1
|
Abu-Helo A, Daubeuf F, Tranchant T, Lehalle C, Elhabazi K, Zeder-Lutz G, Kugler V, Lugnier C, Frossard N, Lecat S, Simonin F. GPRASP1 deletion in mice abrogates adverse side effects associated with chronic stimulation of Beta2-adrenoceptor. Biomed Pharmacother 2025; 187:118073. [PMID: 40286598 DOI: 10.1016/j.biopha.2025.118073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 04/02/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
GPCR associated sorting protein 1 (GPRASP1) interacts with numerous GPCRs including the Beta2-adrenoceptor (B2AR) and has been proposed to be involved in adaptations associated with chronic stimulation of those receptors. In clinic, long acting B2AR agonists (LABAs) such as formoterol are used in the treatment of asthma as potent bronchodilators but with adverse side effects including the development of tolerance and airway hyperresponsiveness upon chronic administration. Here, we investigated the role of GPRASP1 on B2AR activity and on B2AR agonists-associated side effects in vitro and in vivo. To this purpose, we set-up a model of chronic formoterol administration in mouse leading to B2AR down-regulation as well as to the development of airway hyperreactivity and bronchodilator tolerance and studied the phenotype of GPRASP1 knockout animals. We show in cells that GPRASP1 expression has no impact on agonist-induced B2AR down-regulation but strongly modulate B2AR-associated signalling. Moreover, wild-type mice chronically treated with formoterol developed airway hyperresponsiveness to methacholine and bronchodilator tolerance to formoterol that were absent in GPRASP1 KO mice while B2AR down-regulation was similar in both genotypes. These adverse side effects were correlated with an increase in the number of cells and in collagen levels in the lungs of wild-type but not of GPRASP1 KO mice. Collectively, our data show that GPRASP1 is critically involved in adaptations to chronic activation of B2AR that leads to lung tissue remodelling, development of bronchial hyperresponsiveness and bronchodilator tolerance to B2AR agonist formoterol and could therefore represent an interesting target to limit side effects associated with LABAs.
Collapse
Affiliation(s)
- Alaa Abu-Helo
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| | - François Daubeuf
- Plateforme de Chimie Biologique Intégrative de Strasbourg, UAR 3286, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| | - Thibaud Tranchant
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| | - Christine Lehalle
- Plateforme de Chimie Biologique Intégrative de Strasbourg, UAR 3286, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| | - Khadija Elhabazi
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France
| | - Gabrielle Zeder-Lutz
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| | - Valérie Kugler
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| | - Claire Lugnier
- Section de Structures Biologiques, Pharmacologie et Enzymologie, CNRS/Unistra, CRBS, UR, Strasbourg 3072, France.
| | - Nelly Frossard
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| | - Sandra Lecat
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| | - Frédéric Simonin
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, Illkirch, France.
| |
Collapse
|
2
|
Tan C, Zhou H, Xiong Q, Xian X, Liu Q, Zhang Z, Xu J, Yao H. Cromolyn sodium reduces LPS-induced pulmonary fibrosis by inhibiting the EMT process enhanced by MC-derived IL-13. Respir Res 2025; 26:3. [PMID: 39762844 PMCID: PMC11706190 DOI: 10.1186/s12931-024-03045-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 11/17/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Sepsis is a systemic inflammatory response caused by infection. When this inflammatory response spreads to the lungs, it can lead to acute lung injury (ALI) or more severe acute respiratory distress syndrome (ARDS). Pulmonary fibrosis is a potential complication of these conditions, and the early occurrence of pulmonary fibrosis is associated with a higher mortality rate. The underlying mechanism of ARDS-related pulmonary fibrosis remains unclear. METHODS To evaluate the role of mast cell in sepsis-induced pulmonary fibrosis and elucidate its molecular mechanism. We investigated the level of mast cell and epithelial-mesenchymal transition(EMT) in LPS-induced mouse model and cellular model. We also explored the influence of cromolyn sodium and mast cell knockout on pulmonary fibrosis. Additionally, we explored the effect of MC-derived IL-13 on the EMT and illustrated the relationship between mast cell and pulmonary fibrosis. RESULTS Mast cell was up-regulated in the lung tissues of the pulmonary fibrotic mouse model compared to control groups. Cromolyn sodium and mast cell knockout decreased the expression of EMT-related protein and IL-13, alleviated the symptoms of pulmonary fibrosis in vivo and in vitro. The PI3K/AKT/mTOR signaling was activated in fibrotic lung tissue, whereas Cromolyn sodium and mast cell knockout inhibited this pathway. CONCLUSION The expression level of mast cell is increased in fibrotic lungs. Cromolyn sodium intervention and mast cell knockout alleviate the symptoms of pulmonary fibrosis probably via the PI3K/AKT/mTOR signaling pathway. Therefore, mast cell inhibition is a potential therapeutic target for sepsis-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Cheng Tan
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Wuxi, 214002, Jiangsu Province, China
| | - Hang Zhou
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, 210011, Jiangsu Province, China
| | - Qiangfei Xiong
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, 210011, Jiangsu Province, China
| | - Xian Xian
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, 210011, Jiangsu Province, China
| | - Qiyuan Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, 210011, Jiangsu Province, China
| | - Zexin Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, 210011, Jiangsu Province, China
| | - Jingjing Xu
- Department of Anesthesiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Wuxi, 214002, Jiangsu Province, China.
| | - Hao Yao
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, 210011, Jiangsu Province, China.
| |
Collapse
|
3
|
Atiakshin D, Kostin A, Alekhnovich A, Volodkin A, Ignatyuk M, Klabukov I, Baranovskii D, Buchwalow I, Tiemann M, Artemieva M, Medvedeva N, LeBaron TW, Noda M, Medvedev O. The Role of Mast Cells in the Remodeling Effects of Molecular Hydrogen on the Lung Local Tissue Microenvironment under Simulated Pulmonary Hypertension. Int J Mol Sci 2024; 25:11010. [PMID: 39456794 PMCID: PMC11507233 DOI: 10.3390/ijms252011010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Molecular hydrogen (H2) has antioxidant, anti-inflammatory, and anti-fibrotic effects. In a rat model simulating pulmonary fibrotic changes induced by monocrotaline-induced pulmonary hypertension (MPH), we had previously explored the impact of inhaled H2 on lung inflammation and blood pressure. In this study, we further focused the biological effects of H2 on mast cells (MCs) and the parameters of the fibrotic phenotype of the local tissue microenvironment. MPH resulted in a significantly increased number of MCs in both the pneumatic and respiratory parts of the lungs, an increased number of tryptase-positive MCs with increased expression of TGF-β, activated interaction with immunocompetent cells (macrophages and plasma cells) and fibroblasts, and increased MC colocalization with a fibrous component of the extracellular matrix of connective tissue. The alteration in the properties of the MC population occurred together with intensified collagen fibrillogenesis and an increase in the integral volume of collagen and elastic fibers of the extracellular matrix of the pulmonary connective tissue. The exposure of H2 together with monocrotaline (MCT), despite individual differences between animals, tended to decrease the intrapulmonary MC population and the severity of the fibrotic phenotype of the local tissue microenvironment compared to changes in animals exposed to the MCT effect alone. In addition, the activity of collagen fibrillogenesis associated with MCs and the expression of TGF-β and tryptase in MCs decreased, accompanied by a reduction in the absolute and relative content of reticular and elastic fibers in the lung stroma. Thus, with MCT exposure, inhaled H2 has antifibrotic effects involving MCs in the lungs of rats. This reveals the unknown development mechanisms of the biological effects of H2 on the remodeling features of the extracellular matrix under inflammatory background conditions of the tissue microenvironment.
Collapse
Affiliation(s)
- Dmitrii Atiakshin
- RUDN University, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (A.K.); (A.A.); (A.V.); (M.I.); (I.B.); (M.N.); (O.M.)
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Andrey Kostin
- RUDN University, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (A.K.); (A.A.); (A.V.); (M.I.); (I.B.); (M.N.); (O.M.)
| | - Alexander Alekhnovich
- RUDN University, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (A.K.); (A.A.); (A.V.); (M.I.); (I.B.); (M.N.); (O.M.)
| | - Artem Volodkin
- RUDN University, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (A.K.); (A.A.); (A.V.); (M.I.); (I.B.); (M.N.); (O.M.)
| | - Michael Ignatyuk
- RUDN University, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (A.K.); (A.A.); (A.V.); (M.I.); (I.B.); (M.N.); (O.M.)
| | - Ilya Klabukov
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva st. 4, 249036 Obninsk, Russia (D.B.)
| | - Denis Baranovskii
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva st. 4, 249036 Obninsk, Russia (D.B.)
| | - Igor Buchwalow
- RUDN University, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (A.K.); (A.A.); (A.V.); (M.I.); (I.B.); (M.N.); (O.M.)
- Institute for Hematopathology, Fangdieckstr. 75a, 22547 Hamburg, Germany;
| | - Markus Tiemann
- Institute for Hematopathology, Fangdieckstr. 75a, 22547 Hamburg, Germany;
| | - Marina Artemieva
- Faculty of Biology, Lomonosov Moscow State University, Leninskie Gory 1-12, 119234 Moscow, Russia; (M.A.); (N.M.)
| | - Nataliya Medvedeva
- Faculty of Biology, Lomonosov Moscow State University, Leninskie Gory 1-12, 119234 Moscow, Russia; (M.A.); (N.M.)
| | - Tyler W. LeBaron
- Department of Kinesiology and Outdoor Recreation, Southern Utah University, Cedar City, UT 84720, USA;
- Molecular Hydrogen Institute, Cedar City, UT 84720, USA
| | - Mami Noda
- RUDN University, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (A.K.); (A.A.); (A.V.); (M.I.); (I.B.); (M.N.); (O.M.)
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science, Xi’an Jiaotong University, Xi’an 710049, China
| | - Oleg Medvedev
- RUDN University, 6 Miklukho-Maklaya St, 117198 Moscow, Russia; (A.K.); (A.A.); (A.V.); (M.I.); (I.B.); (M.N.); (O.M.)
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Prospect 27-1, 119991 Moscow, Russia
| |
Collapse
|
4
|
Naia Fioretto M, Maciel FA, Barata LA, Ribeiro IT, Basso CBP, Ferreira MR, Dos Santos SAA, Mattos R, Baptista HS, Portela LMF, Padilha PM, Felisbino SL, Scarano WR, Zambrano E, Justulin LA. Impact of maternal protein restriction on the proteomic landscape of male rat lungs across the lifespan. Mol Cell Endocrinol 2024; 592:112348. [PMID: 39218056 DOI: 10.1016/j.mce.2024.112348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
The developmental origins of healthy and disease (DOHaD) concept has demonstrated a higher rate of chronic diseases in the adult population of individuals whose mothers experienced severe maternal protein restriction (MPR). Using proteomic and in silico analyses, we investigated the lung proteomic profile of young and aged rats exposed to MPR during pregnancy and lactation. Our results demonstrated that MPR lead to structural and immune system pathways changes, and this outcome is coupled with a rise in the PI3k-AKT-mTOR signaling pathway, with increased MMP-2 activity, and CD8 expression in the early life, with long-term effects with aging. This led to the identification of commonly or inversely differentially expressed targets in early life and aging, revealing dysregulated pathways related to the immune system, stress, muscle contraction, tight junctions, and hemostasis. We identified three miRNAs (miR-378a-3p, miR-378a-5p, let-7a-5p) that regulate four proteins (ACTN4, PPIA, HSPA5, CALM1) as probable epigenetic lung marks generated by MPR. In conclusion, MPR impacts the lungs early in life, increasing the possibility of long-lasting negative outcomes for respiratory disorders in the offspring.
