1
|
Subali D, Kurniawan R, Surya R, Lee IS, Chung S, Ko SJ, Moon M, Choi J, Park MN, Taslim NA, Hardinsyah H, Nurkolis F, Kim B, Kim KI. Revealing the mechanism and efficacy of natural products on treating the asthma: Current insights from traditional medicine to modern drug discovery. Heliyon 2024; 10:e32008. [PMID: 38882318 PMCID: PMC11176852 DOI: 10.1016/j.heliyon.2024.e32008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/18/2024] Open
Abstract
Asthma remains a significant global health challenge, demanding innovative approaches to treatment. Traditional medicine has a rich history of using natural products to alleviate asthmatic symptoms. However, transitioning from these traditional remedies to modern drug discovery approaches has provided fresh insights into the mechanisms and effectiveness of these natural products. This study provides our comprehensive review, which examines the current state of knowledge in the treatment of asthma. It delves into the mechanisms through which natural products ameliorate asthma symptoms, and it discusses their potential in the development of novel therapeutic interventions. Our analysis reveals that natural products, traditionally employed for asthma relief, exhibit diverse mechanisms of action. These include anti-inflammatory, bronchodilatory, immunomodulatory effects, and reducing gene expression. In the context of modern drug discovery, these natural compounds serve as valuable candidates for the development of novel asthma therapies. The transition from traditional remedies to modern drug discovery represents a promising avenue for asthma treatment. Our review highlights the substantial efficacy of natural products in managing asthma symptoms, underpinned by well-defined mechanisms of action. By bridging the gap between traditional and contemporary approaches, we contribute to the growing body of knowledge in the field, emphasizing the potential of natural products in shaping the future of asthma therapy.
Collapse
Affiliation(s)
- Dionysius Subali
- Department of Biotechnology, Faculty of Biotechnology, Atma Jaya Catholic University of Indonesia, Jakarta, 12930, Indonesia
| | - Rudy Kurniawan
- Diabetes Connection Care, Eka Hospital Bumi Serpong Damai, Tangerang, 15321, Indonesia
| | - Reggie Surya
- Department of Food Technology, Faculty of Engineering, Bina Nusantara University, Jakarta, 11480, Indonesia
| | - In-Seon Lee
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Acupuncture & Meridian Science Research Center, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Sanghyun Chung
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Kyung Hee Myungbo Clinic of Korean Medicine, Hwaseong-si, 18466, Republic of Korea
| | - Seok-Jae Ko
- Department of Gastroenterology, College of Korean Medicine, Kyung Hee University, Seoul, 05253, Republic of Korea
| | - Myunghan Moon
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jinwon Choi
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Nurpudji Astuti Taslim
- Division of Clinical Nutrition, Department of Nutrition, Faculty of Medicine, Hasanuddin University, Makassar, 90245, Indonesia
| | - Hardinsyah Hardinsyah
- Division of Applied Nutrition, Department of Community Nutrition, Faculty of Human Ecology, IPB University, Bogor, 16680, Indonesia
| | - Fahrul Nurkolis
- Department of Biological Sciences, Faculty of Sciences and Technology, State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta, 55281, Indonesia
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Kwan-Il Kim
- Division of Allergy, Immune and Respiratory System, Department of Internal Medicine, College of Korean Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
2
|
Ohm B, Moneke I, Jungraithmayr W. Targeting cluster of differentiation 26 / dipeptidyl peptidase 4 (CD26/DPP4) in organ fibrosis. Br J Pharmacol 2023; 180:2846-2861. [PMID: 36196001 DOI: 10.1111/bph.15967] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/07/2022] [Accepted: 09/29/2022] [Indexed: 11/28/2022] Open
Abstract
Cluster of differentiation 26 (CD26)/dipeptidyl peptidase 4 (DPP4) is an exopeptidase that is expressed as a transmembrane protein in many organs but also present in a circulating soluble form. Beyond its enzymatic and costimulatory activity, CD26/DPP4 is involved in the pathogenesis of chronic fibrotic diseases across many organ types, such as liver cirrhosis, kidney fibrosis and lung fibrosis. Organ fibrosis is associated with a high morbidity and mortality, and there are no causative therapies that can effectively attenuate the progress of the disease. Growing evidence suggests that inhibiting CD26/DPP4 can modulate the profibrotic tissue microenvironment and thus reduce fibrotic changes within affected organs. This review summarizes the role of CD26/DPP4 in fibroproliferative disorders and highlights new opportunities for an antifibrotic treatment by CD26/DPP4 inhibition. As a major advantage, CD26/DPP4 inhibitors have been in safe and routine clinical use in type 2 diabetes for many years and thus qualify for repurposing to repurpose as a promising therapeutic against fibrosis. LINKED ARTICLES: This article is part of a themed issue on Translational Advances in Fibrosis as a Therapeutic Target. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.22/issuetoc.
Collapse
Affiliation(s)
- Birte Ohm
- Department of Thoracic Surgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Isabelle Moneke
- Department of Thoracic Surgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wolfgang Jungraithmayr
- Department of Thoracic Surgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
3
|
Ortiz-Zapater E, Signes-Costa J, Montero P, Roger I. Lung Fibrosis and Fibrosis in the Lungs: Is It All about Myofibroblasts? Biomedicines 2022; 10:biomedicines10061423. [PMID: 35740444 PMCID: PMC9220162 DOI: 10.3390/biomedicines10061423] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 12/15/2022] Open
Abstract
In the lungs, fibrosis is a growing clinical problem that results in shortness of breath and can end up in respiratory failure. Even though the main fibrotic disease affecting the lung is idiopathic pulmonary fibrosis (IPF), which affects the interstitial space, there are many fibrotic events that have high and dangerous consequences for the lungs. Asthma, chronic obstructive pulmonary disease (COPD), excessive allergies, clearance of infection or COVID-19, all are frequent diseases that show lung fibrosis. In this review, we describe the different kinds of fibrosis and analyse the main types of cells involved-myofibroblasts and other cells, like macrophages-and review the main fibrotic mechanisms. Finally, we analyse present treatments for fibrosis in the lungs and highlight potential targets for anti-fibrotic therapies.
Collapse
Affiliation(s)
- Elena Ortiz-Zapater
- Department of Biochemistry and Molecular Biology, Faculty of Medicine-IIS INCLIVA, University of Valencia, 46010 Valencia, Spain
- Correspondence:
| | | | - Paula Montero
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (P.M.); (I.R.)
| | - Inés Roger
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (P.M.); (I.R.)
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, 28029 Madrid, Spain
| |
Collapse
|
4
|
Foster BM, Langsten KL, Mansour A, Shi L, Kerr BA. Tissue distribution of stem cell factor in adults. Exp Mol Pathol 2021; 122:104678. [PMID: 34450114 PMCID: PMC8516741 DOI: 10.1016/j.yexmp.2021.104678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/22/2021] [Accepted: 08/22/2021] [Indexed: 11/22/2022]
Abstract
Stem cell factor (SCF) is an essential cytokine during development and is necessary for gametogenesis, hematopoiesis, mast cell development, stem cell function, and melanogenesis. Here, we measure SCF concentration and distribution in adult humans and mice using gene expression analysis, tissue staining, and organ protein lysates. We demonstrate continued SCF expression in many cell types and tissues into adulthood. Tissues with high expression in adult humans included stomach, spleen, kidney, lung, and pancreas. In mice, we found high SCF expression in the esophagus, ovary, uterus, kidney, and small intestine. Future studies may correlate our findings of increased, organ-specific SCF concentrations within adult tissues with increased risk of SCF/CD117-related disease.
Collapse
Affiliation(s)
- Brittni M Foster
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States of America
| | - Kendall L Langsten
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States of America
| | - Ammar Mansour
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States of America
| | - Lihong Shi
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States of America
| | - Bethany A Kerr
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, United States of America; Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, United States of America.
| |
Collapse
|
5
|
Abstract
Abstract
Purpose of Review
In this brief review, we will highlight important observational and experimental data in the literature that address the origin of scar-forming cells in lung fibrosis.
Recent Findings
Several cellular sources of activated scar-forming cells (myofibroblasts) have been postulated including alveolar epithelial cells; circulating fibrocytes; and lung stromal cell subpopulations including resident fibroblasts, pericytes, and resident mesenchymal stem cells. Recent advances in lineage-tracing models, however, fail to provide experimental evidence for epithelial and fibrocyte origins of lung myofibroblasts. Resident mesenchymal cells of the lung, which include various cell types including resident fibroblasts, pericytes, and resident mesenchymal stem cells, appear to be important sources of myofibroblasts in murine models of lung injury and fibrosis.
Summary
Lung myofibroblasts likely originate from multiple sources of lung-resident mesenchymal cells. Their relative contributions may vary depending on the type of injury. Although lineage-tracing experiments have failed to show significant contribution from epithelial cells or fibrocytes, they may play important functional roles in myofibroblast activation through paracrine signaling.
