1
|
Erice PA, Huang X, Seasock MJ, Robertson MJ, Tung HY, Perez-Negron MA, Lotlikar SL, Corry DB, Kheradmand F, Rodriguez A. Downregulation of Mirlet7 miRNA family promotes Tc17 differentiation and emphysema via de-repression of RORγt. eLife 2024; 13:RP92879. [PMID: 38722677 PMCID: PMC11081633 DOI: 10.7554/elife.92879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024] Open
Abstract
Environmental air irritants including nanosized carbon black (nCB) can drive systemic inflammation, promoting chronic obstructive pulmonary disease (COPD) and emphysema development. The let-7 microRNA (Mirlet7 miRNA) family is associated with IL-17-driven T cell inflammation, a canonical signature of lung inflammation. Recent evidence suggests the Mirlet7 family is downregulated in patients with COPD, however, whether this repression conveys a functional consequence on emphysema pathology has not been elucidated. Here, we show that overall expression of the Mirlet7 clusters, Mirlet7b/Mirlet7c2 and Mirlet7a1/Mirlet7f1/Mirlet7d, are reduced in the lungs and T cells of smokers with emphysema as well as in mice with cigarette smoke (CS)- or nCB-elicited emphysema. We demonstrate that loss of the Mirlet7b/Mirlet7c2 cluster in T cells predisposed mice to exaggerated CS- or nCB-elicited emphysema. Furthermore, ablation of the Mirlet7b/Mirlet7c2 cluster enhanced CD8+IL17a+ T cells (Tc17) formation in emphysema development in mice. Additionally, transgenic mice overexpressing Mirlet7g in T cells are resistant to Tc17 and CD4+IL17a+ T cells (Th17) development when exposed to nCB. Mechanistically, our findings reveal the master regulator of Tc17/Th17 differentiation, RAR-related orphan receptor gamma t (RORγt), as a direct target of Mirlet7 in T cells. Overall, our findings shed light on the Mirlet7/RORγt axis with Mirlet7 acting as a molecular brake in the generation of Tc17 cells and suggest a novel therapeutic approach for tempering the augmented IL-17-mediated response in emphysema.
Collapse
Affiliation(s)
- Phillip A Erice
- Immunology Graduate Program, Baylor College of MedicineHoustonUnited States
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of MedicineHoustonUnited States
| | - Xinyan Huang
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of MedicineHoustonUnited States
| | - Matthew J Seasock
- Immunology Graduate Program, Baylor College of MedicineHoustonUnited States
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of MedicineHoustonUnited States
| | - Matthew J Robertson
- Dan Duncan Comprehensive Cancer Center, Baylor College of MedicineHoustonUnited States
| | - Hui-Ying Tung
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
| | - Melissa A Perez-Negron
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of MedicineHoustonUnited States
| | - Shivani L Lotlikar
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of MedicineHoustonUnited States
| | - David B Corry
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of MedicineHoustonUnited States
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Center for Translational Research on Inflammatory Diseases, Michael E Debakey, Baylor College of MedicineHoustonUnited States
| | - Farrah Kheradmand
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Center for Translational Research on Inflammatory Diseases, Michael E Debakey, Baylor College of MedicineHoustonUnited States
- Department of Medicine, Section of Pulmonary and Critical Care, Baylor College of MedicineHoustonUnited States
| | - Antony Rodriguez
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of MedicineHoustonUnited States
- Center for Translational Research on Inflammatory Diseases, Michael E Debakey, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
2
|
Erice PA, Huang X, Seasock MJ, Robertson MJ, Tung HY, Perez-Negron MA, Lotlikar SL, Corry DB, Kheradmand F, Rodriguez A. Downregulation of Let-7 miRNA promotes Tc17 differentiation and emphysema via de-repression of RORγt. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.12.562059. [PMID: 37905101 PMCID: PMC10614797 DOI: 10.1101/2023.10.12.562059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Environmental air irritants including nanosized carbon black (nCB) can drive systemic inflammation, promoting chronic obstructive pulmonary disease (COPD) and emphysema development. The let-7 family of miRNAs is associated with IL-17-driven T cell inflammation, a canonical signature of lung inflammation. Recent evidence suggests the let-7 family is downregulated in patients with COPD, however, whether this repression conveys a functional consequence on emphysema pathology has not been elucidated. Here we show that overall expression of the let-7 miRNA clusters, let-7b/let-7c2 and let-7a1/let-7f1/let-7d, are reduced in the lungs and T cells of smokers with emphysema as well as in mice with cigarette smoke (CS)- or nCB-elicited emphysema. We demonstrate that loss of the let-7b/let-7c2-cluster in T cells predisposed mice to exaggerated CS- or nCB-elicited emphysema. Furthermore, ablation of the let-7b/let-7c2-cluster enhanced CD8+IL17a+ T cells (Tc17) formation in emphysema development in mice. Additionally, transgenic mice overexpressing let-7 in T cells are resistant to Tc17 and CD4+IL17a+ T cells (Th17) development when exposed to nCB. Mechanistically, our findings reveal the master regulator of Tc17/Th17 differentiation, RAR-related orphan receptor gamma t (RORγt), as a direct target of let-7 miRNA in T cells. Overall, our findings shed light on the let-7/RORγt axis with let-7 acting as a molecular brake in the generation of Tc17 cells and suggests a novel therapeutic approach for tempering the augmented IL-17-mediated response in emphysema.
Collapse
Affiliation(s)
- Phillip A Erice
- Immunology Graduate Program, Baylor College of Medicine, Houston, TX, 77030
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of Medicine Houston TX, 77030
| | - Xinyan Huang
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of Medicine Houston TX, 77030
- Current address, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University. Guangzhou, Guangdong Province, P.R. China
| | - Matthew J Seasock
- Immunology Graduate Program, Baylor College of Medicine, Houston, TX, 77030
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of Medicine Houston TX, 77030
| | - Matthew J Robertson
- Dan Duncan Comprehensive Cancer Center, Baylor College of Medicine Houston, TX, 77030
| | - Hui-Ying Tung
- Department of Pathology and Immunology, Baylor College of Medicine Houston, TX, 77030
| | - Melissa A Perez-Negron
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of Medicine Houston TX, 77030
| | - Shivani L Lotlikar
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of Medicine Houston TX, 77030
| | - David B Corry
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of Medicine Houston TX, 77030
- Department of Pathology and Immunology, Baylor College of Medicine Houston, TX, 77030
- Center for Translational Research on Inflammatory Diseases, Michael E. Debakey, Baylor College of Medicine, Houston, TX, 77030
| | - Farrah Kheradmand
- Department of Pathology and Immunology, Baylor College of Medicine Houston, TX, 77030
- Department of Medicine, Section of Pulmonary and Critical Care, Baylor College of Medicine. Houston, TX, 77030
- Center for Translational Research on Inflammatory Diseases, Michael E. Debakey, Baylor College of Medicine, Houston, TX, 77030
| | - Antony Rodriguez
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of Medicine Houston TX, 77030
- Center for Translational Research on Inflammatory Diseases, Michael E. Debakey, Baylor College of Medicine, Houston, TX, 77030
| |
Collapse
|
3
|
Kheradmand F, Zhang Y, Corry DB. Contribution of adaptive immunity to human COPD and experimental models of emphysema. Physiol Rev 2023; 103:1059-1093. [PMID: 36201635 PMCID: PMC9886356 DOI: 10.1152/physrev.00036.2021] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 02/01/2023] Open
Abstract
The pathophysiology of chronic obstructive pulmonary disease (COPD) and the undisputed role of innate immune cells in this condition have dominated the field in the basic research arena for many years. Recently, however, compelling data suggesting that adaptive immune cells may also contribute to the progressive nature of lung destruction associated with COPD in smokers have gained considerable attention. The histopathological changes in the lungs of smokers can be limited to the large or small airways, but alveolar loss leading to emphysema, which occurs in some individuals, remains its most significant and irreversible outcome. Critically, however, the question of why emphysema progresses in a subset of former smokers remained a mystery for many years. The recognition of activated and organized tertiary T- and B-lymphoid aggregates in emphysematous lungs provided the first clue that adaptive immune cells may play a crucial role in COPD pathophysiology. Based on these findings from human translational studies, experimental animal models of emphysema were used to determine the mechanisms through which smoke exposure initiates and orchestrates adaptive autoreactive inflammation in the lungs. These models have revealed that T helper (Th)1 and Th17 subsets promote a positive feedback loop that activates innate immune cells, confirming their role in emphysema pathogenesis. Results from genetic studies and immune-based discoveries have further provided strong evidence for autoimmunity induction in smokers with emphysema. These new findings offer a novel opportunity to explore the mechanisms underlying the inflammatory landscape in the COPD lung and offer insights for development of precision-based treatment to halt lung destruction.
