPS-induced acute respiratory distress syndrome in mice. Int Immunopharmacol 2024; 133:112060. [PMID: 38652970 DOI: 10.1016/j.intimp.2024.112060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/22/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024]
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by lung tissue oedema and inflammatory cell infiltration, with limited therapeutic interventions available. Receptor-interacting protein kinase 1 (RIPK1), a critical regulator of cell death and inflammation implicated in many diseases, is not fully understood in the context of ARDS. In this study, we employed RIP1 kinase-inactivated (Rip1K45A/K45A) mice and two distinct RIPK1 inhibitors to investigate the contributions of RIP1 kinase activity in lipopolysaccharide (LPS)-induced ARDS pathology. Our results indicated that RIPK1 kinase inactivation, achieved through both genetic and chemical approaches, significantly attenuated LPS-induced ARDS pathology, as demonstrated by reduced polymorphonuclear neutrophil percentage (PMN%) in alveolar lavage fluid, expression of inflammatory and fibrosis-related factors in lung tissues, as well as histological examination. Results by tunnel staining and qRT-PCR analysis indicated that RIPK1 kinase activity played a role in regulating cell apoptosis and inflammation induced by LPS administration in lung tissue. In summary, employing both pharmacological and genetic approaches, this study demonstrated that targeted RIPK1 kinase inactivation attenuates the pathological phenotype induced by LPS inhalation in an ARDS mouse model. This study enhances our understanding of the therapeutic potential of RIPK1 kinase modulation in ARDS, providing insights for the pathogenesis of ARDS.
Collapse
Affiliation(s)
- Emmauel Mago
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China
| | - Xunan Zhao
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China
| | - Weigao Zhang
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China
| | - Qianchao Shao
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China
| | - Peiqi Li
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China
| | - Shuxian Huang
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China
| | - Xinyu Ding
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China
| | - Hu Liu
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China
| | - Tingzhe Sun
- School of Life Sciences, Anqing Normal University, Anqing 246133, Anhui, China
| | - Fei He
- Department of Emergency Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China.
| | - Dan Weng
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, 200 Xiaolingwei Street, Nanjing 210094, China.
| |
Collapse
|
10
|
Hashimoto M, Kojima Y, Sakamoto T, Ozato Y, Nakano Y, Abe T, Hosoda K, Saito H, Higuchi S, Hisamatsu Y, Toshima T, Yonemura Y, Masuda T, Hata T, Nagayama S, Kagawa K, Goto Y, Utou M, Gamachi A, Imamura K, Kuze Y, Zenkoh J, Suzuki A, Takahashi K, Niida A, Hirose H, Hayashi S, Koseki J, Fukuchi S, Murakami K, Yoshizumi T, Kadomatsu K, Tobo T, Oda Y, Uemura M, Eguchi H, Doki Y, Mori M, Oshima M, Shibata T, Suzuki Y, Shimamura T, Mimori K. Spatial and single-cell colocalisation analysis reveals MDK-mediated immunosuppressive environment with regulatory T cells in colorectal carcinogenesis. EBioMedicine 2024; 103:105102. [PMID: 38614865 PMCID: PMC11121171 DOI: 10.1016/j.ebiom.2024.105102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/15/2024] Open
Abstract
BACKGROUND Cell-cell interaction factors that facilitate the progression of adenoma to sporadic colorectal cancer (CRC) remain unclear, thereby hindering patient survival. METHODS We performed spatial transcriptomics on five early CRC cases, which included adenoma and carcinoma, and one advanced CRC. To elucidate cell-cell interactions within the tumour microenvironment (TME), we investigated the colocalisation network at single-cell resolution using a deep generative model for colocalisation analysis, combined with a single-cell transcriptome, and assessed the clinical significance in CRC patients. FINDINGS CRC cells colocalised with regulatory T cells (Tregs) at the adenoma-carcinoma interface. At early-stage carcinogenesis, cell-cell interaction inference between colocalised adenoma and cancer epithelial cells and Tregs based on the spatial distribution of single cells highlighted midkine (MDK) as a prominent signalling molecule sent from tumour epithelial cells to Tregs. Interaction between MDK-high CRC cells and SPP1+ macrophages and stromal cells proved to be the mechanism underlying immunosuppression in the TME. Additionally, we identified syndecan4 (SDC4) as a receptor for MDK associated with Treg colocalisation. Finally, clinical analysis using CRC datasets indicated that increased MDK/SDC4 levels correlated with poor overall survival in CRC patients. INTERPRETATION MDK is involved in the immune tolerance shown by Tregs to tumour growth. MDK-mediated formation of the TME could be a potential target for early diagnosis and treatment of CRC. FUNDING Japan Society for the Promotion of Science (JSPS) Grant-in-Aid for Science Research; OITA Cancer Research Foundation; AMED under Grant Number; Japan Science and Technology Agency (JST); Takeda Science Foundation; The Princess Takamatsu Cancer Research Fund.
Collapse
Affiliation(s)
- Masahiro Hashimoto
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, 874-0838, Japan; Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
| | - Yasuhiro Kojima
- Division of Computational Bioscience, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Takeharu Sakamoto
- Department of Cancer Biology, Institute of Biomedical Science, Kansai Medical University, Hirakata, 573-1010, Japan.
| | - Yuki Ozato
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, 874-0838, Japan; Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
| | - Yusuke Nakano
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, 874-0838, Japan; Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
| | - Tadashi Abe
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, 874-0838, Japan
| | - Kiyotaka Hosoda
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, 874-0838, Japan
| | - Hideyuki Saito
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, 874-0838, Japan; Department of General Surgical Science, Gastroenterological Surgery, Gunma University Graduate School of Medicine, Maebashi, 371-8511, Japan
| | - Satoshi Higuchi
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, 874-0838, Japan; Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
| | - Yuichi Hisamatsu
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, 874-0838, Japan
| | - Takeo Toshima
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, 874-0838, Japan
| | - Yusuke Yonemura
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, 874-0838, Japan
| | - Takaaki Masuda
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, 874-0838, Japan
| | - Tsuyoshi Hata
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
| | - Satoshi Nagayama
- Department of Surgery, Uji-Tokushukai Medical Center, Uji, 611-0041, Japan
| | - Koichi Kagawa
- Department of Gastroenterology, Shin Beppu Hospital, Beppu, 874-8538, Japan
| | - Yasuhiro Goto
- Department of Gastroenterology, Shin Beppu Hospital, Beppu, 874-8538, Japan
| | - Mitsuaki Utou
- Department of Pathology, Kyushu University Beppu Hospital, Beppu, 874-0838, Japan
| | - Ayako Gamachi
- Department of Pathology, Oita Oka Hospital, Oita, 870-0192, Japan
| | - Kiyomi Imamura
- Laboratory of Systems Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, 277-8561, Japan
| | - Yuta Kuze
- Laboratory of Systems Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, 277-8561, Japan
| | - Junko Zenkoh
- Laboratory of Systems Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, 277-8561, Japan
| | - Ayako Suzuki
- Laboratory of Systems Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, 277-8561, Japan
| | - Kazuki Takahashi
- Laboratory of Molecular Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Atsushi Niida
- Laboratory of Molecular Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Haruka Hirose
- Division of Systems Biology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Shuto Hayashi
- Division of Systems Biology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Jun Koseki
- Division of Systems Biology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Satoshi Fukuchi
- Department of Gastroenterological Medicine, Almeida Memorial Hospital, Oita, 870-1195, Japan
| | - Kazunari Murakami
- Department of Gastroenterology, Oita University Hospital, Yufu, 879-5593, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Taro Tobo
- Department of Pathology, Kyushu University Beppu Hospital, Beppu, 874-0838, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Kyushu University Hospital, Fukuoka, 812-8582, Japan
| | - Mamoru Uemura
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
| | - Masaki Mori
- Tokai University School of Medicine, Isehara, 259-1193, Japan
| | - Masanobu Oshima
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Tatsuhiro Shibata
- Laboratory of Molecular Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Yutaka Suzuki
- Laboratory of Systems Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, 277-8561, Japan
| | - Teppei Shimamura
- Division of Systems Biology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan; Department of Computational and Systems Biology, Medical Research Insitute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-0034, Japan.
| | - Koshi Mimori
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, 874-0838, Japan.
| |
Collapse
|
11
|
Monaco CF, Davis JS. Mechanisms of angioregression of the corpus luteum. Front Physiol 2023; 14:1254943. [PMID: 37841308 PMCID: PMC10568036 DOI: 10.3389/fphys.2023.1254943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
The corpus luteum is a transient ovarian endocrine gland that produces the progesterone necessary for the establishment and maintenance of pregnancy. The formation and function of this gland involves angiogenesis, establishing the tissue with a robust blood flow and vast microvasculature required to support production of progesterone. Every steroidogenic cell within the corpus luteum is in direct contact with a capillary, and disruption of angiogenesis impairs luteal development and function. At the end of a reproductive cycle, the corpus luteum ceases progesterone production and undergoes rapid structural regression into a nonfunctional corpus albicans in a process initiated and exacerbated by the luteolysin prostaglandin F2α (PGF2α). Structural regression is accompanied by complete regression of the luteal microvasculature in which endothelial cells die and are sloughed off into capillaries and lymphatic vessels. During luteal regression, changes in nitric oxide transiently increase blood flow, followed by a reduction in blood flow and progesterone secretion. Early luteal regression is marked by an increased production of cytokines and chemokines and influx of immune cells. Microvascular endothelial cells are sensitive to released factors during luteolysis, including thrombospondin, endothelin, and cytokines like tumor necrosis factor alpha (TNF) and transforming growth factor β 1 (TGFB1). Although PGF2α is known to be a vasoconstrictor, endothelial cells do not express receptors for PGF2α, therefore it is believed that the angioregression occurring during luteolysis is mediated by factors downstream of PGF2α signaling. Yet, the exact mechanisms responsible for angioregression in the corpus luteum remain unknown. This review describes the current knowledge on angioregression of the corpus luteum and the roles of vasoactive factors released during luteolysis on luteal vasculature and endothelial cells of the microvasculature.
Collapse
Affiliation(s)
- Corrine F. Monaco
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, United States
| | - John S. Davis
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, United States
- US Department of Veterans Affairs Nebraska-Western Iowa Healthcare System, Omaha, NE, United States
| |
Collapse
|
12
|
Pulito-Cueto V, Remuzgo-Martínez S, Genre F, Atienza-Mateo B, Mora-Cuesta VM, Iturbe-Fernández D, Lera-Gómez L, Mora-Gil MS, Portilla V, Corrales A, Blanco R, Cifrián JM, González-Gay MA, López-Mejías R. E-Selectin, ICAM-1, and ET-1 Biomarkers Address the Concern of the Challenging Diagnosis of Interstitial Lung Disease in Patients with Autoimmune Diseases. Int J Mol Sci 2023; 24:12518. [PMID: 37569893 PMCID: PMC10420063 DOI: 10.3390/ijms241512518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/01/2023] [Accepted: 08/05/2023] [Indexed: 08/13/2023] Open
Abstract
Interstitial lung disease (ILD) constitutes the most critical comorbidity in autoimmune diseases (ADs) and its early diagnosis remains a challenge for clinicians. Accordingly, we evaluated whether E-selectin, ICAM-1, and ET-1, key molecules in endothelial damage, could be useful biomarkers for the detection of AD-ILD+. We recruited patients with rheumatoid arthritis (RA)-ILD+ (n = 21) and systemic sclerosis (SSc)-ILD+ (n = 21). We included comparison groups of patients: RA-ILD- (n = 25), SSc-ILD- (n = 20), and idiopathic pulmonary fibrosis (IPF) (n = 21). Serum levels of these proteins were determined by ELISA. E-selectin, ICAM-1, and ET-1 serum levels were increased in RA-ILD+ and IPF patients in comparison to RA-ILD- patients. Additionally, SSc-ILD+ and IPF patients exhibited higher ICAM-1 levels than those with SSc-ILD-. The ability of E-selectin, ICAM-1, and ET-1 to discriminate RA-ILD+ from RA-ILD- patients, and ICAM-1 to distinguish SSc-ILD+ from SSc-ILD- patients was confirmed using ROC curve analysis. Furthermore, elevated levels of ET-1 and E-selectin correlated with lung function decline in RA-ILD+ and SSc-ILD+ patients, respectively. In conclusion, our findings support the relevant role of E-selectin, ICAM-1, and ET-1 in RA-ILD+ patients as well as of ICAM-1 in SSc-ILD+ patients, constituting potential screening blood biomarkers of ILD in AD. Moreover, this study suggests ET-1 and E-selectin as possible indicators of worsening lung function in RA-ILD+ and SSc-ILD+ patients, respectively.