Collapse
|
6
|
Rasky A, Habiel DM, Morris S, Schaller M, Moore BB, Phan S, Kunkel SL, Phillips M, Hogaboam C, Lukacs NW. Inhibition of the stem cell factor 248 isoform attenuates the development of pulmonary remodeling disease. Am J Physiol Lung Cell Mol Physiol 2019; 318:L200-L211. [PMID: 31747308 DOI: 10.1152/ajplung.00114.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Stem cell factor (SCF) and its receptor c-kit have been implicated in inflammation, tissue remodeling, and fibrosis. Ingenuity Integrated Pathway Analysis of gene expression array data sets showed an upregulation of SCF transcripts in idiopathic pulmonary fibrosis (IPF) lung biopsies compared with tissue from nonfibrotic lungs that are further increased in rapid progressive disease. SCF248, a cleavable isoform of SCF, was abundantly and preferentially expressed in human lung fibroblasts and fibrotic mouse lungs relative to the SCF220 isoform. In fibroblast-mast cell coculture studies, blockade of SCF248 using a novel isoform-specific anti-SCF248 monoclonal antibody (anti-SCF248), attenuated the expression of COL1A1, COL3A1, and FN1 transcripts in cocultured IPF but not normal lung fibroblasts. Administration of anti-SCF248 on days 8 and 12 after bleomycin instillation in mice significantly reduced fibrotic lung remodeling and col1al, fn1, acta2, tgfb, and ccl2 transcript expression. In addition, bleomycin increased numbers of c-kit+ mast cells, eosinophils, and ILC2 in lungs of mice, whereas they were not significantly increased in anti-SCF248-treated animals. Finally, mesenchymal cell-specific deletion of SCF significantly attenuated bleomycin-mediated lung fibrosis and associated fibrotic gene expression. Collectively, these data demonstrate that SCF is upregulated in diseased IPF lungs and blocking SCF248 isoform significantly ameliorates fibrotic lung remodeling in vivo suggesting that it may be a therapeutic target for fibrotic lung diseases.
Collapse
Affiliation(s)
- Andrew Rasky
- Department of Pathology, University of Michigan, Ann Arbor, Michigan.,Opsidio, LLC, Bryn Mawr, Pennsylvania
| | | | - Susan Morris
- Department of Pathology, University of Michigan, Ann Arbor, Michigan.,Opsidio, LLC, Bryn Mawr, Pennsylvania
| | - Matthew Schaller
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Bethany B Moore
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Sem Phan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Steven L Kunkel
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | | | - Cory Hogaboam
- Cedars-Sinai Medical Center, Los Angeles, California
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
7
|
Abstract
KIT is a receptor tyrosine kinase that after binding to its ligand stem cell factor activates signaling cascades linked to biological processes such as proliferation, differentiation, migration and cell survival. Based on studies performed on SCF and/or KIT mutant animals that presented anemia, sterility, and/or pigmentation disorders, KIT signaling was mainly considered to be involved in the regulation of hematopoiesis, gametogenesis, and melanogenesis. More recently, novel animal models and ameliorated cellular and molecular techniques have led to the discovery of a widen repertoire of tissue compartments and functions that are being modulated by KIT. This is the case for the lung, heart, nervous system, gastrointestinal tract, pancreas, kidney, liver, and bone. For this reason, the tyrosine kinase inhibitors that were originally developed for the treatment of hemato-oncological diseases are being currently investigated for the treatment of non-oncological disorders such as asthma, rheumatoid arthritis, and alzheimer's disease, among others. The beneficial effects of some of these tyrosine kinase inhibitors have been proven to depend on KIT inhibition. This review will focus on KIT expression and regulation in healthy and pathologic conditions other than cancer. Moreover, advances in the development of anti-KIT therapies, including tyrosine kinase inhibitors, and their application will be discussed.
Collapse
|
8
|
Aghasafari P, George U, Pidaparti R. A review of inflammatory mechanism in airway diseases. Inflamm Res 2018; 68:59-74. [PMID: 30306206 DOI: 10.1007/s00011-018-1191-2] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 09/12/2018] [Accepted: 09/27/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Inflammation in the lung is the body's natural response to injury. It acts to remove harmful stimuli such as pathogens, irritants, and damaged cells and initiate the healing process. Acute and chronic pulmonary inflammation are seen in different respiratory diseases such as; acute respiratory distress syndrome, chronic obstructive pulmonary disease (COPD), asthma, and cystic fibrosis (CF). FINDINGS In this review, we found that inflammatory response in COPD is determined by the activation of epithelial cells and macrophages in the respiratory tract. Epithelial cells and macrophages discharge transforming growth factor-β (TGF-β), which trigger fibroblast proliferation and tissue remodeling. Asthma leads to airway hyper-responsiveness, obstruction, mucus hyper-production, and airway-wall remodeling. Cytokines, allergens, chemokines, and infectious agents are the main stimuli that activate signaling pathways in epithelial cells in asthma. Mutation of the CF transmembrane conductance regulator (CFTR) gene results in CF. Mutations in CFTR influence the lung epithelial innate immune function that leads to exaggerated and ineffective airway inflammation that fails to abolish pulmonary pathogens. We present mechanistic computational models (based on ordinary differential equations, partial differential equations and agent-based models) that have been applied in studying the complex physiological and pathological mechanisms of chronic inflammation in different airway diseases. CONCLUSION The scope of the present review is to explore the inflammatory mechanism in airway diseases and highlight the influence of aging on airways' inflammation mechanism. The main goal of this review is to encourage research collaborations between experimentalist and modelers to promote our understanding of the physiological and pathological mechanisms that control inflammation in different airway diseases.
Collapse
Affiliation(s)
| | - Uduak George
- College of Engineering, University of Georgia, Athens, GA, USA.,Department of Mathematics and Statistics, San Diego State University, San Diego, CA, USA
| | | |
Collapse
|
9
|
Bagher M, Larsson-Callerfelt AK, Rosmark O, Hallgren O, Bjermer L, Westergren-Thorsson G. Mast cells and mast cell tryptase enhance migration of human lung fibroblasts through protease-activated receptor 2. Cell Commun Signal 2018; 16:59. [PMID: 30219079 PMCID: PMC6139170 DOI: 10.1186/s12964-018-0269-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 08/27/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Mast cells may activate fibroblasts and contribute to remodeling processes in the lung. However, the mechanism behind these actions needs to be further investigated. Fibroblasts are major regulators of on-going remodeling processes. Protease activated receptor 2 (PAR2) expressed by fibroblasts may be activated by serine proteases, such as the mast cell mediator tryptase. The objective in this study was to investigate the effects of mast cells and specifically mast cell tryptase on fibroblast migration and the role of PAR2 activation. METHODS Human lung fibroblasts (HFL-1) were cultured together with human peripheral blood-derived mast cells or LAD2 mast cells and stimulated with either conditioned medium from LAD2 cells or tryptase. Analyses of immunological stimulation of mast cells by IgE/anti IgE in the co-culture system were also performed. The importance of PAR2 activation by mast cells and mast cell tryptase for the migratory effects of fibroblasts was investigated by pre-treatment with the PAR2 antagonist P2pal-18S. The expression of PAR2 was analyzed on fibroblasts and mast cells. RESULTS The migratory capacity of HFL-1 cells was enhanced by blood-derived mast cells (p < 0.02), LAD2 cells (p < 0.001), conditioned medium (p < 0.05) and tryptase (p < 0.006). P2pal-18S decreased the induced migration caused by mast cells (p < 0.001) and tryptase (p < 0.001) and the expression of PAR2 was verified in HFL-1 cells. Mast cells immunologically stimulated with IgE/Anti IgE had no further effects on fibroblast migration. CONCLUSIONS Mast cells and the mast cell mediator tryptase may have crucial roles in inducing lung fibroblast migration via PAR-2 activation, which may contribute to remodeling processes in chronic lung diseases.
Collapse
Affiliation(s)
- Mariam Bagher
- Unit of Lung Biology, Department of Experimental Medical Sciences, Lund University, BMC C12, 221 84, Lund, Sweden. .,Department of Respiratory Medicine and Allergology, Skåne University Hospital, Lund University, Lund, Sweden.
| | | | - Oskar Rosmark
- Unit of Lung Biology, Department of Experimental Medical Sciences, Lund University, BMC C12, 221 84, Lund, Sweden
| | - Oskar Hallgren
- Department of Respiratory Medicine and Allergology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Leif Bjermer
- Department of Respiratory Medicine and Allergology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Gunilla Westergren-Thorsson
- Unit of Lung Biology, Department of Experimental Medical Sciences, Lund University, BMC C12, 221 84, Lund, Sweden
| |
Collapse
|
10
|
Kim SW, Kim JH, Park CK, Kim TJ, Lee SY, Kim YK, Kwon SS, Rhee CK, Yoon HK. Effect of roflumilast on airway remodelling in a murine model of chronic asthma. Clin Exp Allergy 2017; 46:754-63. [PMID: 26542330 DOI: 10.1111/cea.12670] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 09/16/2015] [Accepted: 10/17/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND Airway remodelling is associated with irreversible, or partially reversible, airflow obstruction and ultimately unresponsiveness to asthma therapies such as corticosteroids. Roflumilast is a selective phosphodiesterase-4 inhibitor that has an anti-inflammatory effect in chronic obstructive pulmonary disease (COPD). OBJECTIVE The objective of this study was to study the effect of roflumilast on airway inflammation and remodelling in a murine model of chronic asthma. METHODS BALB/c mice sensitized to ovalbumin (OVA) were chronically exposed to intranasal OVA administration twice a week for additional 3 months. Roflumilast was administered orally during the intranasal OVA challenge. A lung fibroblast cell line was used in the proliferation assay. RESULTS Compared with control mice, mice chronically exposed to OVA developed eosinophilic airway inflammation, airway hyper-responsiveness (AHR), and exhibited features of airway remodelling. Administration of roflumilast significantly inhibited airway inflammation and AHR. Roflumilast also significantly decreased goblet cell hyperplasia and pulmonary fibrosis, which are parameters of airway remodelling. The levels of interleukin (IL)-4, IL-5, and IL-13 in the bronchoalveolar lavage (BAL) fluids were significantly lower in the roflumilast group. In vitro, roflumilast significantly inhibited stem cell factor (SCF)-induced cell proliferation of fibroblasts. The SCF concentration and mRNA expression in a murine model also significantly decreased with roflumilast treatment. CONCLUSIONS These results suggest that the administration of roflumilast regulates airway inflammation, AHR, and airway remodelling in a model of chronic asthma. The beneficial effects from roflumilast may be related to the SCF/c-kit pathway.
