1
|
Okuno D, Akiyama Y, Sakamoto N, Tokito T, Yura H, Kido T, Ishimoto H, Ishimatsu Y, Takemoto S, Takazono T, Okamura H, Nishino T, Tanaka Y, Mukae H. Antifibrotic effects of IL-12-treated natural killer cells on collagen type I regulation in lung fibroblasts. Exp Cell Res 2025; 449:114584. [PMID: 40328414 DOI: 10.1016/j.yexcr.2025.114584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 04/05/2025] [Accepted: 04/29/2025] [Indexed: 05/08/2025]
Abstract
Idiopathic pulmonary fibrosis is a chronic fibrotic lung disease with limited treatment options, making the development of new therapies crucial. We previously demonstrated that γδ T cells, a subset of immune effector cells, exhibit antifibrotic properties. We have also shown that natural killer (NK) cells, another class of immune effectors, can be efficiently expanded in culture using interleukin-2 (IL-2) and interleukin-18 (IL-18). This study examined the effects of interleukin-12 (IL-12)-stimulated NK cells, expanded with IL-2 and IL-18, on type I collagen and α-smooth muscle actin (αSMA) expression in pulmonary fibroblasts. IL-12-stimulated human NK cells exhibited reduced cytotoxicity toward pulmonary fibroblasts while retaining their proliferative capacity. Co-culture of IL-12-stimulated NK cells with fibroblasts significantly suppressed type I collagen and αSMA expression, even without direct cell contact, indicating the involvement of soluble factors. Supernatants from IL-12-stimulated NK cells partially inhibited the expression of these antifibrotic factors, suggesting a dual mechanism: direct cell-cell interaction and soluble factor secretion. Interferon-γ (IFN-γ) in the supernatant significantly increased, and neutralizing anti-IFN-γ monoclonal antibody partially reversed type I collagen and αSMA suppression. Similarly, IL-12-stimulated murine NK cells suppressed type I collagen in mouse pulmonary fibroblasts. These findings suggest that IL-12-stimulated NK cells inhibit the expression of fibrosis-associated molecules via contact-dependent and -independent mechanisms, supporting their potential for adoptive cell therapy in pulmonary fibrosis.
Collapse
Affiliation(s)
- Daisuke Okuno
- Department of Respiratory Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Yoshiko Akiyama
- Department of Respiratory Medicine, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Noriho Sakamoto
- Department of Respiratory Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.
| | - Takatomo Tokito
- Department of Respiratory Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Hirokazu Yura
- Department of Respiratory Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Takashi Kido
- Department of Respiratory Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Hiroshi Ishimoto
- Department of Respiratory Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Yuji Ishimatsu
- Department of Nursing, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Shinnosuke Takemoto
- Department of Respiratory Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Takahiro Takazono
- Department of Infectious disease, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Haruki Okamura
- Department of Tumor Cell Therapy, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya, 663-8501, Japan
| | - Tomoya Nishino
- Department of Nephrology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Yoshimasa Tanaka
- Center for Medical Innovation, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| |
Collapse
|
2
|
Hassan M, Ali AS, Zubairi ABS, Padhani ZA, Kirmani S, Ahmad H, Fatmi Z, Das JK. Gene polymorphisms and risk of idiopathic pulmonary fibrosis: a systematic review and meta-analysis. Monaldi Arch Chest Dis 2024. [PMID: 39480160 DOI: 10.4081/monaldi.2024.2952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 09/09/2024] [Indexed: 11/02/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) has been widely hypothesized to occur as a result of an interplay between a nexus of environmental and genetic risk factors. However, not much is known about the genetic aspect of this disease. The objective of this review was to identify the genetic polymorphisms associated with the risk of developing IPF. We searched PubMed, EBSCO CINAHL Plus, Web of Science, and Wiley Cochrane Library databases for studies on risk factors of IPF published between March 2000 and November 2023. Studies with an IPF diagnosis based only on the American Thoracic Society and the European Respiratory Society guidelines were included. Thirty-one case-control studies were included with 3997 IPF and 20,925 non-IPF subjects. Two of the studies enrolled biopsy-proven IPF patients; 13 studies diagnosed IPF on the basis of clinical and high-resolution computed tomography (HRCT) findings; and 14 studies diagnosed based on both biopsy and clinical and HRCT findings. 16 studies with MUC5B rs35705950, IL-4 rs2243250, IL-4 rs2070874, and tumor necrosis factor α (TNFα)-308 were eligible for meta-analysis. The allele contrast model (T versus G) for MUC5B rs35705950 revealed statistically significant association of T allele with the risk of IPF [odds ratio (OR) 3.84, 95% confidence interval (CI) 3.20 to 4.61, adjusted p<0.0001), as was the allele contrast model for Asian (OR 2.83, 95% CI 1.51 to 5.32, adjusted p=0.009) and Caucasian (OR 4.11, 95% CI 3.56 to 4.75, adjusted p<0.0001). The allele contrast models for IL-4 rs2243250, IL-4 rs2070874, and TNFα-308 did not demonstrate any significant association with IPF. This review suggests an association of MUC5B rs35705950 T allele with the risk of developing IPF. To our knowledge, this study is the first to aggregate several genetic polymorphisms associated with IPF.
Collapse
Affiliation(s)
- Maryam Hassan
- Department of Medicine, Aga Khan University Hospital, Karachi
| | | | - Ali Bin Sarwar Zubairi
- Department of Medicine, Aga Khan University Hospital, Karachi, Pakistan; Department of Medicine, Southern Illinois University School of Medicine, Springfield, IL
| | - Zahra Ali Padhani
- Faculty of Health and Medical Sciences, School of Public Health, University of Adelaide
| | - Salman Kirmani
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi
| | - Huzaifa Ahmad
- Department of Medicine, Aga Khan University Hospital, Karachi
| | - Zafar Fatmi
- Department of Community Health Sciences, Aga Khan University Hospital, Karachi
| | - Jai K Das
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi; Institute of Global Health and Development, Aga Khan University, Karachi
| |
Collapse
|
3
|
Fu C, Tian X, Wu S, Chu X, Cheng Y, Wu X, Yang W. Role of telomere dysfunction and immune infiltration in idiopathic pulmonary fibrosis: new insights from bioinformatics analysis. Front Genet 2024; 15:1447296. [PMID: 39346776 PMCID: PMC11427275 DOI: 10.3389/fgene.2024.1447296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/29/2024] [Indexed: 10/01/2024] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease characterized by unexplained irreversible pulmonary fibrosis. Although the etiology of IPF is unclear, studies have shown that it is related to telomere length shortening. However, the prognostic value of telomere-related genes in IPF has not been investigated. Methods We utilized the GSE10667 and GSE110147 datasets as the training set, employing differential expression analysis and weighted gene co-expression network analysis (WGCNA) to screen for disease candidate genes. Then, we used consensus clustering analysis to identify different telomere patterns. Next, we used summary data-based mendelian randomization (SMR) analysis to screen core genes. We further evaluated the relationship between core genes and overall survival and lung function in IPF patients. Finally, we performed immune infiltration analysis to reveal the changes in the immune microenvironment of IPF. Results Through differential expression analysis and WGCNA, we identified 35 significant telomere regulatory factors. Consensus clustering analysis revealed two distinct telomere patterns, consisting of cluster A (n = 26) and cluster B (n = 19). Immune infiltration analysis revealed that cluster B had a more active immune microenvironment, suggesting its potential association with IPF. Using GTEx eQTL data, our SMR analysis identified two genes with potential causal associations with IPF, including GPA33 (PSMR = 0.0013; PHEIDI = 0.0741) and MICA (PSMR = 0.0112; PHEIDI = 0.9712). We further revealed that the expression of core genes is associated with survival time and lung function in IPF patients. Finally, immune infiltration analysis revealed that NK cells were downregulated and plasma cells and memory B cells were upregulated in IPF. Further correlation analysis showed that GPA33 expression was positively correlated with NK cells and negatively correlated with plasma cells and memory B cells. Conclusion Our study provides a new perspective for the role of telomere dysfunction and immune infiltration in IPF and identifies potential therapeutic targets. Further research may reveal how core genes affect cell function and disease progression, providing new insights into the complex mechanisms of IPF.
Collapse
Affiliation(s)
- Chenkun Fu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xin Tian
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shuang Wu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xiaojuan Chu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yiju Cheng
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Respiratory and Critical Care Medicine, The Fourth People’s Hospital of Guiyang, Guiyang, China
| | - Xiao Wu
- Department of Critical Care Medicine, The Second People’s Hospital of Guiyang, Guiyang, China
| | - Wengting Yang
- Department of Critical Care Medicine, The Second People’s Hospital of Guiyang, Guiyang, China
| |
Collapse
|
4
|
Yang W, Lecuona E, Wu Q, Liu X, Sun H, Alam H, Nadig SN, Bharat A. The role of lung-restricted autoantibodies in the development of primary and chronic graft dysfunction. FRONTIERS IN TRANSPLANTATION 2023; 2:1237671. [PMID: 38993924 PMCID: PMC11235341 DOI: 10.3389/frtra.2023.1237671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/18/2023] [Indexed: 07/13/2024]
Abstract
Lung transplantation is a life-saving treatment for both chronic end-stage lung diseases and acute respiratory distress syndrome, including those caused by infectious agents like COVID-19. Despite its increasing utilization, outcomes post-lung transplantation are worse than other solid organ transplants. Primary graft dysfunction (PGD)-a condition affecting more than half of the recipients post-transplantation-is the chief risk factor for post-operative mortality, transplant-associated multi-organ dysfunction, and long-term graft loss due to chronic rejection. While donor-specific antibodies targeting allogenic human leukocyte antigens have been linked to transplant rejection, the role of recipient's pre-existing immunoglobulin G autoantibodies against lung-restricted self-antigens (LRA), like collagen type V and k-alpha1 tubulin, is less understood in the context of lung transplantation. Recent studies have found an increased risk of PGD development in lung transplant recipients with LRA. This review will synthesize past and ongoing research-utilizing both mouse models and human subjects-aimed at unraveling the mechanisms by which LRA heightens the risk of PGD. Furthermore, it will explore prospective approaches designed to mitigate the impact of LRA on lung transplant patients.