Collapse
Affiliation(s)
- Verónica Pulito-Cueto
- Immunopathology Group, Marqués de Valdecilla University Hospital-Marqués de Valdecilla Research Institute (IDIVAL), 39011 Santander, Spain; (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (M.S.M.-G.); (V.P.); (A.C.); (R.B.); (J.M.C.); (R.L.-M.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - Sara Remuzgo-Martínez
- Marqués de Valdecilla Research Institute (IDIVAL), 39011 Santander, Spain; (S.R.-M.); (F.G.); (M.A.G.-G.)
| | - Fernanda Genre
- Marqués de Valdecilla Research Institute (IDIVAL), 39011 Santander, Spain; (S.R.-M.); (F.G.); (M.A.G.-G.)
| | - Belén Atienza-Mateo
- Immunopathology Group, Marqués de Valdecilla University Hospital-Marqués de Valdecilla Research Institute (IDIVAL), 39011 Santander, Spain; (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (M.S.M.-G.); (V.P.); (A.C.); (R.B.); (J.M.C.); (R.L.-M.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - Víctor M. Mora-Cuesta
- Immunopathology Group, Marqués de Valdecilla University Hospital-Marqués de Valdecilla Research Institute (IDIVAL), 39011 Santander, Spain; (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (M.S.M.-G.); (V.P.); (A.C.); (R.B.); (J.M.C.); (R.L.-M.)
- Department of Pneumology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - David Iturbe-Fernández
- Immunopathology Group, Marqués de Valdecilla University Hospital-Marqués de Valdecilla Research Institute (IDIVAL), 39011 Santander, Spain; (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (M.S.M.-G.); (V.P.); (A.C.); (R.B.); (J.M.C.); (R.L.-M.)
- Department of Pneumology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - Leticia Lera-Gómez
- Department of Microbiology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain;
| | - María Sebastián Mora-Gil
- Immunopathology Group, Marqués de Valdecilla University Hospital-Marqués de Valdecilla Research Institute (IDIVAL), 39011 Santander, Spain; (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (M.S.M.-G.); (V.P.); (A.C.); (R.B.); (J.M.C.); (R.L.-M.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - Virginia Portilla
- Immunopathology Group, Marqués de Valdecilla University Hospital-Marqués de Valdecilla Research Institute (IDIVAL), 39011 Santander, Spain; (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (M.S.M.-G.); (V.P.); (A.C.); (R.B.); (J.M.C.); (R.L.-M.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - Alfonso Corrales
- Immunopathology Group, Marqués de Valdecilla University Hospital-Marqués de Valdecilla Research Institute (IDIVAL), 39011 Santander, Spain; (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (M.S.M.-G.); (V.P.); (A.C.); (R.B.); (J.M.C.); (R.L.-M.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - Ricardo Blanco
- Immunopathology Group, Marqués de Valdecilla University Hospital-Marqués de Valdecilla Research Institute (IDIVAL), 39011 Santander, Spain; (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (M.S.M.-G.); (V.P.); (A.C.); (R.B.); (J.M.C.); (R.L.-M.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - José M. Cifrián
- Immunopathology Group, Marqués de Valdecilla University Hospital-Marqués de Valdecilla Research Institute (IDIVAL), 39011 Santander, Spain; (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (M.S.M.-G.); (V.P.); (A.C.); (R.B.); (J.M.C.); (R.L.-M.)
- Department of Pneumology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
- School of Medicine, Universidad de Cantabria, 39011 Santander, Spain
| | - Miguel A. González-Gay
- Marqués de Valdecilla Research Institute (IDIVAL), 39011 Santander, Spain; (S.R.-M.); (F.G.); (M.A.G.-G.)
- School of Medicine, Universidad de Cantabria, 39011 Santander, Spain
- Department of Rheumatology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
| | - Raquel López-Mejías
- Immunopathology Group, Marqués de Valdecilla University Hospital-Marqués de Valdecilla Research Institute (IDIVAL), 39011 Santander, Spain; (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (M.S.M.-G.); (V.P.); (A.C.); (R.B.); (J.M.C.); (R.L.-M.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| |
Collapse
|
13
|
Fließer E, Lins T, Berg JL, Kolb M, Kwapiszewska G. The endothelium in lung fibrosis: a core signaling hub in disease pathogenesis? Am J Physiol Cell Physiol 2023; 325:C2-C16. [PMID: 37184232 DOI: 10.1152/ajpcell.00097.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/05/2023] [Accepted: 05/05/2023] [Indexed: 05/16/2023]
Abstract
Pulmonary fibrosis (PF) is a progressive chronic lung disease characterized by excessive deposition of extracellular matrix (ECM) and structural destruction, associated with a severe 5-year mortality rate. The onset of the disease is thought to be triggered by chronic damage to the alveolar epithelium. Since the pulmonary endothelium is an important component of the alveolar-capillary niche, it is also affected by the initial injury. In addition to ensuring proper gas exchange, the endothelium has critical functional properties, including regulation of vascular tone, inflammatory responses, coagulation, and maintenance of vascular homeostasis and integrity. Recent single-cell analyses have shown that shifts in endothelial cell (EC) subtypes occur in PF. Furthermore, the increased vascular remodeling associated with PF leads to deteriorated outcomes for patients, underscoring the importance of the vascular bed in PF. To date, the causes and consequences of endothelial and vascular involvement in lung fibrosis are poorly understood. Therefore, it is of great importance to investigate the involvement of EC and the vascular system in the pathogenesis of the disease. In this review, we will outline the current knowledge on the role of the pulmonary vasculature in PF, in terms of abnormal cellular interactions, hyperinflammation, vascular barrier disorders, and an altered basement membrane composition. Finally, we will summarize recent advances in extensive therapeutic research and discuss the significant value of novel therapies targeting the endothelium.
Collapse
Affiliation(s)
- Elisabeth Fließer
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Thomas Lins
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Johannes Lorenz Berg
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Martin Kolb
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
- Institute for Lung Health, Member of the German Lung Center (DZL), Cardiopulmonary Institute (CPI), Giessen, Germany
| |
Collapse
|
14
|
Hua HS, Wen HC, Lee HS, Weng CM, Yuliani FS, Kuo HP, Chen BC, Lin CH. Endothelin-1 induces connective tissue growth factor expression in human lung fibroblasts by disrupting HDAC2/Sin3A/MeCP2 corepressor complex. J Biomed Sci 2023; 30:40. [PMID: 37312162 DOI: 10.1186/s12929-023-00931-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 05/20/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Reduction of histone deacetylase (HDAC) 2 expression and activity may contribute to amplified inflammation in patients with severe asthma. Connective tissue growth factor (CTGF) is a key mediator of airway fibrosis in severe asthma. However, the role of the HDAC2/Sin3A/methyl-CpG-binding protein (MeCP) 2 corepressor complex in the regulation of CTGF expression in lung fibroblasts remains unclear. METHODS The role of the HDAC2/Sin3A/MeCP2 corepressor complex in endothelin (ET)-1-stimulated CTGF production in human lung fibroblasts (WI-38) was investigated. We also evaluated the expression of HDAC2, Sin3A and MeCP2 in the lung of ovalbumin-induced airway fibrosis model. RESULTS HDAC2 suppressed ET-1-induced CTGF expression in WI-38 cells. ET-1 treatment reduced HDAC2 activity and increased H3 acetylation in a time-dependent manner. Furthermore, overexpression of HDAC2 inhibited ET-1-induced H3 acetylation. Inhibition of c-Jun N-terminal kinase, extracellular signal-regulated kinase, or p38 attenuated ET-1-induced H3 acetylation by suppressing HDAC2 phosphorylation and reducing HDAC2 activity. Overexpression of both Sin3A and MeCP2 attenuated ET-1-induced CTGF expression and H3 acetylation. ET-1 induced the disruption of the HDAC2/Sin3A/MeCP2 corepressor complex and then prompted the dissociation of HDAC2, Sin3A, and MeCP2 from the CTGF promoter region. Overexpression of HDAC2, Sin3A, or MeCP2 attenuated ET-1-stimulated AP-1-luciferase activity. Moreover, Sin3A- or MeCP2-suppressed ET-1-induced H3 acetylation and AP-1-luciferase activity were reversed by transfection of HDAC2 siRNA. In an ovalbumin-induced airway fibrosis model, the protein levels of HDAC2 and Sin3A were lower than in the control group; however, no significant difference in MeCP2 expression was observed. The ratio of phospho-HDAC2/HDAC2 and H3 acetylation in the lung tissue were higher in this model than in the control group. Overall, without stimulation, the HDAC2/Sin3A/MeCP2 corepressor complex inhibits CTGF expression by regulating H3 deacetylation in the CTGF promoter region in human lung fibroblasts. With ET-1 stimulation, the HDAC2/Sin3A/MeCP2 corepressor complex is disrupted and dissociated from the CTGF promoter region; this is followed by AP-1 activation and the eventual initiation of CTGF production. CONCLUSIONS The HDAC2/Sin3A/MeCP2 corepressor complex is an endogenous inhibitor of CTGF in lung fibroblasts. Additionally, HDAC2 and Sin3A may be of greater importance than MeCP2 in the pathogenesis of airway fibrosis.
Collapse
Affiliation(s)
- Hung-Sheng Hua
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 110, Taiwan
| | - Heng-Ching Wen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 110, Taiwan
| | - Hong-Sheng Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 110, Taiwan
- Research Center of Thoracic Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Ming Weng
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 110, Taiwan
- Research Center of Thoracic Medicine, Taipei Medical University, Taipei, Taiwan
| | - Fara Silvia Yuliani
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Han-Pin Kuo
- Division of Thoracic Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Research Center of Thoracic Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Thoracic Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Bing-Chang Chen
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 110, Taiwan.
- Research Center of Thoracic Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Chien-Huang Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 110, Taiwan.
- Research Center of Thoracic Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
15
|
Docosahexaenoic Acid Attenuates Radiation-Induced Myocardial Fibrosis by Inhibiting the p38/ET-1 Pathway in Cardiomyocytes. Int J Radiat Oncol Biol Phys 2023; 115:1229-1243. [PMID: 36529557 DOI: 10.1016/j.ijrobp.2022.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/24/2022] [Accepted: 11/01/2022] [Indexed: 12/23/2022]
Abstract
PURPOSE Radiation-induced myocardial fibrosis (RIMF) is a severe delayed complication of thoracic irradiation (IR). Endothelin-1 (ET-1) is critical in cardiac fibroblast activation, and docosahexaenoic acid (DHA) is protective against various cardiac diseases. This study aimed to explore the roles of ET-1 in RIMF and the potential of DHA in preventing RIMF. METHODS AND MATERIALS Hematoxylin and eosin, sirius red, and Masson trichrome staining were carried out to evaluate the histopathologic conditions in mouse models. Enzyme-linked immunosorbent assays were used to detect the concentration of ET-1 in serum and cell supernatants. Western blotting, immunofluorescence, and immunohistochemistry were used to assess the protein levels. The phenotypic alterations of cardiac fibroblasts were evaluated by cell proliferation/migration assays and α-smooth muscle actin (α-SMA) detection. RESULTS Radiation increased ET-1 expression and secretion by increasing p38 phosphorylation in cardiomyocytes, and ET-1 markedly promoted the activation of cardiac fibroblasts, which were characterized by enhanced fibroblast proliferation, migration, and α-SMA expression. Cardiomyocyte-derived ET-1 mediated radiation-induced fibroblast activation by targeting the PI3K-AKT and MEK-ERK pathways in fibroblasts. DHA suppressed ET-1 levels by blocking p38 signaling in cardiomyocytes and significantly attenuated the activation of cardiac fibroblasts induced by the IR/ET-1 axis. Importantly, DHA decreased collagen deposition and α-SMA expression, alleviating cardiac fibrosis caused by radiation in mouse models. CONCLUSIONS Our findings demonstrate that radiation facilitates cardiac fibroblast activation by enhancing p38/ET-1 signaling in cardiomyocytes, revealing the IR/p38/ET-1 regulatory axis in RIMF for the first time. DHA effectively inhibits fibroblast activation by targeting p38/ET-1 and can be recognized as a promising protective agent against RIMF.
Collapse
|
16
|
Morales E, Sandino J, Galindo M. Lupus nephropathy beyond immunosuppression: Searching for nephro and cardioprotection. FRONTIERS IN NEPHROLOGY 2023; 3:1105676. [PMID: 37675340 PMCID: PMC10479677 DOI: 10.3389/fneph.2023.1105676] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/10/2023] [Indexed: 09/08/2023]
Abstract
Renal involvement in systemic lupus erythematosus (SLE) represents one of the most frequent organ manifestations, often leading to end-stage kidney disease (ESKD). Several therapies have been tested in patients with lupus nephritis (LN) to prevent further organ damage. The effectiveness of immunosuppressive therapy as a treatment for LN is abundant, supported by multiple clinical trials that have shown its efficacy in preventing the development of chronic kidney disease (CKD). In addition to immunosuppressive therapy, several traditional and recent therapies aimed at nephroprotection in patients with proteinuric chronic kidney disease are gaining importance in the setting of LN. Thus, immunosuppressive therapy should be accompanied by nephro- and cardioprotective measures to control cardiovascular risk factors and proteinuria to ensure a better renal prognosis. Despite this, the literature on these specific measures is relatively scarce, with recommendations focused on the blockade of the renin-angiotensin-aldosterone system (RAAS). This review explores the pharmacological options available for cardiovascular and renal protection outside the usual treatment schemes.