Collapse
Affiliation(s)
- Farrah Kheradmand
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
- Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas
| | - Yun Zhang
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - David B Corry
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
- Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas
| |
Collapse
|
4
|
Li CX, Gao J, Sköld CM, Wheelock ÅM. miRNA-mRNA-protein dysregulated network in COPD in women. Front Genet 2022; 13:1010048. [PMID: 36468026 PMCID: PMC9712209 DOI: 10.3389/fgene.2022.1010048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/10/2022] [Indexed: 10/14/2023] Open
Abstract
Rationale: Chronic obstructive pulmonary disease (COPD) is a complex disease caused by a multitude of underlying mechanisms, and molecular mechanistic modeling of COPD, especially at a multi-molecular level, is needed to facilitate the development of molecular diagnostic and prognostic tools and efficacious treatments. Objectives: To investigate the miRNA-mRNA-protein dysregulated network to facilitate prediction of biomarkers and disease subnetwork in COPD in women. Measurements and Results: Three omics data blocks (mRNA, miRNA, and protein) collected from BAL cells from female current-smoker COPD patients, smokers with normal lung function, and healthy never-smokers were integrated with miRNA-mRNA-protein regulatory networks to construct a COPD-specific dysregulated network. Furthermore, downstream network topology, literature annotation, and functional enrichment analysis identified both known and novel disease-related biomarkers and pathways. Both abnormal regulations in miRNA-induced mRNA transcription and protein translation repression play roles in COPD. Finally, the let-7-AIFM1-FKBP1A pathway is highlighted in COPD pathology. Conclusion: For the first time, a comprehensive miRNA-mRNA-protein dysregulated network of primary immune cells from the lung related to COPD in females was constructed to elucidate specific biomarkers and disease pathways. The multi-omics network provides a new molecular insight from a multi-molecular aspect and highlights dysregulated interactions. The highlighted let-7-AIFM1-FKBP1A pathway also indicates new hypotheses of COPD pathology.
Collapse
Affiliation(s)
- Chuan Xing Li
- Respiratory Medicine Unit, Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jing Gao
- Respiratory Medicine Unit, Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Heart and Lung Centre, Department of Pulmonary Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - C. Magnus Sköld
- Respiratory Medicine Unit, Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Åsa M. Wheelock
- Respiratory Medicine Unit, Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
5
|
McQuiston A, Scott D, Nord D, Langerude L, Pelaez A, Machuca T, Mehta A, Chrisie JD, Angel P, Atkinson C. Pro-inflammatory IgG1 N-glycan signature correlates with primary graft dysfunction onset in COPD patients. Transpl Immunol 2021; 71:101491. [PMID: 34767945 DOI: 10.1016/j.trim.2021.101491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/05/2021] [Accepted: 11/06/2021] [Indexed: 11/25/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide. The pathogenesis of COPD is complex; however, recent studies suggest autoimmune changes, characterized by the presence of autoantibodies to elastin and collagen, may contribute to disease status. COPD patients make up approximately 30% of all lung transplants (LTx) annually, however, little is known regarding the relationship between COPD-related autoantibodies and LTx outcomes. We hypothesized that COPD patients that undergo LTx and develop primary graft dysfunction (PGD) have altered circulating autoantibody levels and phenotypic changes as compared those COPD-LTx recipients that do not develop PGD. We measured total immunoglobulin and circulating elastin and collagen autoantibody levels in a cohort of COPD lung transplant recipients pre- and post-LTx. No significant differences were seen in total, elastin, or collagen IgM, IgG, IgG1, IgG2, IgG3, and IgG4 antibodies between PGD+ and PGD- recipients. Antibody function can be greatly altered by glycosylation changes to the antibody Fc region and recent studies have reported altered IgG glycosylation profiles in COPD patients. We therefore utilized a novel mass spectrometry-based multiplexed N-glycoprotein imaging approach and measured changes in IgG-specific antibody N-glycan structures. COPD-LTx recipients who developed PGD had significantly increased IgG1 N-glycan signatures as compared PGD- recipients. In conclusion, we show that immunoglobulin and autoreactive antibody levels are not significantly different in COPD LTx recipients that develop PGD. However, using a novel IgG glycomic analysis we were able to demonstrate multiple significant increases in IgG1 specific N-glycan signatures that were predictive of PGD development. Taken together, these data represent a potential novel method for identifying COPD patients at risk for PGD development and may provide clues to mechanisms by which antibody N-glycan signatures could contribute to antibody-mediated PGD pathogenesis.
Collapse
Affiliation(s)
- Alexander McQuiston
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Danielle Scott
- Department of Surgery, University of Florida, Gainesville, FL, USA
| | - Dianna Nord
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Logan Langerude
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Andres Pelaez
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Tiago Machuca
- Department of Surgery, University of Florida, Gainesville, FL, USA
| | - Anand Mehta
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Jason D Chrisie
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peggi Angel
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA.
| | - Carl Atkinson
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
6
|
Gu BH, Choi JC, Shen YH, Song LZ, Scheurer ME, Luong A, Rodriguez A, Woodruff P, Koth L, Corry DB, Kheradmand F, LeMaire SA. Elastin-Specific Autoimmunity in Smokers With Thoracic Aortic Aneurysm and Dissection is Independent of Chronic Obstructive Pulmonary Disease. J Am Heart Assoc 2020; 8:e011671. [PMID: 30957625 PMCID: PMC6507218 DOI: 10.1161/jaha.118.011671] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Thoracic aortic aneurysm ( TAA ) and dissection ( TAD ) are characterized by progressive disorganization of the aortic wall matrix, including elastin, a highly immunogenic molecule. Whether acquired autoimmune responses can be detected in TAA / TAD patients who are smokers is unknown. The objectives of this study were to determine whether TAA / TAD smokers have increased T-cell responses to human elastin fragments, and to determine whether autoimmune responses in TAA / TAD smokers are dependent on chronic obstructive pulmonary disease. Methods and Results In a cross-sectional study (N=86), we examined peripheral blood CD 4+ T cell responses to elastin fragments in never-, former-, or current-smokers with or without TAA / TAD . CD 4+ T cells were co-cultured with irradiated autologous peripheral blood CD 1a+/ CD 14+ antigen presenting cells pulsed with or without elastin fragments to measure cytokine production. Baseline plasma concentration of anti-elastin antibodies and elastin-degrading enzymes (eg, matrix metalloproteinase-9, and -12, and neutrophil elastase) were measured in the same cohort. elastin fragment-specific CD 4+ T cell expression of interferon-γ, and anti-elastin antibodies were dependent on history of smoking in TAA / TAD patients but were independent of chronic obstructive pulmonary disease. Matrix metalloproteinase-9, and -12, and neutrophil elastase plasma concentrations were also significantly elevated in ever-smokers with TAA / TAD . Conclusions Cigarette smoke is associated with loss of self-tolerance and induction of elastin-specific autoreactive T- and B-cell responses in patients with TAA / TAD . Development of peripheral blood biomarkers to track immunity to self-antigens could be used to identify and potentially prognosticate susceptibility to TAA / TAD in smokers.