Collapse
Affiliation(s)
- Wenbin Yang
- Division of Thoracic Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Emilia Lecuona
- Division of Thoracic Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Qiang Wu
- Division of Thoracic Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Xianpeng Liu
- Division of Thoracic Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Haiying Sun
- Division of Thoracic Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Hasan Alam
- Division of Trauma & Acute Care Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Satish N. Nadig
- Division of Abdominal Transplant, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Ankit Bharat
- Division of Thoracic Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
5
|
Yang L, Xia H, Gilbertsen A, Smith K, Racila E, Bitterman PB, Henke CA. IL-8 concurrently promotes idiopathic pulmonary fibrosis mesenchymal progenitor cell senescence and PD-L1 expression enabling escape from immune cell surveillance. Am J Physiol Lung Cell Mol Physiol 2023; 324:L849-L862. [PMID: 37121574 PMCID: PMC10228676 DOI: 10.1152/ajplung.00028.2023] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/24/2023] [Indexed: 05/02/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease. We discovered fibrogenic mesenchymal progenitor cells (MPCs) in the lungs of IPF patients that display cell-autonomous fibrogenicity and drive fibrotic progression. In a study of the IPF MPC nuclear proteome, we identified DNA damage as one of the most altered functions in IPF MPCs. In prior work we found that IL-8 drives IPF MPC self-renewal. IL-8 can promote replicative stress and DNA damage and induce senescence through the CXCR2 receptor. We hypothesized that IL-8 promotes DNA damage-mediated senescence in IPF MPCs. We show that IL-8 induces DNA damage and promotes IPF MPC senescence. We discovered that IL-8 concurrently promotes senescence and upregulation of the programmed death ligand 1 (PD-L1) in a CXCR2-dependent manner. Disruption of programmed cell death protein-1 (PD-1)-PD-L1 interaction promotes natural killer (NK) cell killing of IPF MPCs in vitro and arrests IPF MPC-mediated experimental lung fibrosis in vivo. Immunohistochemical (IHC) analysis of IPF lung tissue identified PD-L1-expressing IPF MPCs codistributing with NK cells and β-galactosidase-positive cells. Our data indicate that IL-8 simultaneously promotes IPF MPC DNA damage-induced senescence and high PD-L1 expression, enabling IPF MPCs to elude immune cell-targeted removal. Disruption of PD-1-PD-L1 interaction may limit IPF MPC-mediated fibrotic progression.NEW & NOTEWORTHY Here we show that IL-8 concurrently promotes senescence and upregulation of PD-L1 in IPF MPCs. IHC analysis identifies the presence of senescent IPF MPCs intermingled with NK cells in the fibroblastic focus, suggesting that senescent MPCs elude immune cell surveillance. We demonstrate that disruption of PD-1/PD-L1 interaction promotes NK cell killing of IPF MPCs and arrests IPF MPC-mediated experimental lung fibrosis. Disruption of PD-1/PD-L1 interaction may be one means to limit fibrotic progression.
Collapse
Affiliation(s)
- Libang Yang
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States
| | - Hong Xia
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States
| | - Adam Gilbertsen
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States
| | - Karen Smith
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States
| | - Emil Racila
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States
| | - Peter B Bitterman
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States
| | - Craig A Henke
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States
| |
Collapse
|
6
|
Huo R, Huang X, Yang Y, Yang Y, Lin J. Potential of resveratrol in the treatment of interstitial lung disease. Front Pharmacol 2023; 14:1139460. [PMID: 37089962 PMCID: PMC10117935 DOI: 10.3389/fphar.2023.1139460] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/09/2023] [Indexed: 04/08/2023] Open
Abstract
Interstitial lung disease (ILD) is a heterogeneous group of diseases characterized by lung injury caused by lung fibroblast proliferation, interstitial inflammation, and fibrosis. Different cell signal transduction pathways are activated in response to various proinflammatory or fibrotic cytokines, such as IL-6, and these cytokines are increased in different ILDs. The overexpressed cytokines and growth factors in ILD can activate TGF-β/Smad2/3/4, NF-κB, and JAK/STAT signal transduction pathways, promote the activation of immune cells, increase the release of pro-inflammatory and pro-fibrotic factors, differentiate fibroblasts into myofibroblasts, and promote the occurrence and development of ILD. This finding suggests the importance of signal transduction pathways in patients with ILD. Recent evidence suggests that resveratrol (RSV) attenuates excessive inflammation and pulmonary fibrosis by inhibiting the TGF-β/Smad2/3/4, NF-κB, and JAK/STAT signal transduction pathways and overactivation of immune cells. In this review, advances in lung protection and the underlying mechanisms of RSV are summarized, and the potential efficacy of RSV as a promising treatment option for ILD is highlighted.
Collapse
Affiliation(s)
| | | | | | | | - Jinying Lin
- Department of Rheumatology and Immunology, Guangxi Academy of Medical Sciences, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
7
|
Franklin M, Connolly E, Hussell T. Recruited and Tissue-Resident Natural Killer Cells in the Lung During Infection and Cancer. Front Immunol 2022; 13:887503. [PMID: 35844626 PMCID: PMC9284027 DOI: 10.3389/fimmu.2022.887503] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/17/2022] [Indexed: 11/23/2022] Open
Abstract
Natural killer (NK) cells are an important component of the innate immune system, and have a key role in host defense against infection and in tumor surveillance. Tumors and viruses employ remarkably similar strategies to avoid recognition and killing by NK cells and so much can be learnt by comparing NK cells in these disparate diseases. The lung is a unique tissue environment and immune cells in this organ, including NK cells, exist in a hypofunctional state to prevent activation against innocuous stimuli. Upon infection, rapid NK cell infiltration into the lung occurs, the amplitude of which is determined by the extent of inflammation and damage. Activated NK cells kill infected cells and produce pro-inflammatory cytokines and chemokines to recruit cells of the adaptive immune system. More recent evidence has shown that NK cells also play an additional role in resolution of inflammation. In lung cancer however, NK cell recruitment is impaired and those that are present have reduced functionality. The majority of lung NK cells are circulatory, however recently a small population of tissue-resident lung NK cells has been described. The specific role of this subset is yet to be determined, but they show similarity to resident memory T cell subsets. Whether resident or recruited, NK cells are important in the control of pulmonary infections, but equally, can drive excessive inflammation if not regulated. In this review we discuss how NK cells are recruited, controlled and retained in the specific environment of the lung in health and disease. Understanding these mechanisms in the context of infection may provide opportunities to promote NK cell recruitment and function in the lung tumor setting.
Collapse
|
8
|
Huang Y, Oldham JM, Ma SF, Unterman A, Liao SY, Barros AJ, Bonham CA, Kim JS, Vij R, Adegunsoye A, Strek ME, Molyneaux PL, Maher TM, Herazo-Maya JD, Kaminski N, Moore BB, Martinez FJ, Noth I. Blood Transcriptomics Predicts Progression of Pulmonary Fibrosis and Associated Natural Killer Cells. Am J Respir Crit Care Med 2021; 204:197-208. [PMID: 33689671 PMCID: PMC8650792 DOI: 10.1164/rccm.202008-3093oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 03/08/2021] [Indexed: 11/16/2022] Open
Abstract
Rationale: Disease activity in idiopathic pulmonary fibrosis (IPF) remains highly variable, poorly understood, and difficult to predict. Objectives: To identify a predictor using short-term longitudinal changes in gene expression that forecasts future FVC decline and to characterize involved pathways and cell types. Methods: Seventy-four patients from COMET (Correlating Outcomes with Biochemical Markers to Estimate Time-Progression in IPF) cohort were dichotomized as progressors (≥10% FVC decline) or stable. Blood gene-expression changes within individuals were calculated between baseline and 4 months and regressed with future FVC status, allowing determination of expression variations, sample size, and statistical power. Pathway analyses were conducted to predict downstream effects and identify new targets. An FVC predictor for progression was constructed in COMET and validated using independent cohorts. Peripheral blood mononuclear single-cell RNA-sequencing data from healthy control subjects were used as references to characterize cell type compositions from bulk peripheral blood mononuclear RNA-sequencing data that were associated with FVC decline. Measurements and Main Results: The longitudinal model reduced gene-expression variations within stable and progressor groups, resulting in increased statistical power when compared with a cross-sectional model. The FVC predictor for progression anticipated patients with future FVC decline with 78% sensitivity and 86% specificity across independent IPF cohorts. Pattern recognition receptor pathways and mTOR pathways were downregulated and upregulated, respectively. Cellular deconvolution using single-cell RNA-sequencing data identified natural killer cells as significantly correlated with progression. Conclusions: Serial transcriptomic change predicts future FVC decline. An analysis of cell types involved in the progressor signature supports the novel involvement of natural killer cells in IPF progression.
