1
|
Luo M, Tang Y, Qiu J, Yang K, Dai W, Fang E. Proteomic insights into COPD pathogenesis and therapeutic targets: a causal analysis of circulating proteins. Front Med (Lausanne) 2025; 12:1529495. [PMID: 40421300 PMCID: PMC12104083 DOI: 10.3389/fmed.2025.1529495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 04/28/2025] [Indexed: 05/28/2025] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a preventable and treatable condition, yet current therapies do not halt disease progression, highlighting the need for novel treatments. Using an integrated approach combining proteome-wide Mendelian randomization and Bayesian colocalization analysis, this study identifies novel therapeutic targets for COPD. This study explores causal links between circulating proteins and COPD, identifies potential drug targets, and examines the role of lifestyle factors. Methods Using a large proteomics database and a public dataset of 394,244 samples (21,617 COPD cases and 372,627 controls), 4,907 proteins were associated with COPD. Causality was assessed using Bayesian colocalization and proteome-wide Mendelian Randomization (MR), and protein-protein interactions were mapped via STRING. Therapeutic potential was evaluated by identifying drug targets. Additionally, plasma protein expression validation was performed using Western blot experiments in recruited COPD patients and healthy controls. Results MR identified 18 proteins linked to COPD, with 11 accelerating disease onset. Strong colocalization evidence was found for MMP12, ASM, KLC1, NPNT and SNX1, none of which overlap with current COPD drug targets. Western blot analysis validated these findings in plasma samples, showing significantly increased expression of MMP12 and ASM, and decreased expression of NPNT and SNX1 in COPD patients compared to healthy controls, while KLC1 showed no significant difference. Notably, MMP12 was negatively associated with grain and nut intake and positively correlated with smoking (p < 0.05). Conclusion This study identifies potential treatment targets and provides evidence linking specific plasma proteins to COPD risk. Additionally, lifestyle changes may modulate key proteins affecting COPD risk. These findings suggest new avenues for COPD prevention and treatment strategies.
Collapse
Affiliation(s)
- Min Luo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Ying Tang
- Department of Pathology, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jing Qiu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Kai Yang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - WenJing Dai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - EnRong Fang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Kim CY, Park B, Jung JY, Kim JH, Nam CM, An J, Won S, Kim YS. Genome wide interaction study of genetic variants associated with lung function decline. Sci Rep 2025; 15:9824. [PMID: 40118907 PMCID: PMC11928451 DOI: 10.1038/s41598-025-93147-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 03/05/2025] [Indexed: 03/24/2025] Open
Abstract
Some genetic variants are associated with lung function decline and chronic obstructive pulmonary disease (COPD), but functional studies are necessary to confirm causality. We investigated the genetic susceptibility-associated lung function decline with or without COPD, using data from a community-based cohort (N = 8554). A genome-wide interaction study was conducted to identify the association between genetic variants and pulmonary function, and the way variants relate to lung impairment in accordance with smoking status and amount was examined. We further used a linear mixed model to examine the association and interaction to time effect. We found annual mean FEV1 declines of 41.7 mL for men and 33.4 mL for women, and the annual rate of decline in FEV1 was the fastest for current smokers. We also found a previously identified locus near FAM13A, the most significant SNPs from the results of two likelihood ratio tests for FEV1/FVC (P = 1.56 × 10-10). These selected SNPs were located in the upstream region of FAM13A on chromosome 4 and had similar minor allele frequencies (MAFs). Furthermore, we found that certain SNPs tended to have lower FEV1/FVC values, and lung function decreased much faster with time interactions. The SNP most associated with lung function decline was the rs75679995 SNP on chromosome 7, and those SNPs located within the TAD of the DNAH11 region and the eQTL of rs9991425 revealed a higher expression of MFAP3L and AADAT genes (P = 2.28 × 10-7 and 2.01 × 10-6, respectively). This is the first study to investigate gene-time interactions in lung function decline as a risk factor for COPD in the Korean population. In addition to replicating previously known signals for FAM13A, we identified two genomic regions (DNAH11, AADAT) that are potentially involved in gene-environment interactions, warranting further investigation to confirm their roles.
Collapse
Affiliation(s)
- Chi Young Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Boram Park
- Department of Public Health Sciences, School of Public Health, Seoul National University, Kwanak-Ro Kwanak-Gu, Seoul, 151-742, South Korea
| | - Ji Ye Jung
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Je Hyeong Kim
- Division of Pulmonology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea
| | - Chung Mo Nam
- Department of Preventive Medicine and Public Health, Yonsei University College of Medicine, Seoul, Korea
| | - Jaehoon An
- Department of Public Health Sciences, School of Public Health, Seoul National University, Kwanak-Ro Kwanak-Gu, Seoul, 151-742, South Korea
- RexSoft Corps, Seoul, South Korea
| | - Sungho Won
- Department of Public Health Sciences, School of Public Health, Seoul National University, Kwanak-Ro Kwanak-Gu, Seoul, 151-742, South Korea.
- Institute of Health and Environment, Seoul National University, Seoul, South Korea.
- RexSoft Corps, Seoul, South Korea.
| | - Young Sam Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Fricke-Galindo I, García-Carmona S, Bautista-Becerril B, Pérez-Rubio G, Buendia-Roldan I, Chávez-Galán L, Nava-Quiroz KJ, Alanis-Ponce J, Reséndiz-Hernández JM, Blanco-Aguilar E, Erives-Sedano JI, Méndez-Velasco Y, Osuna-Espinoza GE, Salvador-Hernández F, Segura-Castañeda R, Solano-Candia UN, Falfán-Valencia R. Genetic Variants in Genes Related to Lung Function and Interstitial Lung Diseases Are Associated with Worse Outcomes in Severe COVID-19 and Lung Performance in the Post-COVID-19 Condition. Int J Mol Sci 2025; 26:2046. [PMID: 40076669 PMCID: PMC11900979 DOI: 10.3390/ijms26052046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Genetic variants related to susceptibility to chronic respiratory conditions such as interstitial lung disease (ILD) could share critical pathways in the pathogenesis of COVID-19 and be implicated in COVID-19 outcomes and post-COVID-19. We aimed to identify the participation of genetic variants in lung function and ILD genes in severe COVID-19 outcomes and post-COVID-19 condition. We studied 936 hospitalized patients with COVID-19. The requirement of invasive mechanical ventilation (IMV) and the acute respiratory distress syndrome (ARDS) classification were considered. The mortality was assessed as the in-hospital death. The post-COVID-19 group included 102 patients evaluated for pulmonary function tests four times during the year after discharge. Five variants (FAM13A rs2609255, DSP rs2076295, TOLLIP rs111521887, TERT rs2736100, and THSD4 rs872471) were genotyped using TaqMan assays. A multifactor dimensionality reduction method (MDR) was performed for epistasis estimation. The TERT rs2736100 and THSD4 rs872471 variants were associated with differential risk for ARDS severity (moderate vs. severe, CC + CA, p = 0.044, OR = 0.66, 95% CI = 0.44-0.99; and GG p = 0.034, OR = 2.22, 95% CI = 1.04-4.72, respectively). These variants and FAM13A rs2609255 were also related to pulmonary function post-COVID-19. The MDR analysis showed differential epistasis and correlation of the genetic variants included in this study. The well-known variants in recognized genes related to pulmonary function worsening and interstitial disorders are related to the severity and mortality of COVID-19 and lung performance in the post-COVID-19 condition.
Collapse
Affiliation(s)
- Ingrid Fricke-Galindo
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (S.G.-C.); (B.B.-B.); (G.P.-R.); (J.A.-P.)
| | - Salvador García-Carmona
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (S.G.-C.); (B.B.-B.); (G.P.-R.); (J.A.-P.)
| | - Brandon Bautista-Becerril
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (S.G.-C.); (B.B.-B.); (G.P.-R.); (J.A.-P.)
| | - Gloria Pérez-Rubio
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (S.G.-C.); (B.B.-B.); (G.P.-R.); (J.A.-P.)
| | - Ivette Buendia-Roldan
- Translational Research Laboratory on Aging and Pulmonary Fibrosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Leslie Chávez-Galán
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Karol J. Nava-Quiroz
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (S.G.-C.); (B.B.-B.); (G.P.-R.); (J.A.-P.)
| | - Jesús Alanis-Ponce
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (S.G.-C.); (B.B.-B.); (G.P.-R.); (J.A.-P.)
| | - Juan M. Reséndiz-Hernández
- Laboratorio Clínico, Centro Especializado de Atención a Personas con Discapacidad Visual, Instituto de Salud del Estado de México, Naucalpan 53000, Mexico State, Mexico;
| | - Esther Blanco-Aguilar
- Facultad de Medicina Benemérita, Universidad Autónoma de Puebla, Puebla de Zaragoza 72420, Puebla, Mexico;
| | - Jessica I. Erives-Sedano
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez 32310, Chihuahua, Mexico;
| | - Yashohara Méndez-Velasco
- Unidad Académica Profesional Chimalhuacán, Universidad Autónoma del Estado de México, Nezahualcóyotl 56353, Mexico State, Mexico;
| | - Grecia E. Osuna-Espinoza
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán Rosales 80030, Sinaloa, Mexico;
| | - Fidel Salvador-Hernández
- Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City 07320, Mexico;
| | - Rubén Segura-Castañeda
- Facultad Interdisciplinaria de Ciencias Biológicas y de Salud, Universidad de Sonora, Hermosillo Sonora 83000, Sonora, Mexico;
| | - Uriel N. Solano-Candia
- Facultad de Biología, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58000, Michoacán, Mexico;
| | - Ramcés Falfán-Valencia
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (I.F.-G.); (S.G.-C.); (B.B.-B.); (G.P.-R.); (J.A.-P.)
| |
Collapse
|
4
|
Liao SX, Wang YW, Sun PP, Xu Y, Wang TH. Prospects of neutrophilic implications against pathobiology of chronic obstructive pulmonary disease: Pharmacological insights and technological advances. Int Immunopharmacol 2025; 144:113634. [PMID: 39577220 DOI: 10.1016/j.intimp.2024.113634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/03/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a prevalent chronic inflammatory condition that affects the lungs globally. A key feature of this inflammatory response is the migration and aggregation of polymorphonuclear neutrophils (PMNs). The presence of neutrophilic inflammation within the airways is as distinguishing characteristic of COPD. As research advances, PMNs and their products emerge as central players in the airway inflammatory cascade of COPD patients. Their involvement in phagocytosis, degranulation, and the formation of neutrophil extracellular traps (NETs) significantly contributes to the pathogenesis of COPD. Moreover, studies have shown that excessive biological activities of neutrophils in the lungs can result in airway epithelial injury, emphysema, and mucus hypersecretion. Currently, there is growing empirical support for the moderate targeting neutrophils in the clinical management of COPD. This article delves into the pivotal role of neutrophils in COPD, emphasizing the urgency for novel therapeutic approaches that specifically target neutrophils. Additionally, it explores the potential of utilizing single-cell RNA sequencing to further investigate neutrophils and relevant risk genes as potential biomarkers for COPD treatment. By elucidating these mechanisms, this review aims to pave the way for future strategies to modulate neutrophil function, thereby addressing the pressing need for more effective COPD therapies.
Collapse
Affiliation(s)
- Shi-Xia Liao
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
| | - Yan-Wen Wang
- West China Clinical Medical College, Sichuan University, Chengdu 610041, China
| | - Peng-Peng Sun
- Department of Osteopathy, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
| | - Yang Xu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Ting-Hua Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; Institute of Neurological Disease, West China Hospital, Sichuan University & The Research Units of West China, Chinese Academy of Medical Sciences, Chengdu 610041, China.
| |
Collapse
|
5
|
Axelsson GT, Jonmundsson T, Woo Y, Frick EA, Aspelund T, Loureiro JJ, Orth AP, Jennings LL, Gudmundsson G, Emilsson V, Gudmundsdottir V, Gudnason V. Proteomic associations with forced expiratory volume: a Mendelian randomisation study. Respir Res 2024; 25:44. [PMID: 38238732 PMCID: PMC10797790 DOI: 10.1186/s12931-023-02587-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 10/30/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND A decline in forced expiratory volume (FEV1) is a hallmark of respiratory diseases that are an important cause of morbidity among the elderly. While some data exist on biomarkers that are related to FEV1, we sought to do a systematic analysis of causal relations of biomarkers with FEV1. METHODS Data from the population-based AGES-Reykjavik study were used. Serum proteomic measurements were done using 4782 DNA aptamers (SOMAmers). Data from 1479 participants with spirometric data were used to assess the association of SOMAmer measurements with FEV1 using linear regression. Bi-directional two-sample Mendelian randomisation (MR) analyses were done to assess causal relations of observationally associated SOMAmers with FEV1, using genotype and SOMAmer data from 5368 AGES-Reykjavik participants and genetic associations with FEV1 from a publicly available GWAS (n = 400,102). RESULTS In observational analyses, 530 SOMAmers were associated with FEV1 after multiple testing adjustment (FDR < 0.05). The most significant were Retinoic Acid Receptor Responder 2 (RARRES2), R-Spondin 4 (RSPO4) and Alkaline Phosphatase, Placental Like 2 (ALPPL2). Of the 257 SOMAmers with genetic instruments available, eight were associated with FEV1 in MR analyses. Three were directionally consistent with the observational estimate, Thrombospondin 2 (THBS2), Endoplasmic Reticulum Oxidoreductase 1 Beta (ERO1B) and Apolipoprotein M (APOM). THBS2 was further supported by a colocalization analysis. Analyses in the reverse direction, testing whether changes in SOMAmer levels were caused by changes in FEV1, were performed but no significant associations were found after multiple testing adjustments. CONCLUSIONS In summary, this large scale proteogenomic analyses of FEV1 reveals circulating protein markers of FEV1, as well as several proteins with potential causality to lung function.