Collapse
Affiliation(s)
- S W Kim
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - J H Kim
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - C K Park
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - T J Kim
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - S Y Lee
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Y K Kim
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - S S Kwon
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - C K Rhee
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - H K Yoon
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
11
|
Ngkelo A, Richart A, Kirk JA, Bonnin P, Vilar J, Lemitre M, Marck P, Branchereau M, Le Gall S, Renault N, Guerin C, Ranek MJ, Kervadec A, Danelli L, Gautier G, Blank U, Launay P, Camerer E, Bruneval P, Menasche P, Heymes C, Luche E, Casteilla L, Cousin B, Rodewald HR, Kass DA, Silvestre JS. Mast cells regulate myofilament calcium sensitization and heart function after myocardial infarction. J Exp Med 2017; 213:1353-74. [PMID: 27353089 PMCID: PMC4925026 DOI: 10.1084/jem.20160081] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 05/12/2016] [Indexed: 11/24/2022] Open
Abstract
Ngkelo et al. use a mast cell–deficient mouse model to reveal a protective role of mast cells in myocardial infarction, through regulation of the cardiac contractile machinery. Acute myocardial infarction (MI) is a severe ischemic disease responsible for heart failure and sudden death. Inflammatory cells orchestrate postischemic cardiac remodeling after MI. Studies using mice with defective mast/stem cell growth factor receptor c-Kit have suggested key roles for mast cells (MCs) in postischemic cardiac remodeling. Because c-Kit mutations affect multiple cell types of both immune and nonimmune origin, we addressed the impact of MCs on cardiac function after MI, using the c-Kit–independent MC-deficient (Cpa3Cre/+) mice. In response to MI, MC progenitors originated primarily from white adipose tissue, infiltrated the heart, and differentiated into mature MCs. MC deficiency led to reduced postischemic cardiac function and depressed cardiomyocyte contractility caused by myofilament Ca2+ desensitization. This effect correlated with increased protein kinase A (PKA) activity and hyperphosphorylation of its targets, troponin I and myosin-binding protein C. MC-specific tryptase was identified to regulate PKA activity in cardiomyocytes via protease-activated receptor 2 proteolysis. This work reveals a novel function for cardiac MCs modulating cardiomyocyte contractility via alteration of PKA-regulated force–Ca2+ interactions in response to MI. Identification of this MC-cardiomyocyte cross-talk provides new insights on the cellular and molecular mechanisms regulating the cardiac contractile machinery and a novel platform for therapeutically addressable regulators.
Collapse
Affiliation(s)
- Anta Ngkelo
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Adèle Richart
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Jonathan A Kirk
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD 212015
| | - Philippe Bonnin
- INSERM, U965, Hôpital Lariboisière-Fernand-Widal, Assistance Publique Hôpitaux de Paris, F-75010 Paris, France
| | - Jose Vilar
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Mathilde Lemitre
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Pauline Marck
- INSERM, UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, F-31004 Toulouse, France
| | - Maxime Branchereau
- INSERM, UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, F-31004 Toulouse, France
| | - Sylvain Le Gall
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Nisa Renault
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Coralie Guerin
- National Cytometry Platform, Department of Infection and Immunity, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg
| | - Mark J Ranek
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD 212015
| | - Anaïs Kervadec
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Luca Danelli
- Laboratoire d'Excellence INFLAMEX, Université Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France INSERM, U1149, F-75018 Paris, France Centre National de la Recherche Scientifique (CNRS) ERL 8252, F-75018 Paris, France
| | - Gregory Gautier
- Laboratoire d'Excellence INFLAMEX, Université Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France INSERM, U1149, F-75018 Paris, France
| | - Ulrich Blank
- Laboratoire d'Excellence INFLAMEX, Université Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France INSERM, U1149, F-75018 Paris, France Centre National de la Recherche Scientifique (CNRS) ERL 8252, F-75018 Paris, France
| | - Pierre Launay
- Laboratoire d'Excellence INFLAMEX, Université Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France INSERM, U1149, F-75018 Paris, France
| | - Eric Camerer
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Patrick Bruneval
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France Hôpital European George Pompidou, Assistance Publique Hôpitaux de Paris, F-75015 Paris, France
| | - Philippe Menasche
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France Hôpital European George Pompidou, Assistance Publique Hôpitaux de Paris, F-75015 Paris, France
| | - Christophe Heymes
- INSERM, UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, F-31004 Toulouse, France
| | - Elodie Luche
- STROMALab, Etablissement Français du Sang, INSERM U1031, CNRS ERL 5311, Université de Toulouse, F-31004 Toulouse, France
| | - Louis Casteilla
- STROMALab, Etablissement Français du Sang, INSERM U1031, CNRS ERL 5311, Université de Toulouse, F-31004 Toulouse, France
| | - Béatrice Cousin
- STROMALab, Etablissement Français du Sang, INSERM U1031, CNRS ERL 5311, Université de Toulouse, F-31004 Toulouse, France
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, D-69120 Heidelberg, Germany
| | - David A Kass
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD 212015
| | - Jean-Sébastien Silvestre
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| |
Collapse
|
12
|
Association of stem cell factor gene expression with severity and atopic state in patients with bronchial asthma. Respir Res 2017; 18:21. [PMID: 28100228 PMCID: PMC5241923 DOI: 10.1186/s12931-017-0504-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/12/2017] [Indexed: 12/21/2022] Open
Abstract
Background Bronchial asthma is a chronic inflammatory and remodeling disorder of the airways, in which many cells, cellular elements, and cytokines play important roles. Stem cell factor (SCF) may contribute to the inflammatory changes occurring in asthma. We aimed to show the expression of SCF gene in patients with asthma as a means of diagnosis and its association with severity and atopic state in these patients. Methods This study was carried out on 80 subjects, 50 asthmatic patients and 30 age and gender matched healthy control persons. They were subjected to full history taking, general and local chest examination, spirometric measurements (pre and post broncodilators) using a spirometer, serum IgE, and real time PCR for assessment of SCF mRNA expression. Results This study showed significant difference between the studied groups regarding pulmonary function tests (P < 0.001). Asthmatic patients had significant higher SCF expression compared to control (P < 0.001), also atopic patients vs non atopic (P = 0.03) and severe asthmatic patients vs mild ones (P < 0.001). SCF expression at cut off point (0.528) is sufficient to discriminate asthmatic patients from control while at cut off point (1.84) for discrimination of atopic patients from non-atopic patients and at cut off point (1.395) for discrimination of severe asthmatic patients from mild ones. A significant negative correlation between SCF expression and inhaled steroid while significant positive correlation with serum IgE was found. Conclusion Measuring SCF mRNA expression can be used as an efficient marker for evaluation of atopy and detection of severity of bronchial asthma.
Collapse
|
13
|
Serelaxin improves the therapeutic efficacy of RXFP1-expressing human amnion epithelial cells in experimental allergic airway disease. Clin Sci (Lond) 2016; 130:2151-2165. [PMID: 27647937 DOI: 10.1042/cs20160328] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/12/2016] [Indexed: 11/17/2022]
Abstract
Current asthma therapies primarily target airway inflammation (AI) and suppress episodes of airway hyperresponsiveness (AHR) but fail to treat airway remodelling (AWR), which can develop independently of AI and contribute to irreversible airway obstruction. The present study compared the anti-remodelling and therapeutic efficacy of human bone marrow-derived mesenchymal stem cells (MSCs) to that of human amnion epithelial stem cells (AECs) in the setting of chronic allergic airways disease (AAD), in the absence or presence of an anti-fibrotic (serelaxin; RLX). Female Balb/c mice subjected to the 9-week model of ovalbumin (OVA)-induced chronic AAD, were either vehicle-treated (OVA alone) or treated with MSCs or AECs alone [intranasally (i.n.)-administered with 1×106 cells once weekly], RLX alone (i.n.-administered with 0.8 mg/ml daily) or a combination of MSCs or AECs and RLX from weeks 9-11 (n=6/group). Measures of AI, AWR and AHR were then assessed. OVA alone exacerbated AI, epithelial damage/thickness, sub-epithelial extracellular matrix (ECM) and total collagen deposition, markers of collagen turnover and AHR compared with that in saline-treated counterparts (all P<0.01 compared with saline-treated controls). RLX or AECs (but not MSCs) alone normalized epithelial thickness and partially diminished the OVA-induced fibrosis and AHR by ∼40-50% (all P<0.05 compared with OVA alone). Furthermore, the combination treatments normalized epithelial thickness, measures of fibrosis and AHR to that in normal mice, and significantly decreased AI. Although AECs alone demonstrated greater protection against the AAD-induced AI, AWR and AHR, compared with that of MSCs alone, combining RLX with MSCs or AECs reversed airway fibrosis and AHR to an even greater extent.