Collapse
Affiliation(s)
- Enrique Morales
- Department of Nephrology, University Hospital “12 de Octubre”, Madrid, Spain
- Research Institute of University Hospital “12 de Octubre” (imas12), Madrid, Spain
- Department of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Justo Sandino
- Department of Nephrology, University Hospital “12 de Octubre”, Madrid, Spain
- Research Institute of University Hospital “12 de Octubre” (imas12), Madrid, Spain
| | - María Galindo
- Research Institute of University Hospital “12 de Octubre” (imas12), Madrid, Spain
- Department of Medicine, Complutense University of Madrid, Madrid, Spain
- Department of Rheumatology, University Hospital “12 de Octubre”, Madrid, Spain
| |
Collapse
|
17
|
Endothelin-1 as a Biomarker of Idiopathic Pulmonary Fibrosis and Interstitial Lung Disease Associated with Autoimmune Diseases. Int J Mol Sci 2023; 24:ijms24021275. [PMID: 36674789 PMCID: PMC9862125 DOI: 10.3390/ijms24021275] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/23/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
The aim of this study was to determine the role of endothelin-1 (ET-1), a molecule involved in multiple vascular and fibrosing abnormalities, as a biomarker of interstitial lung disease (ILD), as well as its use for the differential diagnosis between idiopathic pulmonary fibrosis (IPF) and ILD associated with autoimmune diseases (AD-ILD), using a large and well-defined cohort of patients with ILD. A total of 112 patients with IPF, 91 patients with AD-ILD (28 rheumatoid arthritis (RA), 26 systemic sclerosis, 20 idiopathic inflammatory myositis and 17 interstitial pneumonia with autoimmune features) and 44 healthy controls were included. ET-1 serum levels were determined by enzyme-linked immunosorbent assay. A significant increase in ET-1 levels was found in patients with IPF compared to controls. Likewise, AD-ILD patients also showed higher ET-1 levels than controls when the whole cohort was stratified by the type of AD. Similar ET-1 levels were found in IPF and AD-ILD patients, regardless of the underlying AD. Interestingly, increased ET-1 levels were correlated with worse lung function in IPF and RA-ILD patients. Our study supports that serum ET-1 may be useful as a biomarker of ILD, although it could not help in the differential diagnosis between IPF and AD-ILD. Moreover, ET-1 levels may be associated with ILD severity.
Collapse
|
18
|
Septembre-Malaterre A, Boina C, Douanier A, Gasque P. Deciphering the Antifibrotic Property of Metformin. Cells 2022; 11:cells11244090. [PMID: 36552855 PMCID: PMC9777391 DOI: 10.3390/cells11244090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Fibrosis is a chronic progressive and incurable disease leading to organ dysfunction. It is characterized by the accumulation of extracellular matrix proteins produced by mesenchymal stem cells (MSCs) differentiating into myofibroblasts. Given the complexity of its pathophysiology, the search for effective treatments for fibrosis is of paramount importance. Metformin, a structural dimethyl analog of the galegine guanide extracted from the "French Lilac" (Fabaceae Galega officinalis), is the most widely used antidiabetic drug, recently recognized for its antifibrotic effects through ill-characterized mechanisms. The in vitro model of TGF-β1-induced fibrosis in human primary pulmonary mesenchymal stem cells (HPMSCs), identified as CD248+ and CD90+ cells, was used to study the effects of metformin extracts. These effects were tested on the expression of canonical MSC differentiation markers, immune/inflammatory factors and antioxidative stress molecules using qRT-PCR (mRNA, miRNA), immunofluorescence and ELISA experiments. Interestingly, metformin is able to reduce/modulate the expression of different actors involved in fibrosis. Indeed, TGF-β1 effects were markedly attenuated by metformin, as evidenced by reduced expression of three collagen types and Acta2 mRNAs. Furthermore, metformin attenuated the effects of TGF-β1 on the expression of PDGF, VEGF, erythropoietin, calcitonin and profibrotic miRs, possibly by controlling the expression of several key TGF/Smad factors. The expression of four major fibrogenic MMPs was also reduced by metformin treatment. In addition, metformin controlled MSC differentiation into lipofibroblasts and osteoblasts and had the ability to restore redox balance via the Nox4/Nrf2, AMP and Pi3K pathways. Overall, these results show that metformin is a candidate molecule for antifibrotic effect and/or aiming to combat the development of chronic inflammatory diseases worldwide.
Collapse
Affiliation(s)
- Axelle Septembre-Malaterre
- Unité de Recherche, EPI ‘Etudes en Pharmaco-Immunologie’, Université de la Réunion, Allée des Topazes, CS11021, 97400 Saint Denis, France
- Laboratoire D’immunologie Clinique et Expérimentale de la Zone de L’océan Indien (LICE-OI), CHU La Réunion Site Félix Guyon Allée des Topazes, CS11021, 97400 Saint Denis, France
- Correspondence:
| | - Chailas Boina
- Unité de Recherche, EPI ‘Etudes en Pharmaco-Immunologie’, Université de la Réunion, Allée des Topazes, CS11021, 97400 Saint Denis, France
- Laboratoire D’immunologie Clinique et Expérimentale de la Zone de L’océan Indien (LICE-OI), CHU La Réunion Site Félix Guyon Allée des Topazes, CS11021, 97400 Saint Denis, France
| | - Audrey Douanier
- Unité de Recherche, EPI ‘Etudes en Pharmaco-Immunologie’, Université de la Réunion, Allée des Topazes, CS11021, 97400 Saint Denis, France
- Laboratoire D’immunologie Clinique et Expérimentale de la Zone de L’océan Indien (LICE-OI), CHU La Réunion Site Félix Guyon Allée des Topazes, CS11021, 97400 Saint Denis, France
| | - Philippe Gasque
- Unité de Recherche, EPI ‘Etudes en Pharmaco-Immunologie’, Université de la Réunion, Allée des Topazes, CS11021, 97400 Saint Denis, France
- Laboratoire D’immunologie Clinique et Expérimentale de la Zone de L’océan Indien (LICE-OI), CHU La Réunion Site Félix Guyon Allée des Topazes, CS11021, 97400 Saint Denis, France
| |
Collapse
|
19
|
Shakil F, Snijder J, Salvatore MM. Why is UIP peripheral? Expert Rev Respir Med 2022; 16:907-915. [PMID: 36066423 DOI: 10.1080/17476348.2022.2119131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The radiology pattern associated with IPF is called UIP. It is unique because unlike any other form of fibrosis it is peripheral in its distribution. We investigated the peripheral nature of UIP and why it was a key feature of IPF the deadliest of the ILDS. AREAS COVERED It is not enough to say that UIP is peripheral but instead as scientists we must ask ourselves why it is peripheral. This review dives into the published hypothesis that includes vascular insult, tensile forces, microaspiration, and inflammation and looks at the pros and cons for each argument, and ultimately comes to its own conclusion. PubMed searches using the below keywords were used to identify papers that described pathogenesis of IPF with regard to a particular theory. EXPERT OPINION In this paper, we will review four ideas that support why UIP is peripheral and propose the most likely explanation given what is currently known about the pathophysiology of IPF.
Collapse
Affiliation(s)
- Faariah Shakil
- Department of Radiology, Columbia University Irving Medical Center, New York, USA
| | - Juan Snijder
- Department of Radiology, Columbia University Irving Medical Center, New York, USA
| | - Mary M Salvatore
- Department of Radiology, Columbia University Irving Medical Center, New York, USA
| |
Collapse
|
20
|
Molecular pathways and role of epigenetics in the idiopathic pulmonary fibrosis. Life Sci 2022; 291:120283. [PMID: 34998839 DOI: 10.1016/j.lfs.2021.120283] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/19/2021] [Accepted: 12/27/2021] [Indexed: 12/12/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease with unknown etiological factors that can progress to other dangerous diseases like lung cancer. Environmental and genetic predisposition are the two major etiological or risk factors involved in the pathology of the IPF. Among the environmental risk factors, smoking is one of the major causes for the development of IPF. Epigenetic pathways like nucleosomes remodeling, DNA methylation, histone modifications and miRNA mediated genes play a crucial role in development of IPF. Mutations in the genes make the epigenetic factors as important drug targets in IPF. Transcriptional changes due to environmental factors are also involved in the progression of IPF. The mutations in human telomerase reverse transcriptase (hTERT) have shown decreased life expectancy in IPF patients. The TERT-gene is highly expressed in chronic smokers and makes the role of epigenetics evident. Drug like nintedanib acts through vascular endothelial growth factor receptors (VEGFR), while drug pirfenidone acts through transforming growth factor (TGF), which is useful in IPF. Gefitinib, a tyrosine kinase inhibitor of EGFR, is useful as an anti-fibrosis agent in preclinical models. Newer drugs such as Celgene-CC90001 and FibroGen-FG-3019 are currently under investigations acts through the modulating epigenetic mechanisms. Thus, the study on epigenetics opens a wide window for the discovery of newer drugs. This study provides an elementary analysis of multiple regulators of epigenetics and their roles associated with the pathology of IPF. Further, this review also includes epigenetic drugs under development in preclinical and clinical stages.
Collapse
|
21
|
Shi X, Chen Y, Liu Q, Mei X, Liu J, Tang Y, Luo R, Sun D, Ma Y, Wu W, Tu W, Zhao Y, Xu W, Ke Y, Jiang S, Huang Y, Zhang R, Wang L, Chen Y, Xia J, Pu W, Zhu H, Zuo X, Li Y, Xu J, Gao F, Wei D, Chen J, Yin W, Wang Q, Dai H, Yang L, Guo G, Cui J, Song N, Zou H, Zhao S, Distler JH, Jin L, Wang J. LDLR dysfunction induces LDL accumulation and promotes pulmonary fibrosis. Clin Transl Med 2022; 12:e711. [PMID: 35083881 PMCID: PMC8792399 DOI: 10.1002/ctm2.711] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/27/2021] [Accepted: 01/05/2022] [Indexed: 12/15/2022] Open
Abstract
Treatments for pulmonary fibrosis (PF) are ineffective because its molecular pathogenesis and therapeutic targets are unclear. Here, we show that the expression of low-density lipoprotein receptor (LDLR) was significantly decreased in alveolar type II (ATII) and fibroblast cells, whereas it was increased in endothelial cells from systemic sclerosis-related PF (SSc-PF) patients and idiopathic PF (IPF) patients compared with healthy controls. However, the plasma levels of low-density lipoprotein (LDL) increased in SSc-PF and IPF patients. The disrupted LDL-LDLR metabolism was also observed in four mouse PF models. Upon bleomycin (BLM) treatment, Ldlr-deficient (Ldlr-/-) mice exhibited remarkably higher LDL levels, abundant apoptosis, increased fibroblast-like endothelial and ATII cells and significantly earlier and more severe fibrotic response compared to wild-type mice. In vitro experiments revealed that apoptosis and TGF-β1 production were induced by LDL, while fibroblast-like cell accumulation and ET-1 expression were induced by LDLR knockdown. Treatment of fibroblasts with LDL or culture medium derived from LDL-pretreated endothelial or epithelial cells led to obvious fibrotic responses in vitro. Similar results were observed after LDLR knockdown operation. These results suggest that disturbed LDL-LDLR metabolism contributes in various ways to the malfunction of endothelial and epithelial cells, and fibroblasts during pulmonary fibrogenesis. In addition, pharmacological restoration of LDLR levels by using a combination of atorvastatin and alirocumab inhibited BLM-induced LDL elevation, apoptosis, fibroblast-like cell accumulation and mitigated PF in mice. Therefore, LDL-LDLR may serve as an important mediator in PF, and LDLR enhancing strategies may have beneficial effects on PF.