Collapse
Affiliation(s)
- Gisli Thor Axelsson
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland
- Department of Internal Medicine, Landspitali University Hospital, 101, Reykjavik, Iceland
| | - Thorarinn Jonmundsson
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland
| | - Youngjae Woo
- Novartis Biomedical Research, Cambridge, MA, 02139, USA
| | | | - Thor Aspelund
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland
| | | | - Anthony P Orth
- Novartis Institutes for Biomedical Research, San Diego, CA, 92121, USA
| | | | - Gunnar Gudmundsson
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland
- Department of Respiratory Medicine and Sleep, Landspitali University Hospital, 108, Reykjavik, Iceland
| | - Valur Emilsson
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland
| | - Valborg Gudmundsdottir
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland.
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland.
| | - Vilmundur Gudnason
- Icelandic Heart Association, Holtasmari 1, 201, Kopavogur, Iceland.
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland.
| |
Collapse
|
6
|
Zhang M, Hei R, Zhou Z, Xiao W, Liu X, Chen Y. Macrophage polarization involved the inflammation of chronic obstructive pulmonary disease by S1P/HDAC1 signaling. Am J Cancer Res 2023; 13:4478-4489. [PMID: 37818082 PMCID: PMC10560935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 09/04/2023] [Indexed: 10/12/2023] Open
Abstract
Globally, chronic obstructive pulmonary disease (COPD) is the cause of high morbidity and mortality, and constitutes a huge public health burden. Previous studies have reported that inflammation is closely related to COPD, but its potential mechanism is still unclear. Since the polarization of macrophages is involved in regulating inflammation, we assume that COPD changes the polarization of macrophages. To verify this, we investigated the relationship between the expression of S1PR1, HADC1, and inflammatory macrophages in COPD patients via flow cytometry, qRT-PCR, and western blot analysis. We found that macrophages of COPD individuals differentiated into M1 phenotype, and the expression of S1PR1 increased and HDAC1 decreased. S1PR1 also inhibits the expression of HDAC1, so S1PR1/HDAC1 signal regulates the polarization of macrophages. The results of the study put forward new ideas of the pathogenesis of COPD, and also proposed the possible treatment options.
Collapse
Affiliation(s)
- Min Zhang
- Department of Pulmonary Critical Care Medicine, The 1st Affiliated Hospital of Shenzhen UniversityShenzhen 518035, Guangdong, PR China
| | - Ruoxuan Hei
- Department of Clinical Diagnose, The Second Affiliated Hospital of The Air Force Military Medical UniversityNo. 569 Xinsi Road, Xi’an 710038, Shaanxi, PR China
| | - Zhou Zhou
- Department of Pulmonary and Critical Care Medicine, Southern University of Science and Technology HospitalShenzhen 518102, Guangdong, PR China
| | - Wendi Xiao
- Department of Pulmonary Critical Care Medicine, The 1st Affiliated Hospital of Shenzhen UniversityShenzhen 518035, Guangdong, PR China
| | - Xi Liu
- Department of Pulmonary Critical Care Medicine, The 1st Affiliated Hospital of Shenzhen UniversityShenzhen 518035, Guangdong, PR China
| | - Yanwei Chen
- Department of Pulmonary Critical Care Medicine, The 1st Affiliated Hospital of Shenzhen UniversityShenzhen 518035, Guangdong, PR China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of The Air Force Military Medical UniversityNo. 569 Xinsi Road, Xi’an 710038, Shaanxi, PR China
| |
Collapse
|
7
|
Axelsson GT, Jonmundsson T, Woo YJ, Frick EA, Aspelund T, Loureiro JJ, Orth AP, Jennings LL, Gudmundsson G, Emilsson V, Gudmundsdottir V, Gudnason V. Proteomic associations with forced expiratory volume - a Mendelian randomisation study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.30.23292035. [PMID: 37425696 PMCID: PMC10327250 DOI: 10.1101/2023.06.30.23292035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
A decline in forced expiratory volume (FEV1) is a hallmark of obstructive respiratory diseases, an important cause of morbidity among the elderly. While some data exist on biomarkers that are related to FEV1, we sought to do a systematic analysis of causal relations of biomarkers with FEV1. Data from the general population-based AGES-Reykjavik study were used. Proteomic measurements were done using 4,782 DNA aptamers (SOMAmers). Data from 1,648 participants with spirometric data were used to assess the association of SOMAmer measurements with FEV1 using linear regression. Bi-directional Mendelian randomisation (MR) analyses were done to assess causal relations of observationally associated SOMAmers with FEV1, using genotype and SOMAmer data from 5,368 AGES-Reykjavik participants and genetic associations with FEV1 from a publicly available GWAS (n = 400,102). In observational analyses, 473 SOMAmers were associated with FEV1 after multiple testing adjustment. The most significant were R-Spondin 4, Alkaline Phosphatase, Placental Like 2 and Retinoic Acid Receptor Responder 2. Of the 235 SOMAmers with genetic data, eight were associated with FEV1 in MR analyses. Three were directionally consistent with the observational estimate, Thrombospondin 2 (THBS2), Endoplasmic Reticulum Oxidoreductase 1 Beta and Apolipoprotein M. THBS2 was further supported by a colocalization analysis. Analyses in the reverse direction, testing whether changes in SOMAmer levels were caused by changes in FEV1, were performed but no significant associations were found after multiple testing adjustments. In summary, this large scale proteogenomic analyses of FEV1 reveals protein markers of FEV1, as well as several proteins with potential causality to lung function.
Collapse
|
8
|
Huang CW, Lo SH. Tensins in Kidney Function and Diseases. Life (Basel) 2023; 13:1244. [PMID: 37374025 PMCID: PMC10305691 DOI: 10.3390/life13061244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/11/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Tensins are focal adhesion proteins that regulate various biological processes, such as mechanical sensing, cell adhesion, migration, invasion, and proliferation, through their multiple binding activities that transduce critical signals across the plasma membrane. When these molecular interactions and/or mediated signaling are disrupted, cellular activities and tissue functions are compromised, leading to disease development. Here, we focus on the significance of the tensin family in renal function and diseases. The expression pattern of each tensin in the kidney, their roles in chronic kidney diseases, renal cell carcinoma, and their potentials as prognostic markers and/or therapeutic targets are discussed in this review.
Collapse
Affiliation(s)
- Chien-Wei Huang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Su Hao Lo
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA
| |
Collapse
|
9
|
Lin L, Li J, Song Q, Cheng W, Chen P. The role of HMGB1/RAGE/TLR4 signaling pathways in cigarette smoke-induced inflammation in chronic obstructive pulmonary disease. Immun Inflamm Dis 2022; 10:e711. [PMID: 36301039 PMCID: PMC9552978 DOI: 10.1002/iid3.711] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/10/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common chronic respiratory disease with irreversible and continuous progression. It has become the fifth most burdensome disease and the third most deadly disease globally. Therefore, the prevention and treatment of COPD are urgent, and it is also important to clarify the pathogenesis of it. Smoking is the main and most common risk factor for COPD. Cigarette smoke (CS) can cause lung inflammation and other pathological mechanisms in the airways and lung tissue. Airway inflammation is one of the important mechanisms leading to the pathogenesis of COPD. Recent studies have shown that high mobility group box 1 (HMGB1) is involved in the occurrence and development of respiratory diseases, including COPD. HMGB1 is a typical damage-associated molecular pattern (DAMP) protein, which mainly exerts its activity by binding to the receptor for advanced glycation end products (RAGE) and toll-like receptor 4 (TLR4) and further participate in the process of airway inflammation. Studies have shown that the abnormal expression of HMGB1, RAGE, and TLR4 are related to inflammation in COPD. Herein, we discuss the roles of HMGB1, RAGE, and TLR4 in CS/cigarette smoke extract-induced inflammation in COPD, providing a new target for the diagnosis, treatment and prevention of COPD.
Collapse
Affiliation(s)
- Ling Lin
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Jing Li
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Qing Song
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Wei Cheng
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Ping Chen
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| |
Collapse
|
10
|
Dormoy V, Pons S, Gosset P, Maskos U. [Nicotinic acetylcholine receptor and chronic obstructive pulmonary disease]. Med Sci (Paris) 2022; 38:637-639. [PMID: 36094229 DOI: 10.1051/medsci/2022092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Valérian Dormoy
- Université de Reims Champagne-Ardenne, Inserm, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France
| | - Stéphanie Pons
- Institut Pasteur de Paris, Université Paris Cité, Neurobiologie intégrative des systèmes cholinergiques, CNRS UMR 3571, Paris, France
| | - Philippe Gosset
- Université de Lille, CNRS UMR8204, Inserm U1019, CHRU Lille, Institut Pasteur Lille, Centre d'infection et d'immunité de Lille, Lille, France
| | - Uwe Maskos
- Institut Pasteur de Paris, Université Paris Cité, Neurobiologie intégrative des systèmes cholinergiques, CNRS UMR 3571, Paris, France
| |
Collapse
|
11
|
Pantazopoulos I, Magounaki K, Kotsiou O, Rouka E, Perlikos F, Kakavas S, Gourgoulianis K. Incorporating Biomarkers in COPD Management: The Research Keeps Going. J Pers Med 2022; 12:jpm12030379. [PMID: 35330379 PMCID: PMC8955907 DOI: 10.3390/jpm12030379] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/17/2022] [Accepted: 02/28/2022] [Indexed: 12/17/2022] Open
Abstract
Globally, chronic obstructive pulmonary disease (COPD) remains a major cause of morbidity and mortality, having a significant socioeconomic effect. Several molecular mechanisms have been related to COPD including chronic inflammation, telomere shortening, and epigenetic modifications. Nowadays, there is an increasing need for novel therapeutic approaches for the management of COPD. These treatment strategies should be based on finding the source of acute exacerbation of COPD episodes and estimating the patient’s own risk. The use of biomarkers and the measurement of their levels in conjunction with COPD exacerbation risk and disease prognosis is considered an encouraging approach. Many types of COPD biomarkers have been identified which include blood protein biomarkers, cellular biomarkers, and protease enzymes. They have been isolated from different sources including peripheral blood, sputum, bronchoalveolar fluid, exhaled air, and genetic material. However, there is still not an exclusive biomarker that is used for the evaluation of COPD but rather a combination of them, and this is attributed to disease complexity. In this review, we summarize the clinical significance of COPD-related biomarkers, their association with disease outcomes, and COPD patients’ management. Finally, we depict the various samples that are used for identifying and measuring these biomarkers.
Collapse
Affiliation(s)
- Ioannis Pantazopoulos
- Department of Emergency Medicine, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece
- Correspondence: ; Tel.: +30-6945661525
| | | | - Ourania Kotsiou
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece; (O.K.); (E.R.); (K.G.)
| | - Erasmia Rouka
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece; (O.K.); (E.R.); (K.G.)
| | - Fotis Perlikos
- ICU Department, Henry Dynant Hospital Center, 11526 Athens, Greece;
| | - Sotirios Kakavas
- Critical Care Department, “Sotiria” General Hospital of Chest Diseases, 11527 Athens, Greece;
| | - Konstantinos Gourgoulianis
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece; (O.K.); (E.R.); (K.G.)
| |
Collapse
|
12
|
miR-31-5p modulates cell progression in lung adenocarcinoma through TNS1/p53 axis. Strahlenther Onkol 2022; 198:304-314. [PMID: 35037949 DOI: 10.1007/s00066-021-01895-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 12/17/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To clarify the modulatory mechanism of miR-31-5p in lung adenocarcinoma (LUAD) progression in vivo and in vitro. METHODS The Cancer Genome Atlas (TCGA) database was employed to access LUAD-related miRNA and mRNA expression data. Downstream targets of miR-31-5p were predicted by public databases. The interaction between miR-31-5p and TNS1 was determined by dual-luciferase reporter assay. Quantitative real-time polymerase chain reaction (qRT-PCR) was utilized to measure miR-31-5p and TNS1 expression levels in LUAD cells. Western blot was introduced to test protein expression levels of TNS1, p53, and apoptosis-related proteins. In-vitro functional assays were conducted to evaluate the biological effects of miR-31-5p on cell proliferation, colony formation, migration, and apoptosis. In-vivo tumor xenograft experiment was applied to examine the effects of miR-31-5p on LUAD tumor growth, followed by immunochemistry assays for assessing TNS1 and p53 expression levels in the tumor tissue. RESULTS miR-31-5p was prominently upregulated in LUAD tissue and was identified to present a similar trend in LUAD cell lines H1299, H23, and A549. miR-31-5p overexpression exerted an active role in cell proliferation and migration, but it suppressed cell apoptosis. Additionally, a reverse correlation between miR-31-5p and TNS1 regarding the expression level was identified, and TNS1 was verified to be a direct target of miR-31-5p. Besides, it was further validated by the rescue experiments that the tumor-promoting effects of miR-31-5p on LUAD cell functions were attenuated by TNS1 overexpression to some extent. The results based on the tumor xenograft experiment revealed that LUAD cell growth could be facilitated by miR-31-5p via the TNS1/p53 axis. CONCLUSION miR-31-5p facilitates LUAD cell progression mediated by the TNS1/p53 axis.