Collapse
Affiliation(s)
- Yong Huang
- Division of Pulmonary and Critical Care Medicine, The University of Virginia, Charlottesville, Virginia
| | - Justin M. Oldham
- Division of Pulmonary, Critical Care, and Sleep Medicine, The University of California at Davis, Sacramento, California
| | - Shwu-Fan Ma
- Division of Pulmonary and Critical Care Medicine, The University of Virginia, Charlottesville, Virginia
| | - Avraham Unterman
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Shu-Yi Liao
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - Andrew J. Barros
- Division of Pulmonary and Critical Care Medicine, The University of Virginia, Charlottesville, Virginia
| | - Catherine A. Bonham
- Division of Pulmonary and Critical Care Medicine, The University of Virginia, Charlottesville, Virginia
| | - John S. Kim
- Division of Pulmonary and Critical Care Medicine, The University of Virginia, Charlottesville, Virginia
| | - Rekha Vij
- Section of Pulmonary and Critical Care Medicine and
| | - Ayodeji Adegunsoye
- Section of Pulmonary and Critical Care Medicine and
- Department of Human Genetics, Genetics, Genomic and Systems Biology, University of Chicago, Chicago, Illinois
| | | | - Philip L. Molyneaux
- National Heart and Lung Institute, Imperial College, London, United Kingdom
- Royal Brompton Hospital, London, United Kingdom
| | - Toby M. Maher
- National Heart and Lung Institute, Imperial College, London, United Kingdom
- Royal Brompton Hospital, London, United Kingdom
- Division of Pulmonary, Critical Care and Sleep Medicine, Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Jose D. Herazo-Maya
- Division of Pulmonary, Critical Care, and Sleep Medicine, Tampa General Hospital, University of South Florida, Tampa, Florida
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Bethany B. Moore
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan; and
| | - Fernando J. Martinez
- Internal Medicine, Weill Cornell Medical College, Cornell University, New York, New York
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, The University of Virginia, Charlottesville, Virginia
| |
Collapse
|
9
|
Furukawa H, Oka S, Higuchi T, Shimada K, Hashimoto A, Matsui T, Tohma S. Biomarkers for interstitial lung disease and acute-onset diffuse interstitial lung disease in rheumatoid arthritis. Ther Adv Musculoskelet Dis 2021; 13:1759720X211022506. [PMID: 34211592 PMCID: PMC8216360 DOI: 10.1177/1759720x211022506] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/11/2021] [Indexed: 12/31/2022] Open
Abstract
Interstitial lung disease (ILD) is frequently a complication of rheumatoid arthritis (RA) as an extra-articular manifestation which has a poor prognosis. Acute-onset diffuse ILD (AoDILD) occasionally occurs in RA and includes acute exacerbation of ILD, drug-induced ILD, and Pneumocystis pneumonia. AoDILD also confers a poor prognosis in RA. Previously-established biomarkers for ILD include Krebs von den lungen-6 and surfactant protein-D originally defined in patients with idiopathic pulmonary fibrosis; the sensitivity of these markers for RA-associated ILD (RA-ILD) is low. Although many studies on ILD markers have been performed in idiopathic pulmonary fibrosis, only a few validation studies in RA-ILD or AoDILD have been reported. Biomarkers for RA-ILD and AoDILD are thus still required. Recently, genomic, cytokine, antibody, and metabolomic profiles of RA-ILD or AoDILD have been investigated with the aim of improving biomarkers. In this review, we summarize current preliminary data on these potential biomarkers for RA-ILD or AoDILD. The development of biomarkers on RA-ILD has only just begun. When validated, such candidate biomarkers will provide valuable information on pathogenesis, prognosis, and drug responses in RA-ILD in future.
Collapse
Affiliation(s)
- Hiroshi Furukawa
- Department of Rheumatology, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose 204-8585, Japan
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Minami-ku, Sagamihara, Japan
| | - Shomi Oka
- Department of Rheumatology, National Hospital Organization Tokyo National Hospital, Kiyose, Japan
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Minami-ku, Sagamihara, Japan
| | - Takashi Higuchi
- Department of Rheumatology, National Hospital Organization Tokyo National Hospital, Kiyose, Japan
- Department of Nephrology, Ushiku Aiwa General Hospital, Ushiku, Japan
| | - Kota Shimada
- Department of Rheumatology, National Hospital Organization Sagamihara National Hospital, Minami-ku, Sagamihara, Japan
- Department of Rheumatic Diseases, Tokyo Metropolitan Tama Medical Center, Fuchu, Japan
| | - Atsushi Hashimoto
- Department of Rheumatology, National Hospital Organization Sagamihara National Hospital, Minami-ku, Sagamihara, Japan
- Department of Internal Medicine, Sagami Seikyou Hospital, Minami-ku, Sagamihara, Japan
| | - Toshihiro Matsui
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Minami-ku, Sagamihara, Japan
- Department of Rheumatology, National Hospital Organization Sagamihara National Hospital, Minami-ku, Sagamihara, Japan
| | - Shigeto Tohma
- Department of Rheumatology, National Hospital Organization Tokyo National Hospital, Kiyose, Japan
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Minami-ku, Sagamihara, Japan
| |
Collapse
|
10
|
Sun S, Huang C, Leng D, Chen C, Zhang T, Lei KC, Zhang XD. Gene fusion of IL7 involved in the regulation of idiopathic pulmonary fibrosis. Ther Adv Respir Dis 2021; 15:1753466621995045. [PMID: 33878985 PMCID: PMC8064517 DOI: 10.1177/1753466621995045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Idiopathic pulmonary fibrosis (IPF) is a rare form of immune-mediated interstitial lung disease characterized by progressive pulmonary fibrosis and scarring. The pathogenesis of IPF is still unclear. Gene fusion events exist universally during transcription and show alternated patterns in a variety of lung diseases. Therefore, the comprehension of the function of gene fusion in IPF might shed light on IPF pathogenesis research and facilitate treatment development. Methods: In this study, we included 91 transcriptome datasets from the National Center for Biotechnology Information (NCBI), including 52 IPF patients and 39 healthy controls. We detected fusion events in these datasets and probed gene fusion-associated differential gene expression and functional pathways. To obtain robust results, we corrected the batch bias across different projects. Results: We identified 1550 gene fusion events in all transcriptomes and studied the possible impacts of IL7 = AC083837.1 gene fusion. The two genes locate adjacently in chromosome 8 and share the same promoters. Their fusion is associated with differential expression of 282 genes enriched in six Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and 35 functional gene sets. Gene ontology (GO) enrichment analysis shows that IL7 = AC083837.1 gene fusion is associated with the enrichment of 187 gene sets. The co-expression network of interleukin-7 (IL7) indicates that decreased IL7 expression is associated with many pathways that regulate IPF progress. Conclusion: Based on the results, we conclude that IL7 = AC083837.1 gene fusion might exacerbate fibrosis in IPF via enhancing activities of natural killer cell-mediated cytotoxicity, skin cell apoptosis, and vessel angiogenesis, the interaction of which contributes to the development of fibrosis and the deterioration of respiratory function of IPF patients. Our work unveils the possible roles of gene fusion in regulating IPF and demonstrates that gene fusion investigation is a valid approach in probing immunologic mechanisms and searching potential therapeutic targets for treating IPF. The reviews of this paper are available via the supplemental material section.
Collapse
Affiliation(s)
- Shixue Sun
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Chen Huang
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Dongliang Leng
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Chang Chen
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Teng Zhang
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Kuan Cheok Lei
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Xiaohua Douglas Zhang
- CRDA, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China
| |
Collapse
|
11
|
Calabrese DR, Aminian E, Mallavia B, Liu F, Cleary SJ, Aguilar OA, Wang P, Singer JP, Hays SR, Golden JA, Kukreja J, Dugger D, Nakamura M, Lanier LL, Looney MR, Greenland JR. Natural killer cells activated through NKG2D mediate lung ischemia-reperfusion injury. J Clin Invest 2021; 131:137047. [PMID: 33290276 PMCID: PMC7852842 DOI: 10.1172/jci137047] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 11/25/2020] [Indexed: 12/18/2022] Open
Abstract
Pulmonary ischemia-reperfusion injury (IRI) is a clinical syndrome of acute lung injury that occurs after lung transplantation or remote organ ischemia. IRI causes early mortality and has no effective therapies. While NK cells are innate lymphocytes capable of recognizing injured cells, their roles in acute lung injury are incompletely understood. Here, we demonstrated that NK cells were increased in frequency and cytotoxicity in 2 different IRI mouse models. We showed that NK cells trafficked to the lung tissue from peripheral reservoirs and were more mature within lung tissue. Acute lung ischemia-reperfusion injury was blunted in a NK cell-deficient mouse strain but restored with adoptive transfer of NK cells. Mechanistically, NK cell NKG2D receptor ligands were induced on lung endothelial and epithelial cells following IRI, and antibody-mediated NK cell depletion or NKG2D stress receptor blockade abrogated acute lung injury. In human lung tissue, NK cells were increased at sites of ischemia-reperfusion injury and activated NK cells were increased in prospectively collected human bronchoalveolar lavage in subjects with severe IRI. These data support a causal role for recipient peripheral NK cells in pulmonary IRI via NK cell NKG2D receptor ligation. Therapies targeting NK cells may hold promise in acute lung injury.
Collapse
Affiliation(s)
- Daniel R. Calabrese
- Department of Medicine, University of California, San Francisco, California
- Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Emily Aminian
- Department of Medicine, University of California, San Francisco, California
| | - Benat Mallavia
- Department of Medicine, University of California, San Francisco, California
| | - Fengchun Liu
- Department of Medicine, University of California, San Francisco, California
| | - Simon J. Cleary
- Department of Medicine, University of California, San Francisco, California
| | - Oscar A. Aguilar
- Department of Microbiology and Immunology, University of California, San Francisco, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Ping Wang
- Department of Medicine, University of California, San Francisco, California
| | - Jonathan P. Singer
- Department of Medicine, University of California, San Francisco, California
| | - Steven R. Hays
- Department of Medicine, University of California, San Francisco, California
| | - Jeffrey A. Golden
- Department of Medicine, University of California, San Francisco, California
| | - Jasleen Kukreja
- Department of Surgery, University of California, San Francisco, California
| | - Daniel Dugger
- Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Mary Nakamura
- Department of Medicine, University of California, San Francisco, California
- Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Lewis L. Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Mark R. Looney
- Department of Medicine, University of California, San Francisco, California
| | - John R. Greenland
- Department of Medicine, University of California, San Francisco, California
- Medical Service, Veterans Affairs Health Care System, San Francisco, California
| |
Collapse
|
12
|
Whole-exome sequencing identifies susceptibility genes and pathways for idiopathic pulmonary fibrosis in the Chinese population. Sci Rep 2021; 11:1443. [PMID: 33446833 PMCID: PMC7809470 DOI: 10.1038/s41598-020-80944-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Genetic factors play a role in the risk of idiopathic pulmonary fibrosis (IPF). Specifically, MUC5B rs35705950 non-risk alleles and immunologic aberrations were associated with the IPF’s progression. However, rare genetic variants have not been systematically investigated in Chinese IPF patients. In this study, we aimed to improve understanding of the genetic architecture of IPF in the Chinese population and to assess whether rare protein-coding variants in the immunity pathway genes are enriched in the IPF patients with non-risk alleles at rs35705950. A case–control exome-wide study including 110 IPF patients and 60 matched healthy controls was conducted. rs35705950 was genotyped by Sanger sequencing. To identify genes enriched in IPF, gene-based association analyses were performed. Identified genes were included for further pathway analyses using gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Associations between rs35705950 and genes enriched in the immunity pathway were also tested. 226 genes that were enriched with deleterious variants were identified in IPF patients. Out of them, 36 genes were significantly enriched in GO and KEGG pathways in the IPF. Pathway analyses implicated that these genes were involved in the immune response and cell adhesion. Rare protein-altering variants in genes related to the immunity pathway did not significantly differ between patients with a MUC5B risk allele and individuals without risk allele. We drafted a comprehensive mutational landscape of rare protein-coding variants in the Chinese IPF and identified genes related to immune response and cell adhesion. These results partially explain changes in gene expression involved in the immunity/inflammatory pathways in IPF patients.