Collapse
Affiliation(s)
- Bon-Hee Gu
- 1 Department of Medicine, Pulmonary and Critical Care Baylor College of Medicine Houston TX
| | - Justin C Choi
- 2 Division of Cardiothoracic Surgery Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX
| | - Ying H Shen
- 2 Division of Cardiothoracic Surgery Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX.,3 Cardiovascular Research Institute Baylor College of Medicine Houston TX.,7 Department of Cardiovascular Surgery Texas Heart Institute Houston TX
| | - Li-Zhen Song
- 1 Department of Medicine, Pulmonary and Critical Care Baylor College of Medicine Houston TX
| | - Michael E Scheurer
- 4 Section of Hematology-Oncology Department of Pediatrics Baylor College of Medicine Houston TX
| | - Amber Luong
- 8 Department of Otorhinolaryngology - Head and Neck Surgery McGovern Medical School University of Texas Health Science Center Houston TX
| | - Antony Rodriguez
- 1 Department of Medicine, Pulmonary and Critical Care Baylor College of Medicine Houston TX.,5 Departments of Pathology and Immunology Baylor College of Medicine Houston TX.,6 Biology of Inflammation Center Baylor College of Medicine Houston TX
| | | | - Laura Koth
- 9 University of California San Francisco San Francisco CA
| | - David B Corry
- 1 Department of Medicine, Pulmonary and Critical Care Baylor College of Medicine Houston TX.,5 Departments of Pathology and Immunology Baylor College of Medicine Houston TX.,6 Biology of Inflammation Center Baylor College of Medicine Houston TX.,10 Center for Translational Research in Inflammatory Diseases Michael E. DeBakey VA Houston TX
| | - Farrah Kheradmand
- 1 Department of Medicine, Pulmonary and Critical Care Baylor College of Medicine Houston TX.,3 Cardiovascular Research Institute Baylor College of Medicine Houston TX.,5 Departments of Pathology and Immunology Baylor College of Medicine Houston TX.,6 Biology of Inflammation Center Baylor College of Medicine Houston TX.,10 Center for Translational Research in Inflammatory Diseases Michael E. DeBakey VA Houston TX
| | - Scott A LeMaire
- 2 Division of Cardiothoracic Surgery Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX.,7 Department of Cardiovascular Surgery Texas Heart Institute Houston TX.,11 CHI St Luke's Health-Baylor St Luke's Medical Center Houston TX
| |
Collapse
|
7
|
COPD: To Be or Not to Be, That is the Question. Am J Med 2019; 132:1271-1278. [PMID: 31152719 PMCID: PMC8359778 DOI: 10.1016/j.amjmed.2019.04.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 04/26/2019] [Accepted: 04/28/2019] [Indexed: 01/01/2023]
Abstract
As our knowledge on the natural history of chronic obstructive pulmonary disease (COPD) progresses, a conceptual model simply based on an accelerated decline of lung function in adult life in response to smoking has become inadequate to capture the complexity of this disease, and increasing attention is being given to possible contributions from events or alterations of developmental processes that take place earlier in life. In addition, a remarkable heterogeneity has emerged among the pathobiological mechanisms that are involved in different phenotypes of COPD, suggesting that an effective disease management will require individualized treatment approaches largely based on the underlying biological mechanisms (endotypes). In this review, we will discuss the many faces of COPD from an epidemiological, pathobiological, and clinical standpoint and argue that airflow limitation encompasses a number of manifestations that are too diverse to be still clustered under the same diagnostic label.
Collapse
|
8
|
Gu BH, Sprouse ML, Madison MC, Hong MJ, Yuan X, Tung HY, Landers CT, Song LZ, Corry DB, Bettini M, Kheradmand F. A Novel Animal Model of Emphysema Induced by Anti-Elastin Autoimmunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:349-359. [PMID: 31182478 PMCID: PMC6688643 DOI: 10.4049/jimmunol.1900113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 05/13/2019] [Indexed: 12/21/2022]
Abstract
Loss of immune tolerance to self-antigens can promote chronic inflammation and disrupt the normal function of multiple organs, including the lungs. Degradation of elastin, a highly insoluble protein and a significant component of the lung structural matrix, generates proinflammatory molecules. Elastin fragments (EFs) have been detected in the serum of smokers with emphysema, and elastin-specific T cells have also been detected in the peripheral blood of smokers with emphysema. However, an animal model that could recapitulate T cell-specific autoimmune responses by initiating and sustaining inflammation in the lungs is lacking. In this study, we report an animal model of autoimmune emphysema mediated by the loss of tolerance to elastin. Mice immunized with a combination of human EFs plus rat EFs but not mouse EFs showed increased infiltration of innate and adaptive immune cells to the lungs and developed emphysema. We cloned and expanded mouse elastin-specific CD4+ T cells from the lung and spleen of immunized mice. Finally, we identified TCR sequences from the autoreactive T cell clones, suggesting possible pathogenic TCRs that can cause loss of immune tolerance against elastin. This new autoimmune model of emphysema provides a useful tool to examine the immunological factors that promote loss of immune tolerance to self.
Collapse
Affiliation(s)
- Bon-Hee Gu
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, TX 77030
| | - Maran L Sprouse
- Section of Diabetes and Endocrinology, Department of Pediatrics, McNair Medical Institute, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030
| | - Matthew C Madison
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, TX 77030
| | - Monica J Hong
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, TX 77030
| | - Xiaoyi Yuan
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, TX 77030
| | - Hui-Ying Tung
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, TX 77030
| | - Cameron T Landers
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, TX 77030
| | - Li-Zhen Song
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, TX 77030
| | - David B Corry
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, TX 77030
- Center for Translational Research in Inflammatory Diseases, Michael E. DeBakey VA Medical Center, Houston, TX 77030
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030; and
- Biology of Inflammation Center, Baylor College of Medicine, Houston, TX 77030
| | - Maria Bettini
- Section of Diabetes and Endocrinology, Department of Pediatrics, McNair Medical Institute, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030;
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030; and
- Biology of Inflammation Center, Baylor College of Medicine, Houston, TX 77030
| | - Farrah Kheradmand
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, TX 77030;
- Center for Translational Research in Inflammatory Diseases, Michael E. DeBakey VA Medical Center, Houston, TX 77030
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030; and
- Biology of Inflammation Center, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
9
|
Yuan X, Chang CY, You R, Shan M, Gu BH, Madison MC, Diehl G, Perusich S, Song LZ, Cornwell L, Rossen RD, Wetsel R, Kimal R, Coarfa C, Eltzschig HK, Corry DB, Kheradmand F. Cigarette smoke-induced reduction of C1q promotes emphysema. JCI Insight 2019; 5:124317. [PMID: 31112138 DOI: 10.1172/jci.insight.124317] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Alteration of innate immune cells in the lungs can promote loss of peripheral tolerance that leads to autoimmune responses in cigarette smokers. Development of autoimmunity in smokers with emphysema is also strongly linked to the expansion of autoreactive T helper (Th) cells expressing interferon gamma (Th1), and interleukin 17A (Th17). However, the mechanisms responsible for enhanced self-recognition and reduced immune tolerance in smoker with emphysema remain less clear. Here we show that C1q, a component of the complement protein 1 complex (C1), is downregulated in lung CD1a+ antigen presenting cells (APCs) isolated from emphysematous human, and mouse lung APCs after chronic cigarette smoke exposure. C1q potentiated the function of APCs to differentiate CD4+ T cells to Tregs, while it inhibited Th17 cell development and proliferation. Mice deficient in C1q that were exposed to chronic smoke exhibited exaggerated lung inflammation marked by increased Th17 cells, while reconstitution of C1q in the lungs enhanced Tregs abundance, dampened smoke-induced lung inflammation, and reversed established emphysema. Our findings demonstrate that cigarette smoke-mediated loss of C1q could play a key role in reduced peripheral tolerance, which could be explored to treat emphysema.
