1
|
Gentili M, Glass K, Maiorino E, Hobbs BD, Xu Z, Castaldi PJ, Cho MH, Hersh CP, Qiao D, Morrow JD, Carey VJ, Platig J, Silverman EK. Partial correlation network analysis identifies coordinated gene expression within a regional cluster of COPD genome-wide association signals. PLoS Comput Biol 2024; 20:e1011079. [PMID: 39418301 PMCID: PMC11521246 DOI: 10.1371/journal.pcbi.1011079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 10/29/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a complex disease influenced by well-established environmental exposures (most notably, cigarette smoking) and incompletely defined genetic factors. The chromosome 4q region harbors multiple genetic risk loci for COPD, including signals near HHIP, FAM13A, GSTCD, TET2, and BTC. Leveraging RNA-Seq data from lung tissue in COPD cases and controls, we estimated the co-expression network for genes in the 4q region bounded by HHIP and BTC (~70MB), through partial correlations informed by protein-protein interactions. We identified several co-expressed gene pairs based on partial correlations, including NPNT-HHIP, BTC-NPNT and FAM13A-TET2, which were replicated in independent lung tissue cohorts. Upon clustering the co-expression network, we observed that four genes previously associated to COPD: BTC, HHIP, NPNT and PPM1K appeared in the same network community. Finally, we discovered a sub-network of genes differentially co-expressed between COPD vs controls (including FAM13A, PPA2, PPM1K and TET2). Many of these genes were previously implicated in cell-based knock-out experiments, including the knocking out of SPP1 which belongs to the same genomic region and could be a potential local key regulatory gene. These analyses identify chromosome 4q as a region enriched for COPD genetic susceptibility and differential co-expression.
Collapse
Affiliation(s)
- Michele Gentili
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kimberly Glass
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Enrico Maiorino
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Brian D. Hobbs
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Zhonghui Xu
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Peter J. Castaldi
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- Division of General Internal Medicine and Primary Care, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Michael H. Cho
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Craig P. Hersh
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Dandi Qiao
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jarrett D. Morrow
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Vincent J. Carey
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - John Platig
- Department of Genome Sciences, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Edwin K. Silverman
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| |
Collapse
|
2
|
Sun G, Zhou Y, Han X, Che X, Yu S, Song D, Ma F, Huang L. Potential marker genes for chronic obstructive pulmonary disease revealed based on single-cell sequencing and Mendelian randomization analysis. Aging (Albany NY) 2024; 16:8922-8943. [PMID: 38787375 PMCID: PMC11164476 DOI: 10.18632/aging.205849] [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: 12/05/2023] [Accepted: 04/15/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Progress is being made in the prevention and treatment of chronic obstructive pulmonary disease (COPD), but it is still unsatisfactory. With the development of genetic technology, validated genetic information can better explain COPD. OBJECTIVE The study utilized scRNA-seq and Mendelian randomization analysis of eQTLs to identify crucial genes and potential mechanistic pathways underlying COPD pathogenesis. MEHODS Single-cell sequencing data were used to identify marker genes for immune cells in the COPD process. Data on eQTLs for immune cell marker genes were obtained from the eQTLGen consortium. To estimate the causal effect of marker genes on COPD, we selected an independent cohort (ukb-b-16751) derived from the UK Biobank database for two-sample Mendelian randomization analysis. Subsequently, we performed immune infiltration analysis, gene set enrichment analysis (GSEA), and co-expression network analysis on the key genes. RESULTS The 154 immune cell-associated marker genes identified were mainly involved in pathways such as vacuolar cleavage, positive regulation of immune response and regulation of cell activation. Mendelian randomization analysis screened four pairs of marker genes (GZMH, COTL1, CSTA and CD14) were causally associated with COPD. These four key genes were significantly associated with immune cells. In addition, we have identified potential transcription factors associated with these key genes using the Cistrome database, thus contributing to a deeper understanding of the regulatory network of these gene expressions. CONCLUSIONS This eQTLs Mendelian randomization study identified four key genes (GZMH, COTL1, CSTA, and CD14) causally associated with COPD, providing new insights for prevention and treatment of COPD.
Collapse
Affiliation(s)
- Gang Sun
- General Hospital of Northern Theater Command, Shenyang 110000, Liaoning, China
| | - Yun Zhou
- East China Normal University Wuhu Affiliated Hospital (The Second People’s Hospital of Wuhu City), Wuhu 241000, Anhui, China
| | - Xiaoxiao Han
- The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei 230022, Anhui China
| | - Xiangqian Che
- General Hospital of Northern Theater Command, Shenyang 110000, Liaoning, China
| | - Shuo Yu
- The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei 230022, Anhui China
| | - Di Song
- General Hospital of Northern Theater Command, Shenyang 110000, Liaoning, China
| | - Feifei Ma
- General Hospital of Northern Theater Command, Shenyang 110000, Liaoning, China
| | - Lewei Huang
- General Hospital of Northern Theater Command, Shenyang 110000, Liaoning, China
| |
Collapse
|
3
|
Werder RB, Zhou X, Cho MH, Wilson AA. Breathing new life into the study of COPD with genes identified from genome-wide association studies. Eur Respir Rev 2024; 33:240019. [PMID: 38811034 PMCID: PMC11134200 DOI: 10.1183/16000617.0019-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 02/23/2024] [Indexed: 05/31/2024] Open
Abstract
COPD is a major cause of morbidity and mortality globally. While the significance of environmental exposures in disease pathogenesis is well established, the functional contribution of genetic factors has only in recent years drawn attention. Notably, many genes associated with COPD risk are also linked with lung function. Because reduced lung function precedes COPD onset, this association is consistent with the possibility that derangements leading to COPD could arise during lung development. In this review, we summarise the role of leading genes (HHIP, FAM13A, DSP, AGER and TGFB2) identified by genome-wide association studies in lung development and COPD. Because many COPD genome-wide association study genes are enriched in lung epithelial cells, we focus on the role of these genes in the lung epithelium in development, homeostasis and injury.