Collapse
Affiliation(s)
- Xiangguang Shi
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
| | - Yahui Chen
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
| | - Qingmei Liu
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
| | - Xueqian Mei
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
| | - Jing Liu
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
- Division of RheumatologyHuashan hospital, Fudan UniversityShanghaiP. R. China
| | - Yulong Tang
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
| | - Ruoyu Luo
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
| | - Dayan Sun
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
| | - Yanyun Ma
- MOE Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life SciencesFudan UniversityShanghaiP. R. China
- Institute for Six‐sector EconomyFudan UniversityShanghaiP. R. China
| | - Wenyu Wu
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
| | - Wenzhen Tu
- Division of RheumatologyShanghai TCM‐Integrated HospitalShanghaiP. R. China
| | - Yinhuan Zhao
- Division of RheumatologyShanghai TCM‐Integrated HospitalShanghaiP. R. China
| | - Weihong Xu
- The Clinical Laboratory of Tongren HosipitalShanghai Jiaotong UniversityShanghaiP. R. China
| | - Yuehai Ke
- Department of Pathology and PathophysiologyZhejiang University School of MedicineHangzhouZhejiang ProvinceP. R. China
| | - Shuai Jiang
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
| | - Yan Huang
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
| | - Rui Zhang
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
- Institute for Six‐sector EconomyFudan UniversityShanghaiP. R. China
| | - Lei Wang
- Division of RheumatologyShanghai TCM‐Integrated HospitalShanghaiP. R. China
| | - Yuanyuan Chen
- Division of RheumatologyShanghai TCM‐Integrated HospitalShanghaiP. R. China
| | - Jingjing Xia
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
| | - Weilin Pu
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
| | - Honglin Zhu
- Department of Internal Medicine 3 and Institute for Clinical ImmunologyUniversity of ErlangenNurembergGermany
- Department of Rheumatology, Xiangya HospitalCentral South UniversityChangshaHunan ProvinceP. R. China
| | - Xiaoxia Zuo
- Department of Rheumatology, Xiangya HospitalCentral South UniversityChangshaHunan ProvinceP. R. China
| | - Yisha Li
- Department of Rheumatology, Xiangya HospitalCentral South UniversityChangshaHunan ProvinceP. R. China
| | - Jinhua Xu
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
| | - Fei Gao
- Wuxi Lung Transplant CenterWuxi People's Hospital affiliated to Nanjing Medical UniversityWuxiP. R. China
| | - Dong Wei
- Wuxi Lung Transplant CenterWuxi People's Hospital affiliated to Nanjing Medical UniversityWuxiP. R. China
| | - Jingyu Chen
- Wuxi Lung Transplant CenterWuxi People's Hospital affiliated to Nanjing Medical UniversityWuxiP. R. China
| | - Wenguang Yin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongP. R. China
| | - Qingwen Wang
- Rheumatology and Immunology DepartmentPeking University Shenzhen HospitalShenzhenP. R. China
| | - Huaping Dai
- Department of Pulmonary and Critical Care Medicine, China‐Japan Friendship Hospital; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory MedicineChinese Academy of Medical ScienceBeijingP. R. China
| | - Libing Yang
- Department of Pulmonary and Critical Care Medicine, China‐Japan Friendship Hospital; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory MedicineChinese Academy of Medical ScienceBeijingP. R. China
- School of MedicineTsinghua UniversityBeijingP. R. China
| | - Gang Guo
- Department of Rheumatology and ImmunologyYiling Hospital Affiliated to Hebei Medical UniversityShijiazhuangHebei ProvinceP. R. China
| | - Jimin Cui
- Department of Rheumatology and ImmunologyYiling Hospital Affiliated to Hebei Medical UniversityShijiazhuangHebei ProvinceP. R. China
| | - Nana Song
- Department of Nephrology, Zhongshan Hospital, Fudan UniversityFudan Zhangjiang InstituteShanghaiP. R. China
| | - Hejian Zou
- Division of RheumatologyHuashan hospital, Fudan UniversityShanghaiP. R. China
- Institute of Rheumatology, Immunology and AllergyFudan UniversityShanghaiP. R. China
| | - Shimin Zhao
- Institute of Metabolism and Integrative BiologyFudan UniversityShanghaiP. R. China
| | - Jörg H.W. Distler
- Department of Internal Medicine 3 and Institute for Clinical ImmunologyUniversity of ErlangenNurembergGermany
| | - Li Jin
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
- Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058)Chinese Academy of Medical SciencesShanghaiP. R. China
| | - Jiucun Wang
- Department of Dermatology, Huashan Hospital and State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiP. R. China
- Human Phenome Institute and Collaborative Innovation Center for Genetics and DevelopmentFudan UniversityShanghaiP. R. China
- Institute of Rheumatology, Immunology and AllergyFudan UniversityShanghaiP. R. China
- Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058)Chinese Academy of Medical SciencesShanghaiP. R. China
| |
Collapse
|
22
|
Napolioni V, Bianconi F, Potenza R, Carpi FM, Ludovini V, Picciolini M, Tofanetti FR, Bufalari A, Pallotti S, Poggi C, Anile M, Daddi N, Venuta F, Puma F, Vannucci J. Genome-wide expression of the residual lung reacting to experimental Pneumonectomy. BMC Genomics 2021; 22:881. [PMID: 34872491 PMCID: PMC8650537 DOI: 10.1186/s12864-021-08171-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 11/12/2021] [Indexed: 11/21/2022] Open
Abstract
Background Acute or chronic irreversible respiratory failure may occur in patients undergoing pneumonectomy. Aim of this study was to determine transcriptome expression changes after experimental pneumonectomy in swine model. Experimental left pneumonectomy was performed in five pigs under general anaesthesia. Both the resected and the remaining lung, after 60 post-operative completely uneventful days, underwent genome-wide bulk RNA-Sequencing (RNA-Seq). Results Histological analysis showed dilation of air spaces and rupture of interalveolar septa. In addition, mild inflammation, no fibrosis, radial stretch of the bronchus, strong enlargement of airspaces and thinning of the blood supply were observed. Bioinformatic analyses of bulk RNA-Seq data identified 553 Differentially Expressed Genes (DEGs) at adjusted P-value below 0.001, between pre- and post-pneumonectomy. The top 10 up-regulated DEGs were Edn1, Areg, Havcr2, Gadd45g, Depp1, Cldn4, Atf3, Myc, Gadd45b, Socs3; the top 10 down-regulated DEGs were Obscn, Cdkn2b, ENSSSCG00000015738, Prrt2, Amer1, Flrt3, Efnb2, Tox3, Znf793, Znf365. Leveraging digital cytometry tools, no difference in cellular abundance was found between the two experimental groups, while the analysis of cell type-specific gene expression patterns highlighted a striking predominance of macrophage-specific genes among the DEGs. DAVID-based gene ontology analysis showed a significant enrichment of “Extrinsic apoptotic signaling pathway” (FDR q = 7.60 × 10− 3) and “Response to insulin” (FDR q = 7.60 × 10− 3) genes, along with an enrichment of genes involved as “Negative regulators of DDX58/IFIH1 signaling” (FDR q = 7.50 × 10− 4) found by querying the REACTOME pathway database. Gene network analyses indicated a general dysregulation of gene inter-connections. Conclusion This translational genomics study highlighted the existence both of individual genes, mostly dysregulated in certain cellular populations (e.g., macrophages), and gene-networks involved in pulmonary reaction after left pneumonectomy. Their involvement in lung homeostasis is largely supported by previous studies, carried out both in humans and in other animal models (under homeostatic or disease-related conditions), that adopted candidate-gene approaches. Overall, the present findings represent a preliminary assessment for future, more focused, studies on compensatory lung adaptation, pulmonary regeneration and functional reload. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08171-3.
Collapse
Affiliation(s)
- Valerio Napolioni
- Genomic and Molecular Epidemiology (GAME) Lab., School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | | | - Rossella Potenza
- Department of Thoracic Surgery, University of Perugia Medical School, Perugia, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | | | - Vienna Ludovini
- Department of Medical Oncology, S. Maria Della Misericordia Hospital, Perugia, Italy
| | | | - Francesca R Tofanetti
- Department of Medical Oncology, S. Maria Della Misericordia Hospital, Perugia, Italy
| | - Antonello Bufalari
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Stefano Pallotti
- Genetics and Animal Breeding Group, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Camilla Poggi
- Department of Thoracic Surgery, University of Rome Sapienza, Policlinico Umberto I, Viale del Policlinico 155, 00161, Rome, Italy
| | - Marco Anile
- Department of Thoracic Surgery, University of Rome Sapienza, Policlinico Umberto I, Viale del Policlinico 155, 00161, Rome, Italy
| | - Niccolò Daddi
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Federico Venuta
- Department of Thoracic Surgery, University of Rome Sapienza, Policlinico Umberto I, Viale del Policlinico 155, 00161, Rome, Italy
| | - Francesco Puma
- Department of Thoracic Surgery, University of Perugia Medical School, Perugia, Italy
| | - Jacopo Vannucci
- Department of Thoracic Surgery, University of Rome Sapienza, Policlinico Umberto I, Viale del Policlinico 155, 00161, Rome, Italy.
| |
Collapse
|
23
|
SORAYA ARISTIINTAN, SUZUKI YOKO, MORIMOTO MITSURU, KO CHEMYONGJAY, IKEDA KOJI, HIRATA KENICHI, EMOTO NORIAKI. Protective Effects of Endothelin-2 Expressed in Epithelial Cells on Bleomycin-Induced Pulmonary Fibrosis in Mice. THE KOBE JOURNAL OF MEDICAL SCIENCES 2021; 67:E61-E70. [PMID: 34795157 PMCID: PMC8622215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/05/2021] [Indexed: 06/13/2023]
Abstract
Initially, endothelin (ET)-2 was described as an endothelium-derived vasoconstrictor. However, accumulating evidence suggests the involvement of ET-2 in non-cardiovascular physiology and disease pathophysiology. The deficiency of ET-2 in mice can be lethal, and such mice exhibit a distinct developmental abnormality in the lungs. Nonetheless, the definite role of ET-2 in the lungs remains unclear. The ET-2 isoform, ET-1, promotes pulmonary fibrosis in mice. Although endothelin receptor antagonists (ERAs) show improvements in bleomycin-induced pulmonary fibrosis in mouse models, clinical trials examining ERAs for pulmonary fibrosis treatment have been unsuccessful, even showing harmful effects in patients. We hypothesized that ET-2, which activates the same receptor as ET-1, plays a distinct role in pulmonary fibrosis. In this study, we showed that ET-2 is expressed in the lung epithelium, and ET-2 deletion in epithelial cells of mice results in the exacerbation of bleomycin-induced pulmonary fibrosis. ET-2 knockdown in lung epithelial cell lines resulted in increased apoptosis mediated via oxidative stress induction. In contrast to the effects of ET-1, which induced fibroblast activation, ET-2 hampered fibroblast activation in primary mouse lung fibroblast cells by inhibiting the TGF-β-SMAD2/3 pathway. Our results demonstrated the divergent roles of ET-1 and ET-2 in pulmonary fibrosis pathophysiology and suggested that ET-2, expressed in epithelial cells, exerts protective effects against the development of pulmonary fibrosis in mice.
Collapse
Affiliation(s)
- ARISTI INTAN SORAYA
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakita, Higashinada, Kobe, 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, 650-0017, Japan
| | - YOKO SUZUKI
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakita, Higashinada, Kobe, 658-8558, Japan
| | - MITSURU MORIMOTO
- Laboratory for Lung Development and Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - CHEMYONG JAY KO
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, 2001 South Lincoln Avenue, Urbana, IL 61802, USA
| | - KOJI IKEDA
- Department of Epidemiology for Longevity and Regional Health, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyou, Kyoto 6028566, Japan
| | - KEN-ICHI HIRATA
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, 650-0017, Japan
| | - NORIAKI EMOTO
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakita, Higashinada, Kobe, 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, 650-0017, Japan
| |
Collapse
|
24
|
Dolivo D, Rodrigues A, Sun L, Li Y, Hou C, Galiano R, Hong SJ, Mustoe T. The Na x (SCN7A) channel: an atypical regulator of tissue homeostasis and disease. Cell Mol Life Sci 2021; 78:5469-5488. [PMID: 34100980 PMCID: PMC11072345 DOI: 10.1007/s00018-021-03854-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/15/2021] [Accepted: 05/08/2021] [Indexed: 12/15/2022]
Abstract
Within an articulately characterized family of ion channels, the voltage-gated sodium channels, exists a black sheep, SCN7A (Nax). Nax, in contrast to members of its molecular family, has lost its voltage-gated character and instead rapidly evolved a new function as a concentration-dependent sensor of extracellular sodium ions and subsequent signal transducer. As it deviates fundamentally in function from the rest of its family, and since the bulk of the impressive body of literature elucidating the pathology and biochemistry of voltage-gated sodium channels has been performed in nervous tissue, reports of Nax expression and function have been sparse. Here, we investigate available reports surrounding expression and potential roles for Nax activity outside of nervous tissue. With these studies as justification, we propose that Nax likely acts as an early sensor that detects loss of tissue homeostasis through the pathological accumulation of extracellular sodium and/or through endothelin signaling. Sensation of homeostatic aberration via Nax then proceeds to induce pathological tissue phenotypes via promotion of pro-inflammatory and pro-fibrotic responses, induced through direct regulation of gene expression or through the generation of secondary signaling molecules, such as lactate, that can operate in an autocrine or paracrine fashion. We hope that our synthesis of much of the literature investigating this understudied protein will inspire more research into Nax not simply as a biochemical oddity, but also as a potential pathophysiological regulator and therapeutic target.
Collapse
Affiliation(s)
- David Dolivo
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Adrian Rodrigues
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Lauren Sun
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Yingxing Li
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Chun Hou
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
- Department of Plastic and Cosmetic Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Robert Galiano
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA
| | - Seok Jong Hong
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA.
- , 300 E. Superior St., Chicago, IL, 60611, USA.
| | - Thomas Mustoe
- Department of Surgery, Northwestern University-Feinberg School of Medicine, Chicago, USA.