Collapse
Affiliation(s)
- Matheus Naia Fioretto
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Flávia Alessandra Maciel
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luísa Annibal Barata
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Isabelle Tenori Ribeiro
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Carolina Beatriz Pinheiro Basso
- Molecular Genetics and Bioinformatics Laboratory, Experimental Research Unit - Unipex, School of Medicine, São Paulo State University - Unesp, Botucatu, São Paulo, Brazil
| | - Marcel Rodrigues Ferreira
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Sérgio Alexandre Alcantara Dos Santos
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil; Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, 19111, USA
| | - Renato Mattos
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Hecttor Sebastian Baptista
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luiz Marcos Frediane Portela
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Pedro Magalhães Padilha
- Department of Chemical and Biological Sciences, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Sérgio Luis Felisbino
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Wellerson Rodrigo Scarano
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Elena Zambrano
- Department Reproductive Biology, Salvador Zubirán National Institute of Medical Sciences and Nutrition, Mexico City, Mexico; Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Luis Antonio Justulin
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil.
| |
Collapse
|
5
|
Genovese T, Duranti A, Monaco F, Siracusa R, Fusco R, Impellizzeri D, D’Amico R, Cordaro M, Cuzzocrea S, Di Paola R. Inhibition of Fatty Acid Amide Hydrolase (FAAH) Regulates NF-kb Pathways Reducing Bleomycin-Induced Chronic Lung Inflammation and Pulmonary Fibrosis. Int J Mol Sci 2023; 24:10125. [PMID: 37373275 PMCID: PMC10298572 DOI: 10.3390/ijms241210125] [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: 04/26/2023] [Revised: 06/09/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
The deadly interstitial lung condition known as idiopathic pulmonary fibrosis (IPF) worsens over time and for no apparent reason. The traditional therapy approaches for IPF, which include corticosteroids and immunomodulatory drugs, are often ineffective and can have noticeable side effects. The endocannabinoids are hydrolyzed by a membrane protein called fatty acid amide hydrolase (FAAH). Increasing endogenous levels of endocannabinoid by pharmacologically inhibiting FAAH results in numerous analgesic advantages in a variety of experimental models for pre-clinical pain and inflammation. In our study, we mimicked IPF by administering intratracheal bleomycin, and we administered oral URB878 at a dose of 5 mg/kg. The histological changes, cell infiltration, pro-inflammatory cytokine production, inflammation, and nitrosative stress caused by bleomycin were all reduced by URB878. Our data clearly demonstrate for the first time that the inhibition of FAAH activity was able to counteract not only the histological alteration bleomycin-induced but also the cascade of related inflammatory events.
Collapse
Affiliation(s)
- Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy (R.S.); (D.I.)
| | - Andrea Duranti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Piazza del Rinascimento, 6, 61029 Urbino, Italy
| | - Francesco Monaco
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98166 Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy (R.S.); (D.I.)
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy (R.S.); (D.I.)
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy (R.S.); (D.I.)
| | - Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy (R.S.); (D.I.)
| | - Marika Cordaro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98166 Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy (R.S.); (D.I.)
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy;
| |
Collapse
|
6
|
Atiakshin D, Kostin A, Volodkin A, Nazarova A, Shishkina V, Esaulenko D, Buchwalow I, Tiemann M, Noda M. Mast Cells as a Potential Target of Molecular Hydrogen in Regulating the Local Tissue Microenvironment. Pharmaceuticals (Basel) 2023; 16:817. [PMID: 37375765 DOI: 10.3390/ph16060817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/18/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Knowledge of the biological effects of molecular hydrogen (H2), hydrogen gas, is constantly advancing, giving a reason for the optimism in several healthcare practitioners regarding the management of multiple diseases, including socially significant ones (malignant neoplasms, diabetes mellitus, viral hepatitis, mental and behavioral disorders). However, mechanisms underlying the biological effects of H2 are still being actively debated. In this review, we focus on mast cells as a potential target for H2 at the specific tissue microenvironment level. H2 regulates the processing of pro-inflammatory components of the mast cell secretome and their entry into the extracellular matrix; this can significantly affect the capacity of the integrated-buffer metabolism and the structure of the immune landscape of the local tissue microenvironment. The analysis performed highlights several potential mechanisms for developing the biological effects of H2 and offers great opportunities for translating the obtained findings into clinical practice.
Collapse
Affiliation(s)
- Dmitri Atiakshin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia Named after Patrice Lumumba, 117198 Moscow, Russia
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Andrey Kostin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia Named after Patrice Lumumba, 117198 Moscow, Russia
| | - Artem Volodkin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia Named after Patrice Lumumba, 117198 Moscow, Russia
| | - Anna Nazarova
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia Named after Patrice Lumumba, 117198 Moscow, Russia
| | - Viktoriya Shishkina
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Dmitry Esaulenko
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Igor Buchwalow
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia Named after Patrice Lumumba, 117198 Moscow, Russia
- Institute for Hematopathology, Fangdieckstr. 75a, 22547 Hamburg, Germany
| | - Markus Tiemann
- Institute for Hematopathology, Fangdieckstr. 75a, 22547 Hamburg, Germany
| | - Mami Noda
- Laboratory of Pathophysiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 816-0811, Japan
| |
Collapse
|
7
|
St John AL, Rathore APS, Ginhoux F. New perspectives on the origins and heterogeneity of mast cells. Nat Rev Immunol 2023; 23:55-68. [PMID: 35610312 DOI: 10.1038/s41577-022-00731-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 01/06/2023]
Abstract
Mast cells are immune cells of the haematopoietic lineage that are now thought to have multifaceted functions during homeostasis and in various disease states. Furthermore, while mast cells have been known for a long time to contribute to allergic disease in adults, recent studies, mainly in mice, have highlighted their early origins during fetal development and potential for immune functions, including allergic responses, in early life. Our understanding of the imprinting of mast cells by particular tissues of residence and their potential for regulatory interactions with organ systems such as the peripheral immune, nervous and vascular systems is also rapidly evolving. Here, we discuss the origins of mast cells and their diverse and plastic phenotypes that are influenced by tissue residence. We explore how divergent phenotypes and functions might result from both their hard-wired 'nature' defined by their ontogeny and the 'nurture' they receive within specialized tissue microenvironments.
Collapse
Affiliation(s)
- Ashley L St John
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.
- Department of Pathology, Duke University Medical Center, Durham, NC, USA.
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- SingHealth Duke-NUS Global Health Institute, Singapore, Singapore.
| | - Abhay P S Rathore
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Florent Ginhoux
- Singapore Immunology Network, A*STAR, Singapore, Singapore.
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.
| |
Collapse
|
8
|
Toyoshima S, Okayama Y. Neuro-allergology: Mast cell-nerve cross-talk. Allergol Int 2022; 71:288-293. [PMID: 35688775 DOI: 10.1016/j.alit.2022.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 11/01/2022] Open
Abstract
Mast cells (MCs) are derived from hematopoietic stem cells in the bone marrow, and their maturation is regulated by the tissue environment, such as the skin, lung and gut, leading to host defense. Peripheral nerve fibers located in various tissues are involved in diverse physiological and pathological processes. Anatomical relationships between MCs and nerve fibers were reported to have been observed in various organs. Moreover, MCs are positive for a large number of receptors for classical neurotransmitters (e.g., acetylcholine and corticotropin-releasing hormone) and neuropeptides (e.g., substance P, calcitonin gene-related peptides and hemokinin), and MC's functions are regulated by those nerve-derived factors. Also, histamine and proteases produced and released by MCs modulate nerve fiber functions. This functional cross-talk between MCs and nerve fibers can play physiological and pathological roles. MCs are key effector cells of allergic inflammation, such as atopic dermatitis, airway inflammation and food allergy. Here, we summarize and discuss the molecular mechanisms underlying the functional and anatomical cross-talk between MCs and nerve fibers in allergic inflamed tissues.
Collapse
Affiliation(s)
- Shota Toyoshima
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan; Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Yoshimichi Okayama
- Allergy and Immunology Research Project Team, Research Institute of Medical Science, Center for Medical Education, Nihon University School of Medicine, Tokyo, Japan; Center for Allergy, Nihon University Itabashi Hospital, Tokyo, Japan.
| |
Collapse
|
9
|
Tanaka S, Furuta K. Roles of IgE and Histamine in Mast Cell Maturation. Cells 2021; 10:cells10082170. [PMID: 34440939 PMCID: PMC8392195 DOI: 10.3390/cells10082170] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 11/16/2022] Open
Abstract
Mast cells are activated upon immunoglobulin E (IgE)-mediated antigen stimulation, and release a wide variety of mediators, including histamine to trigger inflammatory responses. The surface expression levels of Fcε receptor I (FcεRI), a high affinity receptor of IgE, were found to be positively regulated by IgE. IgE could protect murine cultured mast cells from apoptotic cell death induced by the deprivation of interleukin-3 and a certain kind of IgE could activate immature mast cells in the absence of antigens, leading to the release of pro-inflammatory cytokines and a transient increase in histamine synthesis. Histamine synthesis in mast cells was found to be required for the maturation of murine connective tissue-type mast cells, raising the possibility that IgE indirectly modulates local mast cell maturation. Although it remains controversial to what extent this concept of "monomeric IgE effects" could have relevance in the modulation of human mast cell functions, the therapeutic effects of anti-IgE antibodies might be accounted for in terms of the decreased serum IgE concentrations. Because drastic increases in serum IgE concentrations are often observed in patients with atopic dermatitis and chronic urticaria, a close investigation of the roles of IgE in mast cell maturation should contribute to development of novel therapeutic approaches for these inflammatory diseases.