Collapse
Affiliation(s)
- Rhiannon B Werder
- Murdoch Children's Research Institute, Melbourne, Australia
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew A Wilson
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Radder JE, Bon J. Multiomics and Multiancestry Approaches: Key Steps to Untangling the Web of Chronic Obstructive Pulmonary Disease Pathogenesis. Ann Am Thorac Soc 2023; 20:1101-1102. [PMID: 37526482 PMCID: PMC10405612 DOI: 10.1513/annalsats.202304-311ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023] Open
Affiliation(s)
- Josiah E Radder
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jessica Bon
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
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
|
Hsieh MJ, Lo YS, Tsai YJ, Ho HY, Lin CC, Chuang YC, Lin SH, Chen MK. FAM13A polymorphisms are associated with a specific susceptibility to clinical progression of oral cancer in alcohol drinkers. BMC Cancer 2023; 23:607. [PMID: 37391706 DOI: 10.1186/s12885-023-11052-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 06/09/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND Single nucleotide polymorphism (SNP) is a genetic variation that occurs when a single nucleotide base in the DNA sequence varies between individuals and is present in at least 1% of the population. Genetic variants in FAM13A are associated with different types of chronic respiratory diseases, including chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), and lung cancer. However, there is little literature on the association of FAM13A genotypes with oral cancer. Therefore, this project will explore the correlation between the FAM13A genotype and the formation of oral cancer. METHODS In this project, we will examine the presence of gene polymorphisms gene polymorphisms of rs1059122, rs3017895, rs3756050, and rs7657817 in the FAM13A gene exon, and combine the expression of these genes to try to clarify the impact of the FAM13A gene polymorphism on oral cancer. First, four loci (rs1059122, rs3017895, rs3756050, and rs7657817) of the FAM13A SNP were genotyped using TaqMan allelic discrimination. RESULTS By estimating OR and AOR, FAM13A exhibited different genotypic variables in four SNPs that were not statistically significant between controls and patients with oral cancer. The results of the general analysis showed that different distributions of allelic types did not affect clinical stage, tumour size, lymph node invasion, distant metastasis, and pathological differentiation status. However, in the alcohol drinking group specifically, patients with the rs3017895 SNP G genotype had a 3.17-fold (95% CI, 1.102-9.116; p = 0.032) increase in the well differentiated state of cells compared to patients with the A allele. CONCLUSIONS Our results suggested that the SNP rs3017895 FAM13A could contribute to oral cancer. More sample studies are needed in the future to confirm our results and more functional studies are needed to investigate their relevant roles in the development of oral cancer.
Collapse
Affiliation(s)
- Ming-Ju Hsieh
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, 500, Taiwan
- Program in Tissue Engineering and Regenerative Medicine, College of Medicine, National Chung Hsing University, Taichung, 402, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan
| | - Yu-Sheng Lo
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, 500, Taiwan
| | - Yun-Jung Tsai
- Department of Surgical Pathology, Changhua Christian Hospital, Changhua, 500, Taiwan
- Translational Pathology Core Laboratory, Changhua Christian Hospital, Changhua, 500, Taiwan
| | - Hsin-Yu Ho
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, 500, Taiwan
| | - Chia-Chieh Lin
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, 500, Taiwan
| | - Yi-Ching Chuang
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, 500, Taiwan
| | - Shu-Hui Lin
- Department of Surgical Pathology, Changhua Christian Hospital, Changhua, 500, Taiwan
- Translational Pathology Core Laboratory, Changhua Christian Hospital, Changhua, 500, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Central Taiwan University of Science and Technology, Taichung, 406, Taiwan
| | - Mu-Kuan Chen
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, No.135, Nanxiao St., Changhua City, Changhua County, 500, Taiwan.
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, 402, Taiwan.
| |
Collapse
|
9
|
Pham KH, Tran NTC, Tran HD, Ngo TH, Tran VD, Ly HHV, Pham NTN, Nguyen T, Nguyen BH, Nguyen KT. Single Nucleotide Polymorphisms of FAM13A Gene in Chronic Obstructive Pulmonary Disease-A Case Control Study in Vietnam. Adv Respir Med 2023; 91:268-277. [PMID: 37366807 DOI: 10.3390/arm91030021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/13/2023] [Accepted: 06/13/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND In 2018, GOLD addressed the issues of genotypes associated with risk factors for COPD. The genome-wide association study (GWAS) demonstrated an association between COPD and several genetic variants of single nucleotide polymorphisms (SNPs) of the FAM13A gene with the risk of COPD. OBJECTIVE To study the single nucleotide polymorphisms rs2869967 and rs17014601 of the FAM13A gene in chronic obstructive pulmonary disease. Subjects and research methods: 80 subjects diagnosed with COPD and 80 subjects determined not to have COPD according to GOLD 2020 criteria; the subjects were clinically examined, interviewed, and identified as possessing single nucleotide polymorphisms using the sanger sequencing method on whole blood samples. RESULTS The male/female ratio of the patient group and the control group was 79/1 and 39/1, respectively. The percentages of C and T alleles of rs2869967 in COPD patients were 50.6% and 49.4%, respectively. The percentages of C and T alleles of rs17014601 in COPD patients were 31.9% and 68.1%, respectively. At rs17014601, the ratio values of alleles T and C in the disease group and the control group were markedly different, making them statistically reliable (p = 0.031). The rate of CT genotype in the group of patients was considerably higher than that of the control group. The TT homozygous genotype had a lower risk of COPD compared with the other genotypes in the dominant model (ORTT/(CC + CT) = 0.441; CI95% = 0.233-0.833); this difference was statistically significant (p = 0.012). CONCLUSIONS With rs17014601, it is characteristic that the frequency of the T allele appears more than the C allele, and the CT heterozygous phenotype accounts for the highest proportion in rs17014601 and rs2869967 recorded in COPD patients. There is an association between the genetic variant of the SNP FAM13A-rs17014601 and the risk of COPD.