Collapse
|
13
|
Karrar A, Rajput B, Hariharan S, Abdelatif D, Houry M, Moosvi A, Ali I, Tan D, Noor S, Esmaeili D, Felix S, Alaparthi L, Otgonsuren M, Lam B, Goodman ZD, Younossi ZM. Major Histocompatibility Complex Class I-Related Chain A Alleles and Histology of Nonalcoholic Fatty Liver Disease. Hepatol Commun 2021; 5:63-73. [PMID: 33437901 PMCID: PMC7789833 DOI: 10.1002/hep4.1610] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 08/11/2020] [Accepted: 08/30/2020] [Indexed: 01/05/2023] Open
Abstract
Major histocompatibility complex class I-related chain A (MICA) is a highly polymorphic gene that modulates immune surveillance by binding to its receptor on natural killer cells, and its genetic polymorphisms have been associated with chronic immune-mediated diseases. The progressive form of nonalcoholic fatty liver disease (NAFLD), nonalcoholic steatohepatitis (NASH), is characterized by accumulation of fat and inflammatory cells in the hepatic parenchyma, potentially leading to liver cell injury and fibrosis. To date, there are no data describing the potential role of MICA in the pathogenesis of NAFLD. Therefore, our aim was to assess the association between MICA polymorphism and NASH and its histologic features. A total of 134 subjects were included. DNA from patients with biopsy-proven NAFLD were genotyped using polymerase chain reaction-sequence-specific oligonucleotide for MICA alleles. Liver biopsies were assessed for histologic diagnosis of NASH and specific pathologic features, including stage of fibrosis and grade of inflammation. Multivariate analysis was performed to draw associations between MICA alleles and the different variables; P ≤ 0.05 was considered significant. Univariate analysis showed that MICA*011 (odds ratio [OR], 7.14; 95% confidence interval [CI], 1.24-41.0; P = 0.04) was associated with a higher risk for histologic NASH. Multivariate analysis showed that MICA*002 was independently associated with a lower risk for focal hepatocyte necrosis (OR, 0.24; 95% CI, 0.08-0.74; P = 0.013) and advanced fibrosis (OR, 0.11; 95% CI, 0.02-0.70; P = 0.019). MICA*017 was independently associated with a higher risk for lymphocyte-mediated inflammation (OR, 5.12; 95% CI, 1.12-23.5; P = 0.035). Conclusion: MICA alleles may be associated with NASH and its histologic features of inflammation and fibrosis. Additional research is required to investigate the potential role of MICA in increased risk or protection against NAFLD.
Collapse
Affiliation(s)
- Azza Karrar
- Betty and Guy Beatty Center for Integrated ResearchInova Fairfax Medical CampusFalls ChurchVAUSA
| | - Bijal Rajput
- Betty and Guy Beatty Center for Integrated ResearchInova Fairfax Medical CampusFalls ChurchVAUSA
| | - Siddharth Hariharan
- Betty and Guy Beatty Center for Integrated ResearchInova Fairfax Medical CampusFalls ChurchVAUSA
| | - Dinan Abdelatif
- Center for Liver DiseasesDepartment of MedicineInova Fairfax HospitalFalls ChurchVAUSA
| | - Mohamad Houry
- Betty and Guy Beatty Center for Integrated ResearchInova Fairfax Medical CampusFalls ChurchVAUSA
| | - Ali Moosvi
- Betty and Guy Beatty Center for Integrated ResearchInova Fairfax Medical CampusFalls ChurchVAUSA
| | - Irfan Ali
- Center for Liver DiseasesDepartment of MedicineInova Fairfax HospitalFalls ChurchVAUSA
| | - Daisong Tan
- Center for Liver DiseasesDepartment of MedicineInova Fairfax HospitalFalls ChurchVAUSA
| | - Sohailla Noor
- Betty and Guy Beatty Center for Integrated ResearchInova Fairfax Medical CampusFalls ChurchVAUSA
| | - Donna Esmaeili
- Betty and Guy Beatty Center for Integrated ResearchInova Fairfax Medical CampusFalls ChurchVAUSA
| | - Sean Felix
- Betty and Guy Beatty Center for Integrated ResearchInova Fairfax Medical CampusFalls ChurchVAUSA
| | - Lakshmi Alaparthi
- Center for Liver DiseasesDepartment of MedicineInova Fairfax HospitalFalls ChurchVAUSA
| | - Munkhzul Otgonsuren
- Betty and Guy Beatty Center for Integrated ResearchInova Fairfax Medical CampusFalls ChurchVAUSA
| | - Brian Lam
- Betty and Guy Beatty Center for Integrated ResearchInova Fairfax Medical CampusFalls ChurchVAUSA.,Center for Liver DiseasesDepartment of MedicineInova Fairfax HospitalFalls ChurchVAUSA
| | - Zachary D Goodman
- Center for Liver DiseasesDepartment of MedicineInova Fairfax HospitalFalls ChurchVAUSA
| | - Zobair M Younossi
- Betty and Guy Beatty Center for Integrated ResearchInova Fairfax Medical CampusFalls ChurchVAUSA.,Center for Liver DiseasesDepartment of MedicineInova Fairfax HospitalFalls ChurchVAUSA
| |
Collapse
|
14
|
Gong W, Guo P, Liu L, Guan Q, Yuan Z. Integrative Analysis of Transcriptome-Wide Association Study and mRNA Expression Profiles Identifies Candidate Genes Associated With Idiopathic Pulmonary Fibrosis. Front Genet 2020; 11:604324. [PMID: 33362862 PMCID: PMC7758323 DOI: 10.3389/fgene.2020.604324] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/17/2020] [Indexed: 12/27/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a type of scarring lung disease characterized by a chronic, progressive, and irreversible decline in lung function. The genetic basis of IPF remains elusive. A transcriptome-wide association study (TWAS) of IPF was performed by FUSION using gene expression weights of three tissues combined with a large-scale genome-wide association study (GWAS) dataset, totally involving 2,668 IPF cases and 8,591 controls. Significant genes identified by TWAS were then subjected to gene ontology (GO) and pathway enrichment analysis. The overlapped GO terms and pathways between enrichment analysis of TWAS significant genes and differentially expressed genes (DEGs) from the genome-wide mRNA expression profiling of IPF were also identified. For TWAS significant genes, protein–protein interaction (PPI) network and clustering modules analyses were further conducted using STRING and Cytoscape. Overall, TWAS identified a group of candidate genes for IPF under the Bonferroni corrected P value threshold (0.05/14929 = 3.35 × 10–6), such as DSP (PTWAS = 1.35 × 10–29 for lung tissue), MUC5B (PTWAS = 1.09 × 10–28 for lung tissue), and TOLLIP (PTWAS = 1.41 × 10–15 for whole blood). Pathway enrichment analysis identified multiple candidate pathways, such as herpes simplex infection (P value = 7.93 × 10–5) and antigen processing and presentation (P value = 6.55 × 10–5). 38 common GO terms and 8 KEGG pathways shared by enrichment analysis of TWAS significant genes and DEGs were identified. In the PPI network, 14 genes (DYNLL1, DYNC1LI1, DYNLL2, HLA-DRB5, HLA-DPB1, HLA-DQB2, HLA-DQA2, HLA-DQB1, HLA-DRB1, POLR2L, CENPP, CENPK, NUP133, and NUP107) were simultaneously detected by hub gene and module analysis. In conclusion, through integrative analysis of TWAS and mRNA expression profiles, we identified multiple novel candidate genes, GO terms and pathways for IPF, which contributes to the understanding of the genetic mechanism of IPF.
Collapse
Affiliation(s)
- Weiming Gong
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ping Guo
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lu Liu
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qingbo Guan
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, China.,Shandong Institute of Endocrine and Metabolic Diseases, Jinan, China
| | - Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
15
|
Borger JG, Lau M, Hibbs ML. The Influence of Innate Lymphoid Cells and Unconventional T Cells in Chronic Inflammatory Lung Disease. Front Immunol 2019; 10:1597. [PMID: 31354734 PMCID: PMC6637857 DOI: 10.3389/fimmu.2019.01597] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/26/2019] [Indexed: 12/11/2022] Open
Abstract
The lungs are continuously subjected to environmental insults making them susceptible to infection and injury. They are protected by the respiratory epithelium, which not only serves as a physical barrier but also a reactive one that can release cytokines, chemokines, and other defense proteins in response to danger signals, and can undergo conversion to protective mucus-producing goblet cells. The lungs are also guarded by a complex network of highly specialized immune cells and their mediators to support tissue homeostasis and resolve integrity deviation. This review focuses on specialized innate-like lymphocytes present in the lung that act as key sensors of lung insults and direct the pulmonary immune response. Included amongst these tissue-resident lymphocytes are innate lymphoid cells (ILCs), which are classified into five distinct subsets (natural killer, ILC1, ILC2, ILC3, lymphoid tissue-inducer cells), and unconventional T cells including natural killer T (NKT) cells, mucosal-associated invariant T (MAIT) cells, and γδ-T cells. While ILCs and unconventional T cells together comprise only a small proportion of the total immune cells in the lung, they have been found to promote lung homeostasis and are emerging as contributors to a variety of chronic lung diseases including pulmonary fibrosis, allergic airway inflammation, and chronic obstructive pulmonary disease (COPD). A particularly intriguing trait of ILCs that has recently emerged is their plasticity and ability to alter their gene expression profiles and adapt their function in response to environmental cues. The malleable nature of these cells may aid in rapid responses to pathogen but may also have downstream pathological consequences. The role of ILC2s in Th2 allergic airway responses is becoming apparent but the contribution of other ILCs and unconventional T cells during chronic lung inflammation is poorly described. This review presents an overview of our current understanding of the involvement of ILCs and unconventional T cells in chronic pulmonary diseases.