Collapse
Affiliation(s)
- Satoshi Tanaka
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Misasagi Nakauchi-cho 5, Yamashina-ku, Kyoto 607-8414, Japan
- Correspondence: ; Tel.: +81-75-595-4667
| | - Kazuyuki Furuta
- Department of Immunobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Tsushima naka 1-1-1, Kita-ku, Okayama 700-8530, Japan;
| |
Collapse
|
10
|
A monoclonal antibody to Siglec-8 suppresses non-allergic airway inflammation and inhibits IgE-independent mast cell activation. Mucosal Immunol 2021; 14:366-376. [PMID: 32814824 PMCID: PMC7946634 DOI: 10.1038/s41385-020-00336-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/30/2020] [Accepted: 07/27/2020] [Indexed: 02/04/2023]
Abstract
In addition to their well characterized role in mediating IgE-dependent allergic diseases, aberrant accumulation and activation of mast cells (MCs) is associated with many non-allergic inflammatory diseases, whereby their activation is likely triggered by non-IgE stimuli (e.g., IL-33). Siglec-8 is an inhibitory receptor expressed on MCs and eosinophils that has been shown to inhibit IgE-mediated MC responses and reduce allergic inflammation upon ligation with a monoclonal antibody (mAb). Herein, we evaluated the effects of an anti-Siglec-8 mAb (anti-S8) in non-allergic disease models of experimental cigarette-smoke-induced chronic obstructive pulmonary disease and bleomycin-induced lung injury in Siglec-8 transgenic mice. Therapeutic treatment with anti-S8 inhibited MC activation and reduced recruitment of immune cells, airway inflammation, and lung fibrosis. Similarly, using a model of MC-dependent, IL-33-induced inflammation, anti-S8 treatment suppressed neutrophil influx, and cytokine production through MC inhibition. Transcriptomic profiling of MCs further demonstrated anti-S8-mediated downregulation of MC signaling pathways induced by IL-33, including TNF signaling via NF-κB. Collectively, these findings demonstrate that ligating Siglec-8 with an antibody reduces non-allergic inflammation and inhibits IgE-independent MC activation, supporting the evaluation of an anti-Siglec-8 mAb as a therapeutic approach in both allergic and non-allergic inflammatory diseases in which MCs play a role.
Collapse
|
11
|
Overed-Sayer C, Miranda E, Dunmore R, Liarte Marin E, Beloki L, Rassl D, Parfrey H, Carruthers A, Chahboub A, Koch S, Güler-Gane G, Kuziora M, Lewis A, Murray L, May R, Clarke D. Inhibition of mast cells: a novel mechanism by which nintedanib may elicit anti-fibrotic effects. Thorax 2020; 75:754-763. [PMID: 32709610 PMCID: PMC7476277 DOI: 10.1136/thoraxjnl-2019-214000] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 04/17/2020] [Accepted: 05/01/2020] [Indexed: 12/31/2022]
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive lung disease which presents a grave prognosis for diagnosed patients. Nintedanib (a triple tyrosine kinase inhibitor) and pirfenidone (unclear mechanism of action) are the only approved therapies for IPF, but have limited efficacy. The pathogenic mechanisms of this disease are not fully elucidated; however, a role for mast cells (MCs) has been postulated. Objectives The aim of this work was to investigate a role for MCs in IPF and to understand whether nintedanib or pirfenidone could impact MC function. Methods and results MCs were significantly elevated in human IPF lung and negatively correlated with baseline lung function (FVC). Importantly, MCs were positively associated with the number of fibroblast foci, which has been linked to increased mortality. Furthermore, MCs were increased in the region immediately surrounding the fibroblast foci, and co-culture studies confirmed a role for MC–fibroblast crosstalk in fibrosis. Nintedanib but not pirfenidone inhibited recombinant stem cell factor (SCF)–induced MC survival. Further evaluation of nintedanib determined that it also inhibited human fibroblast-mediated MC survival. This was likely via a direct effect on ckit (SCF receptor) since nintedanib blocked SCF-stimulated ckit phosphorylation, as well as downstream effects on MC proliferation and cytokine release. In addition, nintedanib ablated the increase in lung MCs and impacted high tissue density frequency (HDFm) in a rat bleomycin model of lung fibrosis. Conclusion Nintedanib inhibits MC survival and activation and thus provides a novel additional mechanism by which this drug may exert anti-fibrotic effects in patients with IPF.
Collapse
Affiliation(s)
- Catherine Overed-Sayer
- Regeneration, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Elena Miranda
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| | - Rebecca Dunmore
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Elena Liarte Marin
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Lorea Beloki
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Doris Rassl
- Royal Papworth Hospital NHS Foundation Trust, Cambridge, Cambridgeshire, UK
| | - Helen Parfrey
- Royal Papworth Hospital NHS Foundation Trust, Cambridge, Cambridgeshire, UK
| | - Alan Carruthers
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Amina Chahboub
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Sofia Koch
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| | - Gülin Güler-Gane
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Cambridge, United Kingdom
| | - Michael Kuziora
- Translational Science, Early Oncology, Oncology Bioinformatics, AstraZeneca, Gaithersburg, Maryland, USA
| | - Arthur Lewis
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| | - Lynne Murray
- Regeneration, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Richard May
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Deborah Clarke
- Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| |
Collapse
|
12
|
Pejler G. Novel Insight into the in vivo Function of Mast Cell Chymase: Lessons from Knockouts and Inhibitors. J Innate Immun 2020; 12:357-372. [PMID: 32498069 DOI: 10.1159/000506985] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 03/03/2020] [Indexed: 12/14/2022] Open
Abstract
Mast cells are now recognized as key players in diverse pathologies, but the mechanisms by which they contribute in such settings are only partially understood. Mast cells are packed with secretory granules, and when they undergo degranulation in response to activation the contents of the granules are expelled to the extracellular milieu. Chymases, neutral serine proteases, are the major constituents of the mast cell granules and are hence released in large amounts upon mast cell activation. Following their release, chymases can cleave one or several of a myriad of potential substrates, and the cleavage of many of these could potentially have a profound impact on the respective pathology. Indeed, chymases have recently been implicated in several pathological contexts, in particular through studies using chymase inhibitors and by the use of chymase-deficient animals. In many cases, chymase has been shown to account for mast cell-dependent detrimental effects in the respective conditions and is therefore emerging as a promising drug target. On the other hand, chymase has been shown to have protective roles in other pathological settings. More unexpectedly, chymase has also been shown to control certain homeostatic processes. Here, these findings are reviewed.
Collapse
Affiliation(s)
- Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden, .,Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden,
| |
Collapse
|
13
|
The Chymase Mouse Mast Cell Protease-4 Regulates Intestinal Cytokine Expression in Mature Adult Mice Infected with Giardia intestinalis. Cells 2020; 9:cells9040925. [PMID: 32283818 PMCID: PMC7226739 DOI: 10.3390/cells9040925] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 12/16/2022] Open
Abstract
Mast cells have been shown to affect the control of infections with the protozoan parasite Giardia intestinalis. Recently, we demonstrated that Giardia excretory-secretory proteins inhibited the activity of the connective tissue mast cell-specific protease chymase. To study the potential role of the chymase mouse mast cell protease (mMCP)-4 during infections with Giardia, mMCP-4+/+ and mMCP-4−/− littermate mice were gavage-infected with G. intestinalis trophozoites of the human assemblage B isolate GS. No significant changes in weight gain was observed in infected young (≈10 weeks old) mMCP-4−/− and mMCP-4+/+ littermate mice. In contrast, infections of mature adult mice (>18 weeks old) caused significant weight loss as compared to uninfected control mice. We detected a more rapid weight loss in mMCP-4−/− mice as compared to littermate mMCP-4+/+ mice. Submucosal mast cell and granulocyte counts in jejunum increased in the infected adult mMCP-4−/− and mMCP-4+/+ mice. This increase was correlated with an augmented intestinal trypsin-like and chymotrypsin-like activity, but the myeloperoxidase activity was constant. Infected mice showed a significantly lower intestinal neutrophil elastase (NE) activity, and in vitro, soluble Giardia proteins inhibited human recombinant NE. Serum levels of IL-6 were significantly increased eight and 13 days post infection (dpi), while intestinal IL-6 levels showed a trend to significant increase 8 dpi. Strikingly, the lack of mMCP-4 resulted in significantly less intestinal transcriptional upregulation of IL-6, TNF-α, IL-25, CXCL2, IL-2, IL-4, IL-5, and IL-10 in the Giardia-infected mature adult mice, suggesting that chymase may play a regulatory role in intestinal cytokine responses.
Collapse
|
14
|
Pejler G. The emerging role of mast cell proteases in asthma. Eur Respir J 2019; 54:13993003.00685-2019. [PMID: 31371445 DOI: 10.1183/13993003.00685-2019] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 07/23/2019] [Indexed: 12/31/2022]
Abstract
It is now well established that mast cells (MCs) play a crucial role in asthma. This is supported by multiple lines of evidence, including both clinical studies and studies on MC-deficient mice. However, there is still only limited knowledge of the exact effector mechanism(s) by which MCs influence asthma pathology. MCs contain large amounts of secretory granules, which are filled with a variety of bioactive compounds including histamine, cytokines, lysosomal hydrolases, serglycin proteoglycans and a number of MC-restricted proteases. When MCs are activated, e.g. in response to IgE receptor cross-linking, the contents of their granules are released to the exterior and can cause a massive inflammatory reaction. The MC-restricted proteases include tryptases, chymases and carboxypeptidase A3, and these are expressed and stored at remarkably high levels. There is now emerging evidence supporting a prominent role of these enzymes in the pathology of asthma. Interestingly, however, the role of the MC-restricted proteases is multifaceted, encompassing both protective and detrimental activities. Here, the current knowledge of how the MC-restricted proteases impact on asthma is reviewed.
Collapse
Affiliation(s)
- Gunnar Pejler
- Dept of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden .,Dept of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
15
|
Succar J, Giatsidis G, Yu N, Hassan K, Khouri R, Gurish MF, Pejler G, Åbrink M, Orgill DP. Mouse Mast Cell Protease-4 Recruits Leukocytes in the Inflammatory Phase of Surgically Wounded Skin. Adv Wound Care (New Rochelle) 2019; 8:469-475. [PMID: 31456904 DOI: 10.1089/wound.2018.0898] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 01/10/2019] [Indexed: 01/29/2023] Open
Abstract
Objective: Mouse mast cell protease-4 (mMCP-4, also known as chymase) has both pro- and anti-inflammatory roles depending on the disease model. However, its effects have not been studied in surgically wounded skin. Given the significant clinical applications of modulating the inflammatory response in wound healing, we examined the role of mMCP-4 and the effect of its inhibitor chymostatin on leukocyte and polymorphonuclear cell (PMN) recruitment in our skin model. Approach: Recruitment was assessed on day-1 postwounding of three groups of mice (n = 10 each): mMCP-4 null mice, wild-type (WT) mice treated with the mMCP-4 inhibitor chymostatin, and WT with no other intervention. Leukocytes were stained with CD-45 cell marker, and PMN cells were stained with chloroacetate esterase. Results: The WT mice had 27 ± 9 leukocytes per field compared with 11 ± 6 for the mMCP-4 nulls, a decrease of 60% (p = 0.03), whereas the chymostatin-injected group had a count comparable with the uninjected WT controls at 24 ± 9. The WT group had a PMN count of 96 ± 12 cells, compared with just 24 ± 8 in the mMCP-4 null group, a decrease of 75% (p = 0.001), whereas the chymostatin-treated group had 60 ± 18 cells, a decrease of 38% compared with the WT group (p = 0.03). Innovation: We showed that the inflammatory process can be influenced by impeding the arrival of PMNs into the surgically injured site using the mMCP-4 inhibitor chymostatin. Conclusion: Chymase contributes to the recruitment of white blood cells in surgically wounded skin.