Collapse
Affiliation(s)
- Khanh Hoang Pham
- Faculty of Medicine, Can Tho University of Medicine and Pharmacy, Can Tho City 900000, Vietnam
| | - Nhung Thi Cam Tran
- Department of Anesthesiology and Resuscitation, Hoan My Cuu Long Hospital, Can Tho City 900000, Vietnam
| | - Hung Do Tran
- Faculty of Medicine, Can Tho University of Medicine and Pharmacy, Can Tho City 900000, Vietnam
| | - Toan Hoang Ngo
- Faculty of Medicine, Can Tho University of Medicine and Pharmacy, Can Tho City 900000, Vietnam
| | - Van De Tran
- Faculty of Medicine, Can Tho University of Medicine and Pharmacy, Can Tho City 900000, Vietnam
| | - Hung Huynh Vinh Ly
- Faculty of Medicine, Can Tho University of Medicine and Pharmacy, Can Tho City 900000, Vietnam
| | - Nga Thi Ngoc Pham
- Faculty of Medicine, Can Tho University of Medicine and Pharmacy, Can Tho City 900000, Vietnam
| | - Thang Nguyen
- Faculty of Medicine, Can Tho University of Medicine and Pharmacy, Can Tho City 900000, Vietnam
| | - Binh Huy Nguyen
- Physiology Department, Hanoi Medical University, Ha Noi 100000, Vietnam
| | - Kien Trung Nguyen
- Faculty of Medicine, Can Tho University of Medicine and Pharmacy, Can Tho City 900000, Vietnam
| |
Collapse
|
10
|
Carlin DE, Larsen SJ, Sirupurapu V, Cho MH, Silverman EK, Baumbach J, Ideker T. Hierarchical association of COPD to principal genetic components of biological systems. PLoS One 2023; 18:e0286064. [PMID: 37228113 DOI: 10.1371/journal.pone.0286064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 05/08/2023] [Indexed: 05/27/2023] Open
Abstract
Many disease-causing genetic variants converge on common biological functions and pathways. Precisely how to incorporate pathway knowledge in genetic association studies is not yet clear, however. Previous approaches employ a two-step approach, in which a regular association test is first performed to identify variants associated with the disease phenotype, followed by a test for functional enrichment within the genes implicated by those variants. Here we introduce a concise one-step approach, Hierarchical Genetic Analysis (Higana), which directly computes phenotype associations against each function in the large hierarchy of biological functions documented by the Gene Ontology. Using this approach, we identify risk genes and functions for Chronic Obstructive Pulmonary Disease (COPD), highlighting microtubule transport, muscle adaptation, and nicotine receptor signaling pathways. Microtubule transport has not been previously linked to COPD, as it integrates genetic variants spread over numerous genes. All associations validate strongly in a second COPD cohort.
Collapse
Affiliation(s)
- Daniel E Carlin
- Department of Medicine, Division of Genetics, University of California San Diego, La Jolla, CA, United States of America
| | | | - Vikram Sirupurapu
- Department of Medicine, Division of Genetics, University of California San Diego, La Jolla, CA, United States of America
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, United States of America
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, United States of America
| | - Jan Baumbach
- Department of Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Trey Ideker
- Department of Medicine, Division of Genetics, University of California San Diego, La Jolla, CA, United States of America
| |
Collapse
|
11
|
Gong L, Bates S, Li Y, Lin X, Wei W, Zhou X. AKT Phosphorylates FAM13A and Promotes Its Degradation via CUL4A/DDB1/DCAF1 E3 Complex. Am J Respir Cell Mol Biol 2023; 68:577-590. [PMID: 36749583 PMCID: PMC10174174 DOI: 10.1165/rcmb.2022-0362oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 02/07/2023] [Indexed: 02/08/2023] Open
Abstract
SNPs within FAM13A (family with sequence similarity 13 member A) gene are significantly associated with chronic obstructive pulmonary disease and lung function in genome-wide association studies (GWAS). However, how FAM13A protein is regulated under physiological and pathological conditions remains largely elusive. Herein, we report that FAM13A is phosphorylated at the serine 312 residue by AKT kinase after cigarette smoke extract treatment and thereby recognized by the CULLIN4A/DCAF1 (DDB1 and CUL4 associated factor 1) E3 ligase complex, rendering the ubiquitination-mediated degradation of FAM13A. More broadly, downregulation of FAM13A protein upon AKT activation, as a general cellular response to acute stress, was also detected in influenza- or naphthalene-injured lungs in mice. Functionally, reduced protein levels of FAM13A lead to accelerated epithelial cell proliferation in murine lungs during the recovery phase after injury. In summary, we characterized a novel molecular mechanism that regulates the stability of FAM13A protein, which enables the fine-tuning of lung epithelial repair after injury. These significant findings will expand our molecular understanding of the regulation of protein stability, which may modulate lung epithelial repair implicated in the development of chronic obstructive pulmonary disease and other lung diseases.
Collapse
Affiliation(s)
- Lu Gong
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts; and
| | - Samuel Bates
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts; and
| | - Yujun Li
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts; and
| | - Xin Lin
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts; and
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts; and
| |
Collapse
|
12
|
Grygiel-Górniak B, Ziółkowska-Suchanek I, Szymkowiak L, Rozwadowska N, Kaczmarek E. The Influence of FAM13A and PPAR-γ2 Gene Polymorphisms on the Metabolic State of Postmenopausal Women. Genes (Basel) 2023; 14:genes14040914. [PMID: 37107672 PMCID: PMC10137345 DOI: 10.3390/genes14040914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Recently, we have observed two significant pandemics caused by communicable (COVID-19) and non-communicable factors (obesity). Obesity is related to a specific genetic background and characterized by immunogenetic features, such as low-grade systemic inflammation. The specific genetic variants include the presence of polymorphism of the Peroxisome Proliferator-Activated Receptors gene (PPAR-γ2; Pro12Ala, rs1801282, and C1431T, rs3856806 polymorphisms), β-adrenergic receptor gene (3β-AR; Trp64Arg, rs4994), and Family With Sequence Similarity 13 Member A gene (FAM13A; rs1903003, rs7671167, rs2869967). This study aimed to analyze the genetic background, body fat distribution, and hypertension risk in obese metabolically healthy postmenopausal women (n = 229, including 105 lean and 124 obese subjects). Each patient underwent anthropometric and genetic evaluations. The study has shown that the highest value of BMI was associated with visceral fat distribution. The analysis of particular genotypes has revealed no differences between lean and obese women except for FAM13A rs1903003 (CC), which was more prevalent in lean patients. The co-existence of the PPAR-γ2 C1431C variant with other FAM13A gene polymorphisms [rs1903003(TT) or rs7671167(TT), or rs2869967(CC)] was related to higher BMI values and visceral fat distribution (WHR > 0.85). The co-association of FAM13A rs1903003 (CC) and 3β-AR Trp64Arg was associated with higher values of systolic (SBP) and diastolic blood pressure (DBP). We conclude that the co-existence of FAM13A variants with C1413C polymorphism of the PPAR-γ2 gene is responsible for body fat amount and distribution.