Collapse
Affiliation(s)
- Xiaoyi Yuan
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, Texas, USA
| | - Cheng-Yen Chang
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, Texas, USA
| | - Ran You
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, Texas, USA
| | - Ming Shan
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, Texas, USA
| | - Bon Hee Gu
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, Texas, USA
| | - Matthew C Madison
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, Texas, USA
| | - Gretchen Diehl
- Department of Molecular Virology and Microbiology Baylor College of Medicine, Houston, Texas, USA
| | - Sarah Perusich
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, Texas, USA
| | - Li-Zhen Song
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, Texas, USA
| | - Lorraine Cornwell
- Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Roger D Rossen
- Center for Translational Research in Inflammatory Diseases, Michael E. DeBakey VA, Houston, Texas, USA
| | - Rick Wetsel
- Institute of Molecular Medicine, UT Health Science Center of Houston, Houston, Texas, USA
| | - Rajapakshe Kimal
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Cristian Coarfa
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Holger K Eltzschig
- Department of Anesthesiology, UT Health Science Center at Houston, Houston, Texas, USA
| | - David B Corry
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, Texas, USA.,Center for Translational Research in Inflammatory Diseases, Michael E. DeBakey VA, Houston, Texas, USA.,Departments of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA.,Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas, USA
| | - Farrah Kheradmand
- Department of Medicine, Pulmonary and Critical Care, Baylor College of Medicine, Houston, Texas, USA.,Center for Translational Research in Inflammatory Diseases, Michael E. DeBakey VA, Houston, Texas, USA.,Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas, USA.,Departments of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA.,Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
10
|
Soriano JB, Polverino F, Cosio BG. What is early COPD and why is it important? Eur Respir J 2018; 52:13993003.01448-2018. [PMID: 30309976 DOI: 10.1183/13993003.01448-2018] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/03/2018] [Indexed: 01/31/2023]
Abstract
There is increasing interest in the origins of chronic obstructive pulmonary disease (COPD), as it is envisaged that preventive efforts and treatment can modify its clinical course. The concept of early COPD is not new, but it has recently regained interest, given new population data, recent cellular and molecular advances and insights from clinical trials. To date, many knowledge gaps in the nature of early COPD still exist, mainly because COPD has always been considered a disease of the elderly, and little attention has been paid to the pathological changes occurring in the lungs of individuals at risk before they develop clinically evident COPD. Future studies should focus on identifying early pathological manifestations of COPD in order to prevent its progression in susceptible individuals. In this review, we aim to summarise what is known on early COPD, from the epidemiological, cellular and clinical perspectives.
Collapse
Affiliation(s)
- Joan B Soriano
- Instituto de Investigación, Hospital Universitario de la Princesa (IISP), Universidad Autónoma de Madrid, Madrid, Spain.,CIBER-Centro de Investigación en Red de Enfermedades Respiratorias, Madrid, Spain
| | - Francesca Polverino
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, USA.,Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | - Borja G Cosio
- CIBER-Centro de Investigación en Red de Enfermedades Respiratorias, Madrid, Spain.,Dept of Respiratory Medicine, Hospital Universitari Son Espases-IdISBa, Palma, Spain
| |
Collapse
|
11
|
Gu BH, Madison MC, Corry D, Kheradmand F. Matrix remodeling in chronic lung diseases. Matrix Biol 2018; 73:52-63. [PMID: 29559389 PMCID: PMC6141350 DOI: 10.1016/j.matbio.2018.03.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 02/08/2018] [Accepted: 03/15/2018] [Indexed: 12/11/2022]
Abstract
Multicellular organisms synthesize and renew components of their subcellular and scaffolding proteins, collectively known as the extracellular matrix molecules (ECMs). In the lung, ECMs maintain tensile strength, elasticity, and dictate the specialized function of multiple cell lineages. These functions are critical in lung homeostatic processes including cellular migration and proliferation during morphogenesis or in response to repair. Alterations in lung ECMs that expose cells to new cryptic fragments, generated in response to endogenous proteinases or exogenous toxins, are associated with the development of several common respiratory diseases. How lung ECMs provide or relay vital signals to epithelial and mesenchymal cells has shed new light on development and progression of several common chronic respiratory diseases. This review will consider how ECMs regulate lung homeostasis and their reorganization under pathological conditions that can modulate the inflammatory diseases asthma, chronic obstructive pulmonary disease (COPD), and idiopathic pulmonary fibrosis (IPF). Better understanding of changes in the distribution of lung ECM could provide novel therapeutic approaches to treat chronic lung diseases.
Collapse
Affiliation(s)
- Bon-Hee Gu
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Matthew C Madison
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Interdepartmental Program in Translational Biology and Molecular Medicine Houston, TX 77030, USA
| | - David Corry
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Interdepartmental Program in Translational Biology and Molecular Medicine Houston, TX 77030, USA; Center for Translational Research in Inflammatory Diseases, Michael E. DeBakey VA, Houston, TX 77030, USA; Biology of Inflammation Center, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Farrah Kheradmand
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Interdepartmental Program in Translational Biology and Molecular Medicine Houston, TX 77030, USA; Center for Translational Research in Inflammatory Diseases, Michael E. DeBakey VA, Houston, TX 77030, USA; Biology of Inflammation Center, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
12
|
Liu Y, O’Brien JL, Ajami NJ, Scheurer ME, Amirian ES, Armstrong G, Tsavachidis S, Thrift AP, Jiao L, Wong MC, Smith DP, Spitz MR, Bondy ML, Petrosino JF, Kheradmand F. Lung tissue microbial profile in lung cancer is distinct from emphysema. Am J Cancer Res 2018; 8:1775-1787. [PMID: 30323970 PMCID: PMC6176189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 02/11/2018] [Indexed: 06/08/2023] Open
Abstract
OBJECTIVES The composition and structure of site-specific microbiota have been investigated as potential biomarkers for a variety of chronic inflammatory diseases and cancers. While many studies have focused on the changes in the airway microbiota using respiratory specimens from patients with various respiratory diseases, more research is needed to explore the microbial profiles within the distal lung parenchyma in smokers with lung cancer and/or emphysema. MATERIALS AND METHODS To describe and contrast lung tissue-associated microbial signatures in smokers with lung cancer and/or emphysema, we employed culture-independent pyrosequencing of 16S rRNA gene hypervariable V4 region and compositional analysis in non-malignant lung tissue samples obtained from 40 heavy smokers, including 10 emphysema-only, 11 lung cancer-only, and 19 with both lung cancer and emphysema. RESULTS AND CONCLUSION The emphysema-only group presented a lower bacterial community evenness defined by a significantly lower Shannon diversity index compared to the lung cancer patients with or without emphysema (P = 0.006). Furthermore, community compositions of lung cancer patients with or without emphysema were characterized by a significantly lower abundance of Proteobacteria (primary the genera Acinetobacter and Acidovorax) and higher prevalence of Firmicutes (Streptococcus) and Bacteroidetes (Prevotella), compared to emphysema-only patients. In conclusion, the lung microbial composition and communities structures of smokers with lung cancer are distinct from the emphysema-only patients. Although preliminary, our findings suggest that lung microbiome changes could be a biomarker of lung cancer that could eventually be used to help screening for the disease.