Collapse
Affiliation(s)
- Julien Succar
- Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery Brigham and Women's Hospital—Harvard Medical School, Boston, Massachusetts
| | - Giorgio Giatsidis
- Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery Brigham and Women's Hospital—Harvard Medical School, Boston, Massachusetts
| | - Nanze Yu
- Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery Brigham and Women's Hospital—Harvard Medical School, Boston, Massachusetts
| | - Kazi Hassan
- Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery Brigham and Women's Hospital—Harvard Medical School, Boston, Massachusetts
| | - Roger Khouri
- Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery Brigham and Women's Hospital—Harvard Medical School, Boston, Massachusetts
| | - Michael F. Gurish
- Human Immunology Center, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital—Harvard Medical School, Boston, Massachusetts
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Magnus Åbrink
- Section of Immunology, Department of Biomedical Sciences & Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Dennis Paul Orgill
- Tissue Engineering and Wound Healing Laboratory, Division of Plastic Surgery, Department of Surgery Brigham and Women's Hospital—Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
16
|
Zaghloul MS, Said E, Suddek GM, Salem HA. Crocin attenuates lung inflammation and pulmonary vascular dysfunction in a rat model of bleomycin-induced pulmonary fibrosis. Life Sci 2019; 235:116794. [DOI: 10.1016/j.lfs.2019.116794] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/18/2019] [Accepted: 08/25/2019] [Indexed: 12/28/2022]
|
17
|
Dell'Italia LJ, Collawn JF, Ferrario CM. Multifunctional Role of Chymase in Acute and Chronic Tissue Injury and Remodeling. Circ Res 2019; 122:319-336. [PMID: 29348253 DOI: 10.1161/circresaha.117.310978] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chymase is the most efficient Ang II (angiotensin II)-forming enzyme in the human body and has been implicated in a wide variety of human diseases that also implicate its many other protease actions. Largely thought to be the product of mast cells, the identification of other cellular sources including cardiac fibroblasts and vascular endothelial cells demonstrates a more widely dispersed production and distribution system in various tissues. Furthermore, newly emerging evidence for its intracellular presence in cardiomyocytes and smooth muscle cells opens an entirely new compartment of chymase-mediated actions that were previously thought to be limited to the extracellular space. This review illustrates how these multiple chymase-mediated mechanisms of action can explain the residual risk in clinical trials of cardiovascular disease using conventional renin-angiotensin system blockade.
Collapse
Affiliation(s)
- Louis J Dell'Italia
- From the Department of Medicine, Division of Cardiology, Birmingham Veteran Affairs Medical Center (L.J.D.), Division of Cardiovascular Disease, Department of Medicine (L.J.D.), and Department of Cell, Developmental and Integrative Biology (J.F.C.), University of Alabama at Birmingham; and Division of Surgical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC (C.M.F.).
| | - James F Collawn
- From the Department of Medicine, Division of Cardiology, Birmingham Veteran Affairs Medical Center (L.J.D.), Division of Cardiovascular Disease, Department of Medicine (L.J.D.), and Department of Cell, Developmental and Integrative Biology (J.F.C.), University of Alabama at Birmingham; and Division of Surgical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC (C.M.F.)
| | - Carlos M Ferrario
- From the Department of Medicine, Division of Cardiology, Birmingham Veteran Affairs Medical Center (L.J.D.), Division of Cardiovascular Disease, Department of Medicine (L.J.D.), and Department of Cell, Developmental and Integrative Biology (J.F.C.), University of Alabama at Birmingham; and Division of Surgical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC (C.M.F.)
| |
Collapse
|
18
|
Legere SA, Haidl ID, Légaré JF, Marshall JS. Mast Cells in Cardiac Fibrosis: New Insights Suggest Opportunities for Intervention. Front Immunol 2019; 10:580. [PMID: 31001246 PMCID: PMC6455071 DOI: 10.3389/fimmu.2019.00580] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 03/04/2019] [Indexed: 12/19/2022] Open
Abstract
Mast cells (MC) are innate immune cells present in virtually all body tissues with key roles in allergic disease and host defense. MCs recognize damage-associated molecular patterns (DAMPs) through expression of multiple receptors including Toll-like receptors and the IL-33 receptor ST2. MCs can be activated to degranulate and release pre-formed mediators, to synthesize and secrete cytokines and chemokines without degranulation, and/or to produce lipid mediators. MC numbers are generally increased at sites of fibrosis. They are potent, resident, effector cells producing mediators that regulate the fibrotic process. The nature of the secretory products produced by MCs depend on micro-environmental signals and can be both pro- and anti-fibrotic. MCs have been repeatedly implicated in the pathogenesis of cardiac fibrosis and in angiogenic responses in hypoxic tissues, but these findings are controversial. Several rodent studies have indicated a protective role for MCs. MC-deficient mice have been reported to have poorer outcomes after coronary artery ligation and increased cardiac function upon MC reconstitution. In contrast, MCs have also been implicated as key drivers of fibrosis. MC stabilization during a hypertensive rat model and an atrial fibrillation mouse model rescued associated fibrosis. Discrepancies in the literature could be related to problems with mouse models of MC deficiency. To further complicate the issue, mice generally have a much lower density of MCs in their cardiac tissue than humans, and as such comparing MC deficient and MC containing mouse models is not necessarily reflective of the role of MCs in human disease. In this review, we will evaluate the literature regarding the role of MCs in cardiac fibrosis with an emphasis on what is known about MC biology, in this context. MCs have been well-studied in allergic disease and multiple pharmacological tools are available to regulate their function. We will identify potential opportunities to manipulate human MC function and the impact of their mediators with a view to preventing or reducing harmful fibrosis. Important therapeutic opportunities could arise from increased understanding of the impact of such potent, resident immune cells, with the ability to profoundly alter long term fibrotic processes.
Collapse
Affiliation(s)
- Stephanie A. Legere
- Departments of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Ian D. Haidl
- Departments of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Jean-François Légaré
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Surgery, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Jean S. Marshall
- Departments of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
19
|
Shimbori C, Upagupta C, Bellaye PS, Ayaub EA, Sato S, Yanagihara T, Zhou Q, Ognjanovic A, Ask K, Gauldie J, Forsythe P, Kolb MRJ. Mechanical stress-induced mast cell degranulation activates TGF-β1 signalling pathway in pulmonary fibrosis. Thorax 2019; 74:455-465. [PMID: 30808717 DOI: 10.1136/thoraxjnl-2018-211516] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 10/29/2018] [Accepted: 11/26/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND The role of mast cells accumulating in idiopathic pulmonary fibrosis (IPF) lungs is unknown. OBJECTIVES We investigated the effect of fibrotic extracellular matrix (ECM) on mast cells in experimental and human pulmonary fibrosis. RESULTS In IPF lungs, mast cell numbers were increased and correlated with disease severity (control vs 60%<FVC<90%, mean difference=-222.7, 95% CI -386.3 to -59.2, p=0.004; control vs FVC<60%, mean difference=-301.7, 95% CI of difference -474.1 to -129.34, p=0.0001; FVC>90% vs 60%<FVC<90%, mean difference=-189.6, 95% CI of difference -353.1 to -26.03, p=0.017; FVC>90% vs FVC<60%, mean difference=-268.6, 95% CI of difference -441.0 to -96.17, p=0.0007). Plasma tryptase levels were increased in IPF and negatively correlated with FVC (control vs FVC<60%, mean difference=-17.12, 95% CI of difference -30.02 to -4.22, p=0.006: correlation curves R=-0.045, p=0.025). In a transforming growth factor (TGF)-β1-induced pulmonary fibrosis model, chymase-positive and tryptase-positive mast cells accumulated in fibrotic lung. Lung tissue was decellularised and reseeded with bone marrow or peritoneum-derived mast cells; cells on fibrotic ECM released more TGF-β1 compared with normal ECM (active TGF-β1: bone marrow-derived mast cell (BMMC)-DL vs BMMC-TGF-β1 p=0.0005, peritoneal mast cell (PMC)-DL vs PMC-TGF-β1 p=0.0003, total TGF-β1: BMMC-DL vs BMMC-TGF-β1 p=0.013, PMC-DL vs PMC-TGF-β1 p=0.001). Mechanical stretch of lungs caused mast cell degranulation; mast cell stabilisers inhibited degranulation (histamine: cont vs doxantrazole p=0.004, β-hexosaminidase: cont vs doxantrazole, mean difference=1.007, 95% CI of difference 0.2700 to 1.744, p=0.007) and TGF-β1 activation (pSmad2/Smad2: cont vs dox p=0.006). Cromoglycate attenuated pulmonary fibrosis in rats (collagen: phosphate-buffered saline (PBS) vs cromoglycate p=0.036, fibrotic area: PBS vs cromoglycate p=0.031). CONCLUSION This study suggests that mast cells may contribute to the progression of pulmonary fibrosis.
Collapse
Affiliation(s)
- Chiko Shimbori
- St Joseph's Healthcare and Department of Medicine, Firestone Institute for Respiratory Health, McMaster University Hamilton, Hamilton, Ontario, Canada
| | - Chandak Upagupta
- St Joseph's Healthcare and Department of Medicine, Firestone Institute for Respiratory Health, McMaster University Hamilton, Hamilton, Ontario, Canada
| | - Pierre-Simon Bellaye
- St Joseph's Healthcare and Department of Medicine, Firestone Institute for Respiratory Health, McMaster University Hamilton, Hamilton, Ontario, Canada
| | - Ehab A Ayaub
- St Joseph's Healthcare and Department of Medicine, Firestone Institute for Respiratory Health, McMaster University Hamilton, Hamilton, Ontario, Canada
| | - Seidai Sato
- St Joseph's Healthcare and Department of Medicine, Firestone Institute for Respiratory Health, McMaster University Hamilton, Hamilton, Ontario, Canada
| | - Toyoshi Yanagihara
- St Joseph's Healthcare and Department of Medicine, Firestone Institute for Respiratory Health, McMaster University Hamilton, Hamilton, Ontario, Canada
| | - Quan Zhou
- St Joseph's Healthcare and Department of Medicine, Firestone Institute for Respiratory Health, McMaster University Hamilton, Hamilton, Ontario, Canada
| | - Alexander Ognjanovic
- St Joseph's Healthcare and Department of Medicine, Firestone Institute for Respiratory Health, McMaster University Hamilton, Hamilton, Ontario, Canada
| | - Kjetil Ask
- St Joseph's Healthcare and Department of Medicine, Firestone Institute for Respiratory Health, McMaster University Hamilton, Hamilton, Ontario, Canada
| | - Jack Gauldie
- St Joseph's Healthcare and Department of Medicine, Firestone Institute for Respiratory Health, McMaster University Hamilton, Hamilton, Ontario, Canada
| | - Paul Forsythe
- St Joseph's Healthcare and Department of Medicine, Firestone Institute for Respiratory Health, McMaster University Hamilton, Hamilton, Ontario, Canada
- McMaster Brain-Body Institute, The Research Institute of St Joseph's Hamilton, Hamilton, Ontario, Canada
| | - Martin R J Kolb
- St Joseph's Healthcare and Department of Medicine, Firestone Institute for Respiratory Health, McMaster University Hamilton, Hamilton, Ontario, Canada
| |
Collapse
|
20
|
Abstract
Fibrosis is a medical condition characterized by an excessive deposition of extracellular matrix compounds such as collagen in tissues. Fibrotic lesions are present in many diseases and can affect all organs. The excessive extracellular matrix accumulation in these conditions can often have serious consequences and in many cases be life-threatening. A typical event seen in many fibrotic conditions is a profound accumulation of mast cells (MCs), suggesting that these cells can contribute to the pathology. Indeed, there is now substantialv evidence pointing to an important role of MCs in fibrotic disease. However, investigations from various clinical settings and different animal models have arrived at partly contradictory conclusions as to how MCs affect fibrosis, with many studies suggesting a detrimental role of MCs whereas others suggest that MCs can be protective. Here, we review the current knowledge of how MCs can affect fibrosis.