Collapse
Affiliation(s)
- Bogna Grygiel-Górniak
- Department of Rheumatology, Rehabilitation and Internal Diseases, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | | | - Lidia Szymkowiak
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland
| | - Natalia Rozwadowska
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland
| | - Elżbieta Kaczmarek
- Department of Bioinformatics and Computational Biology, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| |
Collapse
|
13
|
Higuchi T, Oka S, Furukawa H, Shimada K, Tsunoda S, Ito S, Okamoto A, Katayama M, Saisho K, Shinohara S, Matsui T, Migita K, Nagaoka S, Tohma S. Association of a FAM13A variant with interstitial lung disease in Japanese rheumatoid arthritis. RMD Open 2023; 9:rmdopen-2022-002828. [PMID: 36717188 PMCID: PMC9887688 DOI: 10.1136/rmdopen-2022-002828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/09/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Interstitial lung disease (ILD) occasionally occurs in rheumatoid arthritis (RA) and confers a dismal prognosis. We previously reported that a single-nucleotide variant (SNV) of MUC5B was associated with ILD in RA. However, the pathogenesis of ILD in Japanese patients with RA could not be explained solely by this SNV because its frequency is extremely low in the Japanese population. Here, we examined whether a different idiopathic pulmonary fibrosis susceptibility SNV might be associated with ILD in Japanese patients with RA. METHODS Genotyping of rs2609255 (G/T) in FAM13A was conducted in 208 patients with RA with ILD and 420 without chronic lung disease using TaqMan assays. RESULTS A significant association with usual interstitial pneumonia (UIP) in RA was detected for rs2609255 under the allele model (p=0.0092, Pc=0.0276, OR 1.53, 95% CI 1.12 to 2.11) and recessive model for the G allele (p=0.0003, Pc=0.0009, OR 2.63, 95% CI 1.59 to 4.32). FAM13A rs2609255 was significantly associated with UIP in male patients with RA (p=0.0043, OR 3.65, 95% CI 1.52 to 8.73) under the recessive model. CONCLUSIONS This study is the first to document an association of rs2609255 with ILD in Japanese patients with RA, implicating it in the pathogenesis of UIP, though studies on the function of rs2609255 are warranted.
Collapse
Affiliation(s)
- Takashi Higuchi
- Department of Clinical Research, National Hospital Organization Tokyo National Hospital, Kiyose, Japan,Department of Nephrology, Ushiku Aiwa General Hospital, Ushiku, Japan
| | - Shomi Oka
- Department of Clinical Research, National Hospital Organization Tokyo National Hospital, Kiyose, Japan,Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan
| | - Hiroshi Furukawa
- Department of Clinical Research, National Hospital Organization Tokyo National Hospital, Kiyose, Japan .,Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan
| | - Kota Shimada
- Department of Rheumatic Diseases, Tokyo Metropolitan Tama Medical Center, Fuchu, Japan,Department of Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan
| | - Shinichiro Tsunoda
- Department of Nephrology, Sumitomo Hospital, Osaka, Japan,Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Satoshi Ito
- Department of Rheumatology, Niigata Rheumatic Center, Shibata, Japan
| | - Akira Okamoto
- Department of Rheumatology, National Hospital Organization Himeji Medical Center, Himeji, Japan
| | - Masao Katayama
- Department of Internal Medicine, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Koichiro Saisho
- Tanimura Hospital, Nobeoka, Japan,Department of Orthopedics/Rheumatology, National Hospital Organization Miyakonojo Medical Center, Miyakonojo, Japan
| | | | - Toshihiro Matsui
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan,Department of Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan
| | - Kiyoshi Migita
- Department of Gastroenterology and Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan,Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Japan
| | - Shouhei Nagaoka
- Department of Rheumatology, Yokohama Minami Kyosai Hospital, Yokohama, Japan
| | - Shigeto Tohma
- Department of Clinical Research, National Hospital Organization Tokyo National Hospital, Kiyose, Japan,Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan
| |
Collapse
|
14
|
Multifactorial Diseases of the Heart, Kidneys, Lungs, and Liver and Incident Cancer: Epidemiology and Shared Mechanisms. Cancers (Basel) 2023; 15:cancers15030729. [PMID: 36765688 PMCID: PMC9913123 DOI: 10.3390/cancers15030729] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/05/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Within the aging population, the frequency of cancer is increasing dramatically. In addition, multiple genetic and environmental factors lead to common multifactorial diseases, including cardiovascular disease, chronic kidney disease, chronic obstructive pulmonary disease, and metabolic-associated fatty liver disease. In recent years, there has been a growing awareness of the connection between cancer and multifactorial diseases, as well as how one can affect the other, resulting in a vicious cycle. Although the exact mechanistic explanations behind this remain to be fully explored, some progress has been made in uncovering the common pathologic mechanisms. In this review, we focus on the nature of the link between cancer and common multifactorial conditions, as well as specific shared mechanisms, some of which may represent either preventive or therapeutic targets. Rather than organ-specific interactions, we herein focus on the shared mechanisms among the multifactorial diseases, which may explain the increased cancer risk. More research on this subject will highlight the significance of developing new drugs that target multiple systems rather than just one disease.