Collapse
Affiliation(s)
- Jessica G Borger
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Maverick Lau
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology and Therapeutics, Lung Health Research Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Margaret L Hibbs
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
16
|
Hervier B, Russick J, Cremer I, Vieillard V. NK Cells in the Human Lungs. Front Immunol 2019; 10:1263. [PMID: 31275301 PMCID: PMC6593268 DOI: 10.3389/fimmu.2019.01263] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/17/2019] [Indexed: 12/29/2022] Open
Abstract
The lung offers one of the largest exchange surfaces of the individual with the elements of the environment. As a place of important interactions between self and non-self, the lung is richly endowed in various immune cells. As such, lung natural killer (NK) cells play major effector and immunoregulatory roles to ensure self-integrity. A better understanding of their abilities in health and diseases has been made possible over the past decade thanks to tremendous discoveries in humans and animals. By precisely distinguishing the different NK cell subsets and dissecting the ontogeny and differentiation of NK cells, both blood and tissue-resident NK populations now appear to be much more pleiotropic than previously thought. In light of these recent findings in healthy individuals, this review describes the different lung NK cell populations quantitatively, qualitatively, phenotypically, and functionally. Their identification, immunological diversity, and adaptive capacities are also addressed. For each of these elements, the impact of the mutual interactions of lung NK cells with environmental and microenvironmental factors are questioned in terms of functionality, competence, and adaptive capacities. As pulmonary diseases are major causes of morbidity and mortality worldwide, special attention is also given to the involvement of lung NK cells in various diseases, including infectious, inflammatory, autoimmune, and neoplastic lung diseases. In addition to providing a comprehensive overview of lung NK cell biology, this review also provides insight into the potential of NK cell immunotherapy and the development of targeted biologics.
Collapse
Affiliation(s)
- Baptiste Hervier
- Centre d'Immunologie et des Maladies Infectieuses, Sorbonne Universités, Université Pierre et Marie Curie Université Paris 06, INSERM U1135, CNRS ERL8255, Paris, France
| | - Jules Russick
- Centre de Recherche des Cordeliers, INSERM UMR S1138, Université Pierre et Marie Curie, Sorbonne Universités, Université Pierre et Marie Curie Université Paris 06, Paris, France
| | - Isabelle Cremer
- Centre de Recherche des Cordeliers, INSERM UMR S1138, Université Pierre et Marie Curie, Sorbonne Universités, Université Pierre et Marie Curie Université Paris 06, Paris, France
| | - Vincent Vieillard
- Centre d'Immunologie et des Maladies Infectieuses, Sorbonne Universités, Université Pierre et Marie Curie Université Paris 06, INSERM U1135, CNRS ERL8255, Paris, France
| |
Collapse
|
17
|
Bergeron ME, Stefanov A, Haston CK. Fine mapping of the major bleomycin-induced pulmonary fibrosis susceptibility locus in mice. Mamm Genome 2018; 29:670-679. [PMID: 30173367 PMCID: PMC6182746 DOI: 10.1007/s00335-018-9774-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 08/20/2018] [Indexed: 12/22/2022]
Abstract
Susceptibility to fibrotic lung disease differs among people and among inbred strains of mice exposed to bleomycin where C57BL/6J mice are susceptible and C3H/HeJ mice are spared fibrotic disease. Genetic mapping studies completed in offspring derived from these inbred strains revealed the inheritance of C57BL/6J alleles at loci, including the major locus on chromosome 17, called Blmpf1 bleomycin-induced pulmonary fibrosis 1, to be linked to pulmonary fibrosis in treated mice. In the present study, to reduce the interval of Blmpf1, we bred and phenotyped a panel of subcongenic mice with C3H/HeJ alleles in a C57BL/6J background. Subcongenic mice received bleomycin via osmotic minipump and the fibrosis phenotype was measured histologically. Inheritance of C3H/HeJ alleles from 34.31 to 35.02 Mb was revealed to spare bleomycin-induced pulmonary fibrosis of C57BL/6J mice. From database analysis, 40 protein coding genes have been mapped to this reduced Blmpf1 interval, 18 of which contain C57BL/6J:C3H/HeJ sequence polymorphisms predicted to affect protein structure or to confer allele-dependent expression, and by RT-PCR analysis of lung tissue, we show 6 of these genes to differ in expression between C57BL/6J and C3H/HeJ mice. Genes known to regulate T cell numbers and activation (Btnl family, Notch4) are among the limited list of potential causal variants leading to lung disease in this model and the bronchoalveolar lavage of protected subcongenic mice had fewer lymphocytes, post bleomycin, than did C57BL/6J mice. We conclude that Blmpf1genes contributing to the susceptibility to bleomycin-induced pulmonary fibrosis could alter the adaptive immune response of C57BL/6J mice.
Collapse
Affiliation(s)
| | - Anguel Stefanov
- Meakins-Christie Laboratories McGill University, Montreal, PQ, Canada
| | - Christina K Haston
- Meakins-Christie Laboratories McGill University, Montreal, PQ, Canada. .,2Department of Mathematics, Statistics, Physics, and Computer Science, I.K. Barber School of Arts and Sciences, The University of British Columbia
- Okanagan, ASC 347 - 3187 University Way, Kelowna, BC, V1V 1V7, Canada.
| |
Collapse
|
18
|
Kaur A, Mathai SK, Schwartz DA. Genetics in Idiopathic Pulmonary Fibrosis Pathogenesis, Prognosis, and Treatment. Front Med (Lausanne) 2017; 4:154. [PMID: 28993806 PMCID: PMC5622313 DOI: 10.3389/fmed.2017.00154] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 09/07/2017] [Indexed: 12/14/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF), the most common form of idiopathic interstitial pneumonia (IIP), is characterized by irreversible scarring of the lung parenchyma and progressive decline in lung function leading to eventual respiratory failure. The prognosis of IPF is poor with a median survival of 3–5 years after diagnosis and no curative medical therapies. Although the pathogenesis of IPF is not well understood, there is a growing body of evidence that genetic factors contribute to disease risk. Recent studies have identified common and rare genetic variants associated with both sporadic and familial forms of pulmonary fibrosis, with at least one-third of the risk for developing fibrotic IIP explained by common genetic variants. The IPF-associated genetic loci discovered to date are implicated in diverse biological processes, including alveolar stability, host defense, cell–cell barrier function, and cell senescence. In addition, some common variants have also been associated with distinct clinical phenotypes. Better understanding of how genetic variation plays a role in disease risk and phenotype could identify potential therapeutic targets and inform clinical decision-making. In addition, clinical studies should be designed controlling for the genetic backgrounds of subjects, since clinical outcomes and therapeutic responses may differ by genotype. Further understanding of these differences will allow the development of personalized approaches to the IPF management.
Collapse
Affiliation(s)
- Amarpreet Kaur
- Department of Medicine, University of Colorado Denver School of Medicine, Aurora, CO, United States
| | - Susan K Mathai
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver School of Medicine, Aurora, CO, United States
| | - David A Schwartz
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver School of Medicine, Aurora, CO, United States
| |
Collapse
|
19
|
Osterburg AR, Nelson RL, Yaniv BZ, Foot R, Donica WR, Nashu MA, Liu H, Wikenheiser-Brokamp KA, Moss J, Gupta N, McCormack FX, Borchers MT. NK cell activating receptor ligand expression in lymphangioleiomyomatosis is associated with lung function decline. JCI Insight 2016; 1:e87270. [PMID: 27734028 DOI: 10.1172/jci.insight.87270] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a rare lung disease of women that leads to progressive cyst formation and accelerated loss of pulmonary function. Neoplastic smooth muscle cells from an unknown source metastasize to the lung and drive destructive remodeling. Given the role of NK cells in immune surveillance, we postulated that NK cell activating receptors and their cognate ligands are involved in LAM pathogenesis. We found that ligands for the NKG2D activating receptor UL-16 binding protein 2 (ULBP2) and ULBP3 are localized in cystic LAM lesions and pulmonary nodules. We found elevated soluble serum ULBP2 (mean = 575 pg/ml ± 142) in 50 of 100 subjects and ULBP3 in 30 of 100 (mean = 8,300 pg/ml ± 1,515) subjects. LAM patients had fewer circulating NKG2D+ NK cells and decreased NKG2D surface expression. Lung function decline was associated with soluble NKG2D ligand (sNKG2DL) detection. The greatest rate of decline forced expiratory volume in 1 second (FEV1, -124 ± 30 ml/year) in the 48 months after enrollment (NHLBI LAM Registry) occurred in patients expressing both ULBP2 and ULBP3, whereas patients with undetectable sNKG2DL levels had the lowest rate of FEV1 decline (-32.7 ± 10 ml/year). These data suggest a role for NK cells, sNKG2DL, and the innate immune system in LAM pathogenesis.