Collapse
|
14
|
Royce SG, Shen M, Patel KP, Huuskes BM, Ricardo SD, Samuel CS. Mesenchymal stem cells and serelaxin synergistically abrogate established airway fibrosis in an experimental model of chronic allergic airways disease. Stem Cell Res 2015; 15:495-505. [PMID: 26426509 DOI: 10.1016/j.scr.2015.09.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/03/2015] [Accepted: 09/20/2015] [Indexed: 01/14/2023] Open
Abstract
This study determined if the anti-fibrotic drug, serelaxin (RLN), could augment human bone marrow-derived mesenchymal stem cell (MSC)-mediated reversal of airway remodeling and airway hyperresponsiveness (AHR) associated with chronic allergic airways disease (AAD/asthma). Female Balb/c mice subjected to the 9-week model of ovalbumin (OVA)-induced chronic AAD were either untreated or treated with MSCs alone, RLN alone or both combined from weeks 9-11. Changes in airway inflammation (AI), epithelial thickness, goblet cell metaplasia, transforming growth factor (TGF)-β1 expression, myofibroblast differentiation, subepithelial and total lung collagen deposition, matrix metalloproteinase (MMP) expression, and AHR were then assessed. MSCs alone modestly reversed OVA-induced subepithelial and total collagen deposition, and increased MMP-9 levels above that induced by OVA alone (all p<0.05 vs OVA group). RLN alone more broadly reversed OVA-induced epithelial thickening, TGF-β1 expression, myofibroblast differentiation, airway fibrosis and AHR (all p<0.05 vs OVA group). Combination treatment further reversed OVA-induced AI and airway/lung fibrosis compared to either treatment alone (all p<0.05 vs either treatment alone), and further increased MMP-9 levels. RLN appeared to enhance the therapeutic effects of MSCs in a chronic disease setting; most likely a consequence of the ability of RLN to limit TGF-β1-induced matrix synthesis complemented by the MMP-promoting effects of MSCs.
Collapse
Affiliation(s)
- Simon G Royce
- Fibrosis Laboratory, Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia.
| | - Matthew Shen
- Fibrosis Laboratory, Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Krupesh P Patel
- Fibrosis Laboratory, Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Brooke M Huuskes
- Kidney Regeneration and Stem Cell Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Sharon D Ricardo
- Kidney Regeneration and Stem Cell Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3800, Australia.
| | - Chrishan S Samuel
- Fibrosis Laboratory, Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
15
|
Gallagher KA, Joshi A, Carson WF, Schaller M, Allen R, Mukerjee S, Kittan N, Feldman EL, Henke PK, Hogaboam C, Burant CF, Kunkel SL. Epigenetic changes in bone marrow progenitor cells influence the inflammatory phenotype and alter wound healing in type 2 diabetes. Diabetes 2015; 64:1420-30. [PMID: 25368099 PMCID: PMC4375075 DOI: 10.2337/db14-0872] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Classically activated (M1) macrophages are known to play a role in the development of chronic inflammation associated with impaired wound healing in type 2 diabetes (T2D); however, the mechanism responsible for the dominant proinflammatory (M1) macrophage phenotype in T2D wounds is unknown. Since epigenetic enzymes can direct macrophage phenotypes, we assessed the role of histone methylation in bone marrow (BM) stem/progenitor cells in the programming of macrophages toward a proinflammatory phenotype. We have found that a repressive histone methylation mark, H3K27me3, is decreased at the promoter of the IL-12 gene in BM progenitors and this epigenetic signature is passed down to wound macrophages in a murine model of glucose intolerance (diet-induced obese). These epigenetically "preprogrammed" macrophages result in poised macrophages in peripheral tissue and negatively impact wound repair. We found that in diabetic conditions the H3K27 demethylase Jmjd3 drives IL-12 production in macrophages and that IL-12 production can be modulated by inhibiting Jmjd3. Using human T2D tissue and murine models, we have identified a previously unrecognized mechanism by which macrophages are programmed toward a proinflammatory phenotype, establishing a pattern of unrestrained inflammation associated with nonhealing wounds. Hence, histone demethylase inhibitor-based therapy may represent a novel treatment option for diabetic wounds.
Collapse
Affiliation(s)
| | - Amrita Joshi
- Department of Surgery, University of Michigan, Ann Arbor, MI
| | | | | | - Ronald Allen
- Department of Pathology, University of Michigan, Ann Arbor, MI
| | | | - Nico Kittan
- Department of Pathology, University of Michigan, Ann Arbor, MI
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Peter K Henke
- Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Cory Hogaboam
- Department of Medicine, Cedars-Sinai, Los Angeles, CA
| | | | - Steven L Kunkel
- Department of Pathology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
16
|
Bade G, Khan MA, Srivastava AK, Khare P, Solaiappan KK, Guleria R, Palaniyar N, Talwar A. Serum cytokine profiling and enrichment analysis reveal the involvement of immunological and inflammatory pathways in stable patients with chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis 2014; 9:759-73. [PMID: 25125975 PMCID: PMC4130715 DOI: 10.2147/copd.s61347] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major global health problem. It results from chronic inflammation and causes irreversible airway damage. Levels of different serum cytokines could be surrogate biomarkers for inflammation and lung function in COPD. We aimed to determine the serum levels of different biomarkers in COPD patients, the association between cytokine levels and various prognostic parameters, and the key pathways/networks involved in stable COPD. In this study, serum levels of 48 cytokines were examined by multiplex assays in 30 subjects (control, n=9; COPD, n=21). Relationships between serum biomarkers and forced expiratory volume in 1 second, peak oxygen uptake, body mass index, dyspnea score, and smoking were assessed. Enrichment pathways and network analyses were implemented, using a list of cytokines showing differential expression between healthy controls and patients with COPD by Cytoscape and GeneGo Metacore™ software (Thomson-Reuters Corporation, New York, NY, USA). Concentrations of cutaneous T-cell attracting chemokine, eotaxin, hepatocyte growth factor, interleukin 6 (IL-6), IL-16, and stem cell factor are significantly higher in COPD patients compared with in control patients. Notably, this study identifies stem cell factor as a biomarker for COPD. Multiple regression analysis predicts that cutaneous T-cell-attracting chemokine, eotaxin, IL-6, and stem cell factor are inversely associated with forced expiratory volume in 1 second and peak oxygen uptake change, whereas smoking is related to eotaxin and hepatocyte growth factor changes. Enrichment pathways and network analyses reveal the potential involvement of specific inflammatory and immune process pathways in COPD. Identified network interaction and regulation of different cytokines would pave the way for deeper insight into mechanisms of the disease process.
Collapse
Affiliation(s)
- Geetanjali Bade
- Department of Physiology, All India Institute of Medical Sciences, New Delhi, India
| | - Meraj Alam Khan
- Program in Physiology and Experimental Medicine, The Hospital for Sick Children, Department of Laboratory Medicine and Pathobiology, and Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Parul Khare
- Department of Physiology, All India Institute of Medical Sciences, New Delhi, India
| | | | - Randeep Guleria
- Department of Pulmonary Medicine and Sleep Disorders, All India Institute of Medical Sciences, New Delhi, India
| | - Nades Palaniyar
- Program in Physiology and Experimental Medicine, The Hospital for Sick Children, Department of Laboratory Medicine and Pathobiology, and Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Anjana Talwar
- Department of Physiology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
17
|
Mariñas‐Pardo L, Mirones I, Amor‐Carro Ó, Fraga‐Iriso R, Lema‐Costa B, Cubillo I, Rodríguez Milla MÁ, García‐Castro J, Ramos‐Barbón D. Mesenchymal stem cells regulate airway contractile tissue remodeling in murine experimental asthma. Allergy 2014; 69:730-40. [PMID: 24750069 PMCID: PMC4114550 DOI: 10.1111/all.12392] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2014] [Indexed: 01/10/2023]
Abstract
Background Mesenchymal stem cells may offer therapeutic potential for asthma due to their immunomodulatory properties and host tolerability, yet prior evidence suggests that bloodborne progenitor cells may participate in airway remodeling. Here, we tested whether mesenchymal stem cells administered as anti‐inflammatory therapy may favor airway remodeling and therefore be detrimental. Methods Adipose tissue‐derived mesenchymal stem cells were retrovirally transduced to express green fluorescent protein and intravenously injected into mice with established experimental asthma induced by repeat intranasal house dust mite extract. Controls were house dust mite‐instilled animals receiving intravenous vehicle or phosphate‐buffered saline‐instilled animals receiving mesenchymal stem cells. Data on lung function, airway inflammation, and remodeling were collected at 72 h after injection or after 2 weeks of additional intranasal challenge. Results The mesenchymal stem cells homed to the lungs and rapidly downregulated airway inflammation in association with raised T‐helper‐1 lung cytokines, but such effect declined under sustained allergen challenge despite a persistent presence of mesenchymal stem cells. Conversely, airway hyperresponsiveness and contractile tissue underwent a late reduction regardless of continuous pathogenic stimuli and inflammatory rebound. Tracking of green fluorescent protein did not show mesenchymal stem cell integration or differentiation in airway wall tissues. Conclusions Therapeutic mesenchymal stem cell infusion in murine experimental asthma is free of unwanted pro‐remodeling effects and ameliorates airway hyper‐responsiveness and contractile tissue remodeling. These outcomes support furthering the development of mesenchymal stem cell‐based asthma therapies, although caution and solid preclinical data building are warranted.