Collapse
|
13
|
Guan Q, Tian Y, Zhang Z, Zhang L, Zhao P, Li J. Identification of Potential Key Genes in the Pathogenesis of Chronic Obstructive Pulmonary Disease Through Bioinformatics Analysis. Front Genet 2021; 12:754569. [PMID: 34804123 PMCID: PMC8595135 DOI: 10.3389/fgene.2021.754569] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/13/2021] [Indexed: 12/20/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common respiratory disease with high morbidity and mortality. The etiology of COPD is complex, and the pathogenesis mechanisms remain unclear. In this study, we used rat and human COPD gene expression data from our laboratory and the Gene Expression Omnibus (GEO) database to identify differentially expressed genes (DEGs) between individuals with COPD and healthy individuals. Then, protein–protein interaction (PPI) networks were constructed, and hub genes were identified. Cytoscape was used to construct the co-expressed network and competitive endogenous RNA (ceRNA) networks. A total of 198 DEGs were identified, and a PPI network with 144 nodes and 355 edges was constructed. Twelve hub genes were identified by the cytoHubba plugin in Cytoscape. Of these genes, CCR3, CCL2, COL4A2, VWF, IL1RN, IL2RA, and CCL13 were related to inflammation or immunity, or tissue-specific expression in lung tissue, and their messenger RNA (mRNA) levels were validated by qRT-PCR. COL4A2, VWF, and IL1RN were further verified by the GEO dataset GSE76925, and the ceRNA network was constructed with Cytoscape. These three genes were consistent with COPD rat model data compared with control data, and their dysregulation direction was reversed when the COPD rat model was treated with effective-component compatibility of Bufei Yishen formula III. This bioinformatics analysis strategy may be useful for elucidating novel mechanisms underlying COPD. We pinpointed three key genes that may play a role in COPD pathogenesis and therapy, which deserved to be further studied.
Collapse
Affiliation(s)
- Qingzhou Guan
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China.,Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yange Tian
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China.,Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhenzhen Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China.,Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lanxi Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China.,Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Peng Zhao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China.,Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China.,Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
14
|
An J, Yang T, Dong J, Liao Z, Wan C, Shen Y, Chen L. Identifying miRNA Modules and Related Pathways of Chronic Obstructive Pulmonary Disease Associated Emphysema by Weighted Gene Co-Expression Network Analysis. Int J Chron Obstruct Pulmon Dis 2021; 16:3119-3130. [PMID: 34815668 PMCID: PMC8605490 DOI: 10.2147/copd.s325300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/25/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a heterogeneous chronic inflammatory disease characterized by progressive airflow limitation that causes high morbidity and mortality. MicroRNA, a short-chain noncoding RNA, regulates gene expression at the transcriptional level. microRNA modules with a role in the pathogenesis of COPD may serve as COPD biomarkers. METHODS We downloaded the GSE33336 microarray data set from the Gene Expression Omnibus (GEO) database, the data are derived from 29 lung samples of patients with emphysema undergoing curative resection for lung cancer. We used weighted gene co-expression network analysis (WGCNA) to construct co-expression modules and detect trait-related microRNA modules. We used the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis to predict the biological function of the interest modules, and we screened out candidate hub microRNAs based on their module membership (MM) value and top proteins on the results of the protein-protein interaction (PPI) network. RESULTS Three microRNA modules (royal blue, light yellow and grey60) were highly associated with COPD. Axon guidance, proteoglycans in cancer and mitogen-activated protein kinases (MAPK) signaling pathway were common pathways in these three modules. Keratin18 (KRT18) was the top protein in our study. miR-452, miR-149, miR-133a, miR-181a and miR-421 in hub microRNAs may play a role in COPD. CONCLUSION These findings provide evidence for the role of miRNAs in COPD and identify biomarker candidates.
Collapse
Affiliation(s)
- Jing An
- Department of Respiratory and Critical Care Medicine, Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Ting Yang
- Department of Respiratory and Critical Care Medicine, Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Jiajia Dong
- Department of Respiratory and Critical Care Medicine, Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Zenglin Liao
- Department of Respiratory and Critical Care Medicine, Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Chun Wan
- Department of Respiratory and Critical Care Medicine, Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Yongchun Shen
- Department of Respiratory and Critical Care Medicine, Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Lei Chen
- Department of Respiratory and Critical Care Medicine, Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| |
Collapse
|
15
|
Zhao H, Jiang J, Wang M, Xuan Z. Genome-Wide Identification of m6A-Associated Single-Nucleotide Polymorphisms in Colorectal Cancer. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2021; 14:887-892. [PMID: 34305406 PMCID: PMC8297552 DOI: 10.2147/pgpm.s314373] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/30/2021] [Indexed: 12/15/2022]
Abstract
Background N6-methyladenosine (m6A)-associated single-nucleotide polymorphisms (SNPs) play important roles in cancers, with previous research suggesting potential associations between m6A-SNPs and cancer. However, the relationship between the genetic determinants of m6A modification and colorectal cancer (CRC) remains unclear. Methods An integrative method combining raw data and summary statistics of genome-wide association studies with expression quantitative trait loci (eQTL) and differential expression data was applied to screen potential candidate CRC-associated m6A-SNPs. Results A total of 402 m6A-SNPs were identified as being associated with CRC (p < 0.001), with 98 showing eQTL signals. In particular, three genes were found to harbor CRC-associated m6A-SNPs: rs178184 in NOVA1, rs35782901 in HTR4, and rs60571683 in SLCO1B3. These genes were differentially expressed in at least one publicly available dataset (p < 0.05), with NOVA1 (p = 3.41×10-11) and HTR4 (p = 5.56×10-7) being significantly downregulated in CRC (dataset: GSE89076), and SLCO1B3 was significantly overexpressed (datasets: GSE32323 [p = 3.27×10-5], GSE21510 [p = 1.09×10-6], and GSE89076 [p = 7.63×10-6]). Conclusion This study identified three m6A-SNPs (rs178184, rs35782901, and rs60571683) that may be associated with CRC. However, the lack of analysis of primary CRC samples in order to further elucidate the underlying pathogenesis is a major limitation of this study. Future investigations are needed to validate these CRC-associated m6A-SNPs and explore the m6A-mediated pathogenic mechanism in CRC.
Collapse
Affiliation(s)
- Hongying Zhao
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China
| | - Jinying Jiang
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China
| | - Mingshan Wang
- Departments of Infection Diseases, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China
| | - Zixue Xuan
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
16
|
Walton NI, Zhang X, Soltis AR, Starr J, Dalgard CL, Wilkerson MD, Conrad D, Pollard HB. Tensin 1 (TNS1) is a modifier gene for low body mass index (BMI) in homozygous [F508del]CFTR patients. Physiol Rep 2021; 9:e14886. [PMID: 34086412 PMCID: PMC8176904 DOI: 10.14814/phy2.14886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/02/2021] [Indexed: 11/24/2022] Open
Abstract
Cystic fibrosis (CF) is a life‐limiting autosomal recessive genetic disease caused by variants in the CFTR gene, most commonly by the [F508del] variant. Although CF is a classical Mendelian disease, genetic variants in several modifier genes have been associated with variation of the clinical phenotype for pulmonary and gastrointestinal function and urogenital development. We hypothesized that whole genome sequencing of a well‐phenotyped CF populations might identify novel variants in known, or hitherto unknown, modifier genes. Whole genome sequencing was performed on the Illumina HiSeq X platform for 98 clinically diagnosed cystic fibrosis patient samples from the Adult CF Clinic at the University of California San Diego (UCSD). We compared protein‐coding, non‐silent variants genome wide between CFTR [F508del] homozygotes vs CFTR compound heterozygotes. Based on a single variant score test, we found 3 SNPs in common variants (MAF >5%) that occurred at significantly different rates between homozygous [F508del]CFTR and compound heterozygous [F508del]CFTR patients. The 3 SNPs were all located in one gene on chromosome 2: Tensin 1 (TNS1: rs3796028; rs2571445: and rs918949). We observed significantly lower BMIs in homozygous [F508del]CFTR patients who were also homozygous for Tensin 1 rs918949 (T/T) (p = 0.023) or rs2571445 (G/G) (p = 0.02) variants. The Tensin 1 gene is thus a potential modifier gene for low BMI in CF patients homozygous for the [F508del]CFTR variant.
Collapse
Affiliation(s)
- Nathan I Walton
- The Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Consortium for Health and Military Performance, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Xijun Zhang
- The Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Anthony R Soltis
- The Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Joshua Starr
- The Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Clifton L Dalgard
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Matthew D Wilkerson
- The Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.,The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Douglas Conrad
- Department of Medicine, University of California, San Diego, CA, USA
| | - Harvey B Pollard
- The Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Consortium for Health and Military Performance, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
17
|
Park S, Lee S, Kim Y, Cho S, Kim K, Kim YC, Han SS, Lee H, Lee JP, Joo KW, Lim CS, Kim YS, Kim DK. Kidney function and obstructive lung disease: a bidirectional Mendelian randomisation study. Eur Respir J 2021; 58:13993003.00848-2021. [PMID: 33958431 DOI: 10.1183/13993003.00848-2021] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 04/26/2021] [Indexed: 11/05/2022]
Abstract
BACKGROUND Additional study is warranted to investigate the causal effects between kidney function and obstructive lung disease. METHODS This study was a bidirectional two-sample Mendelian randomisation (MR) analysis. The CKDGen genome-wide association study (GWAS) meta-analysis for estimated glomerular filtration rate (eGFR) including individuals of European ancestry (N=567 460) provided the genetic instrument for kidney function and outcome summary statistics. A GWAS for FEV1/FVC including individuals of European ancestry from the UK Biobank (N=321 047) provided the genetic instrument for FEV1/FVC and outcome data. A polygenic score (PGS) analysis was performed to test the causal estimates from kidney function to binary obstructive lung disease outcomes, including chronic obstructive pulmonary disease (COPD), asthma, and FEV1/FVC<70%, and to perform non-linear MR with individual-level UK Biobank data. RESULTS The causal estimates by summary-level MR indicated that genetically predicted increased kidney function was significantly associated with increased FEV1/FVC Z scores [10% increase in eGFR, beta 0.055 (0.024, 0.086)]. The PGS for increased eGFR showed a significant association with a reduced risk of FEV1/FVC<70% [OR 0.93 (0.87, 0.99)], COPD [OR 0.93 (0.87, 0.99)] and late-onset (≥50 years old) asthma [OR 0.93 (0.88, 0.99)]. The non-linear MR demonstrated that the causal effect from eGFR to FEV1/FVC was apparent in eGFR ranges lower than 60 mL/min/1.73 m2. On the other hand, genetically predicted FEV1/FVC showed nonsignificant causal estimates of eGFR change [beta 0.568% (-0.458, 1.605%)]. CONCLUSION This study supports kidney function impairment would be a causative factor for obstructive lung disease.
Collapse
Affiliation(s)
- Sehoon Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Armed Forces Capital Hospital, Gyeonggi-do, Korea
| | - Soojin Lee
- Division of Nephrology, Department of Internal Medicine, Uijeongbu Eulji University Medical Center, Gyeonggi-do, Korea
| | - Yaerim Kim
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Semin Cho
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kwangsoo Kim
- Transdisciplinary Department of Medicine & Advanced Technology, Seoul National University Hospital, Seoul, Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Seung Seok Han
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Hajeong Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea
| | - Jung Pyo Lee
- Kidney Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Kwon Wook Joo
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Chun Soo Lim
- Kidney Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Yon Su Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.,Kidney Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea .,Kidney Research Institute, Seoul National University, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Li Y, Zhang L, Polverino F, Guo F, Hao Y, Lao T, Xu S, Li L, Pham B, Owen CA, Zhou X. Hedgehog interacting protein (HHIP) represses airway remodeling and metabolic reprogramming in COPD-derived airway smooth muscle cells. Sci Rep 2021; 11:9074. [PMID: 33907231 PMCID: PMC8079715 DOI: 10.1038/s41598-021-88434-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 04/07/2021] [Indexed: 02/05/2023] Open
Abstract
Although HHIP locus has been consistently associated with the susceptibility to COPD including airway remodeling and emphysema in genome-wide association studies, the molecular mechanism underlying this genetic association remains incompletely understood. By utilizing Hhip+/- mice and primary human airway smooth muscle cells (ASMCs), here we aim to determine whether HHIP haploinsufficiency increases airway smooth muscle mass by reprogramming glucose metabolism, thus contributing to airway remodeling in COPD pathogenesis. The mRNA levels of HHIP were compared in normal and COPD-derived ASMCs. Mitochondrial oxygen consumption rate and lactate levels in the medium were measured in COPD-derived ASMCs with or without HHIP overexpression as readouts of glucose oxidative phosphorylation and aerobic glycolysis rates. The proliferation rate was measured in healthy and COPD-derived ASMCs treated with or without 2-DG. Smooth muscle mass around airways was measured by immunofluorescence staining for α-smooth muscle actin (α-SMA) in lung sections from Hhip+/- mice and their wild type littermates, Hhip+/+ mice. Airway remodeling was assessed in Hhip+/- and Hhip+/- mice exposed to 6 months of cigarette smoke. Our results show HHIP inhibited aerobic glycolysis and represses cell proliferation in COPD-derived ASMCs. Notably, knockdown of HHIP in normal ASMCs increased PKM2 activity. Importantly, Hhip+/- mice demonstrated increased airway remodeling and increased intensity of α-SMA staining around airways compared to Hhip+/+ mice. In conclusion, our findings suggest that HHIP represses aerobic glycolysis and ASMCs hyperplasia, which may contribute to the increased airway remodeling in Hhip+/- mice.
Collapse
Affiliation(s)
- Yan Li
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China.
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, China.
| | - Li Zhang
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Francesca Polverino
- Asthma and Airway Disease Research Center, University of Arizona, Medicine, Tucson, AZ, 85724, USA
| | - Feng Guo
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Yuan Hao
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Taotao Lao
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - Shuang Xu
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Lijia Li
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Betty Pham
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Caroline A Owen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
19
|
Liao YC, Lo SH. Tensins - emerging insights into their domain functions, biological roles and disease relevance. J Cell Sci 2021; 134:jcs254029. [PMID: 33597154 PMCID: PMC10660079 DOI: 10.1242/jcs.254029] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Tensins are a family of focal adhesion proteins consisting of four members in mammals (TNS1, TNS2, TNS3 and TNS4). Their multiple domains and activities contribute to the molecular linkage between the extracellular matrix and cytoskeletal networks, as well as mediating signal transduction pathways, leading to a variety of physiological processes, including cell proliferation, attachment, migration and mechanical sensing in a cell. Tensins are required for maintaining normal tissue structures and functions, especially in the kidney and heart, as well as in muscle regeneration, in animals. This Review discusses our current understanding of the domain functions and biological roles of tensins in cells and mice, as well as highlighting their relevance to human diseases.