Collapse
Affiliation(s)
- Andrew R Osterburg
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Rebecca L Nelson
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Benyamin Z Yaniv
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Rachel Foot
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Walter Rf Donica
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Madison A Nashu
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Huan Liu
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Kathryn A Wikenheiser-Brokamp
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,Division of Pathology & Laboratory Medicine and Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Joel Moss
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Nishant Gupta
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Francis X McCormack
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Michael T Borchers
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
20
|
Hervier B, Perez M, Allenbach Y, Devilliers H, Cohen F, Uzunhan Y, Ouakrim H, Dorgham K, Méritet JF, Longchampt E, Stenzel W, Cremer I, Benveniste O, Vieillard V. Involvement of NK Cells and NKp30 Pathway in Antisynthetase Syndrome. THE JOURNAL OF IMMUNOLOGY 2016; 197:1621-30. [DOI: 10.4049/jimmunol.1501902] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 06/29/2016] [Indexed: 11/19/2022]
|
21
|
Fingerlin TE, Zhang W, Yang IV, Ainsworth HC, Russell PH, Blumhagen RZ, Schwarz MI, Brown KK, Steele MP, Loyd JE, Cosgrove GP, Lynch DA, Groshong S, Collard HR, Wolters PJ, Bradford WZ, Kossen K, Seiwert SD, du Bois RM, Garcia CK, Devine MS, Gudmundsson G, Isaksson HJ, Kaminski N, Zhang Y, Gibson KF, Lancaster LH, Maher TM, Molyneaux PL, Wells AU, Moffatt MF, Selman M, Pardo A, Kim DS, Crapo JD, Make BJ, Regan EA, Walek DS, Daniel JJ, Kamatani Y, Zelenika D, Murphy E, Smith K, McKean D, Pedersen BS, Talbert J, Powers J, Markin CR, Beckman KB, Lathrop M, Freed B, Langefeld CD, Schwartz DA. Genome-wide imputation study identifies novel HLA locus for pulmonary fibrosis and potential role for auto-immunity in fibrotic idiopathic interstitial pneumonia. BMC Genet 2016; 17:74. [PMID: 27266705 PMCID: PMC4895966 DOI: 10.1186/s12863-016-0377-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 05/20/2016] [Indexed: 12/18/2022] Open
Abstract
Background Fibrotic idiopathic interstitial pneumonias (fIIP) are a group of fatal lung diseases with largely unknown etiology and without definitive treatment other than lung transplant to prolong life. There is strong evidence for the importance of both rare and common genetic risk alleles in familial and sporadic disease. We have previously used genome-wide single nucleotide polymorphism data to identify 10 risk loci for fIIP. Here we extend that work to imputed genome-wide genotypes and conduct new RNA sequencing studies of lung tissue to identify and characterize new fIIP risk loci. Results We performed genome-wide genotype imputation association analyses in 1616 non-Hispanic white (NHW) cases and 4683 NHW controls followed by validation and replication (878 cases, 2017 controls) genotyping and targeted gene expression in lung tissue. Following meta-analysis of the discovery and replication populations, we identified a novel fIIP locus in the HLA region of chromosome 6 (rs7887 Pmeta = 3.7 × 10−09). Imputation of classic HLA alleles identified two in high linkage disequilibrium that are associated with fIIP (DRB1*15:01 P = 1.3 × 10−7 and DQB1*06:02 P = 6.1 × 10−8). Targeted RNA-sequencing of the HLA locus identified 21 genes differentially expressed between fibrotic and control lung tissue (Q < 0.001), many of which are involved in immune and inflammatory response regulation. In addition, the putative risk alleles, DRB1*15:01 and DQB1*06:02, are associated with expression of the DQB1 gene among fIIP cases (Q < 1 × 10−16). Conclusions We have identified a genome-wide significant association between the HLA region and fIIP. Two HLA alleles are associated with fIIP and affect expression of HLA genes in lung tissue, indicating that the potential genetic risk due to HLA alleles may involve gene regulation in addition to altered protein structure. These studies reveal the importance of the HLA region for risk of fIIP and a basis for the potential etiologic role of auto-immunity in fIIP. Electronic supplementary material The online version of this article (doi:10.1186/s12863-016-0377-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tasha E Fingerlin
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, USA. .,Department of Biostatistics and Informatics, University of Colorado Denver, Aurora, CO, USA.
| | - Weiming Zhang
- Department of Biostatistics and Informatics, University of Colorado Denver, Aurora, CO, USA
| | - Ivana V Yang
- Department of Medicine, School of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Hannah C Ainsworth
- Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Pamela H Russell
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Rachel Z Blumhagen
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, USA.,Department of Biostatistics and Informatics, University of Colorado Denver, Aurora, CO, USA
| | - Marvin I Schwarz
- Department of Medicine, School of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Kevin K Brown
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, USA
| | - Mark P Steele
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - James E Loyd
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Gregory P Cosgrove
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, USA
| | - David A Lynch
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, USA
| | - Steve Groshong
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, USA
| | - Harold R Collard
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Paul J Wolters
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | | | | | | | - Roland M du Bois
- National Heart and Lung Institute, Imperial College, London, UK.,National Institute for Health Research Biomedical Research Unit, Royal Brompton Hospital, London, UK
| | | | - Megan S Devine
- Department of Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Gunnar Gudmundsson
- Landspitali University Hospital and University of Iceland Faculty of Medicine, Reykjavik, Iceland
| | - Helgi J Isaksson
- Landspitali University Hospital and University of Iceland Faculty of Medicine, Reykjavik, Iceland
| | - Naftali Kaminski
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yingze Zhang
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kevin F Gibson
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lisa H Lancaster
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Toby M Maher
- National Heart and Lung Institute, Imperial College, London, UK.,National Institute for Health Research Biomedical Research Unit, Royal Brompton Hospital, London, UK
| | - Philip L Molyneaux
- National Heart and Lung Institute, Imperial College, London, UK.,National Institute for Health Research Biomedical Research Unit, Royal Brompton Hospital, London, UK
| | - Athol U Wells
- National Heart and Lung Institute, Imperial College, London, UK.,National Institute for Health Research Biomedical Research Unit, Royal Brompton Hospital, London, UK
| | - Miriam F Moffatt
- National Heart and Lung Institute, Imperial College, London, UK.,National Institute for Health Research Biomedical Research Unit, Royal Brompton Hospital, London, UK
| | - Moises Selman
- Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | - Annie Pardo
- Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Dong Soon Kim
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - James D Crapo
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, USA
| | - Barry J Make
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, USA
| | - Elizabeth A Regan
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, USA
| | - Dinesha S Walek
- University of Minnesota Genomics Center, University of Minnesota, Minneapolis, MN, USA
| | - Jerry J Daniel
- University of Minnesota Genomics Center, University of Minnesota, Minneapolis, MN, USA
| | - Yoichiro Kamatani
- Fondation Jean Dausset, Centre d'Étude du Polymorphisme Humain, Paris, France
| | - Diana Zelenika
- Commissariat à l'Energie Atomique, Institut Génomique, Centre National de Génotypage, Evry, France
| | - Elissa Murphy
- Department of Medicine, School of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Keith Smith
- Department of Medicine, School of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - David McKean
- Department of Medicine, School of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Brent S Pedersen
- Department of Medicine, School of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Janet Talbert
- Department of Medicine, School of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Julia Powers
- Department of Medicine, School of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Cheryl R Markin
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Kenneth B Beckman
- University of Minnesota Genomics Center, University of Minnesota, Minneapolis, MN, USA
| | - Mark Lathrop
- Fondation Jean Dausset, Centre d'Étude du Polymorphisme Humain, Paris, France.,Commissariat à l'Energie Atomique, Institut Génomique, Centre National de Génotypage, Evry, France
| | - Brian Freed
- Department of Medicine, School of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Carl D Langefeld
- Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - David A Schwartz
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, USA. .,Department of Medicine, School of Medicine, University of Colorado Denver, Aurora, CO, USA. .,Department of Immunology, School of Medicine, University of Colorado Denver, Aurora, CO, USA.
| |
Collapse
|
22
|
Zhou W, Wang Y. Candidate genes of idiopathic pulmonary fibrosis: current evidence and research. Appl Clin Genet 2016; 9:5-13. [PMID: 26893575 PMCID: PMC4745857 DOI: 10.2147/tacg.s61999] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a group of common and lethal forms of idiopathic interstitial pulmonary disease. IPF is characterized by a progressive decline in lung function with a median survival of 2-3 years after diagnosis. Although the pathogenesis of the disease remains unknown, genetic predisposition could play a causal role in IPF. A set of genes have been identified as candidate genes of IPF in the past 20 years. However, the recent technological advances that allow for the analysis of millions of polymorphisms in different subjects have deepened the understanding of the genetic complexity of IPF susceptibility. Genome-wide association studies and whole-genome sequencing continue to reveal the genetic loci associated with IPF risk. In this review, we describe candidate genes on the basis of their functions and aim to gain a better understanding of the genetic basis of IPF. The discovered candidate genes may help to clarify pivotal aspects in the diagnosis, prognosis, and therapies of IPF.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Medical Genetics, Nanjing University School of Medicine, Nanjing, People’s Republic of China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University School of Medicine, Nanjing, People’s Republic of China
| | - Yaping Wang
- Department of Medical Genetics, Nanjing University School of Medicine, Nanjing, People’s Republic of China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University School of Medicine, Nanjing, People’s Republic of China
| |
Collapse
|
23
|
Aquino-Gálvez A, González-Ávila G, Pérez-Rodríguez M, Partida-Rodríguez O, Nieves-Ramírez M, Piña-Ramírez I, Ramírez-Martínez G, Castillejos-López M, Checa M, Ruiz V, Urrea F, Sommer B, Zúñiga J, Selman M. Analysis of heat shock protein 70 gene polymorphisms Mexican patients with idiopathic pulmonary fibrosis. BMC Pulm Med 2015; 15:129. [PMID: 26496868 PMCID: PMC4619986 DOI: 10.1186/s12890-015-0127-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 10/14/2015] [Indexed: 12/13/2022] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease of unknown etiology. Genetic variation within different major histocompatibility complex (MHC) loci contributes to the susceptibility to IPF. The effect of 70 kDa heat shock proteins (HSP70) gene polymorphisms in the susceptibility to IPF is unknown. The aim of this study was to explore the association between HSP70 polymorphisms and IPF susceptibility in the Mexican population. Methods Four HSP70 single nucleotide polymorphisms (SNPs) were evaluated using real time PCR assays in 168 IPF patients and 205 controls: +2763 C>T of HSPA1L (rs2075800), +2437 of HSP HSPA1L A>G (rs2227956), +190 of HSPA1A G>C (rs1043618) and +1267 of HSPA1B G>A (rs1061581). Results The analysis of the recessive model revealed a significant decrease in the frequency of the genotype HSPA1B AA (rs1061581) in IPF patients (OR = 0.27, 95 % CI = 0.13–0.57, Pc = 0.0003) when compared to controls. Using a multivariate logistic regression analysis in a codominant model the HSPA1B (rs1061581) GA and AA genotypes were associated with a lower risk of IPF compared with GG (OR = 0.22, 95 % CI = 0.07–0.65; p = 0.006 and OR = 0.17, 95 % CI = 0.07–0.41; p = <0.001). Similarly, HSPA1L (rs2227956) AG genotype (OR = 0.34, 95 % CI = 0.12–0.99; p = 0.04) and the dominant model AG + GG genotypes were also associated with a lower risk of IPF (OR = 0.24, 95 % CI = 0.08–0.67; p = 0.007). In contrast, the HSPA1L (rs2075800) TT genotype was associated with susceptibility to IPF (OR = 2.52, 95 % CI = 1.32–4.81; p = 0.005). Conclusion Our findings indicate that HSPA1B (rs1061581), HSPA1L (rs2227956) and HSPA1 (rs1043618) polymorphisms are associated with a decreased risk of IPF. Electronic supplementary material The online version of this article (doi:10.1186/s12890-015-0127-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Arnoldo Aquino-Gálvez
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Georgina González-Ávila
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Martha Pérez-Rodríguez
- Hospital General de México, Universidad Nacional Autónoma de México, Laboratorio de Inmunología, Mexico, Mexico.