Collapse
Affiliation(s)
- L. Mariñas‐Pardo
- Respiratory Research Unit Instituto de Investigación Biomédica de A Coruña (INIBIC)/Complexo Hospitalario Universitario A Coruña Spain
- Cellular Biotechnology Unit Instituto de Salud Carlos III Madrid Spain
| | - I. Mirones
- Cellular Biotechnology Unit Instituto de Salud Carlos III Madrid Spain
| | - Ó. Amor‐Carro
- Respiratory Research Unit Instituto de Investigación Biomédica de A Coruña (INIBIC)/Complexo Hospitalario Universitario A Coruña Spain
- Respiratory Department Hospital de la Santa Creu i Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
| | - R. Fraga‐Iriso
- Respiratory Research Unit Instituto de Investigación Biomédica de A Coruña (INIBIC)/Complexo Hospitalario Universitario A Coruña Spain
- Respiratory Department Hospital de la Santa Creu i Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
| | - B. Lema‐Costa
- Respiratory Research Unit Instituto de Investigación Biomédica de A Coruña (INIBIC)/Complexo Hospitalario Universitario A Coruña Spain
| | - I. Cubillo
- Cellular Biotechnology Unit Instituto de Salud Carlos III Madrid Spain
| | | | - J. García‐Castro
- Cellular Biotechnology Unit Instituto de Salud Carlos III Madrid Spain
| | - D. Ramos‐Barbón
- Respiratory Research Unit Instituto de Investigación Biomédica de A Coruña (INIBIC)/Complexo Hospitalario Universitario A Coruña Spain
- Respiratory Department Hospital de la Santa Creu i Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
- Meakins‐Christie Laboratories Department of Medicine McGill University Montreal QCCanada
| |
Collapse
|
18
|
Unwith S, Zhao H, Hennah L, Ma D. The potential role of HIF on tumour progression and dissemination. Int J Cancer 2014; 136:2491-503. [PMID: 24729302 DOI: 10.1002/ijc.28889] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 03/07/2014] [Accepted: 04/03/2014] [Indexed: 12/20/2022]
Abstract
Cancer is the second cause of mortality worldwide, primarily owing to failure to cure metastatic disease. The need to target the metastatic process to reduce mortality is clear and research over the past decade has shown hypoxia-inducible factor-1 (HIF-1) to be one of the promising targets. In order for metastatic disease to be established, multiple steps need to be taken whereby the tumour cells escape into the bloodstream and survive, disseminate and then establish at a premetastatic niche. HIF-1 mediates hypoxia-induced proangiogenic factors such as vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF), which promote extravasation and chemotaxis. The migration of tumour cells is mediated by loss of E-cadherin, which results in a more invasive phenotype; dissemination of the tumour cells by increased vascular permeability and survival in the bloodstream through resistance to apoptosis as well as adhesion at the premetastatic niche are all controlled by factors under the influence of HIF-1. The overexpression of HIF in many aggressive cancer types as well as its role in the establishment of metastatic disease and treatment resistance demonstrate its potential target in therapeutics. Taken together, the role of HIF-1 in cancer and metastatic disease is clear and the need for better treatment targeting metastases is paramount; more aggressive phenotypes with less response to treatment are associated with HIF-1 expression. Our research has shown promise but many questions still remain to be answered.
Collapse
Affiliation(s)
- Sandeep Unwith
- Section of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and, Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, United Kingdom
| | | | | | | |
Collapse
|
19
|
Liu T, Yu H, Ullenbruch M, Jin H, Ito T, Wu Z, Liu J, Phan SH. The in vivo fibrotic role of FIZZ1 in pulmonary fibrosis. PLoS One 2014; 9:e88362. [PMID: 24516640 PMCID: PMC3916640 DOI: 10.1371/journal.pone.0088362] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 01/10/2014] [Indexed: 01/15/2023] Open
Abstract
FIZZ (found in inflammatory zone) 1, a member of a cysteine-rich secreted protein family, is highly induced in lung allergic inflammation and bleomycin induced lung fibrosis, and primarily expressed by airway and type II alveolar epithelial cells. This novel mediator is known to stimulate α-smooth muscle actin and collagen expression in lung fibroblasts. The objective of this study was to investigate the in vivo effects of FIZZ1 on the development of lung fibrosis by evaluating bleomycin-induced pulmonary fibrosis in FIZZ1 deficient mice. FIZZ1 knockout mice exhibited no detectable abnormality. When these mice were treated with bleomycin they exhibited significantly impaired pulmonary fibrosis relative to wild type mice, along with impaired proinflammatory cytokine/chemokine expression. Deficient lung fibroblast activation was also noted in the FIZZ1 knockout mice. Moreover, recruitment of bone marrow-derived cells to injured lung was deficient in FIZZ1 knockout mice. Interestingly in vitro FIZZ1 was shown to have chemoattractant activity for bone marrow cells, including bone marrow-derived dendritic cells. Finally, overexpression of FIZZ1 exacerbated fibrosis. These findings suggested that FIZZ1 exhibited profibrogenic properties essential for bleomycin induced pulmonary fibrosis, as reflected by its ability to induce myofibroblast differentiation and recruit bone marrow-derived cells.
Collapse
Affiliation(s)
- Tianju Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- * E-mail:
| | - Hongfeng Yu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Matthew Ullenbruch
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Hong Jin
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Toshihiro Ito
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Zhe Wu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Jianhua Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Sem H. Phan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| |
Collapse
|
20
|
Overed-Sayer C, Rapley L, Mustelin T, Clarke DL. Are mast cells instrumental for fibrotic diseases? Front Pharmacol 2014; 4:174. [PMID: 24478701 PMCID: PMC3896884 DOI: 10.3389/fphar.2013.00174] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 12/20/2013] [Indexed: 01/17/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal lung disorder of unknown etiology characterized by accumulation of lung fibroblasts and extracellular matrix deposition, ultimately leading to compromised tissue architecture and lung function capacity. IPF has a heterogeneous clinical course; however the median survival after diagnosis is only 3–5 years. The pharmaceutical and biotechnology industry has made many attempts to find effective treatments for IPF, but the disease has so far defied all attempts at therapeutic intervention. Clinical trial failures may arise for many reasons, including disease heterogeneity, lack of readily measurable clinical end points other than overall survival, and, perhaps most of all, a lack of understanding of the underlying molecular mechanisms of the progression of IPF. The precise link between inflammation and fibrosis remains unclear, but it appears that immune cells can promote fibrosis by releasing fibrogenic factors. So far, however, therapeutic approaches targeting macrophages, neutrophils, or lymphocytes have failed to alter disease pathogenesis. A new cell to garner research interest in fibrosis is the mast cell. Increased numbers of mast cells have long been known to be present in pulmonary fibrosis and clinically correlations between mast cells and fibrosis have been reported. More recent data suggests that mast cells may contribute to the fibrotic process by stimulating fibroblasts resident in the lung, thus driving the pathogenesis of the disease. In this review, we will discuss the mast cell and its physiological role in tissue repair and remodeling, as well as its pathological role in fibrotic diseases such as IPF, where the process of tissue repair and remodeling is thought to be dysregulated.
Collapse
Affiliation(s)
| | - Laura Rapley
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune Ltd Cambridge, UK
| | - Tomas Mustelin
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune Ltd Cambridge, UK
| | - Deborah L Clarke
- Department of Respiratory, Inflammation and Autoimmunity, MedImmune Ltd Cambridge, UK
| |
Collapse
|
21
|
Fischer KD, Agrawal DK. Hematopoietic stem and progenitor cells in inflammation and allergy. Front Immunol 2013; 4:428. [PMID: 24363657 PMCID: PMC3849597 DOI: 10.3389/fimmu.2013.00428] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 11/20/2013] [Indexed: 11/13/2022] Open
Abstract
Hematopoietic stem and progenitor cells contribute to allergic inflammation. Pro-inflammatory cytokines that are generated following allergen challenge can impact the differentiation of hematopoietic progenitor cells leading to increased production of effector cells such as eosinophils and basophils, which are key cells involved in the pathogenesis of allergic airway inflammation. Homing of stem cells to the lungs is associated with inflammatory and remodeling changes in asthmatics. Factors that modulate the differentiation and increased migration of stem cells to the site of inflammation in asthma remain to be defined. Stem cells can mature at the site of inflammation in response to inflammatory mediators and other components in the milieu. While the available data suggest that hematopoietic cells traffic to target tissues, the molecular factors underlying in situ differentiation have yet to be specified. Here, we critically evaluate the potential role of hematopoietic progenitors in contributing to the increased immune cell infiltrate in allergic asthma and the factors that drive their differentiation.
Collapse
Affiliation(s)
- Kimberly D Fischer
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine , Omaha, NE , USA
| | - Devendra K Agrawal
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine , Omaha, NE , USA ; Department of Biomedical Sciences, Creighton University School of Medicine , Omaha, NE , USA ; Department of Internal Medicine, Creighton University School of Medicine , Omaha, NE , USA ; Center for Clinical and Translational Science, Creighton University School of Medicine , Omaha, NE , USA
| |
Collapse
|
22
|
Nonaka K, Miyazawa M, Ban S, Aikawa M, Akimoto N, Koyama I, Kita H. Different healing process of esophageal large mucosal defects by endoscopic mucosal dissection between with and without steroid injection in an animal model. BMC Gastroenterol 2013; 13:72. [PMID: 23617935 PMCID: PMC3652745 DOI: 10.1186/1471-230x-13-72] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 04/24/2013] [Indexed: 02/07/2023] Open
Abstract
Background Stricture formation is one of the major complications after endoscopic removal of large superficial squamous cell neoplasms of the esophagus, and local steroid injections have been adopted to prevent it. However, fundamental pathological alterations related to them have not been well analyzed so far. The aim of this study was to analyze the time course of the healing process of esophageal large mucosal defects resulting in stricture formation and its modification by local steroid injection, using an animal model. Methods Esophageal circumferential mucosal defects were created by endoscopic mucosal dissection (ESD) for four pigs. One pig was sacrificed five minutes after the ESD, and other two pigs were followed-up on endoscopy and sacrificed at the time of one week and three weeks after the ESD, respectively. The remaining one pig was followed-up on endoscopy with five times of local steroid injection and sacrificed at the time of eight weeks after the ESD. The esophageal tissues of all pigs were subjected to pathological analyses. Results For the pigs without steroid injection, the esophageal stricture was completed around three weeks after the ESD on both endoscopy and esophagography. Histopathological examination of the esophageal tissues revealed that spindle-shaped α-smooth muscle actin (SMA)-positive myofibroblasts arranged in a parallel fashion and extending horizontally were identified at the ulcer bed one week after the ESD, and increased contributing to formation of the stenotic luminal ridge covered with the regenerated epithelium three weeks after the ESD. The proper muscle layer of the stricture site was thinned with some myocytes which seemingly showed transition to the myofibroblast layer. By contrast, for the pig with steroid injection, esophageal stricture formation was not evident with limited appearance of the spindle-shaped myofibroblasts, instead, appearance of stellate or polygocal SMA-positive stromal cells arranged haphazardly in the persistent granulation tissue of the ulcer site. Conclusions Proliferation of spindle-shaped myofibroblasts arranged in a parallel fashion is likely to play an important role in stricture formation after circumferential mucosal defects by esophageal ESD, which may be related to the thinning of the proper muscle layer in the healing course of the defects. Local steroid injection seems to be effective to prevent the stricture through the modification of this process.