Collapse
|
15
|
Hayden LP, Hobbs BD, Busch R, Cho MH, Liu M, Lopes-Ramos CM, Lomas DA, Bakke P, Gulsvik A, Silverman EK, Crapo JD, Beaty TH, Laird NM, Lange C, DeMeo DL. X chromosome associations with chronic obstructive pulmonary disease and related phenotypes: an X chromosome-wide association study. Respir Res 2023; 24:38. [PMID: 36726148 PMCID: PMC9891756 DOI: 10.1186/s12931-023-02337-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 01/18/2023] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The association between genetic variants on the X chromosome to risk of COPD has not been fully explored. We hypothesize that the X chromosome harbors variants important in determining risk of COPD related phenotypes and may drive sex differences in COPD manifestations. METHODS Using X chromosome data from three COPD-enriched cohorts of adult smokers, we performed X chromosome specific quality control, imputation, and testing for association with COPD case-control status, lung function, and quantitative emphysema. Analyses were performed among all subjects, then stratified by sex, and subsequently combined in meta-analyses. RESULTS Among 10,193 subjects of non-Hispanic white or European ancestry, a variant near TMSB4X, rs5979771, reached genome-wide significance for association with lung function measured by FEV1/FVC ([Formula: see text] 0.020, SE 0.004, p 4.97 × 10-08), with suggestive evidence of association with FEV1 ([Formula: see text] 0.092, SE 0.018, p 3.40 × 10-07). Sex-stratified analyses revealed X chromosome variants that were differentially trending in one sex, with significantly different effect sizes or directions. CONCLUSIONS This investigation identified loci influencing lung function, COPD, and emphysema in a comprehensive genetic association meta-analysis of X chromosome genetic markers from multiple COPD-related datasets. Sex differences play an important role in the pathobiology of complex lung disease, including X chromosome variants that demonstrate differential effects by sex and variants that may be relevant through escape from X chromosome inactivation. Comprehensive interrogation of the X chromosome to better understand genetic control of COPD and lung function is important to further understanding of disease pathology. Trial registration Genetic Epidemiology of COPD Study (COPDGene) is registered at ClinicalTrials.gov, NCT00608764 (Active since January 28, 2008). Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints Study (ECLIPSE), GlaxoSmithKline study code SCO104960, is registered at ClinicalTrials.gov, NCT00292552 (Active since February 16, 2006). Genetics of COPD in Norway Study (GenKOLS) holds GlaxoSmithKline study code RES11080, Genetics of Chronic Obstructive Lung Disease.
Collapse
Affiliation(s)
- Lystra P. Hayden
- grid.38142.3c000000041936754XDivision of Pulmonary Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA ,grid.38142.3c000000041936754XChanning Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, 181 Longwood Ave, Boston, MA 02115 USA
| | - Brian D. Hobbs
- grid.38142.3c000000041936754XChanning Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, 181 Longwood Ave, Boston, MA 02115 USA ,grid.38142.3c000000041936754XDivision of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Robert Busch
- grid.417587.80000 0001 2243 3366Division of Pulmonology, Allergy, and Critical Care, U.S. Food and Drug Administration, Silver Spring, MD USA
| | - Michael H. Cho
- grid.38142.3c000000041936754XChanning Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, 181 Longwood Ave, Boston, MA 02115 USA ,grid.38142.3c000000041936754XDivision of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Ming Liu
- grid.268323.e0000 0001 1957 0327Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, MA USA
| | - Camila M. Lopes-Ramos
- grid.38142.3c000000041936754XChanning Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, 181 Longwood Ave, Boston, MA 02115 USA ,grid.38142.3c000000041936754XDepartment of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - David A. Lomas
- grid.83440.3b0000000121901201UCL Respiratory, University College London, London, UK
| | - Per Bakke
- grid.7914.b0000 0004 1936 7443Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Amund Gulsvik
- grid.7914.b0000 0004 1936 7443Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Edwin K. Silverman
- grid.38142.3c000000041936754XChanning Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, 181 Longwood Ave, Boston, MA 02115 USA ,grid.38142.3c000000041936754XDivision of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - James D. Crapo
- grid.240341.00000 0004 0396 0728Division of Pulmonary Sciences and Critical Care Medicine, National Jewish Health, Denver, CO USA
| | - Terri H. Beaty
- grid.21107.350000 0001 2171 9311Johns Hopkins School of Public Health, Baltimore, MD USA
| | - Nan M. Laird
- grid.38142.3c000000041936754XDepartment of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Christoph Lange
- grid.38142.3c000000041936754XDepartment of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Dawn L. DeMeo
- grid.38142.3c000000041936754XChanning Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, 181 Longwood Ave, Boston, MA 02115 USA ,grid.38142.3c000000041936754XDivision of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| |
Collapse
|
16
|
Mechanisms of Lung Damage and Development of COPD Due to Household Biomass-Smoke Exposure: Inflammation, Oxidative Stress, MicroRNAs, and Gene Polymorphisms. Cells 2022; 12:cells12010067. [PMID: 36611860 PMCID: PMC9818405 DOI: 10.3390/cells12010067] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/28/2022] Open
Abstract
Chronic exposure to indoor biomass smoke from the combustion of solid organic fuels is a major cause of disease burden worldwide. Almost 3 billion people use solid fuels such as wood, charcoal, and crop residues for indoor cooking and heating, accounting for approximately 50% of all households and 90% of rural households globally. Biomass smoke contains many hazardous pollutants, resulting in household air pollution (HAP) exposure that often exceeds international standards. Long-term biomass-smoke exposure is associated with Chronic Obstructive Pulmonary Disease (COPD) in adults, a leading cause of morbidity and mortality worldwide, chronic bronchitis, and other lung conditions. Biomass smoke-associated COPD differs from the best-known cigarette smoke-induced COPD in several aspects, such as a slower decline in lung function, greater airway involvement, and less emphysema, which suggests a different phenotype and pathophysiology. Despite the high burden of biomass-associated COPD, the molecular, genetic, and epigenetic mechanisms underlying its pathogenesis are poorly understood. This review describes the pathogenic mechanisms potentially involved in lung damage, the development of COPD associated with wood-derived smoke exposure, and the influence of genetic and epigenetic factors on the development of this disease.
Collapse
|
17
|
Lahmar Z, Ahmed E, Fort A, Vachier I, Bourdin A, Bergougnoux A. Hedgehog pathway and its inhibitors in chronic obstructive pulmonary disease (COPD). Pharmacol Ther 2022; 240:108295. [PMID: 36191777 DOI: 10.1016/j.pharmthera.2022.108295] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/22/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022]
Abstract
COPD affects millions of people and is now ranked as the third leading cause of death worldwide. This largely untreatable chronic airway disease results in irreversible destruction of lung architecture. The small lung hypothesis is now supported by epidemiological, physiological and clinical studies. Accordingly, the early and severe COPD phenotype carries the most dreadful prognosis and finds its roots during lung growth. Pathophysiological mechanisms remain poorly understood and implicate individual susceptibility (genetics), a large part of environmental factors (viral infections, tobacco consumption, air pollution) and the combined effects of those triggers on gene expression. Genetic susceptibility is most likely involved as the disease is severe and starts early in life. The latter observation led to the identification of Mendelian inheritance via disease-causing variants of SERPINA1 - known as the basis for alpha-1 anti-trypsin deficiency, and TERT. In the last two decades multiple genome wide association studies (GWAS) identified many single nucleotide polymorphisms (SNPs) associated with COPD. High significance SNPs are located in 4q31 near HHIP which encodes an evolutionarily highly conserved physiological inhibitor of the Hedgehog signaling pathway (HH). HHIP is critical to several in utero developmental lung processes. It is also implicated in homeostasis, injury response, epithelial-mesenchymal transition and tumor resistance to apoptosis. A few studies have reported decreased HHIP RNA and protein levels in human adult COPD lungs. HHIP+/- murine models led to emphysema. HH pathway inhibitors, such as vismodegib and sonidegib, are already validated in oncology, whereas other drugs have evidenced in vitro effects. Targeting the Hedgehog pathway could lead to a new therapeutic avenue in COPD. In this review, we focused on the early and severe COPD phenotype and the small lung hypothesis by exploring genetic susceptibility traits that are potentially treatable, thus summarizing promising therapeutics for the future.