| | - Oswaldo Partida-Rodríguez
- Unidad de Investigación Médica en Inmunología, CMN S-XXI Instituto Mexicano del Seguro Social, Mexico, Mexico.
| | - Miriam Nieves-Ramírez
- Unidad de Investigación Médica en Inmunología, CMN S-XXI Instituto Mexicano del Seguro Social, Mexico, Mexico.
| | - Inocencio Piña-Ramírez
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Gustavo Ramírez-Martínez
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Manuel Castillejos-López
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Marco Checa
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Victor Ruiz
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Francisco Urrea
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Bettina Sommer
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Joaquin Zúñiga
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Moisés Selman
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| |
Collapse
|
24
|
Furukawa H, Oka S, Shimada K, Tsuchiya N, Tohma S. Genetics of Interstitial Lung Disease: Vol de Nuit (Night Flight). CLINICAL MEDICINE INSIGHTS-CIRCULATORY RESPIRATORY AND PULMONARY MEDICINE 2015; 9:1-7. [PMID: 26056507 PMCID: PMC4444491 DOI: 10.4137/ccrpm.s23283] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/10/2015] [Accepted: 03/13/2015] [Indexed: 01/03/2023]
Abstract
Interstitial lung disease (ILD) is a chronic, progressive fibrotic lung disease with a dismal prognosis. ILD of unknown etiology is referred to as idiopathic interstitial pneumonia (IIP), which is sporadic in the majority of cases. ILD is frequently accompanied by rheumatoid arthritis (RA), systemic sclerosis (SSc), polymyositis/dermatomyositis (PM/DM), and other autoimmune diseases, and is referred to as collagen vascular disease-associated ILD (CVD-ILD). Susceptibility to ILD is influenced by genetic and environmental factors. Recent advances in radiographic imaging techniques such as high-resolution computed tomography (CT) scanning as well as high-throughput genomic analyses have provided insights into the genetics of ILD. These studies have repeatedly revealed an association between IIP (sporadic and familial) and a single nucleotide polymorphism (SNP) in the promoter region of the mucin 5B (MUC5B). HLA-DRB1*11 alleles have been reported to correlate with ILD in European patients with SSc, whereas in Japanese patients with RA, the HLA-DR2 serological group was identified. The aim of this review is to describe the genetic background of sporadic IIP, CVD-ILD, drug-induced-ILD (DI-ILD), pneumoconiosis, and hypersensitivity pneumonitis. The genetics of ILD is still in progress. However, this information will enhance the understanding of the pathogenesis of ILD and aid the identification of novel therapeutic targets for personalized medicine in future.
Collapse
Affiliation(s)
- Hiroshi Furukawa
- Clinical Research Center for Allergy and Rheumatology, Sagamihara Hospital, National Hospital Organization, Sagamihara, Japan
| | - Shomi Oka
- Clinical Research Center for Allergy and Rheumatology, Sagamihara Hospital, National Hospital Organization, Sagamihara, Japan
| | - Kota Shimada
- Department of Rheumatic Diseases, Tokyo Metropolitan Tama Medical Center, Fuchu, Japan
| | - Naoyuki Tsuchiya
- Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Shigeto Tohma
- Clinical Research Center for Allergy and Rheumatology, Sagamihara Hospital, National Hospital Organization, Sagamihara, Japan
| |
Collapse
|
25
|
Kumar A, Dougherty M, Findlay GM, Geisheker M, Klein J, Lazar J, Machkovech H, Resnick J, Resnick R, Salter AI, Talebi-Liasi F, Arakawa C, Baudin J, Bogaard A, Salesky R, Zhou Q, Smith K, Clark JI, Shendure J, Horwitz MS. Genome sequencing of idiopathic pulmonary fibrosis in conjunction with a medical school human anatomy course. PLoS One 2014; 9:e106744. [PMID: 25192356 PMCID: PMC4156421 DOI: 10.1371/journal.pone.0106744] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 08/02/2014] [Indexed: 12/21/2022] Open
Abstract
Even in cases where there is no obvious family history of disease, genome sequencing may contribute to clinical diagnosis and management. Clinical application of the genome has not yet become routine, however, in part because physicians are still learning how best to utilize such information. As an educational research exercise performed in conjunction with our medical school human anatomy course, we explored the potential utility of determining the whole genome sequence of a patient who had died following a clinical diagnosis of idiopathic pulmonary fibrosis (IPF). Medical students performed dissection and whole genome sequencing of the cadaver. Gross and microscopic findings were more consistent with the fibrosing variant of nonspecific interstitial pneumonia (NSIP), as opposed to IPF per se. Variants in genes causing Mendelian disorders predisposing to IPF were not detected. However, whole genome sequencing identified several common variants associated with IPF, including a single nucleotide polymorphism (SNP), rs35705950, located in the promoter region of the gene encoding mucin glycoprotein MUC5B. The MUC5B promoter polymorphism was recently found to markedly elevate risk for IPF, though a particular association with NSIP has not been previously reported, nor has its contribution to disease risk previously been evaluated in the genome-wide context of all genetic variants. We did not identify additional predicted functional variants in a region of linkage disequilibrium (LD) adjacent to MUC5B, nor did we discover other likely risk-contributing variants elsewhere in the genome. Whole genome sequencing thus corroborates the association of rs35705950 with MUC5B dysregulation and interstitial lung disease. This novel exercise additionally served a unique mission in bridging clinical and basic science education.
Collapse
Affiliation(s)
- Akash Kumar
- University of Washington School of Medicine, Seattle, Washington, United States of America
- Medical Scientist Training Program (MSTP), University of Washington, Seattle, Washington, United States of America
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Max Dougherty
- University of Washington School of Medicine, Seattle, Washington, United States of America
- Medical Scientist Training Program (MSTP), University of Washington, Seattle, Washington, United States of America
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Gregory M. Findlay
- University of Washington School of Medicine, Seattle, Washington, United States of America
- Medical Scientist Training Program (MSTP), University of Washington, Seattle, Washington, United States of America
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Madeleine Geisheker
- University of Washington School of Medicine, Seattle, Washington, United States of America
- Medical Scientist Training Program (MSTP), University of Washington, Seattle, Washington, United States of America
| | - Jason Klein
- University of Washington School of Medicine, Seattle, Washington, United States of America
- Medical Scientist Training Program (MSTP), University of Washington, Seattle, Washington, United States of America
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - John Lazar
- University of Washington School of Medicine, Seattle, Washington, United States of America
- Medical Scientist Training Program (MSTP), University of Washington, Seattle, Washington, United States of America
| | - Heather Machkovech
- University of Washington School of Medicine, Seattle, Washington, United States of America
- Medical Scientist Training Program (MSTP), University of Washington, Seattle, Washington, United States of America
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Jesse Resnick
- University of Washington School of Medicine, Seattle, Washington, United States of America
- Medical Scientist Training Program (MSTP), University of Washington, Seattle, Washington, United States of America
| | - Rebecca Resnick
- University of Washington School of Medicine, Seattle, Washington, United States of America
- Medical Scientist Training Program (MSTP), University of Washington, Seattle, Washington, United States of America
| | - Alexander I. Salter
- University of Washington School of Medicine, Seattle, Washington, United States of America
- Medical Scientist Training Program (MSTP), University of Washington, Seattle, Washington, United States of America
| | - Faezeh Talebi-Liasi
- University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Christopher Arakawa
- University of Washington School of Medicine, Seattle, Washington, United States of America
- Medical Scientist Training Program (MSTP), University of Washington, Seattle, Washington, United States of America
| | - Jacob Baudin
- University of Washington School of Medicine, Seattle, Washington, United States of America
- Medical Scientist Training Program (MSTP), University of Washington, Seattle, Washington, United States of America
| | - Andrew Bogaard
- University of Washington School of Medicine, Seattle, Washington, United States of America
- Medical Scientist Training Program (MSTP), University of Washington, Seattle, Washington, United States of America
| | - Rebecca Salesky
- University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Qian Zhou
- University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Kelly Smith
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - John I. Clark
- Department of Biological Structure, University of Washington, Seattle, Washington, United States of America
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Marshall S. Horwitz
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
26
|
Santangelo S, Scarlata S, Zito A, Chiurco D, Pedone C, Incalzi RA. Genetic background of idiopathic pulmonary fibrosis. Expert Rev Mol Diagn 2014; 13:389-406. [DOI: 10.1586/erm.13.22] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
27
|
Luo L, Li Z, Wu W, Luo G, Xu C, Sun Z, Mei H. Role of MICA antibodies in solid organ transplantation. Clin Transplant 2013; 28:152-60. [PMID: 24372774 DOI: 10.1111/ctr.12295] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2013] [Indexed: 02/02/2023]
Affiliation(s)
- Lei Luo
- Department of Research and Education; Guizhou Province People's Hospital; Guiyang China
| | - Zhengyu Li
- Department of Thoracic Surgery; Guizhou Province People's Hospital; Guiyang China
| | - Weidong Wu
- Department of Gynecology; Guizhou Province People's Hospital; Guiyang China
| | - Guangheng Luo
- Department of Urology Surgery; Guizhou Province People's Hospital; Guiyang China
| | - Chuan Xu
- Department of Thoracic Surgery; Guizhou Province People's Hospital; Guiyang China
| | - Zhaolin Sun
- Department of Urology Surgery; Guizhou Province People's Hospital; Guiyang China
| | - Hong Mei
- Department of Thoracic Surgery; Guizhou Province People's Hospital; Guiyang China
| |
Collapse
|
28
|
Al-Qahtani AA, Al-Anazi M, Abdo AA, Sanai FM, Al-Hamoudi W, Alswat KA, Al-Ashgar HI, Khalaf N, Viswan N, Al-Ahdal MN. Genetic variation at -1878 (rs2596542) in MICA gene region is associated with chronic hepatitis B virus infection in Saudi Arabian patients. Exp Mol Pathol 2013; 95:255-8. [PMID: 23994040 DOI: 10.1016/j.yexmp.2013.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 08/13/2013] [Accepted: 08/14/2013] [Indexed: 12/12/2022]
Abstract
MHC class I polypeptide-related chain A (MICA), mapping to 6p21.33, belongs to the non-classical class I family and its expression is induced by several stress factors including viral infection. A recent genome-wide association study has identified a single nucleotide polymorphism (SNP) of MICA, rs2596542 to be significantly associated with hepatitis C-induced hepatocellular carcinoma (HCC) in a Japanese population. Therefore, this study aims to investigate whether the SNP rs2596542 plays any role in hepatitis B virus (HBV) sero-clearance or in the development of complications associated with chronic HBV such as cirrhosis and/or HCC. TaqMan genotyping assay was used to identify the association of the SNP among 584 normal healthy controls and 777 HBV-infected patients. The patient group was further categorized into inactive carriers (Group I), active carriers (Group II), cirrhosis (Group III) and cirrhosis-HCC (Group IV). Variation at this SNP was found to be significantly more frequent in control subjects than in patients (OR = 0.852; 95% C.I. = 0.730-0.994; p = 0.0415). Also, the SNP was found to have a highly significant association when the inactive carriers were compared to the rest of the patients (OR = 1.308; 95% C.I. = 1.058-1.617; p = 0.0130). The TT genotype was found to occur more frequently among active HBV carriers (groups II, III and IV) when compared to inactive HBV carriers, thus suggesting that the rs2596542-T may be recessively associated with an active HBV infection. However, no significant association was observed in the case of HBV-related cirrhosis or HCC. These findings indicate that the MICA rs2596542 has a significant role in HBV infection.