Collapse
Affiliation(s)
- Yanhong Liu
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of MedicineHouston, TX 77030, USA
- Department of Medicine, Baylor College of MedicineHouston, TX 77030, USA
| | - Jacqueline L O’Brien
- Department of Molecular Virology and Microbiology, Baylor College of MedicineHouston, TX 77030, USA
- The Alkek Center for Metagenomics and Microbiome Research, Baylor College of MedicineHouston, TX 77030, USA
| | - Nadim J Ajami
- Department of Molecular Virology and Microbiology, Baylor College of MedicineHouston, TX 77030, USA
- The Alkek Center for Metagenomics and Microbiome Research, Baylor College of MedicineHouston, TX 77030, USA
| | - Michael E Scheurer
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of MedicineHouston, TX 77030, USA
- Department of Pediatrics, Baylor College of MedicineHouston, TX 77030, USA
| | - E Susan Amirian
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of MedicineHouston, TX 77030, USA
| | - Georgina Armstrong
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of MedicineHouston, TX 77030, USA
| | - Spiridon Tsavachidis
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of MedicineHouston, TX 77030, USA
| | - Aaron P Thrift
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of MedicineHouston, TX 77030, USA
- Department of Medicine, Baylor College of MedicineHouston, TX 77030, USA
| | - Li Jiao
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of MedicineHouston, TX 77030, USA
- Michael E. DeBakey Veterans Affairs Medical Center, Baylor College of MedicineHouston, TX 77030, USA
| | - Matthew C Wong
- Department of Molecular Virology and Microbiology, Baylor College of MedicineHouston, TX 77030, USA
- The Alkek Center for Metagenomics and Microbiome Research, Baylor College of MedicineHouston, TX 77030, USA
| | - Daniel P Smith
- Department of Molecular Virology and Microbiology, Baylor College of MedicineHouston, TX 77030, USA
- The Alkek Center for Metagenomics and Microbiome Research, Baylor College of MedicineHouston, TX 77030, USA
| | - Margaret R Spitz
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of MedicineHouston, TX 77030, USA
- Department of Medicine, Baylor College of MedicineHouston, TX 77030, USA
| | - Melissa L Bondy
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of MedicineHouston, TX 77030, USA
- Department of Medicine, Baylor College of MedicineHouston, TX 77030, USA
| | - Joseph F Petrosino
- Department of Molecular Virology and Microbiology, Baylor College of MedicineHouston, TX 77030, USA
- The Alkek Center for Metagenomics and Microbiome Research, Baylor College of MedicineHouston, TX 77030, USA
| | - Farrah Kheradmand
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of MedicineHouston, TX 77030, USA
- Department of Medicine, Baylor College of MedicineHouston, TX 77030, USA
- Michael E. DeBakey Veterans Affairs Medical Center, Baylor College of MedicineHouston, TX 77030, USA
| |
Collapse
|
13
|
Zhou Y, Horowitz JC, Naba A, Ambalavanan N, Atabai K, Balestrini J, Bitterman PB, Corley RA, Ding BS, Engler AJ, Hansen KC, Hagood JS, Kheradmand F, Lin QS, Neptune E, Niklason L, Ortiz LA, Parks WC, Tschumperlin DJ, White ES, Chapman HA, Thannickal VJ. Extracellular matrix in lung development, homeostasis and disease. Matrix Biol 2018. [PMID: 29524630 DOI: 10.1016/j.matbio.2018.03.005] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The lung's unique extracellular matrix (ECM), while providing structural support for cells, is critical in the regulation of developmental organogenesis, homeostasis and injury-repair responses. The ECM, via biochemical or biomechanical cues, regulates diverse cell functions, fate and phenotype. The composition and function of lung ECM become markedly deranged in pathological tissue remodeling. ECM-based therapeutics and bioengineering approaches represent promising novel strategies for regeneration/repair of the lung and treatment of chronic lung diseases. In this review, we assess the current state of lung ECM biology, including fundamental advances in ECM composition, dynamics, topography, and biomechanics; the role of the ECM in normal and aberrant lung development, adult lung diseases and autoimmunity; and ECM in the regulation of the stem cell niche. We identify opportunities to advance the field of lung ECM biology and provide a set recommendations for research priorities to advance knowledge that would inform novel approaches to the pathogenesis, diagnosis, and treatment of chronic lung diseases.
Collapse
Affiliation(s)
- Yong Zhou
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, United States.
| | - Jeffrey C Horowitz
- Division of Pulmonary and Critical Care Medicine, University of Michigan, United States.
| | - Alexandra Naba
- Department of Physiology & Biophysics, University of Illinois at Chicago, United States.
| | | | - Kamran Atabai
- Lung Biology Center, University of California, San Francisco, United States.
| | | | | | - Richard A Corley
- Systems Toxicology & Exposure Science, Pacific Northwest National Laboratory, United States.
| | - Bi-Sen Ding
- Weill Cornell Medical College, United States.
| | - Adam J Engler
- Sanford Consortium for Regenerative Medicine, University of California, San Diego, United States.
| | - Kirk C Hansen
- Biochemistry & Molecular Genetics, University of Colorado Denver, United States.
| | - James S Hagood
- Pediatric Respiratory Medicine, University of California San Diego, United States.
| | - Farrah Kheradmand
- Division of Pulmonary and Critical Care, Baylor College of Medicine, United States.
| | - Qing S Lin
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, United States.
| | - Enid Neptune
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, United States.
| | - Laura Niklason
- Department of Anesthesiology, Yale University, United States.
| | - Luis A Ortiz
- Division of Environmental and Occupational Health, University of Pittsburgh, United States.
| | - William C Parks
- Department of Medicine, Cedars-Sinai Medical Center, United States.
| | - Daniel J Tschumperlin
- Department of Physiology & Biomedical Engineering, Mayo Clinic College of Medicine, United States.
| | - Eric S White
- Division of Pulmonary and Critical Care Medicine, University of Michigan, United States.
| | - Harold A Chapman
- Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, United States.
| | - Victor J Thannickal
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, United States.
| |
Collapse
|
14
|
Iman M, Rezaei R, Azimzadeh Jamalkandi S, Shariati P, Kheradmand F, Salimian J. Th17/Treg immunoregulation and implications in treatment of sulfur mustard gas-induced lung diseases. Expert Rev Clin Immunol 2017; 13:1173-1188. [PMID: 28994328 DOI: 10.1080/1744666x.2017.1389646] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Sulfur mustard (SM) is an extremely toxic gas used in chemical warfare to cause massive lung injury and death. Victims exposed to SM gas acutely present with inhalational lung injury, but among those who survive, some develop obstructive airway diseases referred to as SM-lung syndrome. Pathophysiologically, SM-lung shares many characteristics with smoking-induced chronic obstructive pulmonary disease (COPD), including airway remodeling, goblet cell metaplasia, and obstructive ventilation defect. Some of the hallmarks of COPD pathogenesis, which include dysregulated lung inflammation, neutrophilia, recruitment of interleukin 17A (IL -17A) expressing CD4+T cells (Th17), and the paucity of lung regulatory T cells (Tregs), have also been described in SM-lung. Areas covered: A literature search was performed using the MEDLINE, EMBASE, and Web of Science databases inclusive of all literature prior to and including May 2017. Expert commentary: Here we review some of the recent findings that suggest a role for Th17 cell-mediated inflammatory changes associated with pulmonary complications in SM-lung and suggest new therapeutic approaches that could potentially alter disease progression with immune modulating biologics that can restore the lung Th17/Treg balance.
Collapse
Affiliation(s)
- Maryam Iman
- a Chemical Injuries Research Center , Baqiyatallah University of Medical Sciences , Tehran , Iran
| | - Ramazan Rezaei
- b Department of Immunology , School of Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | | | - Parvin Shariati
- c Department of Industrial and Environmental Biotechnology , National Institute of Genetic Engineering and Biotechnology , Tehran , Iran
| | - Farrah Kheradmand
- d Center for Translational Research in Inflammatory Diseases, Michael E. DeBakey VA, & Department of Medicine , Pulmonary and Critical Care, Baylor College of Medicine , Houston , TX , USA
| | - Jafar Salimian
- a Chemical Injuries Research Center , Baqiyatallah University of Medical Sciences , Tehran , Iran
| |
Collapse
|
15
|
Meghraoui-Kheddar A, Pierre A, Sellami M, Audonnet S, Lemaire F, Le Naour R. Elastin receptor (S-gal) occupancy by elastin peptides modulates T-cell response during murine emphysema. Am J Physiol Lung Cell Mol Physiol 2017; 313:L534-L547. [DOI: 10.1152/ajplung.00465.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 05/04/2017] [Accepted: 05/27/2017] [Indexed: 11/22/2022] Open
Abstract
Chronic obstructive pulmonary disease and emphysema are associated with increased elastin peptides (EP) production because of excessive breakdown of lung connective tissue. We recently reported that exposure of mice to EP elicited hallmark features of emphysema. EP effects are largely mediated through a receptor complex that includes the elastin-binding protein spliced-galactosidase (S-gal). In previous studies, we established a correlation between cytokine production and S-gal protein expression in EP-treated immune cells. In this study, we investigated the S-gal-dependent EP effects on T-helper (Th) and T-cytotoxic (Tc) responses during murine EP-triggered pulmonary inflammation. C57BL/6J mice were endotracheally instilled with the valine-glycine-valine-alanine-proline-glycine (VGVAPG) elastin peptide, and, 21 days after treatment, local and systemic T-lymphocyte phenotypes were analyzed at cytokine and transcription factor expression levels by multicolor flow cytometry. Exposure of mice to the VGVAPG peptide resulted in a significant increase in the proportion of the CD4+ and CD8+ T cells expressing the cytokines IFN-γ or IL-17a and the transcription factors T-box expressed in T cells or retinoic acid-related orphan receptor-γt (RORγt) without effects on IL-4 and Gata-binding protein 3 to DNA sequence [A/T]GATA[A/G] expression. These effects were maximized when each T-cell subpopulation was challenged ex vivo with EP, and they were inhibited in vivo when an analogous peptide antagonizing the EP/S-gal interactions was instilled together with the VGVAPG peptide. This study demonstrates that, during murine emphysema, EP-S-gal interactions contribute to a Th-1 and Th-17 proinflammatory T-cell response combined with a Tc-1 response. Our study also highlights the S-gal receptor as a putative pharmacological target to modulate such an immune response.