- , 737 N. Michigan Ave., Chicago, IL, 60611, USA.
| |
Collapse
|
25
|
Wu Q, Liu Y, Xie Y, Wei S, Liu Y. Identification of Potential ceRNA Network and Patterns of Immune Cell Infiltration in Systemic Sclerosis-Associated Interstitial Lung Disease. Front Cell Dev Biol 2021; 9:622021. [PMID: 34222222 PMCID: PMC8248550 DOI: 10.3389/fcell.2021.622021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 05/26/2021] [Indexed: 02/05/2023] Open
Abstract
Purpose Systemic sclerosis-associated interstitial lung disease (SSc-ILD) is one of the most severe complications of systemic sclerosis (SSc) and is the leading cause of SSc-related deaths. However, the precise pathogenesis of pulmonary fibrosis in SSc-ILD remains unknown. This study aimed to evaluate the competing endogenous RNA (ceRNA) regulatory network and immune cell infiltration patterns in SSc-ILD. Methods One microRNA (miRNA) and three messenger RNA (mRNA) microarray datasets were obtained from the Gene Expression Omnibus (GEO) database. Then, the differentially expressed miRNAs (DEmiRs) and mRNAs (DEMs) between SSc-ILD patients and normal controls were identified, respectively, followed by the prediction of the target genes and target lncRNAs of DEmiRs. The overlapping genes between DEmiRs target genes and DEMs were identified as core mRNAs to construct the ceRNA network. In addition, the “Cell Type Identification by Estimating Relative Subsets of Known RNA Transcripts (CIBERSORT)” algorithm was used to analyze the composition of infiltrating immune cells in lung tissues of SSc-ILD patients and controls, and differentially expressed immune cells were recognized. The correlation between immune cells and core mRNAs was evaluated by Pearson correlation analysis. Results Totally, 42 SSc-ILD lung tissues and 18 normal lung tissues were included in this study. We identified 35 DEmiRs and 142 DEMs and predicted 1,265 target genes of DEmiRs. Then, 9 core mRNAs related to SSc-ILD were recognized, which were the overlapping genes between DEmiRs target genes and DEMs. Meanwhile, 9 DEmiRs related to core mRNAs were identified reversely, and their target lncRNAs were predicted. In total, 9 DEmiRs, 9 core mRNAs, and 51 predicted lncRNAs were integrated to construct the ceRNA regulatory network of SSc-ILD. In addition, 9 types of immune cells were differentially expressed in lung tissues between SSc-ILD patients and controls. Some core mRNAs, such as COL1A1, FOS, and EDN1, were positively or negatively correlated with the number of infiltrating immune cells. Conclusion This is the first comprehensive study to construct the potential ceRNA regulatory network and analyze the composition of infiltrating immune cells in lung tissues of SSc-ILD patients, which improves our understanding of the pathogenesis of SSc-ILD.
Collapse
Affiliation(s)
- Qiuhong Wu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Liu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Xie
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Shixiong Wei
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.,Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China.,Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
26
|
New insights into the Hippo/YAP pathway in idiopathic pulmonary fibrosis. Pharmacol Res 2021; 169:105635. [PMID: 33930530 DOI: 10.1016/j.phrs.2021.105635] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease characterised by an inexorable decline in lung function. The development of IPF involves multiple positive feedback loops; and a strong support role of the Hippo/YAP signalling pathway, which is essential for regulating cell proliferation and organ size, in IPF pathogenesis has been unveiled recently in cell and animal models. YAP/TAZ contributes to both pulmonary fibrosis and alveolar regeneration via the conventional Hippo/YAP signalling pathway, G protein-coupled receptor signalling, and mechanotransduction. Selectively inhibiting YAP/TAZ in lung fibroblasts may inhibit fibroblast proliferation and extracellular matrix deposition, while activating YAP/TAZ in alveolar epithelial cells may promote alveolar regeneration. In this review, we explore, for the first time, the bidirectional and cell-specific regulation of the Hippo/YAP pathway in IPF pathogenesis and discuss recent research progress and future prospects of IPF treatment based on Hippo/YAP signalling, thus providing a basis for the development of new therapeutic strategies to alleviate or even reverse IPF.
Collapse
|
27
|
Haak AJ, Kostallari E, Sicard D, Ligresti G, Choi KM, Caporarello N, Jones DL, Tan Q, Meridew J, Diaz Espinosa AM, Aravamudhan A, Maiers JL, Britt RD, Roden AC, Pabelick CM, Prakash YS, Nouraie SM, Li X, Zhang Y, Kass DJ, Lagares D, Tager AM, Varelas X, Shah VH, Tschumperlin DJ. Selective YAP/TAZ inhibition in fibroblasts via dopamine receptor D1 agonism reverses fibrosis. Sci Transl Med 2020; 11:11/516/eaau6296. [PMID: 31666402 DOI: 10.1126/scitranslmed.aau6296] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 03/01/2019] [Accepted: 09/04/2019] [Indexed: 01/18/2023]
Abstract
Tissue fibrosis is characterized by uncontrolled deposition and diminished clearance of fibrous connective tissue proteins, ultimately leading to organ scarring. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) have recently emerged as pivotal drivers of mesenchymal cell activation in human fibrosis. Therapeutic strategies inhibiting YAP and TAZ have been hindered by the critical role that these proteins play in regeneration and homeostasis in different cell types. Here, we find that the Gαs-coupled dopamine receptor D1 (DRD1) is preferentially expressed in lung and liver mesenchymal cells relative to other resident cells of these organs. Agonism of DRD1 selectively inhibits YAP/TAZ function in mesenchymal cells and shifts their phenotype from profibrotic to fibrosis resolving, reversing in vitro extracellular matrix stiffening and in vivo tissue fibrosis in mouse models. Aromatic l-amino acid decarboxylase [DOPA decarboxylase (DDC)], the enzyme responsible for the final step in biosynthesis of dopamine, is decreased in the lungs of subjects with idiopathic pulmonary fibrosis, and its expression inversely correlates with disease severity, consistent with an endogenous protective role for dopamine signaling that is lost in pulmonary fibrosis. Together, these findings establish a pharmacologically tractable and cell-selective approach to targeting YAP/TAZ via DRD1 that reverses fibrosis in mice.
Collapse
Affiliation(s)
- Andrew J Haak
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Enis Kostallari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Delphine Sicard
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Giovanni Ligresti
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Kyoung Moo Choi
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Nunzia Caporarello
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Dakota L Jones
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Qi Tan
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Jeffrey Meridew
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Ana M Diaz Espinosa
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Aja Aravamudhan
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Jessica L Maiers
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Rodney D Britt
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA.,Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester MN 55905, USA.,Abigail Wexner Research Institute at Nationwide Children's Hospital and Department of Pediatrics, Ohio State University, Columbus, OH 43215, USA
| | - Anja C Roden
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester MN 55905, USA
| | - Christina M Pabelick
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA.,Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester MN 55905, USA
| | - Y S Prakash
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA.,Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester MN 55905, USA
| | - Seyed Mehdi Nouraie
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease and Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xiaoyun Li
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease and Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Yingze Zhang
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease and Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Daniel J Kass
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease and Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - David Lagares
- Division of Pulmonary and Critical Care Medicine, Fibrosis Research Center, and Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Andrew M Tager
- Division of Pulmonary and Critical Care Medicine, Fibrosis Research Center, and Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Vijay H Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
28
|
|
29
|
Ezhilarasan D. Endothelin-1 in portal hypertension: The intricate role of hepatic stellate cells. Exp Biol Med (Maywood) 2020; 245:1504-1512. [PMID: 32791849 DOI: 10.1177/1535370220949148] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
IMPACT STATEMENT Portal hypertension is pathologically defined as increase of portal venous pressure, mainly due to chronic liver diseases such as fibrosis and cirrhosis. In fibrotic liver, activated hepatic stellate cells increase their contraction in response to endothelin-1 (ET-1) via autocrine and paracrine stimulation from liver sinusoidal endothelial cells and injured hepatocytes. Clinical studies are limited with ET receptor antagonists in cirrhotic patients with portal hypertension. Hence, studies are needed to find molecules that block ET-1 synthesis. Accumulation of extracellular matrix proteins in the perisinusoidal space, tissue contraction, and alteration in blood flow are prominent during portal hypertension. Therefore, novel matrix modulators should be tested experimentally as well as in clinical studies. Specifically, tumor necrosis factor-α, transforming growth factor-β1, Wnt, Notch, rho-associated protein kinase 1 signaling antagonists, and peroxisome proliferator-activated receptor α and γ, interferon-γ and sirtuin 1 agonists should be tested elaborately against cirrhosis patients with portal hypertension.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Biomedical Research Unit and Laboratory Animal Centre, Saveetha Dental College, 194347Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 600 077, India
| |
Collapse
|
30
|
Gaikwad AV, Eapen MS, McAlinden KD, Chia C, Larby J, Myers S, Dey S, Haug G, Markos J, Glanville AR, Sohal SS. Endothelial to mesenchymal transition (EndMT) and vascular remodeling in pulmonary hypertension and idiopathic pulmonary fibrosis. Expert Rev Respir Med 2020; 14:1027-1043. [PMID: 32659128 DOI: 10.1080/17476348.2020.1795832] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and irreversible fibrotic disease associated with respiratory failure. The disease remains idiopathic, but repeated alveolar epithelium injury, disruption of alveolar-capillary integrity, abnormal vascular repair, and pulmonary vascular remodeling are considered possible pathogenic mechanisms. Also, the development of comorbidities such as pulmonary hypertension (PH) could further impact disease outcome, quality of life and survival rates in IPF. AREAS COVERED The current review provides a comprehensive literature survey of the mechanisms involved in the development and manifestations of IPF and their links to PH pathology. This review also provides the current understanding of molecular mechanisms that link the two pathologies and will specifically decipher the role of endothelial to mesenchymal transition (EndMT) along with the possible triggers of EndMT. The possibility of targeting EndMT as a therapeutic option in IPF is discussed. EXPERT OPINION With a steady increase in prevalence and mortality, IPF is no longer considered a rare disease. Thus, it is of utmost importance and urgency that the underlying profibrotic pathways and mechanisms are fully understood, to enable the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Archana Vijay Gaikwad
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania , Launceston, Australia
| | - Mathew Suji Eapen
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania , Launceston, Australia
| | - Kielan D McAlinden
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania , Launceston, Australia
| | - Collin Chia
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania , Launceston, Australia.,Department of Respiratory Medicine, Launceston General Hospital , Launceston, Australia
| | - Josie Larby
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania , Launceston, Australia.,Department of Respiratory Medicine, Launceston General Hospital , Launceston, Australia
| | - Stephen Myers
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania , Launceston, Australia
| | - Surajit Dey
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania , Launceston, Australia
| | - Greg Haug
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania , Launceston, Australia.,Department of Respiratory Medicine, Launceston General Hospital , Launceston, Australia
| | - James Markos
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania , Launceston, Australia.,Department of Respiratory Medicine, Launceston General Hospital , Launceston, Australia
| | - Allan R Glanville
- Lung Transplant Unit, Department of Thoracic Medicine, St Vincent's Hospital , Sydney, Australia
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania , Launceston, Australia
| |
Collapse
|
31
|
Bochenek ML, Leidinger C, Rosinus NS, Gogiraju R, Guth S, Hobohm L, Jurk K, Mayer E, Münzel T, Lankeit M, Bosmann M, Konstantinides S, Schäfer K. Activated Endothelial TGFβ1 Signaling Promotes Venous Thrombus Nonresolution in Mice Via Endothelin-1: Potential Role for Chronic Thromboembolic Pulmonary Hypertension. Circ Res 2019; 126:162-181. [PMID: 31747868 DOI: 10.1161/circresaha.119.315259] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RATIONALE Chronic thromboembolic pulmonary hypertension (CTEPH) is characterized by defective thrombus resolution, pulmonary artery obstruction, and vasculopathy. TGFβ (transforming growth factor-β) signaling mutations have been implicated in pulmonary arterial hypertension, whereas the role of TGFβ in the pathophysiology of CTEPH is unknown. OBJECTIVE To determine whether defective TGFβ signaling in endothelial cells contributes to thrombus nonresolution and fibrosis. METHODS AND RESULTS Venous thrombosis was induced by inferior vena cava ligation in mice with genetic deletion of TGFβ1 in platelets (Plt.TGFβ-KO) or TGFβ type II receptors in endothelial cells (End.TGFβRII-KO). Pulmonary endarterectomy specimens from CTEPH patients were analyzed using immunohistochemistry. Primary human and mouse endothelial cells were studied using confocal microscopy, quantitative polymerase chain reaction, and Western blot. Absence of TGFβ1 in platelets did not alter platelet number or function but was associated with faster venous thrombus resolution, whereas endothelial TGFβRII deletion resulted in larger, more fibrotic and higher vascularized venous thrombi. Increased circulating active TGFβ1 levels, endothelial TGFβRI/ALK1 (activin receptor-like kinase), and TGFβRI/ALK5 expression were detected in End.TGFβRII-KO mice, and activated TGFβ signaling was present in vessel-rich areas of CTEPH specimens. CTEPH-endothelial cells and murine endothelial cells lacking TGFβRII simultaneously expressed endothelial and mesenchymal markers and transcription factors regulating endothelial-to-mesenchymal transition, similar to TGFβ1-stimulated endothelial cells. Mechanistically, increased endothelin-1 levels were detected in TGFβRII-KO endothelial cells, murine venous thrombi, or endarterectomy specimens and plasma of CTEPH patients, and endothelin-1 overexpression was prevented by inhibition of ALK5, and to a lesser extent of ALK1. ALK5 inhibition and endothelin receptor antagonization inhibited mesenchymal lineage conversion in TGFβ1-exposed human and murine endothelial cells and improved venous thrombus resolution and pulmonary vaso-occlusions in End.TGFβRII-KO mice. CONCLUSIONS Endothelial TGFβ1 signaling via type I receptors and endothelin-1 contribute to mesenchymal lineage transition and thrombofibrosis, which were prevented by blocking endothelin receptors. Our findings may have relevant implications for the prevention and management of CTEPH.