Collapse
Affiliation(s)
- Yi-Chun Liao
- Department of Biochemical Science and Technology, National Taiwan University, Taipei 10617, Taiwan
| | - Su Hao Lo
- Department of Biochemistry and Molecular Medicine, University of California-Davis, Sacramento, CA 95817, USA
| |
Collapse
|
20
|
Duan J, Wang L, Shang L, Yang S, Wu H, Huang Y, Miao Y. miR-152/TNS1 axis inhibits non-small cell lung cancer progression through Akt/mTOR/RhoA pathway. Biosci Rep 2021; 41:BSR20201539. [PMID: 33269380 PMCID: PMC7785040 DOI: 10.1042/bsr20201539] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/08/2020] [Accepted: 06/08/2020] [Indexed: 12/30/2022] Open
Abstract
AIM The purpose of the present study was to explore the function and mechanism of tensin 1 (TNS1) in non-small cell lung cancer (NSCLC) progression. METHODS The expression of TNS1 in NSCLC cells and tissues was assessed by RT-PCR and Western blot. Besides, Kaplan-Meier survival analysis was recruited to explore the association between TNS1 and NSCLC. Cell growth was analyzed by MTT and flow cytometry assay, while cell metastasis was determined by wound healing and transwell assays. The targeting relationship between TNS1 and miR-152 was assessed by luciferase activity assays. And Western blot was employed to determine the expression of related proteins of Akt/mTOR/RhoA pathway. RESULTS TNS1 level was boosted in NSCLC cells and tissues, related to the prognosis of NSCLC patients. Furthermore, it was proved that TNS1 promoted the growth and metastasis of NSCLC cells via Akt/mTOR/RhoA pathway. And miR-152 targeted TNS1 to affect the progression of NSCLC. CONCLUSION miR-152/TNS1 axis inhibits the progression of NSCLC by Akt/mTOR/RhoA pathway.
Collapse
Affiliation(s)
- Jinjin Duan
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, P.R. China
| | - Li Wang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, P.R. China
| | - Liqun Shang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, P.R. China
| | - Shumei Yang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, P.R. China
| | - Hua Wu
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, P.R. China
| | - Yongcheng Huang
- Department of Pathology, Xi’an Central Hospital, Xi’an, Shaanxi 7100033, P.R. China
| | - Yi Miao
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, P.R. China
| |
Collapse
|
21
|
Sharma A, Kaur S, Sarkar M, Sarin BC, Changotra H. The AGE-RAGE Axis and RAGE Genetics in Chronic Obstructive Pulmonary Disease. Clin Rev Allergy Immunol 2020; 60:244-258. [PMID: 33170477 DOI: 10.1007/s12016-020-08815-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2020] [Indexed: 12/25/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a heterogeneous group of lung diseases limiting the airflow due to narrowing of airways, chronic bronchitis and emphysema that leads to difficulties in breathing. Chronic inflammation is another important characteristic of COPD which leads to immune cell infiltration and helps in the alveolar destruction. Pathology of COPD is driven by various environmental and genetic factors. COPD is mainly associated with the inhalation of toxic agents mainly the cigarette smoke. Receptor for advanced glycation end products (RAGE) has emerged as a pattern recognition receptor and is a multiligand receptor expressed moderately in various cells, tissues and highly in the lungs throughout life. RAGE recognizes various ligands produced by cigarette smoke and its role has been implicated in the pathogenesis of COPD. RAGE ligands have been reported to accumulate in the lungs of patients with COPD. RAGE is a membrane receptor but its truncated form i.e. soluble RAGE (sRAGE) mainly functions as a contender of RAGE and inhibits various RAGE dependent cell signalling. Among the various ligands of RAGE, advanced glycation end products (AGEs) are majorly linked with COPD. Accumulated AGE triggers downstream RAGE-AGE axis in COPD. Moreover, RAGE genetics has long been known to play a vital role in the pathology of various airway diseases including COPD and this gene contains an associated locus. A reliable biomarker is needed for the management of this disease. sRAGE has an inverse correlation with the RAGE showed its importance as a valuable marker in COPD. This review is focused on the role of RAGE, sRAGE, RAGE axis and RAGE genetics in COPD.
Collapse
Affiliation(s)
- Ambika Sharma
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173 234, India
| | - Sargeet Kaur
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173 234, India
| | - Malay Sarkar
- Department of Pulmonary Medicine, Indira Gandhi Medical College, Shimla, Himachal Pradesh, 171 001, India
| | - B C Sarin
- Department of Chest and TB, Sri Guru Ram Das Institute of Medical Sciences and Research, Vallah, Amritsar, 143 501, India
| | - Harish Changotra
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173 234, India.
| |
Collapse
|
22
|
Do-Umehara HC, Chen C, Zhang Q, Misharin AV, Abdala-Valencia H, Casalino-Matsuda SM, Reyfman PA, Anekalla KR, Gonzalez-Gonzalez FJ, Sala MA, Peng C, Wu P, Wong CCL, Kalhan R, Bharat A, Perlman H, Ridge KM, Sznajder JI, Sporn PHS, Chandel NS, Yu J, Fu X, Petrache I, Tuder R, Budinger GRS, Liu J. Epithelial cell-specific loss of function of Miz1 causes a spontaneous COPD-like phenotype and up-regulates Ace2 expression in mice. SCIENCE ADVANCES 2020; 6:eabb7238. [PMID: 32851183 PMCID: PMC7428331 DOI: 10.1126/sciadv.abb7238] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 07/02/2020] [Indexed: 05/19/2023]
Abstract
Cigarette smoking, the leading cause of chronic obstructive pulmonary disease (COPD), has been implicated as a risk factor for severe disease in patients infected with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here we show that mice with lung epithelial cell-specific loss of function of Miz1, which we identified as a negative regulator of nuclear factor κB (NF-κB) signaling, spontaneously develop progressive age-related changes resembling COPD. Furthermore, loss of Miz1 up-regulates the expression of Ace2, the receptor for SARS-CoV-2. Concomitant partial loss of NF-κB/RelA prevented the development of COPD-like phenotype in Miz1-deficient mice. Miz1 protein levels are reduced in the lungs from patients with COPD, and in the lungs of mice exposed to chronic cigarette smoke. Our data suggest that Miz1 down-regulation-induced sustained activation of NF-κB-dependent inflammation in the lung epithelium is sufficient to induce progressive lung and airway destruction that recapitulates features of COPD, with implications for COVID-19.
Collapse
Affiliation(s)
- Hanh Chi Do-Umehara
- Department of Surgery, College of Medicine and University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Cong Chen
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Qiao Zhang
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Alexander V. Misharin
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Hiam Abdala-Valencia
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - S. Marina Casalino-Matsuda
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Paul A. Reyfman
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kishore R. Anekalla
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Francisco J. Gonzalez-Gonzalez
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Marc A. Sala
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Chao Peng
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, SARI, CAS, Shanghai 201210, China
| | - Ping Wu
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, SARI, CAS, Shanghai 201210, China
| | | | - Ravi Kalhan
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ankit Bharat
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Division of Thoracic Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Harris Perlman
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Karen M. Ridge
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jacob I. Sznajder
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Peter H. S. Sporn
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| | - Navdeep S. Chandel
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jindan Yu
- Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Xiangdong Fu
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093-0651, USA; Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA 92093-0651, USA
| | - Irina Petrache
- National Jewish Health, 1400 Jackson Street, Molly Blank Building, J203, Denver, CO 80206, USA
- University of Colorado at Denver Health Sciences Center, Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, Denver, CO 80206, USA
| | - Rubin Tuder
- University of Colorado at Denver Health Sciences Center, Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, Denver, CO 80206, USA
| | - G. R. Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| | - Jing Liu
- Department of Surgery, College of Medicine and University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
23
|
De Cunto G, Cavarra E, Bartalesi B, Lucattelli M, Lungarella G. Innate Immunity and Cell Surface Receptors in the Pathogenesis of COPD: Insights from Mouse Smoking Models. Int J Chron Obstruct Pulmon Dis 2020; 15:1143-1154. [PMID: 32547002 PMCID: PMC7246326 DOI: 10.2147/copd.s246219] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/03/2020] [Indexed: 12/23/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is mainly associated with smoking habit. Inflammation is the major initiating process whereby neutrophils and monocytes are attracted into the lung microenvironment by external stimuli present in tobacco leaves and in cigarette smoke, which promote chemotaxis, adhesion, phagocytosis, release of superoxide anions and enzyme granule contents. A minority of smokers develops COPD and different molecular factors, which contribute to the onset of the disease, have been put forward. After many years of research, the pathogenesis of COPD is still an object of debate. In vivo models of cigarette smoke-induced COPD may help to unravel cellular and molecular mechanisms underlying the pathogenesis of COPD. The mouse represents the most favored animal choice with regard to the study of immune mechanisms due to its genetic and physiological similarities to humans, the availability of a large variability of inbred strains, the presence in the species of several genetic disorders analogous to those in man, and finally on the possibility to create models “made-to-measure” by genetic manipulation. The review outlines the different response of mouse strains to cigarette smoke used in COPD studies while retaining a strong focus on their relatability to human patients. These studies reveal the importance of innate immunity and cell surface receptors in the pathogenesis of pulmonary injury induced by cigarette smoking. They further advance the way in which we use wild type or genetically manipulated strains to improve our overall understanding of a multifaceted disease such as COPD. The structural and functional features, which have been found in the different strains of mice after chronic exposure to cigarette smoke, can be used in preclinical studies to develop effective new therapeutic agents for the different phenotypes in human COPD.
Collapse
Affiliation(s)
- Giovanna De Cunto
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Eleonora Cavarra
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Barbara Bartalesi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Monica Lucattelli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Giuseppe Lungarella
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
24
|
Wilson AC, Kumar PL, Lee S, Parker MM, Arora I, Morrow JD, Wouters EFM, Casaburi R, Rennard SI, Lomas DA, Agusti A, Tal-Singer R, Dransfield MT, Wells JM, Bhatt SP, Washko G, Thannickal VJ, Tiwari HK, Hersh CP, Castaldi PJ, Silverman EK, McDonald MLN. Heme metabolism genes Downregulated in COPD Cachexia. Respir Res 2020; 21:100. [PMID: 32354332 PMCID: PMC7193359 DOI: 10.1186/s12931-020-01336-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/11/2020] [Indexed: 11/27/2022] Open
Abstract
INTRODUCTION Cachexia contributes to increased mortality and reduced quality of life in Chronic Obstructive Pulmonary Disease (COPD) and may be associated with underlying gene expression changes. Our goal was to identify differential gene expression signatures associated with COPD cachexia in current and former smokers. METHODS We analyzed whole-blood gene expression data from participants with COPD in a discovery cohort (COPDGene, N = 400) and assessed replication (ECLIPSE, N = 114). To approximate the consensus definition using available criteria, cachexia was defined as weight-loss > 5% in the past 12 months or low body mass index (BMI) (< 20 kg/m2) and 1/3 criteria: decreased muscle strength (six-minute walk distance < 350 m), anemia (hemoglobin < 12 g/dl), and low fat-free mass index (FFMI) (< 15 kg/m2 among women and < 17 kg/m2 among men) in COPDGene. In ECLIPSE, cachexia was defined as weight-loss > 5% in the past 12 months or low BMI and 3/5 criteria: decreased muscle strength, anorexia, abnormal biochemistry (anemia or high c-reactive protein (> 5 mg/l)), fatigue, and low FFMI. Differential gene expression was assessed between cachectic and non-cachectic subjects, adjusting for age, sex, white blood cell counts, and technical covariates. Gene set enrichment analysis was performed using MSigDB. RESULTS The prevalence of COPD cachexia was 13.7% in COPDGene and 7.9% in ECLIPSE. Fourteen genes were differentially downregulated in cachectic versus non-cachectic COPD patients in COPDGene (FDR < 0.05) and ECLIPSE (FDR < 0.05). DISCUSSION Several replicated genes regulating heme metabolism were downregulated among participants with COPD cachexia. Impaired heme biosynthesis may contribute to cachexia development through free-iron buildup and oxidative tissue damage.
Collapse
Affiliation(s)
- Ava C Wilson
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Preeti L Kumar
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sool Lee
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Margaret M Parker
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Itika Arora
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jarrett D Morrow
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Emiel F M Wouters
- Centre of expertise for chronic organ failure, Horn, the Netherlands
| | - Richard Casaburi
- Rehabilitation Clinical Trials Center, Los Angeles Biomedical Research Institute at Harbor Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Stephen I Rennard
- Department of Medicine, Nebraska Medical Center, Omaha, NE, USA
- BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - David A Lomas
- UCL Respiratory, Division of Medicine, University College London, London, UK
| | - Alvar Agusti
- Fundació Investigació Sanitària Illes Balears (FISIB), Ciber Enfermedades Respiratorias (CIBERES), Barcelona, Catalunya, Spain
- Thorax Institute, Hospital Clinic, IDIBAPS, University of Barcelona, Barcelona, Spain
| | | | - Mark T Dransfield
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J Michael Wells
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Surya P Bhatt
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - George Washko
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Victor J Thannickal
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hemant K Tiwari
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Peter J Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Merry-Lynn N McDonald
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA.