Collapse
Affiliation(s)
- Peter Bradding
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, Leicester, UK
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.,Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
21
|
El-Kashef DH. Nicorandil ameliorates pulmonary inflammation and fibrosis in a rat model of silicosis. Int Immunopharmacol 2018; 64:289-297. [PMID: 30223191 DOI: 10.1016/j.intimp.2018.09.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/11/2018] [Accepted: 09/13/2018] [Indexed: 10/28/2022]
Abstract
Nicorandil, an antianginal and potassium channel opener agent, has different useful impacts on cardiovascular and respiratory systems. Its effect against silicosis has not been discussed yet, therefore, this is an attempt to decide whether nicorandil can reduce silica-induced lung injury in rats. Silica model was induced by intranasal instillation of silica dust once. Rats were given nicorandil for 56 days after exposure to silica. Results showed that nicorandil significantly alleviated silica-induced inflammation as it decreased the elevated levels of total and differential cell counts, pulmonary edema (revealed by decreased lung/body weight ratio and W/D weight ratio), LDH and total protein levels in BALF. Notably, nicorandil decreased collagen deposition as evidenced by reduction in levels of hydroxyproline and collagen in lung tissues as well as obvious alleviation in silica-induced fibrosis in histopathological findings. Nicorandil effectively reduced the increased expression of NF-κB and iNOS and decreased MPO levels in lung tissues. Moreover, nicorandil abolished oxidative and nitrosative stress via reducing levels of pulmonary MDA and NOx concomitant with elevating levels of pulmonary GSH and SOD. Meanwhile, nicorandil decreased the levels of TNF-α and TGF-β, up regulated Nrf-2 and HO-1 levels in BALF suggesting antioxidant, anti-inflammatory and antifibrotic properties. In summary, nicorandil can confer protection against silica-induced lung inflammation and fibrosis. This impact might be due to its ability to down regulate the production of inflammatory and fibrotic cytokines in addition to restoring oxidant/antioxidant balance.
Collapse
Affiliation(s)
- Dalia H El-Kashef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
22
|
Paivandy A, Sandelin M, Igelström H, Landelius P, Janson C, Melo FR, Pejler G. Induction of Human Lung Mast Cell Apoptosis by Granule Permeabilization: A Novel Approach for Targeting Mast Cells. Front Immunol 2017; 8:1645. [PMID: 29230220 PMCID: PMC5711769 DOI: 10.3389/fimmu.2017.01645] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 11/10/2017] [Indexed: 12/22/2022] Open
Abstract
Mast cells are implicated as detrimental players in inflammatory lung diseases, particularly asthma. Mast cells respond to activating stimuli by releasing a wide panel of pro-inflammatory compounds that can contribute profoundly to the pathology, and there is currently an unmet need for strategies that efficiently ameliorate harmful effects of mast cells under such conditions. Here, we sought to evaluate a novel concept for targeting human lung mast cells, by assessing the possibility of selectively depleting the lung mast cells by induction of apoptosis. For this purpose, we used lysosomotropic agents, i.e., compounds that are known to permeabilize the secretory granules of mast cells, thereby releasing the contents of the granules into the cytosol. Either intact human lung tissue, purified human lung mast cells or mixed populations of human lung cells were incubated with the lysosomotropic agents mefloquine or siramesine, followed by measurement of apoptosis, reactive oxygen species (ROS) production, and release of cytokines. We show that human lung mast cells were highly susceptible to apoptosis induced by this strategy, whereas other cell populations of the lung were largely refractory. Moreover, we demonstrate that apoptosis induced by this mode is dependent on the production of ROS and that the treatment of lung tissue with lysosomotropic agents causes a decrease in the release of pathogenic cytokines. We conclude that selective apoptosis of human lung mast cells can be accomplished by administration of lysosomotropic agents, thus introducing the possibility of using such drugs as novel therapeutics in the treatment of inflammatory lung disorders such as asthma.
Collapse
Affiliation(s)
- Aida Paivandy
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Martin Sandelin
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Per Landelius
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Christer Janson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Fabio R Melo
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.,Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
23
|
El-Mohandes EM, Moustafa AM, Khalaf HA, Hassan YF. The role of mast cells and macrophages in amiodarone induced pulmonary fibrosis and the possible attenuating role of atorvastatin. Biotech Histochem 2017; 92:467-480. [PMID: 28836856 DOI: 10.1080/10520295.2017.1350750] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Amiodarone (AM) is an effective anti-arrhythmic drug. We investigated the role of mast cells and macrophages on AM induced pulmonary fibrosis and the action of atorvastatin on this fibrosis. Rats were allocated into four groups; negative control (1), positive control (2), 30 mg/kg body weight/day AM (3) and AM + 10 mg/kg/day atorvastatin (4). Lungs were harvested and prepared for histology and immunohistochemistry. Hematoxylin and eosin stained sections of group 3 exhibited disorganized lung architecture. We found cellular debris in the lumen of both intrapulmonary bronchi and bronchioles with partial disruption of the thickened epithelial lining and mononuclear cellular infiltration into the lamina propria. We also observed thickening of the epithelial lining and the smooth muscle layer. Congested, dilated and thickened blood capillaries and thickened inter-alveolar septa were observed with mononuclear cellular infiltrates in the lung of group 3. Most alveoli were collapsed, but some dilated ones were detected. In some alveoli, type ІІ pneumocytes were increased, while type I cells were decreased. We observed significant increases in the amount of collagen in the thickened inter-alveolar septa, around bronchioles and around blood capillaries in sections from group 3. We found a significant increase in mast cells and alveolar macrophages in group 3 compared to group 1. Mast cells and macrophages appear to play important roles in AM induced pulmonary fibrosis. Atorvastatin appears to attenuate this condition.
Collapse
Affiliation(s)
- E M El-Mohandes
- a Histology and Cell Biology Department, Faculty of Medicine , Mansoura University , Egypt
| | - A M Moustafa
- a Histology and Cell Biology Department, Faculty of Medicine , Mansoura University , Egypt
| | - H A Khalaf
- a Histology and Cell Biology Department, Faculty of Medicine , Mansoura University , Egypt
| | - Y F Hassan
- a Histology and Cell Biology Department, Faculty of Medicine , Mansoura University , Egypt
| |
Collapse
|
24
|
Daubeuf F, Becker J, Aguilar-Pimentel JA, Ebel C, Hrabě de Angelis M, Hérault Y, Frossard N. A Fast, Easy, and Customizable Eight-Color Flow Cytometric Method for Analysis of the Cellular Content of Bronchoalveolar Lavage Fluid in the Mouse. ACTA ACUST UNITED AC 2017. [PMID: 28628216 DOI: 10.1002/cpmo.26] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The cell composition of bronchoalveolar lavage fluid (BAL) is an important indicator of airway inflammation. It is commonly determined by cytocentrifuging leukocytes on slides, then staining, identifying, and counting them as eosinophils, neutrophils, macrophages, or lymphocytes according to morphological criteria under light microscopy, where it is not always easy to distinguish macrophages from lymphocytes. We describe here a one-step, easy-to-use, and easy-to-customize 8-color flow cytometric method for performing differential cell count and comparing it to morphological counts on stained cytospins. This method identifies BAL cells by a simultaneous one-step immunolabeling procedure using antibodies to identify T cells, B cells, neutrophils, eosinophils, and macrophages. Morphological analysis of flow-sorted cell subsets is used to validate this protocol. An important advantage of this basic flow cytometry protocol is the ability to customize it by the addition of antibodies to study receptor expression at leukocyte cell surfaces and identify subclasses of inflammatory cells as needed. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- François Daubeuf
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR 7200, Illkirch Cedex, France
| | - Julien Becker
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), Illkirch-Graffenstaden, France
| | - Juan Antonio Aguilar-Pimentel
- German Mouse Clinic, Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
| | - Claudine Ebel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
| | - Yann Hérault
- CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), Illkirch-Graffenstaden, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Nelly Frossard
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR 7200, Illkirch Cedex, France
| |
Collapse
|
25
|
|
26
|
Ramachandran R, Altier C, Oikonomopoulou K, Hollenberg MD. Proteinases, Their Extracellular Targets, and Inflammatory Signaling. Pharmacol Rev 2016; 68:1110-1142. [PMID: 27677721 DOI: 10.1124/pr.115.010991] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Given that over 2% of the human genome codes for proteolytic enzymes and their inhibitors, it is not surprising that proteinases serve many physiologic-pathophysiological roles. In this context, we provide an overview of proteolytic mechanisms regulating inflammation, with a focus on cell signaling stimulated by the generation of inflammatory peptides; activation of the proteinase-activated receptor (PAR) family of G protein-coupled receptors (GPCR), with a mechanism in common with adhesion-triggered GPCRs (ADGRs); and by proteolytic ion channel regulation. These mechanisms are considered in the much wider context that proteolytic mechanisms serve, including the processing of growth factors and their receptors, the regulation of matrix-integrin signaling, and the generation and release of membrane-tethered receptor ligands. These signaling mechanisms are relevant for inflammatory, neurodegenerative, and cardiovascular diseases as well as for cancer. We propose that the inflammation-triggering proteinases and their proteolytically generated substrates represent attractive therapeutic targets and we discuss appropriate targeting strategies.