Collapse
Affiliation(s)
- Magdalena L Bochenek
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Christiane Leidinger
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany
| | - Nico S Rosinus
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Rajinikanth Gogiraju
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Stefan Guth
- Thoracic Surgery, Kerckhoff Clinic, Bad Nauheim, Germany (S.G., E.M.)
| | - Lukas Hobohm
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany
| | - Eckhard Mayer
- Thoracic Surgery, Kerckhoff Clinic, Bad Nauheim, Germany (S.G., E.M.).,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Thomas Münzel
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Mareike Lankeit
- Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany.,Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité -University Medicine, Berlin, Germany (M.L.)
| | - Markus Bosmann
- Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany.,Department of Medicine, Boston University School of Medicine, MA (M.B.)
| | - Stavros Konstantinides
- Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany.,Department of Cardiology, Democritus University of Thrace, Alexandroupolis, Greece (S.K.)
| | - Katrin Schäfer
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| |
Collapse
|
32
|
Comeglio P, Filippi S, Sarchielli E, Morelli A, Cellai I, Corno C, Adorini L, Vannelli GB, Maggi M, Vignozzi L. Therapeutic effects of the selective farnesoid X receptor agonist obeticholic acid in a monocrotaline-induced pulmonary hypertension rat model. J Endocrinol Invest 2019; 42:951-965. [PMID: 30674010 DOI: 10.1007/s40618-019-1009-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 01/11/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Activation of the farnesoid X receptor (FXR), a member of the nuclear receptor steroid superfamily, leads to anti-inflammatory and anti-fibrotic effects in several tissues, including the lung. We have recently demonstrated a protective effect of the farnesoid X receptor (FXR) agonist obeticholic acid (OCA) in rat models of monocrotaline (MCT)-induced pulmonary arterial hypertension (PAH) and bleomycin-induced pulmonary fibrosis. The aim of the present study was to investigate whether the positive effects of OCA treatment could be exerted also in established MCT-induced PAH, i.e., starting treatment 2 weeks after MCT administration. METHODS Rats with MCT-induced PAH were treated, 2 weeks after MCT administration, with OCA or tadalafil for two additional weeks. Pulmonary functional tests were performed at week 2 (before treatment) and four (end of treatment). At the same time points, lung morphological features and expression profile of genes related to smooth muscle relaxation/contraction and tissue remodeling were also assessed. RESULTS 2 weeks after MCT-induced injury, the treadmill resistance (a functional parameter related to pulmonary hypertension) was significantly decreased. At the same time point, we observed right ventricular hypertrophy and vascular remodeling, with upregulation of genes related to inflammation. At week 4, we observed a further worsening of the functional and morphological parameters, accompanied by dysregulation of inflammatory and extracellular matrix markers mRNA expression. Administration of OCA (3 or 10 mg/kg/day), starting 2 weeks after MCT-induced injury, significantly improved pulmonary function, effectively normalizing the exercise capacity. OCA also reverted most of the lung alterations, with a significant reduction of lung vascular wall thickness, right ventricular hypertrophy, and restoration of the local balance between relaxant and contractile pathways. Markers of remodeling pathways were also normalized by OCA treatment. Notably, results with OCA treatment were similar, or even superior, to those obtained with tadalafil, a recently approved treatment for pulmonary hypertension. CONCLUSIONS The results of this study demonstrate a significant therapeutic effect of OCA in established MCT-induced PAH, improving exercise capacity associated with reduction of right ventricular hypertrophy and lung vascular remodeling. Thus, OCA dosing in a therapeutic protocol restores the balance between relaxant and contractile pathways in the lung, promoting cardiopulmonary protective actions in MCT-induced PAH.
Collapse
Affiliation(s)
- P Comeglio
- Sexual Medicine and Andrology Unit, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy
| | - S Filippi
- Interdepartmental Laboratory of Functional and Cellular Pharmacology of Reproduction, Department of NEUROFARBA, University of Florence, Florence, Italy
| | - E Sarchielli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - A Morelli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - I Cellai
- Sexual Medicine and Andrology Unit, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy
| | - C Corno
- Sexual Medicine and Andrology Unit, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy
| | - L Adorini
- Intercept Pharmaceuticals, New York, NY, USA
| | - G B Vannelli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - M Maggi
- Sexual Medicine and Andrology Unit, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy
- I.N.B.B. (Istituto Nazionale Biostrutture E Biosistemi), Rome, Italy
| | - L Vignozzi
- Sexual Medicine and Andrology Unit, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy.
- I.N.B.B. (Istituto Nazionale Biostrutture E Biosistemi), Rome, Italy.
| |
Collapse
|
33
|
Kwapiszewska G, Gungl A, Wilhelm J, Marsh LM, Thekkekara Puthenparampil H, Sinn K, Didiasova M, Klepetko W, Kosanovic D, Schermuly RT, Wujak L, Weiss B, Schaefer L, Schneider M, Kreuter M, Olschewski A, Seeger W, Olschewski H, Wygrecka M. Transcriptome profiling reveals the complexity of pirfenidone effects in idiopathic pulmonary fibrosis. Eur Respir J 2018; 52:13993003.00564-2018. [DOI: 10.1183/13993003.00564-2018] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/03/2018] [Indexed: 11/05/2022]
Abstract
Despite the beneficial effects of pirfenidone in treating idiopathic pulmonary fibrosis (IPF), it remains unclear if lung fibroblasts (FB) are the main therapeutic target.To resolve this question, we employed a comparative transcriptomic approach and analysed lung homogenates (LH) and FB derived from IPF patients treated with or without pirfenidone.In FB, pirfenidone therapy predominantly affected growth and cell division pathways, indicating a major cellular metabolic shift. In LH samples, pirfenidone treatment was mostly associated with inflammation-related processes. In FB and LH, regulated genes were over-represented in the Gene Ontology node “extracellular matrix”. We identified lower expression of cell migration-inducing and hyaluronan-binding protein (CEMIP) in both LH and FB from pirfenidone-treated IPF patients. Plasma levels of CEMIP were elevated in IPF patients compared to healthy controls and decreased after 7 months of pirfenidone treatment. CEMIP expression in FB was downregulated in a glioma-associated oncogene homologue-dependent manner and CEMIP silencing in IPF FB reduced collagen production and attenuated cell proliferation and migration.Cumulatively, our approach indicates that pirfenidone exerts beneficial effects via its action on multiple pathways in both FB and other pulmonary cells, through its ability to control extracellular matrix architecture and inflammatory reactions.
Collapse
|
34
|
Karampitsakos T, Tzouvelekis A, Chrysikos S, Bouros D, Tsangaris I, Fares WH. Pulmonary hypertension in patients with interstitial lung disease. Pulm Pharmacol Ther 2018; 50:38-46. [PMID: 29605286 DOI: 10.1016/j.pupt.2018.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 03/12/2018] [Accepted: 03/28/2018] [Indexed: 01/20/2023]
Abstract
Interstitial lung diseases (ILDs) comprise a broad and heterogeneous group of more than two hundred diseases with common functional characteristics. Their diagnosis and management require a multidisciplinary approach. This multidisciplinary approach involves the assessment of comorbid conditions including pulmonary hypertension (PH) that exerts a dramatic impact on survival. The current World Health Organization (WHO) classification of PH encompasses many of the interstitial lung diseases into WHO Group 3, while sarcoidosis, Pulmonary Langerhans Cell Histiocytosis and lymphangioleiomyomatosis are placed into WHO Group 5 as diseases with unclear or multifactorial mechanisms. Connective tissue diseases could span any of the 5 WHO groups based on the primary phenotype into which they manifest. Interestingly, several challenging phenotypes present with features that overlap between two or more WHO PH groups. Currently, PH-specific treatment is recommended only for patients classified into WHO Group 1 PH. The lack of specific treatment for other groups, including PH in the setting of ILD, reflects the poor outcomes of these patients. Thus, identification of the optimal strategy for ILD patients with PH remains an amenable need. This review article provides a brief overview of biomarkers indicative of vascular remodeling in interstitial lung disease, summarizes the current state of knowledge regarding patients with PH and ILD and highlights future perspectives that remain to be addressed.
Collapse
Affiliation(s)
| | - Argyrios Tzouvelekis
- First Academic Department of Pneumonology, Hospital for Thoracic Diseases, "Sotiria", Medical School, National and Kapodistrian University of Athens, Athens, Greece; Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Athens, Greece
| | - Serafeim Chrysikos
- 5(th) Department of Pneumonology, Hospital for Thoracic Diseases, "Sotiria", Athens, Greece
| | - Demosthenes Bouros
- First Academic Department of Pneumonology, Hospital for Thoracic Diseases, "Sotiria", Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Iraklis Tsangaris
- Second Critical Care Department, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Wassim H Fares
- Section of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
35
|
Hosseinzadeh A, Javad-Moosavi SA, Reiter RJ, Hemati K, Ghaznavi H, Mehrzadi S. Idiopathic pulmonary fibrosis (IPF) signaling pathways and protective roles of melatonin. Life Sci 2018; 201:17-29. [PMID: 29567077 DOI: 10.1016/j.lfs.2018.03.032] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/13/2018] [Accepted: 03/15/2018] [Indexed: 12/19/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by the progressive loss of lung function due to tissue scarring. A variety of pro-inflammatory and pro-fibrogenic factors including interleukin‑17A, transforming growth factor β, Wnt/β‑catenin, vascular endothelial growth factor, platelet-derived growth factor, fibroblast growth factors, endotelin‑1, renin angiotensin system and impaired caveolin‑1 function are involved in the IPF pathogenesis. Current therapies for IPF have some limitations and this highlights the need for effective therapeutic agents to treat this fatal disease. Melatonin and its metabolites are broad-spectrum antioxidants that not only remove reactive oxygen and nitrogen species by radical scavenging but also up-regulate the expression and activity of endogenous antioxidants. Via these actions, melatonin and its metabolites modulate a variety of molecular pathways in different pathophysiological conditions. Herein, we review the signaling pathways involved in the pathophysiology of IPF and the potentially protective effects of melatonin on these pathways.
Collapse
Affiliation(s)
- Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health, San Antonio, TX, USA
| | - Karim Hemati
- Department of Anesthesiology, Iran University of Medical Sciences, Tehran, Iran; Department of Anesthesiology, Ilam University of Medical Sciences, Ilam, Iran
| | - Habib Ghaznavi
- Department of Pharmacology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
36
|
Chang W, Lajko M, Fawzi AA. Endothelin-1 is associated with fibrosis in proliferative diabetic retinopathy membranes. PLoS One 2018; 13:e0191285. [PMID: 29351334 PMCID: PMC5774761 DOI: 10.1371/journal.pone.0191285] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/02/2018] [Indexed: 01/09/2023] Open
Abstract
Purpose To characterize the relationship between endothelin-1 and fibrosis in epiretinal membranes in proliferative diabetic retinopathy and explore the role of endothelial-mesenchymal transition in these membranes. Methods Membranes were obtained from eyes undergoing pars plana vitrectomy for complicated proliferative diabetic retinopathy or idiopathic epiretinal membrane. Through standard immunohistochemical techniques, we labeled membranes to explore the distribution of endothelin-1 and endothelin receptor B, comparing proliferative diabetic retinopathy and idiopathic epiretinal membranes. In addition, membranes were also labeled with markers for fibroblasts, endothelial, and glial cells and studied with confocal laser scanning microscopy. The intensity of endothelin-1 labeling was quantified using standard image analysis software. Results Fourteen membranes were included in the analysis, nine from eyes with proliferative diabetic retinopathy and five idiopathic membranes. Flatmount diabetic membranes showed co-localization of endothelin-1 with S100A4 and CD31. Immunohistochemistry and quantitative analysis of cross-sectional membranes showed significantly higher endothelin-1 labeling in proliferative diabetic retinopathy membranes compared to idiopathic membranes (p<0.05). Diabetic membranes showed more elements staining positive for S100A4 compared to idiopathic membranes. Conclusion Epiretinal membrane formation in proliferative diabetic retinopathy involves higher tissue levels of endothelin-1 and fibroblastic activity. Furthermore, endothelin-1, endothelial and fibroblastic staining appear to be correlated, suggestive of endothelial-to-mesenchymal transition in proliferative diabetic retinopathy.
Collapse
Affiliation(s)
- William Chang
- Department of Ophthalmology, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States of America
| | - Michelle Lajko
- Department of Ophthalmology, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States of America
| | - Amani A. Fawzi
- Department of Ophthalmology, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States of America
- * E-mail:
| |
Collapse
|
37
|
Duan Z, Jia Y, Zhang J, Long L, Tang L. Endothelin-1-induced expression of α-smooth muscle actin in human myometrial fibroblasts. J Obstet Gynaecol Res 2017; 44:540-546. [PMID: 29271089 DOI: 10.1111/jog.13539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 09/27/2017] [Indexed: 11/28/2022]
Abstract
AIM We aimed to investigate the potential role of endothelin-1 (ET-1) in the regulation of the expression of α-smooth muscle actin (α-SMA) in human myometrial fibroblasts. METHODS Primary myometrial fibroblasts were obtained from myometrium and were identified by immunocytochemical staining. Then, 1 × 107 cells were treated with ET-1 at a concentration of 0.1, 1.0, 10.0, or 100.0 nM for 24 h. To investigate the time course effects of ET-1 on the growth of fibroblasts, 1 × 107 cells were treated with 10.0 nM ET-1 for 6, 12, 24, and 48 h. Real-time quantitative PCR (qPCR) and Western blot analysis were used to determine the expression of α-SMA mRNA and protein, respectively. RESULTS Human myometrial fibroblasts were identified by immunohistochemistry. Compared with the control group, the expression levels of α-SMA mRNA and protein were identified in cells treated with ET-1 at a concentration of 0.1, 1.0, 10.0, or 100.0 nM for 24 h (P < 0.05). ET-1 treatment affected the expression of α-SMA mRNA and protein in a dose-dependent manner (P < 0.05). The induction of α-SMA mRNA and protein expression increased from 6 to 48 h. CONCLUSION The results show that ET-1 induces the expression of α-SMA in human myometrium in vitro.