Collapse
Affiliation(s)
- Ahmed A Al-Qahtani
- Department of Infection and Immunity, Research Center, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia; Liver Disease Research Center, King Saud University, Riyadh, Saudi Arabia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Wytrychowski K, Hans-Wytrychowska A, Nowakowska B. Familial idiopathic pulmonary fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 788:363-7. [PMID: 23835999 DOI: 10.1007/978-94-007-6627-3_49] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease of unknown etiology, with an appearance of usual interstitial pneumonia on lung biopsy. To-date, about a 100 families diagnosed with IPF have been described. Familial IPF is defined as histologically confirmed IPF occurring in two or more members of a family. Familial pulmonary fibrosis is hereditary, most probably as a feature which is autosomal dominant with variable penetration. Since 2002, we have been following two families with IPF, referred to in the present article as A and B. The patients in Family A included brother, sister, and sister's daughter. We examined two closest relatives of the patients in family A who are healthy. The patients in Family B included father and his two children. In Family B, we examined six other closest relatives, all of whom proved healthy. In all cases, IPF diagnosis was confirmed histologically. We examined human leukocyte antigen (HLA) alleles in both families, including antigens Class I (locus A, B, and C) and Class II (locus DR). On the basis of the results obtained it is impossible to determine the relation between major histocompatibility complex (MHC) polymorphisms and the incidence of the disease.
Collapse
Affiliation(s)
- K Wytrychowski
- Department of Internal Diseases, Gerontology and Allergology, Wroclaw Medical University, 4 Pasteura St, 50-369, Wroclaw, Poland,
| | | | | |
Collapse
|
30
|
Abstract
There is clear evidence that environmental exposures and genetic predisposition contribute to the pathogenesis of idiopathic pulmonary fibrosis (IPF). Cigarette smoking increases the risk of developing IPF several-fold, as do other exposures such as metal-fume and wood-dust exposure. Occupations that increase the risk of IPF are agricultural work, hairdressing, and stone polishing, supporting the role of environmental exposure in disease pathogenesis. Genetic predisposition to IPF is evident from its familial aggregation and the fact that pulmonary fibrosis develops in several rare genetic disorders. Mutations in surfactant proteins lead to pulmonary fibrosis and are associated with endoplasmic reticulum stress in alveolar type II epithelial cells. Mutations in telomerase have been found in several families with IPF, and shortened telomeres are found in sporadic cases of IPF. A common variant in mucin 5B predisposes to both familial and sporadic IPF and is present in the majority of cases, indicating sporadic IPF occurs in those with genetic predisposition.
Collapse
Affiliation(s)
- Mark P Steele
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA.
| | | |
Collapse
|
31
|
Association of major histocompatibility complex class 1 chain-related gene a dimorphism with type 1 diabetes and latent autoimmune diabetes in adults in the Algerian population. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:557-61. [PMID: 22323559 DOI: 10.1128/cvi.05473-11] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Major histocompatibility complex class I chain-related gene A (MICA-129) dimorphism was investigated in 73 autoimmune diabetes patients (type 1 diabetes and latent autoimmune diabetes in adults) and 75 controls from Algeria. Only MICA-129 Val allele and MICA-129 Val/Val genotype frequencies were higher among patients than in the control group. Statistical analysis of the estimated extended HLA-DR-DQ-MICA haplotypes shown that individual effects of MICA alleles on HLA-DQ2-DR3-MICA-129 Val/Val and HLA-DQ8-DR4-MICA-129 Val/Val haplotypes were significantly higher in patients than in the control groups. These preliminary data might suggest a relevant role of MICA-129 Val/Val single nucleotide polymorphism (weak/weak binders of NKG2D receptor) in the pathogenesis of T1D and LADA.
Collapse
|
32
|
Ding Q, Luckhardt T, Hecker L, Zhou Y, Liu G, Antony VB, deAndrade J, Thannickal VJ. New insights into the pathogenesis and treatment of idiopathic pulmonary fibrosis. Drugs 2011; 71:981-1001. [PMID: 21668038 PMCID: PMC3955181 DOI: 10.2165/11591490-000000000-00000] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common and lethal of the idiopathic interstitial pneumonias. There are currently no effective pharmacological therapies approved for the treatment of IPF. Despite the focus on targeting fibrogenic pathways, recent clinical trials have been largely disappointing. Progress is being made in elucidating key cellular processes and molecular pathways critical to IPF pathogenesis, and this should facilitate the development of more effective therapeutics for this recalcitrant disease. Emerging pathobiological concepts include the role of aging and cellular senescence, oxidative stress, endoplasmic reticulum stress, cellular plasticity, microRNAs and mechanotransduction. Therapeutic approaches that target molecular pathways to modulate aberrant cellular phenotypes and promote tissue homeostasis in the lung must be developed. Heterogeneity in biological and clinical phenotypes of IPF warrants a personalized medicine approach to diagnosis and treatment of this lung disorder.
Collapse
Affiliation(s)
- Qiang Ding
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham
| | - Tracy Luckhardt
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham
| | - Louise Hecker
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham
| | - Yong Zhou
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham
| | - Gang Liu
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham
| | - Veena B. Antony
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham
| | - Joao deAndrade
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham
| | - Victor J. Thannickal
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham
| |
Collapse
|
33
|
Angaswamy N, Saini D, Ramachandran S, Nath DS, Phelan D, Hachem R, Trulock E, Patterson GA, Mohanakumar T. Development of antibodies to human leukocyte antigen precedes development of antibodies to major histocompatibility class I-related chain A and are significantly associated with development of chronic rejection after human lung transplantation. Hum Immunol 2010; 71:560-5. [PMID: 20211214 PMCID: PMC2874120 DOI: 10.1016/j.humimm.2010.02.021] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Revised: 02/09/2010] [Accepted: 02/18/2010] [Indexed: 01/06/2023]
Abstract
The development of antibodies (Abs) to major histocompatibility (MHC) class I-related chain A (MICA) and human leukocyte antigen (HLA) and their role in the immunopathogenesis of chronic rejection (bronchiolitis obliterans syndrome [BOS]) after human lung transplantation (LTx) was analyzed. Sera from 80 LTx recipients were analyzed for anti-MICA and anti-HLA Abs using Luminex and flow PRA (panel reactive assay). Development of Abs either to MICA alone or MICA and HLA together significantly correlated (p < 0.01) with development of BOS. Kinetic analysis in the post-LTx period revealed that development of anti-HLA Abs (7.6 +/- 4.7 months) preceded the development of anti-MICA Abs (10.0 +/- 3.5 months). Abs to MICA alleles (*001 and *009) developed approximately 6 months after LTx and peak titers were present at the time of clinical diagnosis of BOS (16.3 +/- 2.7 months). The development of Abs to both MICA and HLA was strongly associated with the development of BOS thereby suggesting a synergistic effect. Furthermore, immune response to mismatched HLA can lead to development of Abs to other MHC related antigens expressed on the airway epithelial cells. Cumulatively, these immune responses contribute to the pathogenesis of chronic rejection following human LTx.
Collapse
Affiliation(s)
- Nataraju Angaswamy
- Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | - Deepti Saini
- Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | | | - Dilip S. Nath
- Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | - Donna Phelan
- Barnes-Jewish Hospital, Washington University School of Medicine, St. Louis, MO
| | - Ramsey Hachem
- Division of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Elbert Trulock
- Division of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - G. Alexander Patterson
- Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO
| | - T. Mohanakumar
- Department of Surgery, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
34
|
Abstract
The lungs are a major site of entry of pathogens into the body and thus require rapid and effective innate responses to prevent pathogens establishing infection and to limit their spread. Additionally, the immune response in the lung must be tightly regulated such that pathogens are cleared, but immunopathology and chronic inflammation are prevented. In this review, I consider the role of natural killer (NK) cells in pulmonary infection and inflammation, specifically their contributions to influenza, tuberculosis, asthma and chronic obstructive pulmonary disease (COPD), which are major causes of morbidity and mortality world-wide. Despite evidence of the importance of NK cells in these diseases, there are still major gaps in our understanding of how their function is regulated in this unique tissue environment. Understanding how different beneficial and detrimental effector functions of NK cells are triggered will be crucial if NK cells are to be exploited therapeutically in respiratory disease.
Collapse
Affiliation(s)
- Fiona J Culley
- Respiratory Medicine, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London W21PG, UK.
| |
Collapse
|