Collapse
Affiliation(s)
- Z Lahmar
- Department of Respiratory Diseases, CHU de Montpellier, Montpellier, France
| | - E Ahmed
- Department of Respiratory Diseases, CHU de Montpellier, Montpellier, France; PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France
| | - A Fort
- PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France
| | - I Vachier
- Department of Respiratory Diseases, CHU de Montpellier, Montpellier, France; PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France
| | - A Bourdin
- Department of Respiratory Diseases, CHU de Montpellier, Montpellier, France; PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France
| | - A Bergougnoux
- PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France; Laboratoire de Génétique Moléculaire et de Cytogénomique, CHU de Montpellier, Montpellier, France.
| |
Collapse
|
18
|
Kim W, Hecker J, Barr RG, Boerwinkle E, Cade B, Correa A, Dupuis J, Gharib SA, Lange L, London SJ, Morrison AC, O'Connor GT, Oelsner EC, Psaty BM, Vasan RS, Redline S, Rich SS, Rotter JI, Yu B, Lange C, Manichaikul A, Zhou JJ, Sofer T, Silverman EK, Qiao D, Cho MH. Assessing the contribution of rare genetic variants to phenotypes of chronic obstructive pulmonary disease using whole-genome sequence data. Hum Mol Genet 2022; 31:3873-3885. [PMID: 35766891 PMCID: PMC9652112 DOI: 10.1093/hmg/ddac117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/13/2022] [Accepted: 05/16/2021] [Indexed: 01/04/2023] Open
Abstract
RATIONALE Genetic variation has a substantial contribution to chronic obstructive pulmonary disease (COPD) and lung function measurements. Heritability estimates using genome-wide genotyping data can be biased if analyses do not appropriately account for the nonuniform distribution of genetic effects across the allele frequency and linkage disequilibrium (LD) spectrum. In addition, the contribution of rare variants has been unclear. OBJECTIVES We sought to assess the heritability of COPD and lung function using whole-genome sequence data from the Trans-Omics for Precision Medicine program. METHODS Using the genome-based restricted maximum likelihood method, we partitioned the genome into bins based on minor allele frequency and LD scores and estimated heritability of COPD, FEV1% predicted and FEV1/FVC ratio in 11 051 European ancestry and 5853 African-American participants. MEASUREMENTS AND MAIN RESULTS In European ancestry participants, the estimated heritability of COPD, FEV1% predicted and FEV1/FVC ratio were 35.5%, 55.6% and 32.5%, of which 18.8%, 19.7%, 17.8% were from common variants, and 16.6%, 35.8%, and 14.6% were from rare variants. These estimates had wide confidence intervals, with common variants and some sets of rare variants showing a statistically significant contribution (P-value < 0.05). In African-Americans, common variant heritability was similar to European ancestry participants, but lower sample size precluded calculation of rare variant heritability. CONCLUSIONS Our study provides updated and unbiased estimates of heritability for COPD and lung function, and suggests an important contribution of rare variants. Larger studies of more diverse ancestry will improve accuracy of these estimates.
Collapse
Affiliation(s)
- Wonji Kim
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Julian Hecker
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - R Graham Barr
- Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Brian Cade
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Adolfo Correa
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Josée Dupuis
- Department of Biostatistics, Boston University of Public Health, Boston, MA 02118, USA
| | - Sina A Gharib
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Leslie Lange
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Stephanie J London
- Epidemiology Branch, National Institute of Environmental Health Sciences, Department of Health and Human Services, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Alanna C Morrison
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - George T O'Connor
- Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Elizabeth C Oelsner
- Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101, USA
- Departments of Epidemiology and Health Services, University of Washington, Seattle, WA 98101, USA
| | - Ramachandran S Vasan
- Lung and Blood Institute Framingham Heart Study, Boston University and National Heart, Framingham, MA 01702, USA
- Department of Preventive Medicine and Epidemiology, School of Medicine and Public Health, Boston University, Boston, MA 02118, USA
| | - Susan Redline
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Christoph Lange
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, USA
| | - Jin J Zhou
- Department of Epidemiology and Biostatistics, University of Arizona, Tucson, AZ 85721, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorder, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Dandi Qiao
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
19
|
Wang Y, Zhou Q, Chen L, Dong L, Xiong M, Xie X, Zhao L, Xu J, Zheng Z, Wang J, Lu W. Identification of genetic variants of the IL-22 gene in association with an altered risk of COPD susceptibility. THE CLINICAL RESPIRATORY JOURNAL 2022; 16:537-545. [PMID: 35808996 PMCID: PMC9376143 DOI: 10.1111/crj.13517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 05/28/2022] [Accepted: 06/02/2022] [Indexed: 01/13/2023]
Abstract
The incidence of chronic obstructive pulmonary disease (COPD) is related to the interaction between environmental exposure and genetic factors. Far more than 15% of smokers eventually develop COPD. In addition to smoking, genetic susceptibility may be another factor in the development of COPD. IL-22 and its receptors are increased in human and experimental COPD and contribute to pathogenesis. Here, we conducted a case-control study to evaluate the association between IL-22 tag-single nucleotide polymorphisms (SNPs) and COPD risk. Four tag-SNPs (rs2227478, rs2227481, rs2227484 and rs2227485) were identified according to linkage disequilibrium (LD) analysis in 30 healthy controls. A total of 513 COPD cases and 504 controls were recruited to perform an association study between these four tag-SNPs and COPD risk. We found that the "C" allele of rs2227478T>C and the "T" allele of rs2227481C>T were obviously related to decreased COPD susceptibility. Genetic model analysis showed that rs2227478T>C and rs2227481C>T were significantly associated with a decreased risk of COPD under dominant models after adjusting for the above factors. In the recessive model, rs2227485T>C was obviously associated with decreased COPD risk. Our data showed that only rs2227485T>C was associated with a decreased COPD risk after Bonferroni correction. The eQTL analysis showed that rs2227485T>C was significantly associated with IL-22 expression. The pGL4-rs2227485-C gene reporter had a higher promoter activity than pGL4-rs2227485-T. In our study, rs2227485T>C, located in the promoter region of IL-22, was associated with a decreased risk of COPD and increased IL-22 promoter activity, suggesting that this variant might modulate COPD susceptibility.