Collapse
Affiliation(s)
- Zhaoning Duan
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Jia
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Zhang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Long
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liangdan Tang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
38
|
Herrmann FE, Wollin L, Wirth J, Gantner F, Lämmle B, Wex E. Olodaterol shows anti-fibrotic efficacy in in vitro and in vivo models of pulmonary fibrosis. Br J Pharmacol 2017; 174:3848-3864. [PMID: 28810065 DOI: 10.1111/bph.13982] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 08/08/2017] [Accepted: 08/09/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND AND PURPOSE Idiopathic pulmonary fibrosis (IPF) is a fatal respiratory disease characterized by excessive fibroblast activation ultimately leading to scarring of the lungs. Although, the activation of β2 -adrenoceptors (β2 -AR) has been shown to inhibit pro-fibrotic events primarily in cell lines, the role of β2 -adrenoceptor agonists has not yet been fully characterized. The aim of our study was to explore the anti-fibrotic activity of the long-acting β2 -adrenoceptor agonist olodaterol in primary human lung fibroblasts (HLF) and in murine models of pulmonary fibrosis. EXPERIMENTAL APPROACH We assessed the activity of olodaterol to inhibit various pro-fibrotic mechanisms, induced by different pro-fibrotic mediators, in primary HLF from control donors and patients with IPF (IPF-LF). The in vivo anti-fibrotic activity of olodaterol, given once daily by inhalation in either a preventive or therapeutic treatment regimen, was explored in murine models of lung fibrosis induced by either bleomycin or the overexpression of TGF-β1. KEY RESULTS In both HLF and IPF-LF, olodaterol attenuated TGF-β-induced expression of α-smooth muscle actin, fibronectin and endothelin-1 (ET-1), FGF- and PDGF-induced motility and proliferation and TGF-β/ET-1-induced contraction. In vivo olodaterol significantly attenuated the bleomycin-induced increase in lung weight, reduced bronchoalveolar lavage cell counts and inhibited release of pro-fibrotic mediators (TGF-ß, MMP-9 and tissue inhibitor of metalloproteinase-1). Forced vital capacity was increased only with the preventive treatment regimen. In the TGF-β-overexpressing model, olodaterol additionally reduced the Col3A1 mRNA expression. CONCLUSION AND IMPLICATIONS Olodaterol showed anti-fibrotic properties in primary HLF from control and IPF patients and in murine models of lung fibrosis.
Collapse
Affiliation(s)
- Franziska Elena Herrmann
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Lutz Wollin
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Johannes Wirth
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Florian Gantner
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Bärbel Lämmle
- Target Discovery Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Eva Wex
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| |
Collapse
|
39
|
Predescu SA, Zhang J, Bardita C, Patel M, Godbole V, Predescu DN. Mouse Lung Fibroblast Resistance to Fas-Mediated Apoptosis Is Dependent on the Baculoviral Inhibitor of Apoptosis Protein 4 and the Cellular FLICE-Inhibitory Protein. Front Physiol 2017; 8:128. [PMID: 28352235 PMCID: PMC5348516 DOI: 10.3389/fphys.2017.00128] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 02/17/2017] [Indexed: 01/01/2023] Open
Abstract
A characteristic feature of idiopathic pulmonary fibrosis (IPF) is accumulation of apoptotic resistant fibroblasts/myofibroblasts in the fibroblastic foci. As caveolin (Cav)-null mice develop pulmonary fibrosis (PF), we hypothesized that the participating fibroblasts display an apoptosis-resistant phenotype. To test this hypothesis and identify the molecular mechanisms involved we isolated lung fibroblasts from Cav-null mice and examined the expression of several inhibitors of apoptosis (IAPs), of c-FLIP, of Bcl-2 proteins and of the death receptor CD95/Fas. We found significant increase in XIAP and c-FLIP constitutive protein expression with no alteration of Bcl-2 and lower levels of CD95/Fas. The isolated fibroblasts were then treated with the CD95/Fas ligand (FasL) to induce apoptosis. While the morphological and biochemical alterations induced by FasL were similar in wild-type (wt) and Cav-null mouse lung fibroblasts, the time course and the extent of the alterations were greater in the Cav-null fibroblasts. Several salient features of Cav-null fibroblasts response such as loss of membrane potential, fragmentation of the mitochondrial continuum concurrent with caspase-8 activation, and subsequent Bid cleavage, prior to caspase-3 activation were detected. Furthermore, M30 antigen formation, phosphatidylserine expression and DNA fragmentation were caspase-3 dependent. SiRNA-mediated silencing of XIAP and c-FLIP, individually or combined, enhanced the sensitivity of lung fibroblasts to FasL-induced apoptosis. Pharmacological inhibition of Bcl-2 had no effect. Together our findings support a mechanism in which CD95/Fas engagement activates caspase-8, inducing mitochondrial apoptosis through Bid cleavage. XIAP and c-FLIP fine tune this process in a cell-type specific manner.
Collapse
Affiliation(s)
- Sanda A Predescu
- Department of Internal Medicine, Division of Pulmonary and Critical Care, Rush University, Medical College Chicago, IL, USA
| | - Jian Zhang
- Department of Biological Sciences, Columbia University New York, NY, USA
| | - Cristina Bardita
- Department of Internal Medicine, Division of Pulmonary and Critical Care, Rush University, Medical College Chicago, IL, USA
| | - Monal Patel
- Northwestern University Feinberg School of Medicine Chicago, IL, USA
| | - Varun Godbole
- Department of Internal Medicine, Division of Pulmonary and Critical Care, Rush University, Medical College Chicago, IL, USA
| | - Dan N Predescu
- Department of Internal Medicine, Division of Pulmonary and Critical Care, Rush University, Medical College Chicago, IL, USA
| |
Collapse
|
40
|
Wermuth PJ, Li Z, Mendoza FA, Jimenez SA. Stimulation of Transforming Growth Factor-β1-Induced Endothelial-To-Mesenchymal Transition and Tissue Fibrosis by Endothelin-1 (ET-1): A Novel Profibrotic Effect of ET-1. PLoS One 2016; 11:e0161988. [PMID: 27583804 PMCID: PMC5008786 DOI: 10.1371/journal.pone.0161988] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 08/16/2016] [Indexed: 11/29/2022] Open
Abstract
TGF-β-induced endothelial-to-mesenchymal transition (EndoMT) is a newly recognized source of profibrotic activated myofibroblasts and has been suggested to play a role in the pathogenesis of various fibrotic processes. Endothelin-1 (ET-1) has been implicated in the development of tissue fibrosis but its participation in TGF-β-induced EndoMT has not been studied. Here we evaluated the role of ET-1 on TGF-β1-induced EndoMT in immunopurified CD31+/CD102+ murine lung microvascular endothelial cells. The expression levels of α-smooth muscle actin (α-SMA), of relevant profibrotic genes, and of various transcription factors involved in the EndoMT process were assessed employing quantitative RT-PCR, immunofluorescence histology and Western blot analysis. TGF-β1 caused potent induction of EndoMT whereas ET-1 alone had a minimal effect. However, ET-1 potentiated TGF-β1-induced EndoMT and TGF-β1-stimulated expression of mesenchymal cell specific and profibrotic genes and proteins. ET-1 also induced expression of the TGF-β receptor 1 and 2 genes, suggesting a plausible autocrine mechanism to potentiate TGF-β-mediated EndoMT and fibrosis. Stimulation of TGF-β1-induced skin and lung fibrosis by ET-1 was confirmed in vivo in an animal model of TGF-β1-induced tissue fibrosis. These results suggest a novel role for ET-1 in the establishment and progression of tissue fibrosis.
Collapse
Affiliation(s)
- Peter J. Wermuth
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Zhaodong Li
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Fabian A. Mendoza
- Division of Rheumatology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Sergio A. Jimenez
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|
41
|
Shen CH, Wang SC, Wang ST, Lin SM, Wu JD, Lin CT, Liu YW. Evaluation of urinary bladder fibrogenesis in a mouse model of long-term ketamine injection. Mol Med Rep 2016; 14:1880-90. [PMID: 27431428 PMCID: PMC4991691 DOI: 10.3892/mmr.2016.5482] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 06/22/2016] [Indexed: 01/12/2023] Open
Abstract
Long-term ketamine abuse has been shown to affect the lower urinary tract and result in interstitial cystitis-like syndrome. However, the causative mechanism of ketamine-induced dysfunction remains unclear. The present study aimed to investigate the physiological, histological and molecular changes on ketamine-associated cystitis (KC) in a mouse model. Both male and female Balb/c mice were separately distributed into the control group (normal saline) and ketamine group, which received ketamine hydrochloride (100 mg/kg/day) daily by intraperitoneal injection for a total period of 20 weeks. In each group, the urine was analyzed by gas chromatography-mass spectrometry to measure the concentration of ketamine and its metabolites. Urinary frequency and urine volume were examined to investigate the urinary voiding functions. Mice bladders were excised for cDNA microarray and hematoxylin and eosin (HE) staining. The ketamine and metabolites were detected only in ketamine-treated mice urine. The voiding interval was reduced in the male mice group after 20 week ketamine administration. Additionally, the result of cDNA array analysis revealed a number of gene expression levels involved in chronic wound healing response and collagen accumulation, which were closely associated with fibrosis progression in the connective tissue. In HE staining of the bladder tissue, the ketamine-injected mice exhibited prominently denser blood vessel distribution in the submucosal layer. Based on the evidence in the present study, a mechanism that delineates fibrosis formation of urinary bladder induced by the pathogenesis of ketamine abuse can be constructed.
Collapse
Affiliation(s)
- Cheng-Huang Shen
- Department of Urology, Chiayi Christian Hospital, Chiayi 600, Taiwan, R.O.C
| | - Shou-Chieh Wang
- Division of Nephrology, Department of Internal Medicine, Kuang Tien General Hospital, Taichung 437, Taiwan, R.O.C
| | - Shou-Tsung Wang
- Department of Microbiology, Immunology and Biopharmaceuticals, College of Life Sciences, National Chiayi University, Chiayi 600, Taiwan, R.O.C
| | - Shu-Mei Lin
- Department of Food Science, College of Life Sciences, National Chiayi University, Chiayi 600, Taiwan, R.O.C
| | - Jiann-Der Wu
- Department of Pathology, Chiayi Christian Hospital, Chiayi 600, Taiwan, R.O.C
| | - Chang-Te Lin
- Department of Urology, Chiayi Christian Hospital, Chiayi 600, Taiwan, R.O.C
| | - Yi-Wen Liu
- Department of Microbiology, Immunology and Biopharmaceuticals, College of Life Sciences, National Chiayi University, Chiayi 600, Taiwan, R.O.C
| |
Collapse
|
42
|
Wettlaufer SH, Scott JP, McEachin RC, Peters-Golden M, Huang SK. Reversal of the Transcriptome by Prostaglandin E2 during Myofibroblast Dedifferentiation. Am J Respir Cell Mol Biol 2016; 54:114-27. [PMID: 26098591 DOI: 10.1165/rcmb.2014-0468oc] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Myofibroblasts, the major effector cells in pathologic fibrosis, derive from the differentiation of fibroblasts driven by mediators such as transforming growth factor-β1 (TGF-β1) and biomechanical signals. Although the myofibroblast has traditionally been considered a terminally differentiated cell, the lipid mediator prostaglandin E2 (PGE2) has been shown to not only prevent but also reverse myofibroblast differentiation, as characterized by the ability of PGE2 to diminish expression of collagen I and α-smooth muscle actin in established myofibroblasts. Here, we use microarrays to examine the extent of transcriptomic changes that occur during TGF-β1-induced differentiation and PGE2-induced dedifferentiation of myofibroblasts. Normal primary human adult lung fibroblasts were cultured for 24 hours with or without TGF-β1 and treated for 48 hours with PGE2. Gene expression levels were assessed from total RNA on the Affymetrix U219 microarray. TGF-β1 up-regulated 588 genes and down-regulated 689 genes compared with control cells. PGE2 reversed the expression of 363 (62%) of the TGF-β1-up-regulated genes and 345 (50%) of the TGF-β1-down-regulated genes. Genes up-regulated by TGF-β1 and reversed by PGE2 were enriched in annotations for Cell Adhesion, Contractile Fiber, and Actin Binding, whereas genes down-regulated by TGF-β1 but subsequently reversed by PGE2 were enriched in annotations for Glycoprotein, Polysaccharide Binding, and Regulation of Cell Migration. Surprisingly, the genes whose expression was affected by PGE2 differed between TGF-β1-induced myofibroblasts and undifferentiated fibroblasts. These data demonstrate the capacity of PGE2 to effect marked global alterations in the transcriptomic program of differentiated myofibroblasts and emphasize the considerable plasticity of these cells.