Collapse
Affiliation(s)
- Kouichi Nonaka
- Department of Gastroenterology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, 350-1298, Japan
| | | | | | | | | | | | | |
Collapse
|
23
|
Ding L, Dolgachev V, Wu Z, Liu T, Nakashima T, Wu Z, Ullenbruch M, Lukacs NW, Chen Z, Phan SH. Essential role of stem cell factor-c-Kit signalling pathway in bleomycin-induced pulmonary fibrosis. J Pathol 2013; 230:205-14. [PMID: 23401096 DOI: 10.1002/path.4177] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 01/22/2013] [Accepted: 02/01/2013] [Indexed: 01/13/2023]
Abstract
Stem cell factor (SCF) and its receptor c-Kit have been implicated in tissue remodelling and fibrosis. Alveolar fibroblasts from patients with diffuse interstitial fibrosis secrete more SCF. However, its precise role remains unclear. In this study the potential role of the SCF-c-Kit axis in pulmonary fibrosis was examined. Fibrosis was induced by intratracheal instillation of bleomycin (BLM), which caused increased SCF levels in plasma, bronchoalveolar lavage fluid (BALF) and lung tissue, as well as increased expression by lung fibroblasts. These changes were accompanied by increased numbers of bone marrow-derived c-Kit(+) cells in the lung, with corresponding depletion in bone marrow. Both recombinant SCF and lung extracts from BLM-treated animals induced bone-marrow cell migration, which was blocked by c-Kit inhibitor. The migrated cells promoted myofibroblast differentiation when co-cultured with fibroblasts, suggesting a paracrine pathogenic role. Interestingly, lung fibroblast cultures contained a subpopulation of cells that expressed functionally active c-Kit, which were significantly greater and more responsive to SCF induction when isolated from fibrotic lungs, including those from patients with idiopathic pulmonary fibrosis (IPF). This c-Kit(+) subpopulation was αSMA-negative and expressed lower levels of collagen I but significantly higher levels of TGFβ than c-Kit-negative cells. SCF deficiency achieved by intratracheal treatment with neutralizing anti-SCF antibody or by use of Kitl(Sl)/Kitl(Sl-d) mutant mice in vivo resulted in significant reduction in pulmonary fibrosis. Taken together, the SCF-c-Kit pathway was activated in BLM-injured lung and might play a direct role in pulmonary fibrosis by the recruitment of bone marrow progenitor cells capable of promoting lung myofibroblast differentiation.
Collapse
Affiliation(s)
- Lin Ding
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Implication for bone marrow derived stem cells in hepatocyte regeneration after orthotopic liver transplantation. Int J Hepatol 2013; 2013:310612. [PMID: 24109514 PMCID: PMC3784276 DOI: 10.1155/2013/310612] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 08/05/2013] [Accepted: 08/12/2013] [Indexed: 12/15/2022] Open
Abstract
The liver has the outstanding ability to regenerate itself and restore parenchymal tissue after injury. The most common cell source in liver growth/regeneration is replication of preexisting hepatocytes although liver progenitor cells have been postulated to participate in liver regeneration in cases of massive injury. Bone marrow derived hematopoietic stem cells (BM-HSC) have the formal capacity to act as a source for hepatic regeneration under special circumstances; however, the impact of this process in liver tissue maintenance and regeneration remains controversial. Whether BM-HSC are involved in liver regeneration or not would be of particular interest as the cells have been suggested to be an alternative donor source for the treatment of liver failure. Data from murine models of liver disease show that BM-HSC can repopulate liver tissue and restore liver function; however, data obtained from human liver transplantation show only little evidence for liver regeneration by this mechanism. The cell source for liver regeneration seems to depend on the nature of regeneration process and the extent of injury; however, the precise mechanisms still need to be resolved. Current data suggest, that in human orthotopic liver transplantation, liver regeneration by BM-HSC is a rather rare event and therefore not of clinical relevance.
Collapse
|
25
|
Paredes BD, Faccioli LAP, Quintanilha LF, Asensi KD, do Valle CZ, Canary PC, Takiya CM, de Carvalho ACC, Goldenberg RCDS. Bone marrow progenitor cells do not contribute to liver fibrogenic cells. World J Hepatol 2012; 4:274-83. [PMID: 23293712 PMCID: PMC3537161 DOI: 10.4254/wjh.v4.i10.274] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 10/24/2012] [Accepted: 10/26/2012] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the contribution of bone marrow (BM) cells to hepatic fibrosis. METHODS To establish a model of chimerism, C57Bl/6 female mice were subjected to full-body irradiation (7 Gy) resulting in BM myeloablation. BM mononuclear cells obtained from male transgenic mice expressing enhanced green fluorescent protein (GFP) were used for reconstitution. Engraftment was confirmed by flow cytometry. To induce liver injury, chimeric animals received carbon tetrachloride (CCl(4)) 0.5 mL/kg intraperitoneally twice a week for 30 d (CCl(4) 30 d) and age-matched controls received saline (Saline 30 d). At the end of this period, animals were sacrificed for post mortem analysis. Liver samples were stained with hematoxylin and eosin to observe liver architectural changes and with Sirius red for collagen quantification by morphometric analysis. α-smooth muscle actin (α-SMA) was analyzed by confocal microscopy to identify GFP+ cells with myofibroblast (MF) characteristics. Liver tissue, BM and peripheral blood were collected and prepared for flow cytometric analysis using specific markers for detection of hepatic stellate cells (HSCs) and precursors from the BM. RESULTS Injury to the liver induced changes in the hepatic parenchymal architecture, as reflected by the presence of inflammatory infiltrate and an increase in collagen deposition (Saline 30 d = 11.10% ± 1.12% vs CCl(4) 30 d = 12.60% ± 0.73%, P = 0.0329). Confocal microscopy revealed increased reactivity against α-SMA in CCl(4) 30 d compared to Saline 30 d, but there was no co-localization with GFP+ cells, suggesting that cells from BM do not differentiate to MFs. Liver flow cytometric analysis showed a significant increase of CD45+/GFP+ cells in liver tissue (Saline 30 d = 3.2% ± 2.2% vs CCl(4) 30 d = 5.8% ± 1.3%, P = 0.0458), suggesting that this increase was due to inflammatory cell infiltration (neutrophils and monocytes). There was also a significant increase of common myeloid progenitor cells (CD117+/CD45+) in the livers of CCl(4)-treated animals (Saline 30 d = 2.16% ± 1.80% vs CCl(4) 30 d = 5.60% ± 1.30%, P = 0.0142). In addition the GFP-/CD38+/CD45- subpopulation was significantly increased in the CCl(4) 30 d group compared to the Saline 30 d group (17.5% ± 3.9% vs 9.3% ± 2.4%, P = 0.004), indicating that the increase in the activated HSC subpopulation was not of BM origin. CONCLUSION BM progenitor cells do not contribute to fibrosis, but there is a high recruitment of inflammatory cells that stimulates HSCs and MFs of liver origin.
Collapse
Affiliation(s)
- Bruno Diaz Paredes
- Bruno Diaz Paredes, Lanuza Alaby Pinheiro Faccioli, Luiz Fernando Quintanilha, Karina Dutra Asensi, Camila Zaverucha do Valle, Christina Maeda Takiya, Antonio Carlos Campos de Carvalho, Regina Coeli dos Santos Goldenberg, Carlos Chagas Filho Biophysics Institute, Rio de Janeiro 21941-902, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
The potential use of tyrosine kinase inhibitors in severe asthma. Curr Opin Allergy Clin Immunol 2012; 12:68-75. [PMID: 22157153 DOI: 10.1097/aci.0b013e32834ecb4f] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE OF REVIEW Severe asthma comprises heterogeneous phenotypes that share in common a poor response to traditional therapies. Recent and ongoing work with tyrosine kinase inhibitors suggests a potential beneficial role in treatment of severe asthma. RECENT FINDINGS Various receptor and nonreceptor tyrosine kinase pathways contribute to aspects of airway inflammation, airway hyperresponsiveness, and remodeling of asthma. Selective and nonselective tyrosine kinase inhibitors may be useful to block pathways that are pathologically overactive or overexpressed in severe asthma. Recent in-vivo studies have demonstrated the utility of inhibitors against specific tyrosine kinases (epidermal growth factor receptor, c-kit/platelet derived growth factor receptor, vascular endothelial growth factor receptor, spleen tyrosine kinase, and janus kinase) in altering key aspects of severe asthma. SUMMARY Asthma and even severe asthma does not consist of a single phenotype. Targeting key inflammatory and remodeling pathways engaged across subphenotypes with tyrosine kinase inhibitors appears to hold promise.