Collapse
Affiliation(s)
| | - Alexandre Pierre
- EA4683, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France; and
| | - Mehdi Sellami
- EA4683, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France; and
| | - Sandra Audonnet
- Plateau Technique de Cytométrie en Flux, Plateforme Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Flora Lemaire
- EA4683, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France; and
| | - Richard Le Naour
- EA4683, SFR CAP-Santé, Université de Reims Champagne-Ardenne, Reims, France; and
| |
Collapse
|
16
|
Briend E, Ferguson GJ, Mori M, Damera G, Stephenson K, Karp NA, Sethi S, Ward CK, Sleeman MA, Erjefält JS, Finch DK. IL-18 associated with lung lymphoid aggregates drives IFNγ production in severe COPD. Respir Res 2017; 18:159. [PMID: 28830544 PMCID: PMC5568255 DOI: 10.1186/s12931-017-0641-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 08/10/2017] [Indexed: 11/16/2022] Open
Abstract
Background Increased interferon gamma (IFNγ) release occurs in Chronic Obstructive Pulmonary Disease (COPD) lungs. IFNγ supports optimal viral clearance, but if dysregulated could increase lung tissue destruction. Methods The present study investigates which mediators most closely correlate with IFNγ in sputum in stable and exacerbating disease, and seeks to shed light on the spatial requirements for innate production of IFNγ, as reported in mouse lymph nodes, to observe whether such microenvironmental cellular organisation is relevant to IFNγ production in COPD lung. Results We show tertiary follicle formation in severe disease alters the dominant mechanistic drivers of IFNγ production, because cells producing interleukin-18, a key regulator of IFNγ, are highly associated with such structures. Interleukin-1 family cytokines correlated with IFNγ in COPD sputum. We observed that the primary source of IL-18 in COPD lungs was myeloid cells within lymphoid aggregates and IL-18 was increased in severe disease. IL-18 released from infected epithelium or from activated myeloid cells, was more dominant in driving IFNγ when IL-18-producing and responder cells were in close proximity. Conclusions Unlike tight regulation to control infection spread in lymphoid organs, this local interface between IL-18-expressing and responder cell is increasingly supported in lung as disease progresses, increasing its potential to increase tissue damage via IFNγ. Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0641-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Emmanuel Briend
- MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.,Present address: Agenus Ltd, Cambridge, UK
| | | | - Michiko Mori
- Department of Experimental Medical Science, BMC D12, Lund University, SE-221 84, Lund, Sweden
| | - Gautam Damera
- MedImmune LLC, 1 MedImmune Way, Gaithersburg, MD, USA
| | - Katherine Stephenson
- MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.,Present address: University of Nottingham, Nottingham, UK
| | - Natasha A Karp
- Quantitative Biology IMED, AstraZeneca R&D, Cambridge, UK
| | - Sanjay Sethi
- Department of Medicine, University at Buffalo, 3495 Bailey Avenue, Buffalo, NY, 14215, USA
| | - Christine K Ward
- MedImmune LLC, 1 MedImmune Way, Gaithersburg, MD, USA.,Present address: Bristol-Myers Squibb, Princeton, NJ, USA
| | - Matthew A Sleeman
- MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.,Present address: Regeneron Pharmaceuticals Inc, Tarrytown, NY, USA
| | - Jonas S Erjefält
- Department of Experimental Medical Science, BMC D12, Lund University, SE-221 84, Lund, Sweden.,Department of Respiratory Medicine and Allergology, Lund University Hospital, Lund, Sweden
| | - Donna K Finch
- MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.
| |
Collapse
|
17
|
Verhoeven J, Lambrecht A, Verbrugghe P, Herijgers P, Fourneau I. Remnant Epitope Autoimmunity in Human Abdominal Aortic Aneurysm: A Pilot Study with Elastin Peptides. Ann Vasc Surg 2017; 44:408-413. [PMID: 28602894 DOI: 10.1016/j.avsg.2017.05.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 05/28/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a prevalent disease affecting around 5% of the population aged more than 65 years. The exact etiology and physiopathology of AAA still raises questions, and elective surgery is currently the only treatment option for this often progressive disease. In this study, we hypothesized and tested a pathophysiological model that depicts AAA as an inflammation-triggered autoimmune disease with remnant vessel wall peptide fragments as the antigen. METHODS A pilot study with male AAA patients (n = 14) and male controls (n = 8) was conducted. In both study groups, peripheral blood monocytes and plasma were separated from whole blood by centrifugation. An ELISpot test was performed on cultured white blood cells for the presence of elastin-specific T-lymphocytes. An Enzyme-linked immuno sorbent assay (ELISA) was performed on plasma for the presence of elastin-specific IgG molecules. RESULTS ELISpot interferon-gamma secretion in AAA (7.7 ± 9.5%) and control (4.6 ± 3.5%) and ELISA anti-elastin IgG titer in AAA (77.5 ± 17.8%) and control (78.2 ± 31.5%) were not significantly different (P = 0.94 and P = 0.55, respectively). Both results are expressed as a percentage relative to the respective positive and negative control. CONCLUSIONS The results of our pilot study did not indicate a clear and invariable autoimmune process directed against remnant elastin peptide fragments. Further research into the model mechanics and a possible antigen is still necessary. In the mean time, the model as presented here already offers a pathophysiological framework to further research into the possible remnant epitope-driven AAA etiology.
Collapse
Affiliation(s)
- Jelle Verhoeven
- Department of Cardiovascular Sciences, KU Leuven-University of Leuven, Leuven, Belgium.
| | - Alix Lambrecht
- Department of Cardiovascular Sciences, KU Leuven-University of Leuven, Leuven, Belgium
| | - Peter Verbrugghe
- Department of Cardiovascular Sciences, KU Leuven-University of Leuven, Leuven, Belgium
| | - Paul Herijgers
- Department of Cardiovascular Sciences, KU Leuven-University of Leuven, Leuven, Belgium
| | - Inge Fourneau
- Department of Cardiovascular Sciences, KU Leuven-University of Leuven, Leuven, Belgium
| |
Collapse
|
18
|
KHERADMAND FARRAH, YOU R, HEE GU BON, CORRY D. Cigarette Smoke and DNA Cleavage Promote Lung Inflammation and Emphysema. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2017; 128:222-233. [PMID: 28790504 PMCID: PMC5525399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Smoking-related lung diseases are among the most preventable and incurable ailments in the world. Smokers are at increased risk of developing chronic obstructive pulmonary disease that can be further complicated by emphysema and lung cancer. A subset of former smokers shows persistent lung inflammation and progressive loss of lung function, indicating a role for activation of acquired immunity in smoking-induced lung diseases. In addition to the well-established noxious effects of volatile compounds in cigarette smoke, incomplete combustion of tobacco generates nano-sized carbon black (nCB) that accumulate in lung myeloid dendritic cells and macrophages. Experimentally, intra-nasal instillation nCB can cause airway inflammation and emphysema in mice, underscoring their pathogenic role in inflammatory lung diseases. High throughput analyses of macrophages that have engulfed nCB reveal de novo activation of DNA repair enzymes, and histological studies provide evidence for DNA double-stranded breaks. Emphysematous lung myeloid dendritic cells that contain nCB express pro-inflammatory cytokines, and can efficiently differentiate naive CD4 T cells to interferon-g-secreting T helper 1 and interleukin 17A expressing cell subsets. Together these findings indicate that nCB accumulation in lung innate immune cells can initiate and sustain lung inflammation and promote emphysema development.