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
25
|
Genetic profiling for disease stratification in chronic obstructive pulmonary disease and asthma. Curr Opin Pulm Med 2020; 25:317-322. [PMID: 30762612 DOI: 10.1097/mcp.0000000000000568] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW In asthma and chronic obstructive pulmonary disease (COPD), the movement towards genetic profiling with a push towards 'personalized medicine' has been hindered by complex environment--gene interactions and lack of tools to identify clear causal genetic traits. In this review, we will discuss the need for genetic profiling in asthma and COPD, what methods are currently used in the clinics and the recent finding using new sequencing methods. RECENT FINDINGS Over the past 10-15 years, genome-wide association studies analysis of common variants has provide little in the way of new genetic profiling markers for asthma and COPD. Whole exome/genome sequencing has provided a new method to identify lowly abundant alleles, which might have a much higher impact. Although, low population numbers due to high costs has hindered early studies, recent studies have reached genome wide significance. SUMMARY The use of genetic profiling of COPD in the clinic is current limited to the identification of Alpha-1 antitrypsin deficiency, while being absent in asthma. Advances in sequencing technology provide new avenues to identify disease causes or therapy response altering variants that in the short-term will allow for the development of screening procedures for disease to identify patients at risk of developing asthma or COPD.
Collapse
|
26
|
McGivney BA, Han H, Corduff LR, Katz LM, Tozaki T, MacHugh DE, Hill EW. Genomic inbreeding trends, influential sire lines and selection in the global Thoroughbred horse population. Sci Rep 2020; 10:466. [PMID: 31949252 PMCID: PMC6965197 DOI: 10.1038/s41598-019-57389-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023] Open
Abstract
The Thoroughbred horse is a highly valued domestic animal population under strong selection for athletic phenotypes. Here we present a high resolution genomics-based analysis of inbreeding in the population that may form the basis for evidence-based discussion amid concerns in the breeding industry over the increasing use of small numbers of popular sire lines, which may accelerate a loss of genetic diversity. In the most comprehensive globally representative sample of Thoroughbreds to-date (n = 10,118), including prominent stallions (n = 305) from the major bloodstock regions of the world, we show using pan-genomic SNP genotypes that there has been a highly significant decline in global genetic diversity during the last five decades (FIS R2 = 0.942, P = 2.19 × 10-13; FROH R2 = 0.88, P = 1.81 × 10-10) that has likely been influenced by the use of popular sire lines. Estimates of effective population size in the global and regional populations indicate that there is some level of regional variation that may be exploited to improve global genetic diversity. Inbreeding is often a consequence of selection, which in managed animal populations tends to be driven by preferences for cultural, aesthetic or economically advantageous phenotypes. Using a composite selection signals approach, we show that centuries of selection for favourable athletic traits among Thoroughbreds acts on genes with functions in behaviour, musculoskeletal conformation and metabolism. As well as classical selective sweeps at core loci, polygenic adaptation for functional modalities in cardiovascular signalling, organismal growth and development, cellular stress and injury, metabolic pathways and neurotransmitters and other nervous system signalling has shaped the Thoroughbred athletic phenotype. Our results demonstrate that genomics-based approaches to identify genetic outcrosses will add valuable objectivity to augment traditional methods of stallion selection and that genomics-based methods will be beneficial to actively monitor the population to address the marked inbreeding trend.
Collapse
Affiliation(s)
| | - Haige Han
- Plusvital Ltd, The Highline, Dun Laoghaire Business Park, Dublin, Ireland
- UCD School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Leanne R Corduff
- Plusvital Ltd, The Highline, Dun Laoghaire Business Park, Dublin, Ireland
| | - Lisa M Katz
- UCD School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Teruaki Tozaki
- Genetic Analysis Department, Laboratory of Racing Chemistry, Utsunomiya, Tochigi, Japan
| | - David E MacHugh
- UCD School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Emmeline W Hill
- Plusvital Ltd, The Highline, Dun Laoghaire Business Park, Dublin, Ireland.
- UCD School of Agriculture and Food Science, University College Dublin, Dublin, Ireland.
| |
Collapse
|
27
|
Tensin1 expression and function in chronic obstructive pulmonary disease. Sci Rep 2019; 9:18942. [PMID: 31831813 PMCID: PMC6908681 DOI: 10.1038/s41598-019-55405-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 11/25/2019] [Indexed: 11/18/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) constitutes a major cause of morbidity and mortality. Genome wide association studies have shown significant associations between airflow obstruction or COPD with a non-synonymous SNP in the TNS1 gene, which encodes tensin1. However, the expression, cellular distribution and function of tensin1 in human airway tissue and cells are unknown. We therefore examined these characteristics in tissue and cells from controls and people with COPD or asthma. Airway tissue was immunostained for tensin1. Tensin1 expression in cultured human airway smooth muscle cells (HASMCs) was evaluated using qRT-PCR, western blotting and immunofluorescent staining. siRNAs were used to downregulate tensin1 expression. Tensin1 expression was increased in the airway smooth muscle and lamina propria in COPD tissue, but not asthma, when compared to controls. Tensin1 was expressed in HASMCs and upregulated by TGFβ1. TGFβ1 and fibronectin increased the localisation of tensin1 to fibrillar adhesions. Tensin1 and α-smooth muscle actin (αSMA) were strongly co-localised, and tensin1 depletion in HASMCs attenuated both αSMA expression and contraction of collagen gels. In summary, tensin1 expression is increased in COPD airways, and may promote airway obstruction by enhancing the expression of contractile proteins and their localisation to stress fibres in HASMCs.
Collapse
|
28
|
Wu ZY, Chiu CL, Lo E, Lee YRJ, Yamada S, Lo SH. Hyperactivity of Mek in TNS1 knockouts leads to potential treatments for cystic kidney diseases. Cell Death Dis 2019; 10:871. [PMID: 31740667 PMCID: PMC6861224 DOI: 10.1038/s41419-019-2119-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/28/2019] [Accepted: 11/05/2019] [Indexed: 12/29/2022]
Abstract
Cystic kidney disease is the progressive development of multiple fluid-filled cysts that may severely compromise kidney functions and lead to renal failure. TNS1 (tensin-1) knockout mice develop cystic kidneys and die from renal failure. Here, we have established TNS1-knockout MDCK cells and applied 3D culture system to investigate the mechanism leading to cyst formation. Unlike wild-type MDCK cells, which form cysts with a single lumen, TNS1-knockout cysts contain multiple lumens and upregulated Mek/Erk activities. The multiple lumen phenotype and Mek/Erk hyperactivities are rescued by re-expression of wild-type TNS1 but not the TNS1 mutant lacking a fragment essential for its cell–cell junction localization. Furthermore, Mek inhibitor treatments restore the multiple lumens back to single lumen cysts. Mek/Erk hyperactivities are also detected in TNS1-knockout mouse kidneys. Treatment with the Mek inhibitor trametinib significantly reduces the levels of interstitial infiltrates, fibrosis and dilated tubules in TNS1-knockout kidneys. These studies establish a critical role of subcellular localization of TNS1 in suppressing Mek/Erk signaling and maintaining lumenogenesis, and provide potential therapeutic strategies by targeting the Mek/Erk pathway for cystic kidney diseases.
Collapse
Affiliation(s)
- Zong-Ye Wu
- Department of Biochemistry and Molecular Medicine, University of California-Davis, Sacramento, CA, 95817, USA
| | - Chun-Lung Chiu
- Department of Biochemistry and Molecular Medicine, University of California-Davis, Sacramento, CA, 95817, USA
| | - Ethan Lo
- Department of Biochemistry and Molecular Medicine, University of California-Davis, Sacramento, CA, 95817, USA
| | - Yuh-Ru Julie Lee
- Department of Plant Biology, University of California-Davis, Davis, CA, 95616, USA
| | - Soichiro Yamada
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, 95616, USA
| | - Su Hao Lo
- Department of Biochemistry and Molecular Medicine, University of California-Davis, Sacramento, CA, 95817, USA.
| |
Collapse
|
29
|
Liu B, Henry AP, Azimi S, Miller S, Lee FK, Lee JC, Probert K, Kotlikoff MI, Sayers I, Hall IP. Exposure to lipopolysaccharide (LPS) reduces contractile response of small airways from GSTCD-/- mice. PLoS One 2019; 14:e0221899. [PMID: 31513609 PMCID: PMC6742219 DOI: 10.1371/journal.pone.0221899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/17/2019] [Indexed: 11/25/2022] Open
Abstract
Introduction Genome-Wide Association Studies suggest glutathione S transferase C terminal domain (GSTCD) may play a role in development of Chronic Obstructive Pulmonary Disease. We aimed to define the potential role of GSTCD in airway inflammation and contraction using precision cut lung slice (PCLS) from wild-type (GSTCD+/+) and GSTCD knockout mice (GSTCD-/-). Methods PCLS from age and gender matched GSTCD+/+ and GSTCD-/- mice were prepared using a microtome. Contraction was studied after applying either a single dose of Methacholine (Mch) (1 μM) or different doses of Mch (0.001 to 100 μM). Each slice was then treated with lipopolysaccharide (LPS) or vehicle (PBS) for 24 hours. PCLS contraction in the same airway was repeated before and after stimulation. Levels of TNFα production was also measured. Results There were no differences in contraction of PCLS from GSTCD+/+ and GSTCD-/- mice in response to Mch (EC50 of GSTCD+/+ vs GSTCD-/- animals: 100.0±20.7 vs 107.7±24.5 nM, p = 0.855, n = 6 animals/group). However, after LPS treatment, there was a 31.6% reduction in contraction in the GSTCD-/- group (p = 0.023, n = 6 animals). There was no significant difference between PBS and LPS treatment groups in GSTCD+/+ animals. We observed a significant increase in TNFα production induced by LPS in GSTCD-/- lung slices compared to the GSTCD+/+ LPS treated slices. Conclusion GSTCD knockout mice showed an increased responsiveness to LPS (as determined by TNFα production) that was accompanied by a reduced contraction of small airways in PCLS. These data highlight an unrecognised potential function of GSTCD in mediating inflammatory signals that affect airway responses.
Collapse
Affiliation(s)
- Bo Liu
- Division of Respiratory Medicine, & National Institute for Health Medicine, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, England, United Kingdom
| | - Amanda P. Henry
- Division of Respiratory Medicine, & National Institute for Health Medicine, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, England, United Kingdom
- * E-mail:
| | - Sheyda Azimi
- Division of Respiratory Medicine, & National Institute for Health Medicine, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, England, United Kingdom
| | - Suzanne Miller
- Division of Respiratory Medicine, & National Institute for Health Medicine, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, England, United Kingdom
| | - Frank K. Lee
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Jane C. Lee
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Kelly Probert
- Division of Respiratory Medicine, & National Institute for Health Medicine, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, England, United Kingdom
| | - Michael I. Kotlikoff
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Ian Sayers
- Division of Respiratory Medicine, & National Institute for Health Medicine, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, England, United Kingdom
| | - Ian P. Hall
- Division of Respiratory Medicine, & National Institute for Health Medicine, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, England, United Kingdom
| |
Collapse
|
30
|
Ranjan A, Singh A, Walia GK, Sachdeva MP, Gupta V. Genetic underpinnings of lung function and COPD. J Genet 2019; 98:76. [PMID: 31544798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Spirometry based measurement of lung function is a global initiative for chronic obstructive lung disease (GOLD) standard to diagnose chronic obstructive pulmonary disease (COPD), one of the leading causes of mortality worldwide. The environmental and behavioural risk factors for COPD includes tobacco smoking, air pollutants and biomass fuel exposure, which can induce one or more abnormal lung function patterns. While smoking remains the primary risk factor, only 15-20% smokers develop COPD, indicating that the genetic factors are also likely to play a role. According to the study of Global Burden of Disease 2015, ∼174 million people across the world have COPD. From a comprehensive literature search conducted using the 'PubMed' and 'GWAS Catalogue' databases, and reviewing the literature available, only a limited number of studies were identified which had attempted to investigate the genetics of COPD and lung volumes, implying a huge research gap. With the advent of genomewide association studies several genetic variants linked to lung function and COPD, like HHIP, HTR4, ADAM19 and GSTCD etc., have been found and validated in different population groups, suggesting their potential role in determining lung volume and risk for COPD. This article aims at reviewing the present knowledge of the genetics of lung function and COPD.
Collapse
Affiliation(s)
- Astha Ranjan
- Department of Anthropology, University of Delhi, Delhi 110 007, India.
| | | | | | | | | |
Collapse
|
31
|
Ortega VE. Picking the Right Fruit: Intersecting Chronic Obstructive Pulmonary Disease Genome-Wide Association Study Discoveries with Epigenetics. Am J Respir Crit Care Med 2019; 197:1237-1239. [PMID: 29425467 DOI: 10.1164/rccm.201801-0084ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Victor E Ortega
- 1 Department of Internal Medicine and.,2 Center for Precision Medicine Wake Forest School of Medicine Winston-Salem, North Carolina
| |
Collapse
|
32
|
Russell REK, Bafadhel M. What will Happen in the World of COPD 2030? Turk Thorac J 2019; 20:253-257. [PMID: 31390331 DOI: 10.5152/turkthoracj.2019.190307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/07/2019] [Indexed: 11/22/2022]
Abstract
2030 may seem to be a long way into the future, but it's not. We live in a world of relentless rapid change in modern medicine and our approach to our patients with chronic diseases such as chronic obstructive pulmonary disease (COPD) will need to evolve at speed. This review looks at what may occur in society and medicine that will influence the way we manage COPD. The article is the opinion of the authors and is based upon current research at the cutting edge of management with a degree of gazing into a dimly lit crystal ball. COPD is a current epidemic, and this is likely to continue. Legislative efforts to reduce smoking will continue and hopefully accelerate, but this will not be globally accepted or successful. Technological advances will occur that will lead to miniaturization and the rise of near patient testing. This itself will enable a personalised approach to management with the ability to measure rapidly biomarkers which will direct therapy. The blood eosinophil is the most promising of these and is available now. New developments in the identification of disease clusters and phenotypes will also enhance a more personalised approach. Through both these epidemiological studies and also new developments in the understanding of basic mechanisms it is hoped that in the future patients will be given treatments that may fundamentally change the prognosis of COPD. Small molecule and antibody directed therapies may, if given early enough, stop and even possibly reverse the effects of COPD on cells and organs. Of course, the most important step which is achievable now is to ban all tobacco-based products from the world.