Collapse
Affiliation(s)
- Rithwik Ramachandran
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology & Pharmacology (R.R., C.A., M.D.H.) and Department of Medicine (M.D.H.),University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; Department of Pathology and Laboratory Medicine, Toronto Western Hospital, Toronto, Ontario, Canada (K.O.); and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (R.R.)
| | - Christophe Altier
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology & Pharmacology (R.R., C.A., M.D.H.) and Department of Medicine (M.D.H.),University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; Department of Pathology and Laboratory Medicine, Toronto Western Hospital, Toronto, Ontario, Canada (K.O.); and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (R.R.)
| | - Katerina Oikonomopoulou
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology & Pharmacology (R.R., C.A., M.D.H.) and Department of Medicine (M.D.H.),University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; Department of Pathology and Laboratory Medicine, Toronto Western Hospital, Toronto, Ontario, Canada (K.O.); and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (R.R.)
| | - Morley D Hollenberg
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology & Pharmacology (R.R., C.A., M.D.H.) and Department of Medicine (M.D.H.),University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; Department of Pathology and Laboratory Medicine, Toronto Western Hospital, Toronto, Ontario, Canada (K.O.); and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (R.R.)
| |
Collapse
|
27
|
Significant Contribution of Mouse Mast Cell Protease 4 in Early Phases of Experimental Autoimmune Encephalomyelitis. Mediators Inflamm 2016; 2016:9797021. [PMID: 27610007 PMCID: PMC5005578 DOI: 10.1155/2016/9797021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 06/13/2016] [Accepted: 07/21/2016] [Indexed: 02/08/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a mouse model that reproduces cardinal signs of clinical, histopathological, and immunological features found in Multiple Sclerosis (MS). Mast cells are suggested to be involved in the main inflammatory phases occurring during EAE development, possibly by secreting several autacoids and proteases. Among the latter, the chymase mouse mast cell protease 4 (mMCP-4) can contribute to the inflammatory response by producing endothelin-1 (ET-1). The aim of this study was to determine the impact of mMCP-4 on acute inflammatory stages in EAE. C57BL/6 wild type (WT) or mMCP-4 knockout (KO) mice were immunized with MOG35–55 plus complete Freund's adjuvant followed by pertussis toxin. Immunized WT mice presented an initial acute phase characterized by progressive increases in clinical score, which were significantly reduced in mMCP-4 KO mice. In addition, higher levels of spinal myelin were found in mMCP-4 KO as compared with WT mice. Finally, whereas EAE triggered significant increases in brain levels of mMCP-4 mRNA and immunoreactive ET-1 in WT mice, the latter peptide was reduced to basal levels in mMCP-4 KO congeners. Together, the present study supports a role for mMCP-4 in the early inflammatory phases of the disease in a mouse model of MS.
Collapse
|
28
|
Modena BD, Dazy K, White AA. Emerging concepts: mast cell involvement in allergic diseases. Transl Res 2016; 174:98-121. [PMID: 26976119 DOI: 10.1016/j.trsl.2016.02.011] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 02/08/2023]
Abstract
In a process known as overt degranulation, mast cells can release all at once a diverse array of products that are preformed and present within cytoplasmic granules. This occurs typically within seconds of stimulation by environmental factors and allergens. These potent, preformed mediators (ie, histamine, heparin, serotonin, and serine proteases) are responsible for the acute symptoms experienced in allergic conditions such as allergic conjunctivitis, allergic rhinitis, allergy-induced asthma, urticaria, and anaphylaxis. Yet, there is reason to believe that the actions of mast cells are important when they are not degranulating. Mast cells release preformed mediators and inflammatory cytokines for periods after degranulation and even without degranulating at all. Mast cells are consistently seen at sites of chronic inflammation, including nonallergic inflammation, where they have the ability to temper inflammatory processes and shape tissue morphology. Mast cells can trigger actions and chemotaxis in other important immune cells (eg, eosinophils and the newly discovered type 2 innate lymphocytes) that then make their own contributions to inflammation and disease. In this review, we will discuss the many known and theorized contributions of mast cells to allergic diseases, focusing on several prototypical allergic respiratory and skin conditions: asthma, chronic rhinosinusitis, aspirin-exacerbated respiratory disease, allergic conjunctivitis, atopic dermatitis, and some of the more common medication hypersensitivity reactions. We discuss traditionally accepted roles that mast cells play in the pathogenesis of each of these conditions, but we also delve into new areas of discovery and research that challenge traditionally accepted paradigms.
Collapse
Affiliation(s)
- Brian D Modena
- Division of Allergy, Asthma and Immunology, Scripps Clinic, San Diego, Calif; Scripps Translational Science Institute, The Scripps Research Institute, La Jolla, Calif
| | - Kristen Dazy
- Division of Allergy, Asthma and Immunology, Scripps Clinic, San Diego, Calif
| | - Andrew A White
- Division of Allergy, Asthma and Immunology, Scripps Clinic, San Diego, Calif.
| |
Collapse
|
29
|
Veerappan A, Thompson M, Savage AR, Silverman ML, Chan WS, Sung B, Summers B, Montelione KC, Benedict P, Groh B, Vicencio AG, Peinado H, Worgall S, Silver RB. Mast cells and exosomes in hyperoxia-induced neonatal lung disease. Am J Physiol Lung Cell Mol Physiol 2016; 310:L1218-32. [DOI: 10.1152/ajplung.00299.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 04/26/2016] [Indexed: 11/22/2022] Open
Abstract
Chronic lung disease of prematurity (CLD) is a frequent sequela of premature birth and oxygen toxicity is a major associated risk factor. Impaired alveolarization, scarring, and inflammation are hallmarks of CLD. Mast cell hyperplasia is a feature of CLD but the role of mast cells in its pathogenesis is unknown. We hypothesized that mast cell hyperplasia is a consequence of neonatal hyperoxia and contributes to CLD. Additionally, mast cell products may have diagnostic and prognostic value in preterm infants predisposed to CLD. To model CLD, neonatal wild-type and mast cell-deficient mice were placed in an O2 chamber delivering hyperoxic gas mixture [inspired O2 fraction (FiO2) of 0.8] (HO) for 2 wk and then returned to room air (RA) for an additional 3 wk. Age-matched controls were kept in RA (FiO2 of 0.21). Lungs from HO mice had increased numbers of mast cells, alveolar simplification and enlargement, and increased lung compliance. Mast cell deficiency proved protective by preserving air space integrity and lung compliance. The mast cell mediators β-hexosaminidase (β-hex), histamine, and elastase increased in the bronchoalveolar lavage fluid of HO wild-type mice. Tracheal aspirate fluids (TAs) from oxygenated and mechanically ventilated preterm infants were analyzed for mast cell products. In TAs from infants with confirmed cases of CLD, β-hex was elevated over time and correlated with FiO2. Mast cell exosomes were also present in the TAs. Collectively, these data show that mast cells play a significant role in hyperoxia-induced lung injury and their products could serve as potential biomarkers in evolving CLD.
Collapse
Affiliation(s)
- A. Veerappan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - M. Thompson
- Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - A. R. Savage
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - M. L. Silverman
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - W. S. Chan
- Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - B. Sung
- Department of Genetic Medicine, Weill Cornell Medicine, New York, New York; and
| | - B. Summers
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - K. C. Montelione
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - P. Benedict
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - B. Groh
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | - A. G. Vicencio
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - H. Peinado
- Department of Pediatrics, Weill Cornell Medicine, New York, New York
| | - S. Worgall
- Department of Pediatrics, Weill Cornell Medicine, New York, New York
- Department of Genetic Medicine, Weill Cornell Medicine, New York, New York; and
| | - R. B. Silver
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| |
Collapse
|
30
|
Divya T, Dineshbabu V, Soumyakrishnan S, Sureshkumar A, Sudhandiran G. Celastrol enhances Nrf2 mediated antioxidant enzymes and exhibits anti-fibrotic effect through regulation of collagen production against bleomycin-induced pulmonary fibrosis. Chem Biol Interact 2016; 246:52-62. [PMID: 26768587 DOI: 10.1016/j.cbi.2016.01.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 12/01/2015] [Accepted: 01/04/2016] [Indexed: 01/14/2023]
Abstract
Pulmonary fibrosis (PF) is characterized by excessive accumulation of extracellular matrix components in the alveolar region which distorts the normal lung architecture and impairs the respiratory function. The aim of this study is to evaluate the anti-fibrotic effect of celastrol, a quinine-methide tri-terpenoid mainly found in Thunder God Vine root extracts against bleomycin (BLM)-induced PF through the enhancement of antioxidant defense system. A single intratracheal instillation of BLM (3 U/kg.bw) was administered in rats to induce PF. Celastrol (5 mg/kg) was given intraperitoneally, twice a week for a period of 28 days. BLM-induced rats exhibits declined activities of enzymatic and non-enzymatic antioxidants which were restored upon treatment with celastrol. BLM-induced rats show increased total and differential cell counts as compared to control and celastrol treated rats. Histopathological analysis shows increased inflammation and alveolar damage; while assay of hydroxyproline and Masson's trichrome staining shows an increased collagen deposition in BLM-challenged rats that were decreased upon celastrol treatment. Celastrol also reduces inflammation in BLM-induced rats as evidenced by decrease in the expressions of mast cells, Tumor necrosis factor-alpha (TNF- α) and matrix metalloproteinases (MMPs) 2 and 9. Further, Western blot analysis shows that celastrol is a potent inducer of NF-E2-related factor 2 (Nrf2) and it restores the activities of Phase II enzymes such as hemoxygenase-1 (HO-1), glutathione-S-transferase (GSTs) and NADP(H): quinine oxidoreductase (NQO1) which were declined upon BLM administration. The results of this study show evidence on the protective effect of celastrol against BLM-induced PF through its antioxidant and anti-fibrotic effects.
Collapse
Affiliation(s)
- Thomas Divya
- Cell Biology Laboratory, Department of Biochemistry, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Vadivel Dineshbabu
- Cell Biology Laboratory, Department of Biochemistry, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Syamala Soumyakrishnan
- Cell Biology Laboratory, Department of Biochemistry, University of Madras, Guindy Campus, Chennai 600 025, India
| | | | - Ganapasam Sudhandiran
- Cell Biology Laboratory, Department of Biochemistry, University of Madras, Guindy Campus, Chennai 600 025, India.
| |
Collapse
|
31
|
The Protective Effect of Naringin against Bleomycin-Induced Pulmonary Fibrosis in Wistar Rats. Pulm Med 2016; 2016:7601393. [PMID: 26977316 PMCID: PMC4764747 DOI: 10.1155/2016/7601393] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/23/2015] [Accepted: 01/14/2016] [Indexed: 12/22/2022] Open
Abstract
The aim of the current study was to investigate the protective effect of naringin on bleomycin-induced pulmonary fibrosis in rats. Twenty-four Wistar rats randomly divided into four groups (control, bleomycin alone, bleomycin + naringin 40, and bleomycin + naringin 80) were used. Rats were administered a single dose of bleomycin (5 mg/kg; via the tracheal cannula) alone or followed by either naringin 40 mg/kg (orally) or naringin 80 mg/kg (orally) or water (1 mL, orally) for 14 days. Rats and lung tissue were weighed to determine the lung index. TNF-α and IL-1β levels, hydroxyproline content, and malondialdehyde (MDA) levels were assayed. Glutathione peroxidase (GSH-Px) and superoxide dismutase (SOD) activities were determined. Tissue sections were stained with hematoxylin-eosin, Masson's trichrome, and 0.1% toluidine blue. TNF-α, IL-1β, and MDA levels and hydroxyproline content significantly increased (p < 0.01) and GPx and SOD activities significantly decreased in bleomycin group (p < 0.01). Naringin at a dose of 80 mg/kg body weight significantly decreased TNF-α and IL-1β activity, hydroxyproline content, and MDA level (p < 0.01) and increased GPx and SOD activities (p < 0.05). Histological evidence supported the results. These results show that naringin has the potential of reducing the toxic effects of bleomycin and may provide supportive therapy for conventional treatment methods for idiopathic pulmonary fibrosis.