Collapse
Affiliation(s)
- Yan Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Qipeng Zhou
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Lingzhu Chen
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Lian Dong
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Mingmei Xiong
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Xiaohui Xie
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Li Zhao
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Jingyi Xu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Zeguang Zheng
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Jian Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Wenju Lu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
20
|
Li D, Kim W, An J, Kim S, Lee S, Do A, Kim W, Lee S, Yoon D, Lee K, Ha S, Silverman EK, Cho M, Shin C, Won S. Heritability Analyses Uncover Shared Genetic Effects of Lung Function and Change over Time. Genes (Basel) 2022; 13:genes13071261. [PMID: 35886044 PMCID: PMC9316642 DOI: 10.3390/genes13071261] [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: 05/02/2022] [Revised: 07/05/2022] [Accepted: 07/08/2022] [Indexed: 01/27/2023] Open
Abstract
Genetic influence on lung functions has been identified in previous studies; however, the relative longitudinal effects of genetic factors and their interactions with smoking on lung function remain unclear. Here, we identified the longitudinal effects of genetic variants on lung function by determining single nucleotide polymorphism (SNP) heritability and genetic correlations, and by analyzing interactions with smoking. Subject-specific means and annual change rates were calculated for eight spirometric measures obtained from 6622 Korean adults aged 40−69 years every two years for 14 years, and their heritabilities were estimated separately. Statistically significant (p < 0.05) heritability for the subject-specific means of all spirometric measures (8~32%) and change rates of forced expiratory volume in 1 s to forced vital capacity ratio (FEV1/FVC; 16%) and post-bronchodilator FEV1/FVC (17%) were detected. Significant genetic correlations of the change rate with the subject-specific mean were observed for FEV1/FVC (ρg = 0.64) and post-bronchodilator FEV1/FVC (ρg = 0.47). Furthermore, post-bronchodilator FEV1/FVC showed significant heritability of SNP-by-smoking interaction (hGXS2 = 0.4) for the annual change rate. The GWAS also detected genome-wide significant SNPs for FEV1 (rs4793538), FEV1/FVC (rs2704589, rs62201158, and rs9391733), and post-bronchodilator FEV1/FVC (rs2445936). We found statistically significant evidence of heritability role on the change in lung function, and this was shared with the effects on cross-sectional measurements. We also found some evidence of interaction with smoking for the change of lung function.
Collapse
Affiliation(s)
- Donghe Li
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, Korea; (D.L.); (A.D.)
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA 02118, USA
| | - Woojin Kim
- Department of Internal Medicine and Environmental Health Center, School of Medicine, Kangwon National University, Chuncheon 24341, Korea;
| | - Jahoon An
- Department of Public Health Science, Graduate School of Public Health, Seoul National University, Seoul 08826, Korea;
| | - Soriul Kim
- Institute for Human Genomic Study, College of Medicine, Korea University, Seoul 136701, Korea; (S.K.); (S.L.)
| | - Seungku Lee
- Institute for Human Genomic Study, College of Medicine, Korea University, Seoul 136701, Korea; (S.K.); (S.L.)
| | - Ahra Do
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, Korea; (D.L.); (A.D.)
| | - Wonji Kim
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (W.K.); (E.K.S.); (M.C.)
| | - Sanghun Lee
- Department of Medical Consilience, Graduate School, Dankook University, Yongin 16890, Korea;
| | - Dankyu Yoon
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju 28159, Korea;
| | - Kwangbae Lee
- Korea Medical Institute, Seoul 03173, Korea; (K.L.); (S.H.)
| | - Seounguk Ha
- Korea Medical Institute, Seoul 03173, Korea; (K.L.); (S.H.)
| | - Edwin K. Silverman
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (W.K.); (E.K.S.); (M.C.)
| | - Michael Cho
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (W.K.); (E.K.S.); (M.C.)
| | - Chol Shin
- Institute for Human Genomic Study, College of Medicine, Korea University, Seoul 136701, Korea; (S.K.); (S.L.)
- Division of Pulmonary Sleep and Critical Care Medicine, Department of Internal Medicine, Korea University Ansan Hospital, Ansan 15355, Korea
- Correspondence: (C.S.); (S.W.)
| | - Sungho Won
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, Korea; (D.L.); (A.D.)
- Department of Public Health Science, Graduate School of Public Health, Seoul National University, Seoul 08826, Korea;
- Institute of Health and Environment, Seoul National University, Seoul 08826, Korea
- RexSoft Inc., Seoul 08826, Korea
- Correspondence: (C.S.); (S.W.)