Collapse
Affiliation(s)
- FARRAH KHERADMAND
- Correspondence and reprint requests: Farrah Kheradmand, MD, Baylor College of Medicine,
1 Baylor Plaza, Houston, Texas 77030713-798-8622
| | | | | | | |
Collapse
|
19
|
Loss of Peripheral Tolerance in Emphysema. Phenotypes, Exacerbations, and Disease Progression. Ann Am Thorac Soc 2016; 12 Suppl 2:S164-8. [PMID: 26595734 DOI: 10.1513/annalsats.201503-115aw] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Heterogeneity in the development and progression of cigarette smoke-induced lung diseases strongly argues for a need to improve the clinical and phenotypic characterization of patients with chronic obstructive lung disease and emphysema. Smokers with emphysema are at a much higher risk for accelerated loss of lung function, increased cardiovascular morbidity, and development of lung cancer. Recent evidence in human translational studies and animal models suggests that emphysema is associated with activation of specialized antigen-presenting cells and that cigarette smoke can disrupt the induction of immune tolerance in the lungs. Quantitative assessment of cytokines expressed by autoreactive T lymphocytes in response to human lung elastin fragments has shown a strong positive correlation between T helper Type 1 (Th1) and Th17 cells' immune responses and emphysema. In search of factors that could reduce the threshold for induction of autoimmune inflammation, we have discovered that cleavage of complement protein 3 (C3) generates bioactive molecules (e.g., C3a) and activates lung antigen-presenting cells. The autocrine and paracrine function of C3a and its receptor are required in T cell-mediated inflammatory responses to cigarette smoke in both human and preclinical models of emphysema. Targeting upstream molecules that reduce the potential for generation of autoreactive T cells could lead to the development of novel therapeutics to prevent progression of emphysema in smokers.
Collapse
|
20
|
Curtis JL. At the Checkpoint: Lung CD8(+) T Cells, Respiratory Viruses, and Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2016; 193:600-2. [PMID: 26977966 DOI: 10.1164/rccm.201511-2122ed] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Jeffrey L Curtis
- 1 Pulmonary & Critical Care Medicine Section VA Ann Arbor Healthcare System Ann Arbor, Michigan and.,2 Department of Internal Medicine and.,3 Graduate Program in Immunology University of Michigan Health System Ann Arbor, Michigan
| |
Collapse
|
21
|
Wells JM, Gaggar A, Nicod LP. A Fragment in Time. Elastin Peptides and Progression of Emphysema. Am J Respir Crit Care Med 2016; 192:1141-3. [PMID: 26568233 DOI: 10.1164/rccm.201508-1590ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- J Michael Wells
- 1 Division of Pulmonary, Allergy, and Critical Care and.,2 Lung Health Center University of Alabama at Birmingham Birmingham, Alabama and.,3 Birmingham VA Medical Center Birmingham, Alabama
| | - Amit Gaggar
- 1 Division of Pulmonary, Allergy, and Critical Care and.,2 Lung Health Center University of Alabama at Birmingham Birmingham, Alabama and.,3 Birmingham VA Medical Center Birmingham, Alabama
| | - Laurent P Nicod
- 4 Service de Pneumologie University Hospital Lausanne, Switzerland
| |
Collapse
|
22
|
Bhavani S, Tsai CL, Perusich S, Hesselbacher S, Coxson H, Pandit L, Corry DB, Kheradmand F. Clinical and Immunological Factors in Emphysema Progression. Five-Year Prospective Longitudinal Exacerbation Study of Chronic Obstructive Pulmonary Disease (LES-COPD). Am J Respir Crit Care Med 2015; 192:1171-8. [PMID: 26241705 PMCID: PMC4731622 DOI: 10.1164/rccm.201504-0736oc] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/03/2015] [Indexed: 01/18/2023] Open
Abstract
RATIONALE Cross-sectional studies of T-cell responses to self-antigens correlate with baseline emphysema severity. OBJECTIVES We investigated whether clinical and/or immunological factors could predict disease progression, such as emphysema, FEV1, and 6-minute-walk distance (6MWD), in former and active smokers in a 5-year prospective study. METHODS We recruited 224 ever smokers over 40 years of age and with greater than a 15 pack-year smoking history. MEASUREMENTS AND MAIN RESULTS Repeated spirometry, 6MWD, and peripheral blood T-cell cytokine responses to lung elastin fragments were measured. Baseline and repeat chest computed tomography (CT) scans (34 to 65 mo apart) were used to quantify emphysema progression. Of the 141 ever-smokers with baseline and repeat CT scans, the mean (SD) annual rate of change in percent emphysema was +0.46 (0.92), ranging from -1.8 to +4.1. In multivariable analyses, the rate of emphysema progression was greater in subjects who had lower body mass index (BMI) (+0.15 per 5-unit decrease in BMI; 95% confidence interval, +0.03 to +0.29). In active smokers, increased IFN-γ and IL-6 T-cell responses had a positive association with the annual rate of emphysema progression. Male sex and IL-6 T-cell responses to elastin fragments were significantly associated with annual 6MWD decline, whereas IL-13 was associated with an increase in annual 6MWD. CONCLUSIONS The rate of emphysema progression quantified by CT scans among ever-smokers was highly variable; clinical factors and biomarkers explained only some of the variability. Aggressive clinical care that targets active smokers with autoreactive T cells and low BMI may temporize progression of emphysema.
Collapse
Affiliation(s)
| | - Chu-Lin Tsai
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Emergency Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | | | | | - Harvey Coxson
- Department of Radiology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Lavannya Pandit
- Department of Medicine
- Michael E. DeBakey Veterans Affairs Center for Translational Research on Inflammatory Diseases, Houston Texas
| | - David B. Corry
- Department of Medicine
- Department of Pathology and Immunology, and
- Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas
- Michael E. DeBakey Veterans Affairs Center for Translational Research on Inflammatory Diseases, Houston Texas
| | - Farrah Kheradmand
- Department of Medicine
- Department of Pathology and Immunology, and
- Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas
- Michael E. DeBakey Veterans Affairs Center for Translational Research on Inflammatory Diseases, Houston Texas
| |
Collapse
|
23
|
Lu W, You R, Yuan X, Yang T, Samuel ELG, Marcano DC, Sikkema WKA, Tour JM, Rodriguez A, Kheradmand F, Corry DB. The microRNA miR-22 inhibits the histone deacetylase HDAC4 to promote T(H)17 cell-dependent emphysema. Nat Immunol 2015; 16:1185-94. [PMID: 26437241 PMCID: PMC4597310 DOI: 10.1038/ni.3292] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/09/2015] [Indexed: 12/15/2022]
Abstract
Smoking-related emphysema is a chronic inflammatory disease driven by the T(H)17 subset of helper T cells through molecular mechanisms that remain obscure. Here we explored the role of the microRNA miR-22 in emphysema. We found that miR-22 was upregulated in lung myeloid dendritic cells (mDCs) of smokers with emphysema and antigen-presenting cells (APCs) of mice exposed to smoke or nanoparticulate carbon black (nCB) through a mechanism that involved the transcription factor NF-κB. Mice deficient in miR-22, but not wild-type mice, showed attenuated T(H)17 responses and failed to develop emphysema after exposure to smoke or nCB. We further found that miR-22 controlled the activation of APCs and T(H)17 responses through the activation of AP-1 transcription factor complexes and the histone deacetylase HDAC4. Thus, miR-22 is a critical regulator of both emphysema and T(H)17 responses.
Collapse
Affiliation(s)
- Wen Lu
- Department of Pathology &Immunology, Baylor College of Medicine, Houston, Texas, USA
| | - Ran You
- Department of Pathology &Immunology, Baylor College of Medicine, Houston, Texas, USA
| | - Xiaoyi Yuan
- Department of Pathology &Immunology, Baylor College of Medicine, Houston, Texas, USA
| | - Tianshu Yang
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | - James M Tour
- Department of Chemistry, Rice University, Houston, Texas, USA
| | - Antony Rodriguez
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Farrah Kheradmand
- Department of Pathology &Immunology, Baylor College of Medicine, Houston, Texas, USA
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
- Biology of Inflammation Center and the Michael E. DeBakey Virginia Center for Translational Research on Inflammatory Diseases, Houston, Texas, USA
| | - David B Corry
- Department of Pathology &Immunology, Baylor College of Medicine, Houston, Texas, USA
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
- Biology of Inflammation Center and the Michael E. DeBakey Virginia Center for Translational Research on Inflammatory Diseases, Houston, Texas, USA
| |
Collapse
|
24
|
Yuan X, Shan M, You R, Frazier MV, Hong MJ, Wetsel RA, Drouin S, Seryshev A, MD LZS, Cornwell L, Rossen RD, Corry DB, Kheradmand F. Activation of C3a receptor is required in cigarette smoke-mediated emphysema. Mucosal Immunol 2015; 8:874-85. [PMID: 25465103 PMCID: PMC4454642 DOI: 10.1038/mi.2014.118] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 10/23/2014] [Indexed: 02/04/2023]
Abstract
Exposure to cigarette smoke can initiate sterile inflammatory responses in the lung and activate myeloid dendritic cells (mDCs) that induce differentiation of T helper type 1 (Th1) and Th17 cells in the emphysematous lungs. Consumption of complement proteins increases in acute inflammation, but the contribution of complement protein 3 (C3) to chronic cigarette smoke-induced immune responses in the lung is not clear. Here, we show that following chronic exposure to cigarette smoke, C3-deficient (C3(-/-)) mice develop less emphysema and have fewer CD11b(+)CD11c(+) mDCs infiltrating the lungs as compared with wild-type mice. Proteolytic cleavage of C3 by neutrophil elastase releases C3a, which in turn increases the expression of its receptor (C3aR) on lung mDCs. Mice deficient in the C3aR (C3ar(-/-)) partially phenocopy the attenuated responses to chronic smoke observed in C3(-/-) mice. Consistent with a role for C3 in emphysema, C3 and its active fragments are deposited on the lung tissue of smokers with emphysema, and smoke-exposed mice. Together, these findings suggest a critical role for C3a through autocrine/paracrine induction of C3aR in the pathogenesis of cigarette smoke-induced sterile inflammation and provide new therapeutic targets for the treatment of emphysema.