Collapse
Affiliation(s)
- Richard E K Russell
- Respiratory Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.,Lymington New Forest Hospital, Lymington, Hampshire, United Kingdom
| | - Mona Bafadhel
- Respiratory Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
33
|
Henry AP, Probert K, Stewart CE, Thakker D, Bhaker S, Azimi S, Hall IP, Sayers I. Defining a role for lung function associated gene GSTCD in cell homeostasis. Respir Res 2019; 20:172. [PMID: 31370853 PMCID: PMC6676530 DOI: 10.1186/s12931-019-1146-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 07/25/2019] [Indexed: 12/11/2022] Open
Abstract
Genome wide association (GWA) studies have reproducibly identified signals on chromosome 4q24 associated with lung function and COPD. GSTCD (Glutathione S-transferase C-terminal domain containing) represents a candidate causal gene in this locus, however little is currently known about the function of this protein. We set out to further our understanding of the role of GSTCD in cell functions and homeostasis using multiple molecular and cellular approaches in airway relevant cells. Recombinant expression of human GSTCD in conjunction with a GST activity assay did not identify any enzymatic activity for two GSTCD isoforms questioning the assignment of this protein to this family of enzymes. Protein structure analyses identified a potential methyltransferase domain contained within GSTCD, with these enzymes linked to cell viability and apoptosis. Targeted knockdown (siRNA) of GSTCD in bronchial epithelial cells identified a role for GSTCD in cell viability as proliferation rates were not altered. To provide greater insight we completed transcriptomic analyses on cells with GSTCD expression knocked down and identified several differentially expressed genes including those implicated in airway biology; fibrosis e.g. TGFBR1 and inflammation e.g. IL6R. Pathway based transcriptomic analyses identified an over-representation of genes related to adipogenesis which may suggest additional functions for GSTCD. These findings identify potential additional functions for GSTCD in the context of airway biology beyond the hypothesised GST activity and warrant further investigation.
Collapse
Affiliation(s)
- Amanda P Henry
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK.
| | - Kelly Probert
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Ceri E Stewart
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Dhruma Thakker
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Sangita Bhaker
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Sheyda Azimi
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Ian P Hall
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Ian Sayers
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| |
Collapse
|
34
|
Wang C, Cassandras M, Peng T. The Role of Hedgehog Signaling in Adult Lung Regeneration and Maintenance. J Dev Biol 2019; 7:jdb7030014. [PMID: 31323955 PMCID: PMC6787692 DOI: 10.3390/jdb7030014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/29/2019] [Accepted: 07/03/2019] [Indexed: 12/13/2022] Open
Abstract
As a secreted morphogen, Sonic Hedgehog (SHH) determines differential cell fates, behaviors, and functions by forming a gradient of Hedgehog (Hh) activation along an axis of Hh-receptive cells during development. Despite clearly delineated roles for Hh during organ morphogenesis, whether Hh continues to regulate cell fate and behavior in the same fashion in adult organs is less understood. Adult organs, particularly barrier organs interfacing with the ambient environment, are exposed to insults that require renewal of cellular populations to maintain structural integrity. Understanding key aspects of Hh’s ability to generate an organ could translate into conceptual understanding of Hh’s ability to maintain organ homeostasis and stimulate regeneration. In this review, we will summarize the current knowledge about Hh signaling in regulating adult lung regeneration and maintenance, and discuss how alteration of Hh signaling contributes to adult lung diseases.
Collapse
Affiliation(s)
- Chaoqun Wang
- Department of Medicine, Cardiovascular Research Institute, UCSF, San Francisco, CA 94143, USA
| | - Monica Cassandras
- Department of Medicine, Cardiovascular Research Institute, UCSF, San Francisco, CA 94143, USA
| | - Tien Peng
- Department of Medicine, Cardiovascular Research Institute, UCSF, San Francisco, CA 94143, USA.
| |
Collapse
|
35
|
Analysis of genetically driven alternative splicing identifies FBXO38 as a novel COPD susceptibility gene. PLoS Genet 2019; 15:e1008229. [PMID: 31269066 PMCID: PMC6634423 DOI: 10.1371/journal.pgen.1008229] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 07/16/2019] [Accepted: 06/04/2019] [Indexed: 11/19/2022] Open
Abstract
While many disease-associated single nucleotide polymorphisms (SNPs) are associated with gene expression (expression quantitative trait loci, eQTLs), a large proportion of complex disease genome-wide association study (GWAS) variants are of unknown function. Some of these SNPs may contribute to disease by regulating gene splicing. Here, we investigate whether SNPs that are associated with alternative splicing (splice QTL or sQTL) can identify novel functions for existing GWAS variants or suggest new associated variants in chronic obstructive pulmonary disease (COPD). RNA sequencing was performed on whole blood from 376 subjects from the COPDGene Study. Using linear models, we identified 561,060 unique sQTL SNPs associated with 30,333 splice sites corresponding to 6,419 unique genes. Similarly, 708,928 unique eQTL SNPs involving 15,913 genes were detected at 10% FDR. While there is overlap between sQTLs and eQTLs, 55.3% of sQTLs are not eQTLs. Co-localization analysis revealed that 7 out of 21 loci associated with COPD (p<1x10-6) in a published GWAS have at least one shared causal variant between the GWAS and sQTL studies. Among the genes identified to have splice sites associated with top GWAS SNPs was FBXO38, in which a novel exon was discovered to be protective against COPD. Importantly, the sQTL in this locus was validated by qPCR in both blood and lung tissue, demonstrating that splice variants relevant to lung tissue can be identified in blood. Other identified genes included CDK11A and SULT1A2. Overall, these data indicate that analysis of alternative splicing can provide novel insights into disease mechanisms. In particular, we demonstrated that SNPs in a known COPD GWAS locus on chromosome 5q32 influence alternative splicing in the gene FBXO38.
Collapse
|
36
|
John C, Reeve NF, Free RC, Williams AT, Ntalla I, Farmaki AE, Bethea J, Barton LM, Shrine N, Batini C, Packer R, Terry S, Hargadon B, Wang Q, Melbourne CA, Adams EL, Bee CE, Harrington K, Miola J, Brunskill NJ, Brightling CE, Barwell J, Wallace SE, Hsu R, Shepherd DJ, Hollox EJ, Wain LV, Tobin MD. Cohort Profile: Extended Cohort for E-health, Environment and DNA (EXCEED). Int J Epidemiol 2019; 48:678-679j. [PMID: 31062032 PMCID: PMC6659362 DOI: 10.1093/ije/dyz073] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2019] [Indexed: 12/22/2022] Open
Affiliation(s)
- Catherine John
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Nicola F Reeve
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Robert C Free
- NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | | | - Ioanna Ntalla
- Department of Health Sciences, University of Leicester, Leicester, UK
- Department of Clinical Pharmacology, William Harvey Research Institute, Barts & The London Medical School, Queen Mary University of London, Charterhouse Square, London, UK
| | - Aliki-Eleni Farmaki
- Department of Health Sciences, University of Leicester, Leicester, UK
- Department of Population Science and Experimental Medicine, Institute of Cardiovascular Science, University College London, London, UK
| | - Jane Bethea
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Linda M Barton
- Department of Haematology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Nick Shrine
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Chiara Batini
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Richard Packer
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Sarah Terry
- NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Beverley Hargadon
- NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Qingning Wang
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Carl A Melbourne
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Emma L Adams
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Catherine E Bee
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Kyla Harrington
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - José Miola
- Leicester Law School, University of Leicester, Leicester, UK
| | - Nigel J Brunskill
- Department of Cardiovascular Sciences University of Leicester, Leicester, UK
| | - Christopher E Brightling
- NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Julian Barwell
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Susan E Wallace
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Ron Hsu
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - David J Shepherd
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Edward J Hollox
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Louise V Wain
- Department of Health Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Martin D Tobin
- Department of Health Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
37
|
Bermingham ML, Walker RM, Marioni RE, Morris SW, Rawlik K, Zeng Y, Campbell A, Redmond P, Whalley HC, Adams MJ, Hayward C, Deary IJ, Porteous DJ, McIntosh AM, Evans KL. Identification of novel differentially methylated sites with potential as clinical predictors of impaired respiratory function and COPD. EBioMedicine 2019; 43:576-586. [PMID: 30935889 PMCID: PMC6557748 DOI: 10.1016/j.ebiom.2019.03.072] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/25/2019] [Accepted: 03/25/2019] [Indexed: 12/22/2022] Open
Abstract
Background The causes of poor respiratory function and COPD are incompletely understood, but it is clear that genes and the environment play a role. As DNA methylation is under both genetic and environmental control, we hypothesised that investigation of differential methylation associated with these phenotypes would permit mechanistic insights, and improve prediction of COPD. We investigated genome-wide differential DNA methylation patterns using the recently released 850 K Illumina EPIC array. This is the largest single population, whole-genome epigenetic study to date. Methods Epigenome-wide association studies (EWASs) of respiratory function and COPD were performed in peripheral blood samples from the Generation Scotland: Scottish Family Health Study (GS:SFHS) cohort (n = 3781; 274 COPD cases and 2919 controls). In independent COPD incidence data (n = 149), significantly differentially methylated sites (DMSs; p < 3.6 × 10−8) were evaluated for their added predictive power when added to a model including clinical variables, age, sex, height and smoking history using receiver operating characteristic analysis. The Lothian Birth Cohort 1936 (LBC1936) was used to replicate association (n = 895) and prediction (n = 178) results. Findings We identified 28 respiratory function and/or COPD associated DMSs, which mapped to genes involved in alternative splicing, JAK-STAT signalling, and axon guidance. In prediction analyses, we observed significant improvement in discrimination between COPD cases and controls (p < .05) in independent GS:SFHS (p = .016) and LBC1936 (p = .010) datasets by adding DMSs to a clinical model. Interpretation Identification of novel DMSs has provided insight into the molecular mechanisms regulating respiratory function and aided prediction of COPD risk. Further studies are needed to assess the causality and clinical utility of identified associations. Fund Wellcome Trust Strategic Award 10436/Z/14/Z.
Collapse
Affiliation(s)
- Mairead L Bermingham
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK.
| | - Rosie M Walker
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK; Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Stewart W Morris
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Konrad Rawlik
- Division of Genetics and Genomics, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, UK
| | - Yanni Zeng
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK; Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Paul Redmond
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
| | - Heather C Whalley
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
| | - Mark J Adams
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK; Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK; Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Andrew M McIntosh
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK; Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK; Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
| | - Kathryn L Evans
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK; Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
38
|
Pathological Implications of Receptor for Advanced Glycation End-Product ( AGER) Gene Polymorphism. DISEASE MARKERS 2019; 2019:2067353. [PMID: 30863465 PMCID: PMC6378764 DOI: 10.1155/2019/2067353] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/01/2019] [Accepted: 01/15/2019] [Indexed: 12/12/2022]
Abstract
The receptor for advanced glycation end-products (RAGE) is a cell surface transmembrane multiligand receptor, encoded by the AGER gene. RAGE presents many transcripts, is expressed mainly in the lung, and involves multiple pathways (such as NFκB, Akt, p38, and MAP kinases) that initiate and perpetuate an unfavorable proinflammatory state. Due to these numerous functional activities, RAGE is implicated in multiple diseases. AGER is a highly polymorphic gene, with polymorphisms or SNP (single-nucleotide polymorphism) that could be responsible or co-responsible for disease development. This review was designed to shed light on the pathological implications of AGER polymorphisms. Five polymorphisms are described: rs2070600, rs1800624, rs1800625, rs184003, and a 63 bp deletion. The rs2070600 SNP may be associated with the development of human autoimmune disease, diabetes complications, cancer, and lung diseases such as chronic obstructive pulmonary disease and acute respiratory distress syndrome. The rs1800624 SNP involves AGER gene regulation and may be related to reduced risk of heart disease, cancer, Crohn's disease, and type 1 diabetes complications. The rs1800625 SNP may be associated with the development of diabetic retinopathy, cancer, and lupus but may be protective against cardiovascular risk. The rs184003 SNP seems related to coronary artery disease, breast cancer, and diabetes. The 63 bp deletion may be associated with reduced survival from heart diseases during diabetic nephropathy. Here, these potential associations between AGER polymorphisms and the development of diseases are discussed, as there have been conflicting findings on the pathological impact of AGER SNPs in the literature. These contradictory results might be explained by distinct AGER SNP frequencies depending on ethnicity.
Collapse
|
39
|
Sanders KA, Delker DA, Huecksteadt T, Beck E, Wuren T, Chen Y, Zhang Y, Hazel MW, Hoidal JR. RAGE is a Critical Mediator of Pulmonary Oxidative Stress, Alveolar Macrophage Activation and Emphysema in Response to Cigarette Smoke. Sci Rep 2019; 9:231. [PMID: 30659203 PMCID: PMC6338799 DOI: 10.1038/s41598-018-36163-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 11/12/2018] [Indexed: 02/07/2023] Open
Abstract
The receptor for advanced glycation end products (RAGE), a cell membrane receptor, recognizes ligands produced by cigarette smoke (CS) and has been implicated in the pathogenesis of COPD. We demonstrate that deletion or pharmacologic inhibition of RAGE prevents development of CS-induced emphysema. To identify molecular pathways by which RAGE mediates smoking related lung injury we performed unbiased gene expression profiling of alveolar macrophages (AM) obtained from RAGE null and C57BL/6 WT mice exposed to CS for one week or four months. Pathway analysis of RNA expression identified a number of genes integral to the pathogenesis of COPD impacted by the absence of RAGE. Altered expression of antioxidant response genes and lung protein 4-HNE immunostaining suggest attenuated oxidative stress in the RAGE null mice despite comparable CS exposure and lung leukocyte burden as the WT mice. Reduced endoplasmic reticulum stress in response to CS exposure also was observed in the AM from RAGE null mice. These findings provide novel insight into the sources of oxidative stress, macrophage activation, and the pathogenesis of lung disease due to CS exposure.