Collapse
|
27
|
Phan SH. Genesis of the myofibroblast in lung injury and fibrosis. PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY 2012; 9:148-52. [PMID: 22802289 PMCID: PMC5830705 DOI: 10.1513/pats.201201-011aw] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 02/23/2012] [Indexed: 02/07/2023]
Abstract
Tissue injury incites a repair response with a key mesenchymal component that provides the essential connective tissue for subsequent regeneration or pathological fibrosis. The fibroblast is the major mesenchymal cell type to be implicated in this connective tissue response, and it is in its activated or differentiated form that it participates in the repair process. The myofibroblast represents such an activated mesenchymal cell and is a key source of extracellular matrix and inflammatory/fibrogenic cytokines as well as participating in wound contraction. Although successful healing results in gradual disappearance of myofibroblasts, their persistence is associated with chronic and progressive fibrosis. Thus, elucidation of the mechanism involved in the genesis of the myofibroblast should provide insight into both pathogenesis of chronic fibrotic diseases and therapeutic strategies for their management and control. Although the fibroblast is a well-documented progenitor cell for the myofibroblast, recent studies have suggested additional precursor cells that have the potential to give rise to the myofibroblast. Many of the studies focused on mechanisms and factors that regulate induction of α-smooth muscle actin expression, a key and commonly used marker of the myofibroblast. These reveal complex and multifactorial mechanisms involving transcriptional and epigenetic regulation and implicating diverse cell-signaling pathways, including those activated by the potent fibrogenic cytokine transforming growth factor β. Despite these extensive studies, many aspects remain poorly understood, with the suggestion that additional novel mechanisms remain to be discovered. Future studies with the help of newly developed technical advancements should expedite discovery in this direction.
Collapse
Affiliation(s)
- Sem H Phan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA.
| |
Collapse
|
28
|
Xu X, Huang H, Cai M, Qian Y, Han Y, Xiao L, Zhou W, Wang X, Shi B. Serum hematopoietic growth factors as diagnostic and prognostic markers of acute renal allograft rejection: A potential role for serum stem cell factor. Cytokine 2011; 56:779-85. [DOI: 10.1016/j.cyto.2011.09.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 08/31/2011] [Accepted: 09/19/2011] [Indexed: 12/23/2022]
|
29
|
Walker N, Badri L, Wettlaufer S, Flint A, Sajjan U, Krebsbach PH, Keshamouni VG, Peters-Golden M, Lama VN. Resident tissue-specific mesenchymal progenitor cells contribute to fibrogenesis in human lung allografts. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:2461-9. [PMID: 21641374 DOI: 10.1016/j.ajpath.2011.01.058] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Revised: 01/20/2011] [Accepted: 01/28/2011] [Indexed: 01/08/2023]
Abstract
Fibrotic obliteration of the small airways leading to progressive airflow obstruction, termed bronchiolitis obliterans syndrome (BOS), is the major cause of poor outcomes after lung transplantation. We recently demonstrated that a donor-derived population of multipotent mesenchymal stem cells (MSCs) can be isolated from the bronchoalveolar lavage (BAL) fluid of human lung transplant recipients. Herein, we study the organ specificity of these cells and investigate the role of local mesenchymal progenitors in fibrogenesis after lung transplantation. We demonstrate that human lung allograft-derived MSCs uniquely express embryonic lung mesenchyme-associated transcription factors with a 35,000-fold higher expression of forkhead/winged helix transcription factor forkhead box (FOXF1) noted in lung compared with bone marrow MSCs. Fibrotic differentiation of MSCs isolated from normal lung allografts was noted in the presence of profibrotic mediators associated with BOS, including transforming growth factor-β and IL-13. MSCs isolated from patients with BOS demonstrated increased expression of α-SMA and collagen I when compared with non-BOS controls, consistent with a stable in vivo fibrotic phenotype. FOXF1 mRNA expression in the BAL cell pellet correlated with the number of MSCs in the BAL fluid, and myofibroblasts present in the fibrotic lesions expressed FOXF1 by in situ hybridization. These data suggest a key role for local tissue-specific, organ-resident, mesenchymal precursors in the fibrogenic processes in human adult lungs.
Collapse
Affiliation(s)
- Natalie Walker
- Division of Pulmonary and Critical Care, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Schwarz J. Emerging role of c-kit+ progenitor cells in pulmonary hypertension. Am J Respir Crit Care Med 2011; 184:5-7. [PMID: 21737590 DOI: 10.1164/rccm.201104-0664ed] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|
31
|
Knight DA, Rossi FM, Hackett TL. Mesenchymal stem cells for repair of the airway epithelium in asthma. Expert Rev Respir Med 2011; 4:747-58. [PMID: 21128750 DOI: 10.1586/ers.10.72] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The airway epithelium is constantly faced with inflammatory and potentially injurious stimuli. Following damage, rapid repair mechanisms involving proliferation and differentiation of resident progenitor and stem cell pools are necessary in order to maintain a protective barrier. In asthma, evidence pointing to a compromised ability of the epithelium to properly repair and regenerate is rapidly accumulating. The consequences of this are presently unknown but are likely to have a significant impact on lung function. Mesenchymal stem cells have the potential to serve as a universal source for replacement of specific cells in several diseases and thus offer hope as a potential therapeutic intervention for the treatment of the chronic remodeling changes that occur in the asthmatic epithelium. However, controversy exists regarding whether these cells can actually home to and engraft within the airways and contribute to tissue function or whether this mechanism is necessary, since they can have potent paracrine immunomodulatory effects. This article focuses on the current knowledge about specific stem cell populations that may contribute to airway epithelial regeneration and discusses the use of mesenchymal stem cells as a potential therapeutic intervention.
Collapse
Affiliation(s)
- Darryl A Knight
- Providence Heart and Lung Institute at St Paul's Hospital, University of British Columbia, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada.
| | | | | |
Collapse
|
32
|
Rhee CK, Kim JW, Park CK, Kim JS, Kang JY, Kim SJ, Kim SC, Kwon SS, Kim YK, Park SH, Lee SY. Effect of imatinib on airway smooth muscle thickening in a murine model of chronic asthma. Int Arch Allergy Immunol 2011; 155:243-51. [PMID: 21293142 DOI: 10.1159/000321261] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Accepted: 09/15/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Asthma is characterized by airway hyperresponsiveness (AHR), inflammation and remodeling. The tyrosine kinase inhibitor imatinib mesylate was developed to inhibit BCR-ABL kinase activity; however, it also has potent inhibitory activity against the c-Kit and platelet-derived growth factor receptors. The present study aimed to determine whether imatinib suppresses airway smooth muscle (ASM) remodeling and whether its effect is associated with growth factors such as transforming growth factor (TGF)-β1 and stem cell factor (SCF). METHODS We developed a mouse model of airway remodeling, which includes smooth muscle thickening, in which ovalbumin (OVA)-sensitized mice were repeatedly exposed to intranasal OVA administration twice a week for 3 months. Mice were treated with imatinib during the OVA challenge. RESULTS Mice chronically exposed to OVA developed sustained eosinophilic airway inflammation and AHR compared with control mice. In addition, the mice chronically exposed to OVA developed features of airway remodeling, including thickening of the peribronchial smooth muscle layer. Administration of imatinib significantly inhibited the development of AHR, eosinophilic inflammation and, importantly, ASM remodeling in mice chronically exposed to OVA. Imatinib treatment significantly reduced the levels of interleukin-4, -5 and -13. In addition, TGF-β1 and SCF were significantly reduced in the imatinib-treated animals. CONCLUSIONS These results suggest that imatinib administration can prevent not only airway inflammation, but also airway remodeling associated with chronic allergen challenge. Imatinib may provide a clinically attractive therapy for chronic severe asthma.
Collapse
Affiliation(s)
- Chin Kook Rhee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Platelet-activating factor receptor plays a role in lung injury and death caused by Influenza A in mice. PLoS Pathog 2010; 6:e1001171. [PMID: 21079759 PMCID: PMC2974216 DOI: 10.1371/journal.ppat.1001171] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 09/29/2010] [Indexed: 01/03/2023] Open
Abstract
Influenza A virus causes annual epidemics which affect millions of people
worldwide. A recent Influenza pandemic brought new awareness over the health
impact of the disease. It is thought that a severe inflammatory response against
the virus contributes to disease severity and death. Therefore, modulating the
effects of inflammatory mediators may represent a new therapy against Influenza
infection. Platelet activating factor (PAF) receptor (PAFR) deficient mice were
used to evaluate the role of the gene in a model of experimental infection with
Influenza A/WSN/33 H1N1 or a reassortant Influenza A H3N1 subtype. The following
parameters were evaluated: lethality, cell recruitment to the airways, lung
pathology, viral titers and cytokine levels in lungs. The PAFR antagonist
PCA4248 was also used after the onset of flu symptoms. Absence or antagonism of
PAFR caused significant protection against flu-associated lethality and lung
injury. Protection was correlated with decreased neutrophil recruitment, lung
edema, vascular permeability and injury. There was no increase of viral load and
greater recruitment of NK1.1+ cells. Antibody responses were
similar in WT and PAFR-deficient mice and animals were protected from
re-infection. Influenza infection induces the enzyme that synthesizes PAF,
lyso-PAF acetyltransferase, an effect linked to activation of TLR7/8. Therefore,
it is suggested that PAFR is a disease-associated gene and plays an important
role in driving neutrophil influx and lung damage after infection of mice with
two subtypes of Influenza A. Further studies should investigate whether
targeting PAFR may be useful to reduce lung pathology associated with Influenza
A virus infection in humans. Influenza virus causes disease that affects people from different age, gender or
social conditions. The illness spreads easily and affects millions of people
every year. Vaccines are effective preventive approaches, but the high degree of
viral antigenic drift requires annual formulation. Anti-viral drugs are used as
therapy, but are only effective at the very early stages of disease. The main
symptoms that lead to hospitalizations and deaths are associated with the severe
inflammatory host immune response triggered by the virus infection. Our approach
was to decrease the inflammatory events associated with the viral infection by
targeting a molecule, Platelet Activating Factor receptor (PAFR), known to
induce several inflammatory events, including leukocyte recruitment and leakage.