Collapse
|
32
|
Waern I, Karlsson I, Pejler G, Wernersson S. IL-6 and IL-17A degradation by mast cells is mediated by a serglycin:serine protease axis. IMMUNITY INFLAMMATION AND DISEASE 2015; 4:70-9. [PMID: 27042303 PMCID: PMC4768062 DOI: 10.1002/iid3.95] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 11/13/2015] [Accepted: 11/18/2015] [Indexed: 01/04/2023]
Abstract
Mast cells contain large amounts of fully active proteases that are stored in complex with serglycin proteoglycan in their secretory granules. Upon degranulation, such serglycin:protease complexes are released to the extracellular space and can potentially have an impact on the local inflammatory reaction, either through direct effects of serglycin proteoglycan or through effects mediated by its bound proteases. The objective of this study was to address this scenario by investigating the possibility that serglycin‐associated proteases can regulate levels of pro‐inflammatory cytokines. Indeed, we show here that activated cultured peritoneal mast cells from wild type mice efficiently reduced the levels of exogenously administered IL‐6 and IL‐17A, whereas serglycin‐deficient mast cells lacked this ability. Furthermore, our data suggest that the reduction of IL‐6 and IL‐17A concentrations is due to proteolytic degradation mediated by serglycin‐dependent serine proteases. Moreover, we show that activated mast cells have the capacity to release IL‐6 and that the levels of this cytokine in supernatants were markedly higher in cultures of serglycin‐deficient versus serglycin‐sufficient mast cells, suggesting that serglycin‐dependent serine proteases also participate in the regulation of endogenously produced IL‐6. In summary, although the general consensus is that mast cells have a pathogenic impact on inflammatory settings, this study identifies a role for a mast cell‐derived serglycin:serine protease axis in down‐regulating levels of major inflammatory cytokines. These findings support the notion that mast cells could have a dual role in inflammatory settings, by both being able to secrete pathogenic compounds and being able to regulate their levels after release.
Collapse
Affiliation(s)
- Ida Waern
- Department of Anatomy, Physiology, and Biochemistry Swedish University of Agricultural Sciences Uppsala SE-75007 Sweden
| | - Iulia Karlsson
- Department of Anatomy, Physiology, and Biochemistry Swedish University of Agricultural Sciences Uppsala SE-75007 Sweden
| | - Gunnar Pejler
- Department of Anatomy, Physiology, and BiochemistrySwedish University of Agricultural SciencesUppsalaSE-75007Sweden; Department of Medical Biochemistry and MicrobiologyUppsala UniversityUppsalaSE-75123Sweden
| | - Sara Wernersson
- Department of Anatomy, Physiology, and Biochemistry Swedish University of Agricultural Sciences Uppsala SE-75007 Sweden
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW In this review of the literature from 2014 through mid-2015, we examine new data that shed light on how macrophages and other innate immune cells and signals contribute to inflammation, vascular dysfunction, and fibrosis in scleroderma. RECENT FINDINGS Recent human studies have focused on changes early in scleroderma, and linked macrophages to inflammation in skin and progression of lung disease. Plasmacytoid dendritic cells have been implicated in vascular dysfunction. In mice, several factors have been identified that influence macrophage activation and experimental fibrosis. However, emerging data also suggest that myeloid cells can have differential effects in fibrosis. Sustained signaling through different toll-like receptors can lead to inflammation or fibrosis, and these signals can influence both immune and nonimmune cells. SUMMARY There are many types of innate immune cells that can potentially contribute to scleroderma and will be worth exploring in detail. Experimentally dissecting the roles of macrophages based on ontogeny and activation state, and the innate signaling pathways in the tissue microenvironment, may also lead to better understanding of scleroderma pathogenesis.
Collapse
Affiliation(s)
- Jennifer J Chia
- aWeill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program bImmunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences cAutoimmunity and Inflammation Program dAutoimmunity and Inflammation Program and Department of Pediatric Rheumatology, Hospital for Special Surgery eDepartment of Microbiology and Immunology, Weill Cornell Medical College, New York, USA
| | | |
Collapse
|
34
|
Muala A, Rankin G, Sehlstedt M, Unosson J, Bosson JA, Behndig A, Pourazar J, Nyström R, Pettersson E, Bergvall C, Westerholm R, Jalava PI, Happo MS, Uski O, Hirvonen MR, Kelly FJ, Mudway IS, Blomberg A, Boman C, Sandström T. Acute exposure to wood smoke from incomplete combustion--indications of cytotoxicity. Part Fibre Toxicol 2015; 12:33. [PMID: 26511835 PMCID: PMC4625445 DOI: 10.1186/s12989-015-0111-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/21/2015] [Indexed: 12/16/2022] Open
Abstract
Background Smoke from combustion of biomass fuels is a major risk factor for respiratory disease, but the underlying mechanisms are poorly understood. The aim of this study was to determine whether exposure to wood smoke from incomplete combustion would elicit airway inflammation in humans. Methods Fourteen healthy subjects underwent controlled exposures on two separate occasions to filtered air and wood smoke from incomplete combustion with PM1 concentration at 314 μg/m3 for 3 h in a chamber. Bronchoscopy with bronchial wash (BW), bronchoalveolar lavage (BAL) and endobronchial mucosal biopsies was performed after 24 h. Differential cell counts and soluble components were analyzed, with biopsies stained for inflammatory markers using immunohistochemistry. In parallel experiments, the toxicity of the particulate matter (PM) generated during the chamber exposures was investigated in vitro using the RAW264.7 macrophage cell line. Results Significant reductions in macrophage, neutrophil and lymphocyte numbers were observed in BW (p < 0.01, <0.05, <0.05, respectively) following the wood smoke exposure, with a reduction in lymphocytes numbers in BAL fluid (<0.01. This unexpected cellular response was accompanied by decreased levels of sICAM-1, MPO and MMP-9 (p < 0.05, <0.05 and <0.01). In contrast, significant increases in submucosal and epithelial CD3+ cells, epithelial CD8+ cells and submucosal mast cells (p < 0.01, <0.05, <0.05 and <0.05, respectively), were observed after wood smoke exposure. The in vitro data demonstrated that wood smoke particles generated under these incomplete combustion conditions induced cell death and DNA damage, with only minor inflammatory responses. Conclusions Short-term exposure to sooty PAH rich wood smoke did not induce an acute neutrophilic inflammation, a classic hallmark of air pollution exposure in humans. While minor proinflammatory lymphocytic and mast cells effects were observed in the bronchial biopsies, significant reductions in BW and BAL cells and soluble components were noted. This unexpected observation, combined with the in vitro data, suggests that wood smoke particles from incomplete combustion could be potentially cytotoxic. Additional research is required to establish the mechanism of this dramatic reduction in airway leukocytes and to clarify how this acute response contributes to the adverse health effects attributed to wood smoke exposure. Trial registration NCT01488500 Electronic supplementary material The online version of this article (doi:10.1186/s12989-015-0111-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ala Muala
- Department of Public Health and Clinical Medicine, Division of Medicine/Respiratory Medicine, Umeå University, Umeå, Sweden
| | - Gregory Rankin
- Department of Public Health and Clinical Medicine, Division of Medicine/Respiratory Medicine, Umeå University, Umeå, Sweden
| | - Maria Sehlstedt
- Department of Public Health and Clinical Medicine, Division of Medicine/Respiratory Medicine, Umeå University, Umeå, Sweden
| | - Jon Unosson
- Department of Public Health and Clinical Medicine, Division of Medicine/Respiratory Medicine, Umeå University, Umeå, Sweden
| | - Jenny A Bosson
- Department of Public Health and Clinical Medicine, Division of Medicine/Respiratory Medicine, Umeå University, Umeå, Sweden
| | - Annelie Behndig
- Department of Public Health and Clinical Medicine, Division of Medicine/Respiratory Medicine, Umeå University, Umeå, Sweden
| | - Jamshid Pourazar
- Department of Public Health and Clinical Medicine, Division of Medicine/Respiratory Medicine, Umeå University, Umeå, Sweden
| | - Robin Nyström
- Department of Applied Physics and Electronics, Thermochemical Energy Conversion Laboratory, Umeå University, Umeå, Sweden
| | - Esbjörn Pettersson
- Department of Applied Physics and Electronics, Thermochemical Energy Conversion Laboratory, Umeå University, Umeå, Sweden
| | - Christoffer Bergvall
- Department of Environmental Science and Analytical Chemistry, Arrhenius Laboratory, Stockholm University, Stockholm, Sweden
| | - Roger Westerholm
- Department of Environmental Science and Analytical Chemistry, Arrhenius Laboratory, Stockholm University, Stockholm, Sweden
| | - Pasi I Jalava
- Department of Environmental Science, University of Eastern Finland, Kuopio, Finland
| | - Mikko S Happo
- Department of Environmental Science, University of Eastern Finland, Kuopio, Finland
| | - Oskari Uski
- Department of Environmental Science, University of Eastern Finland, Kuopio, Finland
| | | | - Frank J Kelly
- Environmental Research Group, MRC-PHE Centre for Environment and Health, King's College London, London, UK
| | - Ian S Mudway
- Environmental Research Group, MRC-PHE Centre for Environment and Health, King's College London, London, UK
| | - Anders Blomberg
- Department of Public Health and Clinical Medicine, Division of Medicine/Respiratory Medicine, Umeå University, Umeå, Sweden
| | - Christoffer Boman
- Department of Applied Physics and Electronics, Thermochemical Energy Conversion Laboratory, Umeå University, Umeå, Sweden
| | - Thomas Sandström
- Department of Public Health and Clinical Medicine, Division of Medicine/Respiratory Medicine, Umeå University, Umeå, Sweden.
| |
Collapse
|
35
|
Tao W, Su Q, Wang H, Guo S, Chen Y, Duan J, Wang S. Platycodin D attenuates acute lung injury by suppressing apoptosis and inflammation in vivo and in vitro. Int Immunopharmacol 2015; 27:138-47. [DOI: 10.1016/j.intimp.2015.05.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 04/24/2015] [Accepted: 05/04/2015] [Indexed: 12/20/2022]
|
36
|
Kosanovic D, Luitel H, Dahal BK, Cornitescu T, Janssen W, Danser AHJ, Garrelds IM, De Mey JGR, Fazzi G, Schiffers P, Iglarz M, Fischli W, Ghofrani HA, Weissmann N, Grimminger F, Seeger W, Reiss I, Schermuly RT. Chymase: a multifunctional player in pulmonary hypertension associated with lung fibrosis. Eur Respir J 2015; 46:1084-94. [PMID: 26113671 DOI: 10.1183/09031936.00018215] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 04/09/2015] [Indexed: 12/22/2022]
Abstract
Limited literature sources implicate mast-cell mediator chymase in the pathologies of pulmonary hypertension and pulmonary fibrosis. However, there is no evidence on the contribution of chymase to the development of pulmonary hypertension associated with lung fibrosis, which is an important medical condition linked with increased mortality of patients who already suffer from a life-threatening interstitial lung disease.The aim of this study was to investigate the role of chymase in this particular pulmonary hypertension form, by using a bleomycin-induced pulmonary hypertension model.Chymase inhibition resulted in attenuation of pulmonary hypertension and pulmonary fibrosis, as evident from improved haemodynamics, decreased right ventricular remodelling/hypertrophy, pulmonary vascular remodelling and lung fibrosis. These beneficial effects were associated with a strong tendency of reduction in mast cell number and activity, and significantly diminished chymase expression levels. Mechanistically, chymase inhibition led to attenuation of transforming growth factor β1 and matrix-metalloproteinase-2 contents in the lungs. Furthermore, chymase inhibition prevented big endothelin-1-induced vasoconstriction of the pulmonary arteries.Therefore, chymase plays a role in the pathogenesis of pulmonary hypertension associated with pulmonary fibrosis and may represent a promising therapeutic target. In addition, this study may provide valuable insights on the contribution of chymase in the pulmonary hypertension context, in general, regardless of the pulmonary hypertension form.