Collapse
Affiliation(s)
- Karl A Sanders
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Don A Delker
- Division of Gastroenterology, Hepatology, and Nutrition, University of Utah, Salt Lake City, Utah, USA
| | - Tom Huecksteadt
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Emily Beck
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Tanna Wuren
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| | - Yuntian Chen
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Mark W Hazel
- Division of Gastroenterology, Hepatology, and Nutrition, University of Utah, Salt Lake City, Utah, USA
| | - John R Hoidal
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, USA.
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah, USA.
| |
Collapse
|
40
|
Hall R, Hall IP, Sayers I. Genetic risk factors for the development of pulmonary disease identified by genome-wide association. Respirology 2018; 24:204-214. [PMID: 30421854 DOI: 10.1111/resp.13436] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/31/2018] [Accepted: 09/20/2018] [Indexed: 12/17/2022]
Abstract
Chronic respiratory diseases are a major cause of morbidity and mortality. Asthma and chronic obstructive pulmonary disease (COPD) combined affect over 500 million people worldwide. While environmental factors are important in disease progression, asthma and COPD have long been known to be heritable with genetic components playing an important role in the risk of developing disease. Identification of genetic variation contributing to disease progression is important for a number of reasons including identification of risk alleles, understanding underlying disease mechanisms and development of novel therapies. Genome-wide association studies (GWAS) have been successful in identifying many loci associated with lung function, COPD and asthma. In recent years, meta-analyses and improved imputation have facilitated the growth of GWAS in terms of numbers of subjects and the number of single nucleotide polymorphisms (SNP) that can be interrogated. As a consequence, there has been a significant increase in the number of signals associated with asthma, COPD and lung function. SNP that have shown association with lung function reassuringly show a significant overlap with SNP associated with COPD giving a glimpse at pathways that may be involved in COPD mechanisms including genes in, for example, developmental pathways. In asthma, association signals are often in or near genes involved in both adaptive and innate immune response pathways, epithelial cell homeostasis and airway structural changes. The challenges now are translating these genetic signals into a new understanding of lung biology, understanding how variants impact health and disease and how they may provide opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Robert Hall
- Division of Respiratory Medicine, NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Ian P Hall
- Division of Respiratory Medicine, NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Ian Sayers
- Division of Respiratory Medicine, NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| |
Collapse
|
41
|
Prokopenko D, Sakornsakolpat P, Fier HL, Qiao D, Parker MM, McDonald MLN, Manichaikul A, Rich SS, Barr RG, Williams CJ, Brantly ML, Lange C, Beaty TH, Crapo JD, Silverman EK, on behalf of COPDGene Study Investigators* and NHLBI TOPMed Investigators‡. Whole-Genome Sequencing in Severe Chronic Obstructive Pulmonary Disease. Am J Respir Cell Mol Biol 2018; 59:614-622. [PMID: 29949718 PMCID: PMC6236690 DOI: 10.1165/rcmb.2018-0088oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/27/2018] [Indexed: 01/21/2023] Open
Abstract
Genome-wide association studies have identified common variants associated with chronic obstructive pulmonary disease (COPD). Whole-genome sequencing (WGS) offers comprehensive coverage of the entire genome, as compared with genotyping arrays or exome sequencing. We hypothesized that WGS in subjects with severe COPD and smoking control subjects with normal pulmonary function would allow us to identify novel genetic determinants of COPD. We sequenced 821 patients with severe COPD and 973 control subjects from the COPDGene and Boston Early-Onset COPD studies, including both non-Hispanic white and African American individuals. We performed single-variant and grouped-variant analyses, and in addition, we assessed the overlap of variants between sequencing- and array-based imputation. Our most significantly associated variant was in a known region near HHIP (combined P = 1.6 × 10-9); additional variants approaching genome-wide significance included previously described regions in CHRNA5, TNS1, and SERPINA6/SERPINA1 (the latter in African American individuals). None of our associations were clearly driven by rare variants, and we found minimal evidence of replication of genes identified by previously reported smaller sequencing studies. With WGS, we identified more than 20 million new variants, not seen with imputation, including more than 10,000 of potential importance in previously identified COPD genome-wide association study regions. WGS in severe COPD identifies a large number of potentially important functional variants, with the strongest associations being in known COPD risk loci, including HHIP and SERPINA1. Larger sample sizes will be needed to identify associated variants in novel regions of the genome.
Collapse
Affiliation(s)
- Dmitry Prokopenko
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Phuwanat Sakornsakolpat
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Heide Loehlein Fier
- Working Group of Genomic Mathematics, University of Bonn, Bonn, Germany
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts
| | - Dandi Qiao
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Margaret M. Parker
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Merry-Lynn N. McDonald
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
| | - R. Graham Barr
- Department of Medicine, College of Physicians and Surgeons and
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| | - Christopher J. Williams
- Division of Pulmonary Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Mark L. Brantly
- Division of Pulmonary Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida
| | - Christoph Lange
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts
| | - Terri H. Beaty
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland; and
| | | | - Edwin K. Silverman
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - on behalf of COPDGene Study Investigators* and NHLBI TOPMed Investigators‡
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Working Group of Genomic Mathematics, University of Bonn, Bonn, Germany
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
- Department of Medicine, College of Physicians and Surgeons and
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
- Division of Pulmonary Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland; and
- National Jewish Health, Denver, Colorado
| |
Collapse
|
42
|
Mehlig K, Berg C, Björck L, Nyberg F, Olin AC, Rosengren A, Strandhagen E, Torén K, Thelle DS, Lissner L. Cohort Profile: The INTERGENE Study. Int J Epidemiol 2018; 46:1742-1743h. [PMID: 28186561 DOI: 10.1093/ije/dyw332] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2016] [Indexed: 12/17/2022] Open
Affiliation(s)
- Kirsten Mehlig
- Section for Epidemiology and Social Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Christina Berg
- Department of Food and Nutrition, and Sport Science, University of Gothenburg, Gothenburg, Sweden
| | - Lena Björck
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Institute of Health and Care Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Nyberg
- Section for Occupational and Environmental Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Medical Evidence and Observational Research Centre, Global Medical Affairs, AstraZeneca Gothenburg, Mölndal, Sweden
| | - Anna-Carin Olin
- Section for Occupational and Environmental Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Annika Rosengren
- Section for Epidemiology and Social Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elisabeth Strandhagen
- Section for Epidemiology and Social Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kjell Torén
- Section for Occupational and Environmental Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Dag S Thelle
- Section for Epidemiology and Social Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Biostatistics, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Lauren Lissner
- Section for Epidemiology and Social Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
43
|
Shagiwal SS, den Dekker HT, de Jongste JC, Brusselle GG, Jaddoe VWV, Felix JF, Duijts L. Influence of genetic variants on childhood lung function - The Generation R Study. Pediatr Allergy Immunol 2018; 29:589-595. [PMID: 29882296 DOI: 10.1111/pai.12931] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/09/2018] [Indexed: 11/28/2022]
Abstract
Genetic variants associated with adult lung function could already exert the effects on childhood lung function. We aimed to examine the associations of adult lung function-related genetic variants with childhood lung function and asthma, and whether these associations were modified by atopic predisposition, tobacco smoke exposure, or early growth characteristics. In a population-based prospective cohort study among 3347 children, we selected 7 and 20 single nucleotide polymorphisms (SNPs) associated with adult forced expiratory volume in 1 second (FEV1 ) and FEV1 /forced vital capacity (FEV1 /FVC), respectively. Weighted genetic risk scores (GRSs) for FEV1 and FEV1 /FVC were constructed. At age 10, FEV1 , FVC, FEV1 /FVC, forced expiratory flow between 25% and 75% (FEF25-75 ), and forced expiratory flow at 75% (FEF75 ) of FVC were measured, and information on asthma was obtained by parental-reported questionnaires. The FEV1 -GRS was associated with lower childhood FEV1 , FEV1 /FVC, and FEF75 (Z-score (95% CI): -0.03 (-0.05, -0.01), -0.03 (-0.05, -0.01), and -0.04 (-0.05, -0.01), respectively, per additional risk allele). The FEV1 /FVC-GRS was associated with lower childhood FEV1 /FVC and FEF75 (Z-score (95% CI): -0.04 (-0.05, -0.03) and -0.03 (-0.05, -0.02), respectively, per additional risk allele). Effect estimates of FEV1 -GRS with FEF25-75 , FEV1 , FEF75 , and FVC, and of FEV1 /FVC-GRS with FEV1 /FVC and FEF25-75 were stronger among children exposed to non-atopic mothers, smoking during pregnancy or in childhood, or those born with a lower birthweight, respectively (P-values for interaction < .05). Genetic risk scores were not associated with asthma. Adult lung function-related genetic variants were associated with childhood lung function. Maternal atopy, smoking during pregnancy or in childhood, and birthweight modified the observed effects.
Collapse
Affiliation(s)
- Sara S Shagiwal
- The Generation R Study Group, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Division of Respiratory Medicine and Allergology, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Epidemiology, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Herman T den Dekker
- The Generation R Study Group, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Division of Respiratory Medicine and Allergology, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Epidemiology, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Johan C de Jongste
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Guy G Brusselle
- Department of Epidemiology, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Respiratory Medicine and Allergology, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Vincent W V Jaddoe
- The Generation R Study Group, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Epidemiology, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Janine F Felix
- The Generation R Study Group, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Epidemiology, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Liesbeth Duijts
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Respiratory Medicine and Allergology, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
44
|
Burkart KM, Sofer T, London SJ, Manichaikul A, Hartwig FP, Yan Q, Soler Artigas M, Avila L, Chen W, Davis Thomas S, Diaz AA, Hall IP, Horta BL, Kaplan RC, Laurie CC, Menezes AM, Morrison JV, Oelsner EC, Rastogi D, Rich SS, Soto-Quiros M, Stilp AM, Tobin MD, Wain LV, Celedón JC, Barr RG. A Genome-Wide Association Study in Hispanics/Latinos Identifies Novel Signals for Lung Function. The Hispanic Community Health Study/Study of Latinos. Am J Respir Crit Care Med 2018; 198:208-219. [PMID: 29394082 PMCID: PMC6058984 DOI: 10.1164/rccm.201707-1493oc] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 01/30/2018] [Indexed: 12/14/2022] Open
Abstract
RATIONALE Lung function and chronic obstructive pulmonary disease (COPD) are heritable traits. Genome-wide association studies (GWAS) have identified numerous pulmonary function and COPD loci, primarily in cohorts of European ancestry. OBJECTIVES Perform a GWAS of COPD phenotypes in Hispanic/Latino populations to identify loci not previously detected in European populations. METHODS GWAS of lung function and COPD in Hispanic/Latino participants from a population-based cohort. We performed replication studies of novel loci in independent studies. MEASUREMENTS AND MAIN RESULTS Among 11,822 Hispanic/Latino participants, we identified eight novel signals; three replicated in independent populations of European Ancestry. A novel locus for FEV1 in ZSWIM7 (rs4791658; P = 4.99 × 10-9) replicated. A rare variant (minor allele frequency = 0.002) in HAL (rs145174011) was associated with FEV1/FVC (P = 9.59 × 10-9) in a region previously identified for COPD-related phenotypes; it remained significant in conditional analyses but did not replicate. Admixture mapping identified a novel region, with a variant in AGMO (rs41331850), associated with Amerindian ancestry and FEV1, which replicated. A novel locus for FEV1 identified among ever smokers (rs291231; P = 1.92 × 10-8) approached statistical significance for replication in admixed populations of African ancestry, and a novel SNP for COPD in PDZD2 (rs7709630; P = 1.56 × 10-8) regionally replicated. In addition, loci previously identified for lung function in European samples were associated in Hispanic/Latino participants in the Hispanic Community Health Study/Study of Latinos at the genome-wide significance level. CONCLUSIONS We identified novel signals for lung function and COPD in a Hispanic/Latino cohort. Including admixed populations when performing genetic studies may identify variants contributing to genetic etiologies of COPD.