Collapse
Affiliation(s)
- Scott H Wettlaufer
- 1 Division of Pulmonary and Critical Care Medicine in the Department of Internal Medicine and
| | - Jacob P Scott
- 1 Division of Pulmonary and Critical Care Medicine in the Department of Internal Medicine and
| | - Richard C McEachin
- 2 Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Marc Peters-Golden
- 1 Division of Pulmonary and Critical Care Medicine in the Department of Internal Medicine and
| | - Steven K Huang
- 1 Division of Pulmonary and Critical Care Medicine in the Department of Internal Medicine and
| |
Collapse
|
43
|
Chen YC, Chen BC, Yu CC, Lin SH, Lin CH. miR-19a, -19b, and -26b Mediate CTGF Expression and Pulmonary Fibroblast Differentiation. J Cell Physiol 2016; 231:2236-48. [PMID: 26873752 DOI: 10.1002/jcp.25341] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 02/10/2016] [Indexed: 01/01/2023]
Abstract
Although microRNA (miRNA) dysregulation with intracellular signaling cascade disruption has been demonstrated in the pathophysiology of pulmonary fibrosis, the relationship between miRNAs and intracellular signaling cascades in pulmonary fibrosis remains unclear. Using the human embryonic lung fibroblast cell line WI-38, we observed endothelin-1 (ET-1)- and thrombin-induced expression of the differentiation markers α-smooth muscle actin (α-SMA) and vimentin along with increased connective tissue growth factor (CTGF) protein expression. Decreased CTGF protein expression by CTGF siRNA significantly blocked ET-1- and thrombin-induced α-SMA and vimentin expression in WI-38 cells. Activation of the mitogen-activated protein kinases (MAPKs) extracellular signal-regulated kinase ERK, c-Jun N-terminal kinase (JNK), and p38 contributed to ET-1- and thrombin-induced CTGF, α-SMA, and vimentin expression in WI-38 cells. TargetScan Human, miRanda, and PicTar prediction algorithms were used to predict miRNAs with binding sites in the 3' untranslated region (UTR) of CTGF mRNA. miR-19a, -19b, and -26b were candidate miRNAs of CTGF. Direct binding of the candidate miRNAs to the 3'-UTR of CTGF mRNA was verified through luciferase assay by using SV40-promoter-IRES-driven luciferase containing the 3'-UTR of CTGF mRNA as a reporter plasmid. ET-1 and thrombin reduced candidate miRNA levels. Candidate miRNA overexpression significantly suppressed ET-1- and thrombin-induced CTGF expression and reduced α-SMA and vimentin expression in the WI-38 cells. Furthermore, candidate miRNA levels were decreased in the lung tissues of mice with bleomycin-induced pulmonary fibrosis, and intratracheal application of miR-19a, -19b, and 26b reduced the pulmonary fibrotic severity induced by bleomycin. This study is the first to demonstrate crosstalk between MAPK activation and reduction in miR-19a, -19b, and -26b expression leading to lung fibroblast differentiation. J. Cell. Physiol. 231: 2236-2248, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yen-Chou Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Cancer Research Center and Orthopedics Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Bing-Chang Chen
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chung-Chi Yu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shin-Hua Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Huang Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
44
|
Spagnolo P, Wells AU, Collard HR. Pharmacological treatment of idiopathic pulmonary fibrosis: an update. Drug Discov Today 2015; 20:514-24. [DOI: 10.1016/j.drudis.2015.01.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/03/2014] [Accepted: 01/07/2015] [Indexed: 12/22/2022]
|
45
|
Tzouvelekis A, Bonella F, Spagnolo P. Update on therapeutic management of idiopathic pulmonary fibrosis. Ther Clin Risk Manag 2015; 11:359-70. [PMID: 25767391 PMCID: PMC4354471 DOI: 10.2147/tcrm.s69716] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive diffuse parenchymal lung disease of unknown origin, with a mortality rate exceeding that of many cancers. The diagnostic process is complex and relies on the clinician integrating clinical, laboratory, radiological, and histological data. In the last decade, major advances in our understanding of the pathogenesis of IPF have shifted the paradigm from a primarily inflammatory process evolving to fibrosis to a condition driven by aberrant wound healing following alveolar epithelial cell injury that results in scarring of the lung, architectural distortion, and irreversible loss of function. Improved understanding of disease pathogenesis has led to the identification of several therapeutic targets and the design of high-quality clinical trials evaluating novel compounds. However, the results of these studies have been mostly disappointing, probably due to the plethora of mediators, growth factors, and signaling pathways involved in the fibrotic process. Most recently, pirfenidone and nintedanib, two compounds with pleiotropic anti-fibrotic properties, have been proven effective in reducing functional decline and disease progression in IPF. This is a major breakthrough. Nevertheless, we still have a long way to go. In fact, neither pirfenidone nor nintedanib is a cure for IPF, and most patients continue to progress despite treatment. As such, comprehensive care of patients with IPF, including management of concomitant conditions and physical debility, as well as timely referral for lung transplantation, remains essential. Several agents with a high potential are currently being tested, and many more are ready for clinical trials. Their completion is critical for achieving the ultimate goal of curing patients with IPF.
Collapse
Affiliation(s)
- Argyris Tzouvelekis
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Francesco Bonella
- Interstitial and Rare Lung Disease Unit, Ruhrlandklinik, University Hospital, University of Duisburg-Essen, Germany
| | - Paolo Spagnolo
- Medical University Clinic, Canton Hospital Baselland and University of Basel, Liestal, Switzerland
| |
Collapse
|
46
|
Covvey JR, Mancl EE. Recent Evidence for Pharmacological Treatment of Idiopathic Pulmonary Fibrosis. Ann Pharmacother 2014; 48:1611-9. [DOI: 10.1177/1060028014551015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Objective: To describe emerging evidence for the pharmacological treatment of idiopathic pulmonary fibrosis (IPF). Data Sources: A search of PubMed (1966 to July 2014) was performed using the terms idiopathic pulmonary fibrosis and treatment. Study Selection and Data Extraction: Review of articles was restricted to articles in English and relating to placebo-controlled or comparative clinical trial data of recent significance. Evidence statements from the most recent international guidelines and some historical trial data were also included for context. Data Synthesis: Numerous treatment options have been evaluated for IPF. Therapies evaluated in large trials have either resulted in increased mortality (anticoagulation, triple-therapy with N-acetylcysteine [NAC], azathioprine, and prednisone) or demonstrated a lack of efficacy (endothelin receptor antagonists, single-agent NAC). Pirfenidone, a novel antifibrotic and anti-inflammatory agent, has demonstrated efficacy in several recent analyses and is the only approved medication for the treatment of IPF in more than 30 countries outside of the United States, with resubmission to the Food and Drug Administration (FDA) recently made. Nintedanib, a tyrosine kinase inhibitor, has demonstrated encouraging results in phase III studies and has also recently been submitted for FDA approval. Conclusions: Limited options have existed for the treatment of IPF. New evidence suggests that safe and efficacious treatment options for IPF are on the horizon in the form of pirfenidone and nintedanib, although both agents await FDA decisions.
Collapse
Affiliation(s)
- Jordan R. Covvey
- Duquesne University Mylan School of Pharmacy, Pittsburgh, PA, USA
| | | |
Collapse
|
47
|
Abstract
Of the deaths attributed to cancer, 90% are due to metastasis, and treatments that prevent or cure metastasis remain elusive. Emerging data indicate that hypoxia and the extracellular matrix (ECM) might have crucial roles in metastasis. During tumour evolution, changes in the composition and the overall content of the ECM reflect both its biophysical and biological properties and these strongly influence tumour and stromal cell properties, such as proliferation and motility. Originally thought of as independent contributors to metastatic spread, recent studies have established a direct link between hypoxia and the composition and the organization of the ECM, which suggests a new model in which multiple microenvironmental signals might converge to synergistically influence metastatic outcome.
Collapse
Affiliation(s)
- Daniele M Gilkes
- 1] Vascular Program, Institute for Cell Engineering, and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA. [2] Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Gregg L Semenza
- 1] Vascular Program, Institute for Cell Engineering, and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA. [2] Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, Maryland 21218, USA. [3] Departments of Pediatrics, Oncology, Medicine, Radiation Oncology and Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Denis Wirtz
- 1] Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, Maryland 21218, USA. [2] Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21218, USA. [3] Departments of Oncology and Pathology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
48
|
Weng CM, Yu CC, Kuo ML, Chen BC, Lin CH. Endothelin-1 induces connective tissue growth factor expression in human lung fibroblasts by ETAR-dependent JNK/AP-1 pathway. Biochem Pharmacol 2014; 88:402-11. [PMID: 24486572 DOI: 10.1016/j.bcp.2014.01.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 01/18/2014] [Accepted: 01/22/2014] [Indexed: 01/22/2023]
Abstract
Endothelin-1 (ET-1) acts as a key mediator of vasoconstriction and tissue repair. Overproduction of connective tissue growth factor (CTGF) underlies the development of lung fibrosis. ET-1 induces expression of matrix-associated genes in lung fibroblasts, however, little is known about the signaling pathway of CTGF expression caused by ET-1. In this study, we found that ET-1 caused concentration- and time-dependently increases in CTGF expression in human embryonic lung fibroblast cell line (WI-38). ET-1-induced CTGF expression was inhibited by BQ123 (ETAR antagonist), but not BQ788 (ETBR antagonist). Moreover, ET-1-induced CTGF expression was significantly reduced by JNK inhibitor (SP600125), the dominant-negative mutants of JNK1/2 (JNK1/2 DN), and AP-1 inhibitor (curcumin). ET-1 induced phosphorylations of JNK and c-Jun in time-dependent manners. AP-1 luciferase activity was concentration-dependently increased by ET-1, and this effect was attenuated by SP600125. We also found that ET-1-induced CTGF expression was most controlled by the AP-1 binding region of CTGF promoter. ET-1-indiced CTGF luciferase activity was predominately controlled by the sequence -747 to -408 bp upstream of the transcription start site on the human CTGF promoter. Furthermore, ET-1 caused the formation of AP-1-specific DNA-protein complex and the recruitment of c-Jun to the CTGF promoter. Moreover, we found that ET-1 induced α-smooth muscle actin (α-SMA) expression, which was inhibited by BQ123, SP600125, curcumin, and anti-CTGF antibody. These results suggest that ET-1 stimulates expressions of CTGF and α-SMA through ETAR/JNK/AP-1 signaling pathway, and CTGF is required for ET-1-induced α-SMA expression in human lung fibroblasts.
Collapse
Affiliation(s)
- Chih-Ming Weng
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chung-Chi Yu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Min-Liang Kuo
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; Graduate Institute of Biomedical Sciences, College of Life Science, National Taiwan University, Taipei 106, Taiwan
| | - Bing-Chang Chen
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei 11031, Taiwan.
| | - Chien-Huang Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
49
|
Weng CM, Chen BC, Wang CH, Feng PH, Lee MJ, Huang CD, Kuo HP, Lin CH. The Endothelin A Receptor Mediates Fibrocyte Differentiation in Chronic Obstructive Asthma. The Involvement of Connective Tissue Growth Factor. Am J Respir Crit Care Med 2013; 188:298-308. [DOI: 10.1164/rccm.201301-0132oc] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
50
|
Ahmedat AS, Warnken M, Seemann WK, Mohr K, Kostenis E, Juergens UR, Racké K. Pro-fibrotic processes in human lung fibroblasts are driven by an autocrine/paracrine endothelinergic system. Br J Pharmacol 2013; 168:471-87. [PMID: 22935082 DOI: 10.1111/j.1476-5381.2012.02190.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Revised: 07/27/2012] [Accepted: 08/08/2012] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Since endothelin (ET) may act as pro-fibrotic mediator, expression and release of ET isoforms, their receptors and potential pro-fibrotic ET effects were studied in human lung fibroblasts. EXPERIMENTAL APPROACH MRC-5 and primary human lung fibroblasts (phLFb) were cultured. Expression of prepro-ET isoforms was determined by qPCR and release of ET-1 by elisa. ET receptor function was analysed by real-time measurement of dynamic mass redistribution (DMR). Incorporation of [(3) H]-thymidine was determined as measure of proliferation and that of [(3) H]-proline for collagen synthesis. Phospho-p42/44 MAP kinase was determined by Western blot. KEY RESULTS ET-1 is the predominant ET in human lung fibroblasts (hLF), and TGF-β caused a further, selective and sustained up-regulation of ET-1 resulting in increased extracellular ET-1 accumulation. hLFb express mRNA encoding ET-A and ET-B receptors. Expression of both receptors was confirmed at protein level. ET-1 induced marked DMR signals, an effect that involved ET-A and ET-B receptors. Stimulatory effects of ET-1 on hLFb proliferation and collagen synthesis were mediated exclusively via ET-A receptors. ET-1, again via ET-A receptors, induced rapid activation of ERK MAPK, shown to be a crucial cellular signal in ET-1-induced collagen synthesis. ET-1-induced activation of ERK and collagen synthesis was, in contrast to corresponding effect of a muscarinic agonist, largely insensitive to pertussis toxin. CONCLUSIONS AND IMPLICATIONS hLFb are endowed with all elements necessary to build a functional autocrine/paracrine endothelinergic system, which appears to drive pro-fibrotic airway and lung remodelling processes, effects for which only ET-A, but not ET-B receptors appear to be of significance.
Collapse
Affiliation(s)
- A S Ahmedat
- Institute of Pharmacology & Toxicology, University of Bonn, Bonn, Germany
| | | | | | | | | | | | | |
Collapse
|