Collapse
Affiliation(s)
- Djuro Kosanovic
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany Division of Neonatology, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Himal Luitel
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany
| | - Bhola Kumar Dahal
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany Risk Factor Modification Centre (RFMC), St. Michael's Hospital, Toronto, ON, Canada
| | - Teodora Cornitescu
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany
| | - Wiebke Janssen
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany
| | - A H Jan Danser
- Dept of Pharmacology, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Ingrid M Garrelds
- Dept of Pharmacology, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Jo G R De Mey
- Dept of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark Dept of Pharmacology, Maastricht University, Maastricht, The Netherlands
| | - Gregorio Fazzi
- Dept of Pharmacology, Maastricht University, Maastricht, The Netherlands
| | - Paul Schiffers
- Dept of Pharmacology, Maastricht University, Maastricht, The Netherlands
| | - Marc Iglarz
- Actelion Pharmaceuticals Ltd, Allschwill, Switzerland
| | | | - Hossein Ardeschir Ghofrani
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany
| | - Norbert Weissmann
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany
| | - Friedrich Grimminger
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany
| | - Werner Seeger
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research, Bad Nauheim, Germany
| | - Irwin Reiss
- Division of Neonatology, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands Both authors contributed equally
| | - Ralph Theo Schermuly
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany Both authors contributed equally
| |
Collapse
|
37
|
Gaudenzio N, Sibilano R, Starkl P, Tsai M, Galli SJ, Reber LL. Analyzing the Functions of Mast Cells In Vivo Using 'Mast Cell Knock-in' Mice. J Vis Exp 2015:e52753. [PMID: 26068439 DOI: 10.3791/52753] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Mast cells (MCs) are hematopoietic cells which reside in various tissues, and are especially abundant at sites exposed to the external environment, such as skin, airways and gastrointestinal tract. Best known for their detrimental role in IgE-dependent allergic reactions, MCs have also emerged as important players in host defense against venom and invading bacteria and parasites. MC phenotype and function can be influenced by microenvironmental factors that may differ according to anatomic location and/or based on the type or stage of development of immune responses. For this reason, we and others have favored in vivo approaches over in vitro methods to gain insight into MC functions. Here, we describe methods for the generation of mouse bone marrow-derived cultured MCs (BMCMCs), their adoptive transfer into genetically MC-deficient mice, and the analysis of the numbers and distribution of adoptively transferred MCs at different anatomical sites. This method, named the 'mast cell knock-in' approach, has been extensively used over the past 30 years to assess the functions of MCs and MC-derived products in vivo. We discuss the advantages and limitations of this method, in light of alternative approaches that have been developed in recent years.
Collapse
Affiliation(s)
| | | | - Philipp Starkl
- Department of Pathology, Stanford University School of Medicine
| | - Mindy Tsai
- Department of Pathology, Stanford University School of Medicine
| | - Stephen J Galli
- Department of Pathology, Stanford University School of Medicine; Department of Microbiology & Immunology, Stanford University School of Medicine
| | - Laurent L Reber
- Department of Pathology, Stanford University School of Medicine;
| |
Collapse
|
38
|
Reber LL, Sibilano R, Mukai K, Galli SJ. Potential effector and immunoregulatory functions of mast cells in mucosal immunity. Mucosal Immunol 2015; 8:444-63. [PMID: 25669149 PMCID: PMC4739802 DOI: 10.1038/mi.2014.131] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 11/27/2014] [Indexed: 02/04/2023]
Abstract
Mast cells (MCs) are cells of hematopoietic origin that normally reside in mucosal tissues, often near epithelial cells, glands, smooth muscle cells, and nerves. Best known for their contributions to pathology during IgE-associated disorders such as food allergy, asthma, and anaphylaxis, MCs are also thought to mediate IgE-associated effector functions during certain parasite infections. However, various MC populations also can be activated to express functional programs--such as secreting preformed and/or newly synthesized biologically active products--in response to encounters with products derived from diverse pathogens, other host cells (including leukocytes and structural cells), damaged tissue, or the activation of the complement or coagulation systems, as well as by signals derived from the external environment (including animal toxins, plant products, and physical agents). In this review, we will discuss evidence suggesting that MCs can perform diverse effector and immunoregulatory roles that contribute to homeostasis or pathology in mucosal tissues.
Collapse
Affiliation(s)
- Laurent L Reber
- Department of Pathology, Stanford University, School of Medicine, Stanford, California 94305-5324, USA
| | - Riccardo Sibilano
- Department of Pathology, Stanford University, School of Medicine, Stanford, California 94305-5324, USA
| | - Kaori Mukai
- Department of Pathology, Stanford University, School of Medicine, Stanford, California 94305-5324, USA
| | - Stephen J Galli
- Department of Pathology, Stanford University, School of Medicine, Stanford, California 94305-5324, USA,Department of Microbiology & Immunology, Stanford University, School of Medicine, Stanford, California 94305-5324, USA
| |
Collapse
|
39
|
Ningyan G, Xu Y, Hongfei S, Jingjing C, Min C. The role of macrophage migration inhibitory factor in mast cell-stimulated fibroblast proliferation and collagen production. PLoS One 2015; 10:e0122482. [PMID: 25826375 PMCID: PMC4380314 DOI: 10.1371/journal.pone.0122482] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 02/22/2015] [Indexed: 11/19/2022] Open
Abstract
Current clinical and translational studies have shown that mast cell plays a pivotal role in multiple fibrotic diseases including scleroderma. However, the lack of mature human mast cell culture model exhibits a major obstacle for further dissection of cytokines and signaling molecules required for mast cell mediated fibrosis in various diseases. Macrophage Migration Inhibitory Factor is a mast cell released pro-inflammatory cytokine which is deregulated in scleroderma patients and is also involved in non-scleroderma related fibrosis. In the current study, we successfully generated a practical and reliable human mast cell culture system with bone marrow CD34+ hematopietic precursors. The derivative mast cell is normal in terms of both morphology and function as manifested by normal degranulation. More importantly, we were able to show mast cell conditioned medium as well as MIF supplementation augments fibroblast proliferation and collagen synthesis. This positive regulatory effect of mast cell conditioned medium can be dampened by MIF antibody. In addition, MIF-knockdown significantly inhibits pro-fibrotic activities of CD34+ hematopietic precursor derived mast cells. These data strongly suggest that mast cell released MIF is required for mast cell mediated fibrogenic activities. The current manuscript seems to be the first mechanistic report showing the significance of MIF in mast cell mediated fibrosis, which may pave the way for the development of potential MIF-targeted therapy for fibrotic diseases to a further extent. Moreover, we strongly believe mast cell culture and differentiation model as well as corresponding genetic manipulation methodology will be helpful in characterizing novel mast cell based therapeutic targets.
Collapse
Affiliation(s)
- Gu Ningyan
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Yao Xu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Shi Hongfei
- Department of Orthopedics, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Chen Jingjing
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Chen Min
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- * E-mail:
| |
Collapse
|
40
|
Semaan W, Desbiens L, Houde M, Labonté J, Gagnon H, Yamamoto D, Takai S, Laidlaw T, Bkaily G, Schwertani A, Pejler G, Levesque C, Desjardins R, Day R, D’Orléans-Juste P. Chymase inhibitor-sensitive synthesis of endothelin-1 (1–31) by recombinant mouse mast cell protease 4 and human chymase. Biochem Pharmacol 2015; 94:91-100. [DOI: 10.1016/j.bcp.2015.02.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 02/02/2015] [Accepted: 02/02/2015] [Indexed: 10/24/2022]
|
41
|
Galli SJ, Tsai M, Marichal T, Tchougounova E, Reber LL, Pejler G. Approaches for analyzing the roles of mast cells and their proteases in vivo. Adv Immunol 2015; 126:45-127. [PMID: 25727288 DOI: 10.1016/bs.ai.2014.11.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The roles of mast cells in health and disease remain incompletely understood. While the evidence that mast cells are critical effector cells in IgE-dependent anaphylaxis and other acute IgE-mediated allergic reactions seems unassailable, studies employing various mice deficient in mast cells or mast cell-associated proteases have yielded divergent conclusions about the roles of mast cells or their proteases in certain other immunological responses. Such "controversial" results call into question the relative utility of various older versus newer approaches to ascertain the roles of mast cells and mast cell proteases in vivo. This review discusses how both older and more recent mouse models have been used to investigate the functions of mast cells and their proteases in health and disease. We particularly focus on settings in which divergent conclusions about the importance of mast cells and their proteases have been supported by studies that employed different models of mast cell or mast cell protease deficiency. We think that two major conclusions can be drawn from such findings: (1) no matter which models of mast cell or mast cell protease deficiency one employs, the conclusions drawn from the experiments always should take into account the potential limitations of the models (particularly abnormalities affecting cell types other than mast cells) and (2) even when analyzing a biological response using a single model of mast cell or mast cell protease deficiency, details of experimental design are critical in efforts to define those conditions under which important contributions of mast cells or their proteases can be identified.
Collapse
Affiliation(s)
- Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA; Microbiology & Immunology, Stanford University School of Medicine, Stanford, California, USA.
| | - Mindy Tsai
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Thomas Marichal
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA; GIGA-Research and Faculty of Veterinary Medicine, University of Liege, Liege, Belgium
| | - Elena Tchougounova
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Laurent L Reber
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden; Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
42
|
|
43
|
Reber LL, Frossard N. Targeting mast cells in inflammatory diseases. Pharmacol Ther 2014; 142:416-35. [PMID: 24486828 DOI: 10.1016/j.pharmthera.2014.01.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 01/24/2014] [Indexed: 12/24/2022]
Abstract
Although mast cells have long been known to play a critical role in anaphylaxis and other allergic diseases, they also participate in some innate immune responses and may even have some protective functions. Data from the study of mast cell-deficient mice have facilitated our understanding of some of the molecular mechanisms driving mast cell functions during both innate and adaptive immune responses. This review presents an overview of the biology of mast cells and their potential involvement in various inflammatory diseases. We then discuss some of the current pharmacological approaches used to target mast cells and their products in several diseases associated with mast cell activation.
Collapse
Affiliation(s)
- Laurent L Reber
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Nelly Frossard
- Laboratoire d'Innovation Thérapeutique, UMR 7200 CNRS-Université de Strasbourg, Faculté de Pharmacie, France
| |
Collapse
|