Collapse
Affiliation(s)
- Kristin M. Burkart
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Biostatistics, University of Washington School of Public Health, Seattle, Washington
| | - Stephanie J. London
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Fernando P. Hartwig
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Pelotas, Brazil
| | - Qi Yan
- Division of Pulmonary Medicine, Allergy, and Immunology, Department of Pediatrics, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - María Soler Artigas
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - Lydiana Avila
- Division of Pediatric Pulmonology, Hospital Nacional de Niños, San José, Costa Rica
| | - Wei Chen
- Division of Pulmonary Medicine, Allergy, and Immunology, Department of Pediatrics, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sonia Davis Thomas
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina
| | - Alejandro A. Diaz
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ian P. Hall
- Division of Respiratory Medicine, Queen’s Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Bernardo L. Horta
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Pelotas, Brazil
| | | | - Cathy C. Laurie
- Department of Biostatistics, University of Washington School of Public Health, Seattle, Washington
| | - Ana M. Menezes
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Pelotas, Brazil
| | - Jean V. Morrison
- Department of Human Genetics and Statistics, University of Chicago, Chicago, Illinois
| | - Elizabeth C. Oelsner
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Deepa Rastogi
- Department of Pediatrics, Albert Einstein College of Medicine, New York, New York
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Manuel Soto-Quiros
- Division of Pulmonary Medicine, Allergy, and Immunology, Department of Pediatrics, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adrienne M. Stilp
- Department of Biostatistics, University of Washington School of Public Health, Seattle, Washington
| | - Martin D. Tobin
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, United Kingdom
- National Institute for Health Research, Leicester Respiratory Biomedical Research Centre, Glenfield Hospital, Leicester, United Kingdom; and
| | - Louise V. Wain
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, United Kingdom
- National Institute for Health Research, Leicester Respiratory Biomedical Research Centre, Glenfield Hospital, Leicester, United Kingdom; and
| | - Juan C. Celedón
- Division of Pulmonary Medicine, Allergy, and Immunology, Department of Pediatrics, Children’s Hospital of Pittsburgh of University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - R. Graham Barr
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| |
Collapse
|
45
|
Xu J, Shang Y, Cai F, Zhang S, Xiao Z, Wang H, Fan Y, Li T, Sheng S, Fu Y, Chi F, Zhou C. Correlation between lung cancer and the HHIP polymorphisms of chronic obstructive pulmonary disease (COPD) in the Chinese Han population. Genes Immun 2018; 20:273-280. [PMID: 29915314 DOI: 10.1038/s41435-018-0033-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 04/10/2018] [Accepted: 04/17/2018] [Indexed: 11/09/2022]
Abstract
To further investigate the relationship between lung cancer and hedgehog interacting protein (HHIP) polymorphisms of chronic obstructive pulmonary disease (COPD) patients, we conducted a case-control study in a Chinese Han population. Six HHIP SNPs with minor allele frequencies >5% (rs1489758, rs1489759, rs10519717, rs13131837, rs1492820, and rs7689420) were analyzed in 1,017 COPD patients (767 males and 246 females) and 430 non-COPD patients. Using logistic regression analysis, we found that rs7689420 was significantly associated with lung cancer in COPD patients in the Chinese Han population (P < 0.001). The recessive allele of rs7689420 was associated with the occurrence of lung cancer in all COPD patients (odds ratios [OR] of 0.609 and 0.424 for the CT and TT genotypes, respectively) as well as in serious COPD patients (OR of 0.403 and 0.305 for CT and TT, respectively). Additionally, rs1489759 and rs3131837 were associated with lung cancer in various genetic models. rs1489758, rs1489759, and rs10519717 were also associated with lung cancer in serious COPD patients. However, none of the SNPs were significantly associated with lung cancer in mild COPD patients or healthy subjects. Therefore, the HHIP SNPs of COPD patients likely play a role in lung cancer pathology in the Chinese Han population.
Collapse
Affiliation(s)
- Jing Xu
- Department of Respiratory Medicine, Seventh People's Hospital of Shanghai University of TCM, 200137, Shanghai, China.
| | - Yan Shang
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, Second Military Medical University, 200433, Shanghai, China
| | - Feng Cai
- Department of Respiratory Medicine, Seventh People's Hospital of Shanghai University of TCM, 200137, Shanghai, China
| | - Shu Zhang
- Department of Respiratory Medicine, Seventh People's Hospital of Shanghai University of TCM, 200137, Shanghai, China
| | - Zhong Xiao
- Department of Respiratory Medicine, Seventh People's Hospital of Shanghai University of TCM, 200137, Shanghai, China
| | - Haitao Wang
- Department of Respiratory Medicine, Seventh People's Hospital of Shanghai University of TCM, 200137, Shanghai, China
| | - Yanhong Fan
- Department of Respiratory Medicine, Seventh People's Hospital of Shanghai University of TCM, 200137, Shanghai, China
| | - Ting Li
- Department of Respiratory Medicine, Seventh People's Hospital of Shanghai University of TCM, 200137, Shanghai, China
| | - Shuhong Sheng
- Department of Respiratory Medicine, Seventh People's Hospital of Shanghai University of TCM, 200137, Shanghai, China
| | - Youhui Fu
- Department of Respiratory Medicine, Seventh People's Hospital of Shanghai University of TCM, 200137, Shanghai, China
| | - Feng Chi
- Department of Respiratory Medicine, Seventh People's Hospital of Shanghai University of TCM, 200137, Shanghai, China
| | - Chen Zhou
- Department of Respiratory Medicine, Seventh People's Hospital of Shanghai University of TCM, 200137, Shanghai, China
| |
Collapse
|
46
|
Vishweswaraiah S, George L, Purushothaman N, Ganguly K. A candidate gene identification strategy utilizing mouse to human big-data mining: "3R-tenet" in COPD genetic research. Respir Res 2018; 19:92. [PMID: 29871630 PMCID: PMC5989378 DOI: 10.1186/s12931-018-0795-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 04/27/2018] [Indexed: 12/13/2022] Open
Abstract
Background Early life impairments leading to lower lung function by adulthood are considered as risk factors for chronic obstructive pulmonary disease (COPD). Recently, we compared the lung transcriptomic profile between two mouse strains with extreme total lung capacities to identify plausible pulmonary function determining genes using microarray analysis (GSE80078). Advancement of high-throughput techniques like deep sequencing (eg. RNA-seq) and microarray have resulted in an explosion of genomic data in the online public repositories which however remains under-exploited. Strategic curation of publicly available genomic data with a mouse-human translational approach can effectively implement “3R- Tenet” by reducing screening experiments with animals and performing mechanistic studies using physiologically relevant in vitro model systems. Therefore, we sought to analyze the association of functional variations within human orthologs of mouse lung function candidate genes in a publicly available COPD lung RNA-seq data-set. Methods Association of missense single nucleotide polymorphisms, insertions, deletions, and splice junction variants were analyzed for susceptibility to COPD using RNA-seq data of a Korean population (GSE57148). Expression of the associated genes were studied using the Gene Paint (mouse embryo) and Human Protein Atlas (normal adult human lung) databases. The genes were also assessed for replication of the associations and expression in COPD−/mouse cigarette smoke exposed lung tissues using other datasets. Results Significant association (p < 0.05) of variations in 20 genes to higher COPD susceptibility have been detected within the investigated cohort. Association of HJURP, MCRS1 and TLR8 are novel in relation to COPD. The associated ADAM19 and KIT loci have been reported earlier. The remaining 15 genes have also been previously associated to COPD. Differential transcript expression levels of the associated genes in COPD- and/ or mouse emphysematous lung tissues have been detected. Conclusion Our findings suggest strategic mouse-human datamining approaches can identify novel COPD candidate genes using existing datasets in the online repositories. The candidates can be further evaluated for mechanistic role through in vitro studies using appropriate primary cells/cell lines. Functional studies can be limited to transgenic animal models of only well supported candidate genes. This approach will lead to a significant reduction of animal experimentation in respiratory research. Electronic supplementary material The online version of this article (10.1186/s12931-018-0795-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Leema George
- SRM Research Institute, SRM University, Chennai, 603203, India
| | - Natarajan Purushothaman
- Department of Genetic Engineering, School of Bioengineering, Faculty of Engineering and Technology, SRM University, Chennai, 603203, India
| | - Koustav Ganguly
- SRM Research Institute, SRM University, Chennai, 603203, India. .,Work Environment Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Box 287, SE-171 77, Stockholm, Sweden.
| |
Collapse
|
47
|
Kim WJ, Park HJ, Choi YJ, Kwon EY, Kim BM, Lee JH, Chang JH, Lee Kang J, Choi JH. Association between Genetic Variations of MERTK and Chronic Obstructive Pulmonary Disease in Koreans. J Korean Med Sci 2018; 33:e56. [PMID: 29359540 PMCID: PMC5785628 DOI: 10.3346/jkms.2018.33.e56] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/12/2017] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a debilitating lung disease. To date, a large number of clinical studies have been conducted to investigate the association between genetic variations and COPD. However, little is known regarding the genetic susceptibility of Koreans to this disease. MER receptor tyrosine kinase (MERTK) plays important roles in the inhibition of inflammation and in the clearance of apoptotic cells. Here, we investigated the association between genetic variations in MERTK and the development of COPD in Koreans. METHODS We conducted genetic analysis of MERTK using genomic DNA samples from 87 patients with COPD and 88 healthy controls and compared the frequency of each variation or haplotype between the patient and control groups. Subsequently, the effect of each variation was evaluated using in vitro assays. RESULTS Ten variations were identified in this study, four of them for the first time. In addition, we found that the frequency of each variation or haplotype was comparable between the patient and control groups. However, we observed that the frequency for the wild-type haplotype was higher in the control group, compared to that in the group of patients with COPD, in the subgroup analysis of current smokers, although the difference was not statistically significant (P = 0.080). In in vitro assays, we observed that none of the variations affected the activity of the promoter or the expression of MERTK. CONCLUSION Our findings indicate that the susceptibility to COPD is not related to the genetic variations or haplotypes of MERTK in Koreans.
Collapse
Affiliation(s)
- Woo Jin Kim
- Department of Internal Medicine and Environmental Health Center, Kangwon National University, Chuncheon, Korea
| | - Hyo Jin Park
- Department of Pharmacology, Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Yang Ji Choi
- Department of Pharmacology, Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Eun Young Kwon
- Department of Pharmacology, Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Bo Min Kim
- Department of Pharmacology, Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Jin Hwa Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Jung Hyun Chang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Jihee Lee Kang
- Department of Physiology, Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Ji Ha Choi
- Department of Pharmacology, Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, Korea.
| |
Collapse
|
48
|
Lee S, She J, Deng B, Kim J, de Andrade M, Na J, Sun Z, Wampfler JA, Cunningham JM, Wu Y, Limper AH, Aubry MC, Wendt C, Biterman P, Yang P, Lou Z. Multiple-level validation identifies PARK2 in the development of lung cancer and chronic obstructive pulmonary disease. Oncotarget 2018; 7:44211-44223. [PMID: 27329585 PMCID: PMC5190090 DOI: 10.18632/oncotarget.9954] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/09/2016] [Indexed: 12/31/2022] Open
Abstract
An important precursor to lung cancer development is chronic obstructive pulmonary disease (COPD), independent of exposure to tobacco smoke. Both diseases are associated with increased host susceptibility, inflammation, and genomic instability. However, validation of the candidate genes and functional confirmation to test shared genetic contribution and cellular mechanisms to the development of lung cancer in patients with COPD remains underexplored. Here, we show that loss of PARK2 (encoding Parkin) increases the expression of proinflammation factors as well as nuclear NF-κB localization, suggesting a role of PARK2 loss in inflammation. Additional exploration showed that PARK2 deficiency promotes genomic instability and cell transformation. This role of PARK2 in inflammation and chromosome instability provides a potential link among Parkin, COPD and lung cancer. A further comprehensive validation of 114 informative single nucleotide polymorphism (SNP) variants of PARK2, in 2,484 cases and controls with well-defined lung cancer and COPD phenotypes, found rs577876, rs6455728 and rs9346917 (p<0.01) to be significantly associated with lung cancer development in people with COPD. Our findings support the evidence that PARK2 might have a tumor suppressor role in the development of COPD and lung cancer.
Collapse
Affiliation(s)
- SeungBaek Lee
- Division of Oncology Research, Mayo Clinic, Rochester, MN, USA
| | - Jun She
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Bo Deng
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA.,Department of Thoracic Surgery, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - JungJin Kim
- Division of Oncology Research, Mayo Clinic, Rochester, MN, USA
| | - Mariza de Andrade
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Jie Na
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Zhifu Sun
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Jason A Wampfler
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | - Yanhong Wu
- Genomics Shared Resource, Mayo Clinic, Rochester, MN, USA
| | - Andrew H Limper
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Chris Wendt
- Department of Medicine, Pulmonary Division, University of Minnesota, Minneapolis, MN, USA.,Department of Medicine, Pulmonary Section, Minneapolis VA Medical Center, Minneapolis, MN, USA
| | - Peter Biterman
- Department of Medicine, Pulmonary Division, University of Minnesota, Minneapolis, MN, USA
| | - Ping Yang
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Zhenkun Lou
- Division of Oncology Research, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
49
|
van der Plaat DA, de Jong K, Lahousse L, Faiz A, Vonk JM, van Diemen CC, Nedeljkovic I, Amin N, Obeidat M, van Duijn CM, Boezen HM, Postma DS. The Well-Known Gene HHIP and Novel Gene MECR Are Implicated in Small Airway Obstruction. Am J Respir Crit Care Med 2017; 194:1299-1302. [PMID: 27845578 DOI: 10.1164/rccm.201604-0843le] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Diana A van der Plaat
- 1 University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC) Groningen, the Netherlands
| | - Kim de Jong
- 1 University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC) Groningen, the Netherlands
| | - Lies Lahousse
- 2 Erasmus Medical Center Rotterdam, the Netherlands.,3 Ghent University Hospital Ghent, Belgium and
| | - Alen Faiz
- 1 University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC) Groningen, the Netherlands
| | - Judith M Vonk
- 1 University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC) Groningen, the Netherlands
| | - Cleo C van Diemen
- 1 University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC) Groningen, the Netherlands
| | | | - Najaf Amin
- 2 Erasmus Medical Center Rotterdam, the Netherlands
| | | | | | - H Marike Boezen
- 1 University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC) Groningen, the Netherlands
| | - Dirkje S Postma
- 1 University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC) Groningen, the Netherlands
| |
Collapse
|
50
|
What do polymorphisms tell us about the mechanisms of COPD? Clin Sci (Lond) 2017; 131:2847-2863. [PMID: 29203722 DOI: 10.1042/cs20160718] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 10/22/2017] [Accepted: 11/01/2017] [Indexed: 12/11/2022]
Abstract
COPD (chronic obstructive pulmonary disease) is characterized by irreversible lung airflow obstruction. Cigarette smoke is the major risk factor for COPD development. However, only a minority number of smokers develop COPD, and there are substantial variations in lung function among smokers, suggesting that genetic determinants in COPD susceptibility. During the past decade, genome-wide association studies and exome sequencing have been instrumental to identify the genetic determinants of complex traits, including COPD. Focused studies have revealed mechanisms by which genetic variants contribute to COPD and have led to novel insights in COPD pathogenesis. Through functional investigations of causal variants in COPD, from the proteinase-antiproteinase theory to emerging roles of developmental pathways (such as Hedgehog and Wnt pathways) in COPD, we have greatly expanded our understanding on this complex pulmonary disease. In this review, we critically review functional investigations on roles of genetic polymorphisms in COPD, and discuss future challenges and opportunities in discovering novel mechanisms of functional variants.
Collapse
|