We found that PAFR deficient mice or wild type mice treated with a PAFR
antagonist had less pulmonary inflammation, pulmonary injury and lethality rates
when infected by two subtypes of Influenza A virus. In contrast, the immune
response against the virus, as assessed by viral loads and specific antibodies,
were not decreased. Our findings concur with the idea that severe inflammation
plays an important role in flu morbidity and mortality and show that PAFR is a
major driver of the exacerbated inflammation in mice infected with Influenza A
virus.
Collapse
|
34
|
Abstract
Mesenchymal stem cells (MSCs) have been shown to differentiate into a variety of mesenchymal cell types, including fibroblasts, myofibroblasts, osteoblasts, chondroblasts, adipocytes, and myoblasts, as well as epithelial cells. It has been shown that these cells can be recovered from bone marrow as well as umbilical cord blood, and they can be propagated, stored, and administered to animals and patients in clinical trials. It is clear that the cells engraft in the lung, and several laboratories have demonstrated an ameliorating effect in models of acute injury caused by LPS and in chronic lung injury induced by bleomycin and asbestos. However, it is not at all clear under what conditions these cells must be applied to provide an advantage and when using these cells might cause exacerbation of the lung injury. This brief review focuses on the biology of MSCs in vitro, how the cells have been used in some animal models, and the potential for their use in therapeutic strategies for diseases as diverse as lung cancer and interstitial fibrosis.
Collapse
|
35
|
Abstract
There has been an increased understanding, over the past 2 decades, that asthma is a chronic, immunologically mediated condition with a disturbance of the normal airway repair mechanism, which results in inflammatory changes and airway remodeling. The airway inflammation and remodeling together likely explain the clinical manifestations of asthma. The mechanisms by which the external environmental cues, together with the complex genetic actions, propagate the inflammatory process that characterize asthma are beginning to be understood. There is also an evolving awareness of the active participation of structural elements, such as the airway epithelium, airway smooth muscle, and endothelium, in this process. In tandem with this has come the realization that inflammatory cells respond in a coordinated, albeit dysfunctional manner, via an array of complex signaling pathways that facilitate communication between these cells; these structural elements within the lung and the bone marrow serve as reservoirs for and the source of inflammatory cells and their precursors. Although often viewed as separate mechanistic entities, so-called innate and acquired immunity often overlap in the propagation of the asthmatic response. This review examines the newer information on the pathophysiologic characteristics of asthma and focuses on papers published over the past 3 years that have helped to improve current levels of understanding.
Collapse
Affiliation(s)
- Desmond M Murphy
- Firestone Institute for Respiratory Health, St Joseph's Healthcare, McMaster University, Hamilton, ON, Canada
| | | |
Collapse
|
36
|
Miller JD, Lankford SM, Adler KB, Brody AR. Mesenchymal stem cells require MARCKS protein for directed chemotaxis in vitro. Am J Respir Cell Mol Biol 2010; 43:253-8. [PMID: 20224071 DOI: 10.1165/rcmb.2010-0015rc] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) reside within tissues such as bone marrow, cord blood, and dental pulp and can differentiate into other mesenchymal cell types. Differentiated MSCs, called circulating fibrocytes, have been demonstrated in human lungs and migrate to injured lung tissue in experimental models. It is likely that MSCs migrate from the bone marrow to sites of injury by following increasing chemokine concentrations. In the present study, we show that primary mouse bone marrow mesenchymal stem cells (BM-MSCs) exhibit directed chemotaxis through transwell inserts toward increasing concentrations of the chemokines complement component 5a, stromal cell-derived factor-1alpha, and monocyte chemotactic protein-1. Prior research has indicated that myristoylated alanine-rich C kinase substrate (MARCKS) protein is critically important for motility in macrophages, neutrophils, and fibroblasts, and here we investigated a possible role for MARCKS in BM-MSC directed chemotaxis. The presence of MARCKS in these cells as well as in human cord blood MSC was verified by Western blotting, and MARCKS was rapidly phosphorylated in these cells after exposure to chemokines. A synthetic peptide that inhibits MARCKS function attenuated, in a concentration-dependent manner, directed chemotaxis of BM-MSCs, while a missense control peptide had no effect. Our results illustrate, for the first time, that MARCKS protein plays an integral role in BM-MSC-directed chemotaxis in vitro.
Collapse
Affiliation(s)
- Jeffrey D Miller
- Dept. of Molecular Biomedical Sciences, NC State University, Raleigh, 27606, USA
| | | | | | | |
Collapse
|
37
|
Dolgachev V, Petersen BC, Budelsky AL, Berlin AA, Lukacs NW. Pulmonary IL-17E (IL-25) production and IL-17RB+ myeloid cell-derived Th2 cytokine production are dependent upon stem cell factor-induced responses during chronic allergic pulmonary disease. THE JOURNAL OF IMMUNOLOGY 2009; 183:5705-15. [PMID: 19828636 DOI: 10.4049/jimmunol.0901666] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In the present studies local neutralization of allergen-induced stem cell factor (SCF) leads to decreased production of Th2 cytokines, a reduction in inflammation, allergen-specific serum IgE/IgG1, and attenuation of severe asthma-like responses. The local blockade of pulmonary SCF also resulted in a significant reduction of IL-17E (IL-25). Sorted cell populations from the lung indicated that IL-25 was produced from c-kit(+) cells, whereas Th2 cytokine production was primarily from c-kit(-) cell populations. SCF stimulated c-kit(+) eosinophils produced IL-25, whereas bone marrow-derived mast cells did not. Using 4get mice that contain a IL-4-IRES-eGFP that when transcribed coexpress GFP and IL-4, our studies identified cells that comprised a CD11b(+), GR1(+), Ly6C(+/-), c-kit(-), CD4(-), CD11c(-), MHC class II(low) cell population as a source of IL-4 in the lung after chronic allergen challenge. In the bone marrow a similar cell was identified with approximately a third of the IL-4(+) cells also expressing c-kit(+). The pulmonary and bone marrow IL-4(+) cell populations were significantly reduced upon local pulmonary anti-SCF treatment. Subsequently, when IL-25R was examined during the chronic allergen responses the expression was found on the IL-4(+) myeloid cell population that expressed CD11b(+)GR1(+). Interestingly, the IL-25R(+) cells in the bone marrow were also all CD11b(+)GR1(+), similar to the lung cells, but they were also all c-kit(+), potentially suggesting a maturation of the bone marrow cell once it enters the lung and/or is stimulated by SCF. Overall, these studies suggest a complex relationship between SCF, bone marrow-derived IL-25-responsive myeloid cells, Th2 cytokines, and chronic allergic disease.
Collapse
Affiliation(s)
- Vladislav Dolgachev
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | | | |
Collapse
|
38
|
Frid MG, Li M, Gnanasekharan M, Burke DL, Fragoso M, Strassheim D, Sylman JL, Stenmark KR. Sustained hypoxia leads to the emergence of cells with enhanced growth, migratory, and promitogenic potentials within the distal pulmonary artery wall. Am J Physiol Lung Cell Mol Physiol 2009; 297:L1059-72. [PMID: 19767409 DOI: 10.1152/ajplung.90611.2008] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
All forms of chronic pulmonary hypertension (PH) are characterized by structural remodeling of the pulmonary artery (PA) media, a process previously attributed solely to changes in the phenotype of resident smooth muscle cells (SMC). However, recent experimental evidence in both systemic and pulmonary circulations suggests that other cell types, including circulating and local progenitors, contribute significantly to this process. The goal of this study was to determine if hypoxia-induced remodeling of distal PA (dPA) media involves the emergence of cells with phenotypic and functional characteristics distinct from those of resident dPA SMC and fibroblasts. In vivo, in contrast to the phenotypically uniform SMC composition of dPA media in control calves, the remodeled dPA media of neonatal calves with severe hypoxia-induced PH comprised cells exhibiting a distinct phenotype, including the expression of hematopoetic (CD45), leukocytic/monocytic (CD11b, CD14), progenitor (cKit), and motility-associated (S100A4) cell markers. Consistent with these in vivo observations, primary cell cultures isolated from dPA media of hypertensive calves yielded not only differentiated SMC, but also smaller, morphologically rhomboidal (thus termed here "R") cells that transiently expressed CD11b, constitutively expressed the mesenchymal cell marker type I procollagen, expressed high mRNA levels of progenitor cell markers cKit, CD34, CD73, as well as for inflammatory mediators, IL-6 and MCP-1, and, with time in culture, gained expression of a myofibroblast marker, alpha-SM-actin. R cells exhibited highly augmented proliferative, migratory, invasive, and potent promitogenic capabilities, which were due, at least in part, to the production of PDGFs, SDF-1/CXCL12, and S100A4. These data suggest that the cellular mechanisms of dPA remodeling include the emergence of cells with phenotypic and functional characteristics markedly distinct from those of resident dPA cells.
Collapse
Affiliation(s)
- Maria G Frid
- Department of Pediatrics, University of Colorado Denver, Aurora, 80045, USA.
| | | | | | | | | | | | | | | |
Collapse
|