Collapse
Affiliation(s)
- Xiaoyi Yuan
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Ming Shan
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Ran You
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Michael V. Frazier
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Monica Jeongsoo Hong
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Rick A. Wetsel
- Brown Foundation Institute of Molecular Medicine-Research Center for Immunology and Autoimmune Diseases, The University of Texas Medical School at Houston, Houston TX 77030
| | - Scott Drouin
- Brown Foundation Institute of Molecular Medicine-Research Center for Immunology and Autoimmune Diseases, The University of Texas Medical School at Houston, Houston TX 77030
| | - Alexander Seryshev
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | - Li-zhen Song MD
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
| | | | - Roger D Rossen
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Pathology and Immunology, Baylor College of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Michael E DeBakey VA Center for Translational Research on Inflammatory Diseases, Houston TX 77030
| | - David B. Corry
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Pathology and Immunology, Baylor College of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Biology of Inflammation Center, Baylor College of Medicine, Houston TX 77030
- Michael E DeBakey VA Center for Translational Research on Inflammatory Diseases, Houston TX 77030
| | - Farrah Kheradmand
- Department of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Pathology and Immunology, Baylor College of Medicine, The University of Texas Medical School at Houston, Houston TX 77030
- Biology of Inflammation Center, Baylor College of Medicine, Houston TX 77030
- Michael E DeBakey VA Center for Translational Research on Inflammatory Diseases, Houston TX 77030
| |
Collapse
|
25
|
Newton AH, Danahy DB, Chan MA, Benedict SH. Timely blockade of ICAM-1.LFA-1 interaction prevents disease onset in a mouse model of emphysema. Immunotherapy 2015; 7:621-9. [PMID: 26098520 DOI: 10.2217/imt.15.31] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM It is becoming apparent that emphysema is partly driven by self-reactive T cells inducing inflammatory damage. Thus, T cells become targets for therapy similar to other autoimmune diseases. Costimulatory blockade therapy targets disease-specific T cells, rendering them ineffective by blocking a necessary costimulatory event on the T-cell surface. This therapy is tested here in mouse emphysema. MATERIALS & METHODS Peptides representing contact domains of counter receptors LFA-1 and ICAM-1 were used as blockade therapy in elastase-induced emphysema. RESULTS When administered during the first week after disease induction, blockade prevented lung destruction, reduced leukocyte infiltration and inhibited the decrease in T-cell CD4:CD8 ratio, also common in human emphysema. CONCLUSION Costimulatory blockade therapy can affect the progress of emphysema.
Collapse
Affiliation(s)
- Amy H Newton
- Department of Molecular Biosciences, University of Kansas, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA.,Present address: Beirne B Carter Center for Immunology Research, Department of Microbiology, University of Virginia, VA, USA
| | - Derek B Danahy
- Department of Molecular Biosciences, University of Kansas, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA.,Present address: Graduate Program in Immunology, University of Iowa, IA, USA
| | - Marcia A Chan
- Division of Allergy, Asthma & Immunology, Children's Mercy Hospitals & Clinics, MO, USA
| | - Stephen H Benedict
- Department of Molecular Biosciences, University of Kansas, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA
| |
Collapse
|
26
|
Abstract
Matrikines originate from the fragmentation of extracellular matrix proteins and regulate cellular activities by interacting with specific receptors. Matrikines are implicated in inflammation, immune responses, organ development, wound repair, angiogenesis, atherosclerosis, tumor progression and metastasis due to their ability to alter cellular migration, chemotaxis, and mitogenesis. Matrix metalloproteinases (MMPs) degrade extracellular matrix components under normal circumstances and in disease processes. Of the 20 MMPs identified, MMP-1, MMP-2, MMP-8, MMP-9, and MMP-12 have been implicated in regulating the matrikines Val-Gly-Val-Ala-Pro-Gly (elastin peptide) and proline-glycine-proline (PGP). Elastin peptide fragments are generated by elastolytic enzymes and have implications in atherosclerosis, neovascularization, chronic obstructive pulmonary disease, skin disease, as well as tumor invasion and spread. PGP is produced through a multistep pathway that liberates the tripeptide fragment from extracellular collagen. PGP is best described for its role in neutrophil chemotaxis and is implicated in the pathogenesis of corneal ulcers and in chronic lung conditions. In chronic cigarette smoke related lung disease, the PGP pathway can become a self-propagating cycle of inflammation through cigarette-smoke mediated inhibition of leukotriene A4 hydrolase, the enzyme responsible for degrading PGP and halting acute inflammation. This review highlights the roles of MMPs in generating these important matrikines.
Collapse
|
27
|
Snelgrove R, Kheradmand F. Leukotriene A4 hydrolase: the Janus enzyme shows its ugly side in smokers. Am J Respir Crit Care Med 2014; 190:5-7. [PMID: 24983216 DOI: 10.1164/rccm.201405-0975ed] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Robert Snelgrove
- 1 National Heart and Lung Institute Imperial College London London, United Kingdom
| | | |
Collapse
|
28
|
Shan M, You R, Yuan X, Frazier MV, Porter P, Seryshev A, Hong JS, Song LZ, Zhang Y, Hilsenbeck S, Whitehead L, Zarinkamar N, Perusich S, Corry DB, Kheradmand F. Agonistic induction of PPARγ reverses cigarette smoke-induced emphysema. J Clin Invest 2014; 124:1371-81. [PMID: 24569375 DOI: 10.1172/jci70587] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 11/21/2013] [Indexed: 01/12/2023] Open
Abstract
The development of emphysema in humans and mice exposed to cigarette smoke is promoted by activation of an adaptive immune response. Lung myeloid dendritic cells (mDCs) derived from cigarette smokers activate autoreactive Th1 and Th17 cells. mDC-dependent activation of T cell subsets requires expression of the SPP1 gene, which encodes osteopontin (OPN), a pleiotropic cytokine implicated in autoimmune responses. The upstream molecular events that promote SPP1 expression and activate mDCs in response to smoke remain unknown. Here, we show that peroxisome proliferator-activated receptor γ (PPARG/Pparg) expression was downregulated in mDCs of smokers with emphysema and mice exposed to chronic smoke. Conditional knockout of PPARγ in APCs using Cd11c-Cre Pparg(flox/flox) mice led to spontaneous lung inflammation and emphysema that resembled the phenotype of smoke-exposed mice. The inflammatory phenotype of Cd11c-Cre Pparg(flox/flox) mice required OPN, suggesting an antiinflammatory mechanism in which PPARγ negatively regulates Spp1 expression in the lung. A 2-month treatment with a PPARγ agonist reversed emphysema in WT mice despite continual smoke exposure. Furthermore, endogenous PPARγ agonists were reduced in the plasma of smokers with emphysema. These findings reveal a proinflammatory pathway, in which reduced PPARγ activity promotes emphysema, and suggest that targeting this pathway in smokers could prevent and reverse emphysema.
Collapse
|