| |
Collapse
|
21
|
Genetic Variants Associated with Chronic Obstructive Pulmonary Disease Risk: Cumulative Epidemiological Evidence from Meta-Analyses and Genome-Wide Association Studies. Can Respir J 2022; 2022:3982335. [PMID: 35721789 PMCID: PMC9203202 DOI: 10.1155/2022/3982335] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 05/26/2022] [Indexed: 12/03/2022] Open
Abstract
Background Last two decades, many association studies on genetic variants and chronic obstructive pulmonary disease (COPD) risk have been published. But results from different studies are inconsistent. Therefore, we performed this article to systematically evaluate results from previous meta-analyses and genome-wide association studies (GWASs). Material and Methods. Firstly, we retrieved meta-analyses in PubMed, Embase, and China National Knowledge Infrastructure and GWASs in PubMed and GWAS catalog on or before April 7th, 2022. Then, data were extracted and screened. Finally, two main methods—Venice criteria and false-positive report probability test—were used to evaluate significant associations. Results As a result, eighty-eight meta-analyses and 5 GWASs were deemed eligible for inclusion. Fifty variants in 26 genes obtained from meta-analyses were significantly associated with COPD risk. Cumulative epidemiological evidence of an association was graded as strong for 10 variants in 8 genes (GSTM1, CHRNA, ADAM33, SP-D, TNF-α, VDBP, HMOX1, and HHIP), moderate for 6 variants in 5 genes (PI, GSTM1, ADAM33, TNF-α, and VDBP), and weak for 40 variants in 23 genes. Five variants in 4 genes showed convincing evidence of no association with COPD risk in meta-analyses. Additionally, 29 SNPs identified in GWASs were proved to be noteworthy based on the FPRP test. Conclusion In summary, more than half (52.38%) of genetic variants reported in previous meta-analyses showed no association with COPD risk. However, 13 variants in 9 genes had moderate to strong evidence for an association. This article can serve as a useful reference for further studies.
Collapse
|
22
|
Christenson SA, Smith BM, Bafadhel M, Putcha N. Chronic obstructive pulmonary disease. Lancet 2022; 399:2227-2242. [PMID: 35533707 DOI: 10.1016/s0140-6736(22)00470-6] [Citation(s) in RCA: 344] [Impact Index Per Article: 172.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 02/16/2022] [Accepted: 02/25/2022] [Indexed: 12/14/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a major cause of morbidity, mortality, and health-care use worldwide. COPD is caused by exposure to inhaled noxious particles, notably tobacco smoke and pollutants. However, the broad range of factors that increase the risk of development and progression of COPD throughout the life course are increasingly being recognised. Innovations in omics and imaging techniques have provided greater insight into disease pathobiology, which might result in advances in COPD prevention, diagnosis, and treatment. Although few novel treatments have been approved for COPD in the past 5 years, advances have been made in targeting existing therapies to specific subpopulations using new biomarker-based strategies. Additionally, COVID-19 has undeniably affected individuals with COPD, who are not only at higher risk for severe disease manifestations than healthy individuals but also negatively affected by interruptions in health-care delivery and social isolation. This Seminar reviews COPD with an emphasis on recent advances in epidemiology, pathophysiology, imaging, diagnosis, and treatment.
Collapse
Affiliation(s)
- Stephanie A Christenson
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Benjamin M Smith
- Department of Medicine, Columbia University Medical Center, New York, NY, USA; Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Mona Bafadhel
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK; Department of Respiratory Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nirupama Putcha
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
23
|
Cho MH, Hobbs BD, Silverman EK. Genetics of chronic obstructive pulmonary disease: understanding the pathobiology and heterogeneity of a complex disorder. THE LANCET. RESPIRATORY MEDICINE 2022; 10:485-496. [PMID: 35427534 PMCID: PMC11197974 DOI: 10.1016/s2213-2600(21)00510-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/20/2021] [Accepted: 11/09/2021] [Indexed: 12/20/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a deadly and highly morbid disease. Susceptibility to and heterogeneity of COPD are incompletely explained by environmental factors such as cigarette smoking. Family-based and population-based studies have shown that a substantial proportion of COPD risk is related to genetic variation. Genetic association studies have identified hundreds of genetic variants that affect risk for COPD, decreased lung function, and other COPD-related traits. These genetic variants are associated with other pulmonary and non-pulmonary traits, demonstrate a genetic basis for at least part of COPD heterogeneity, have a substantial effect on COPD risk in aggregate, implicate early-life events in COPD pathogenesis, and often involve genes not previously suspected to have a role in COPD. Additional progress will require larger genetic studies with more ancestral diversity, improved profiling of rare variants, and better statistical methods. Through integration of genetic data with other omics data and comprehensive COPD phenotypes, as well as functional description of causal mechanisms for genetic risk variants, COPD genetics will continue to inform novel approaches to understanding the pathobiology of COPD and developing new strategies for management and treatment.
Collapse
Affiliation(s)
- Michael H Cho
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Brian D Hobbs
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Wang M, Zhang S, Sha Q. A computationally efficient clustering linear combination approach to jointly analyze multiple phenotypes for GWAS. PLoS One 2022; 17:e0260911. [PMID: 35482827 PMCID: PMC9049312 DOI: 10.1371/journal.pone.0260911] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 04/13/2022] [Indexed: 11/18/2022] Open
Abstract
There has been an increasing interest in joint analysis of multiple phenotypes in genome-wide association studies (GWAS) because jointly analyzing multiple phenotypes may increase statistical power to detect genetic variants associated with complex diseases or traits. Recently, many statistical methods have been developed for joint analysis of multiple phenotypes in genetic association studies, including the Clustering Linear Combination (CLC) method. The CLC method works particularly well with phenotypes that have natural groupings, but due to the unknown number of clusters for a given data, the final test statistic of CLC method is the minimum p-value among all p-values of the CLC test statistics obtained from each possible number of clusters. Therefore, a simulation procedure needs to be used to evaluate the p-value of the final test statistic. This makes the CLC method computationally demanding. We develop a new method called computationally efficient CLC (ceCLC) to test the association between multiple phenotypes and a genetic variant. Instead of using the minimum p-value as the test statistic in the CLC method, ceCLC uses the Cauchy combination test to combine all p-values of the CLC test statistics obtained from each possible number of clusters. The test statistic of ceCLC approximately follows a standard Cauchy distribution, so the p-value can be obtained from the cumulative density function without the need for the simulation procedure. Through extensive simulation studies and application on the COPDGene data, the results demonstrate that the type I error rates of ceCLC are effectively controlled in different simulation settings and ceCLC either outperforms all other methods or has statistical power that is very close to the most powerful method with which it has been compared.
Collapse
Affiliation(s)
- Meida Wang
- Mathematical Sciences, Michigan Technological University, Houghton, MI, United States of America
| | - Shuanglin Zhang
- Mathematical Sciences, Michigan Technological University, Houghton, MI, United States of America
| | - Qiuying Sha
- Mathematical Sciences, Michigan Technological University, Houghton, MI, United States of America
| |
Collapse
|
25
|
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: 11] [Impact Index Per Article: 5.5] [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
|
26
|
Fixing the GAP: the role of RhoGAPs in cancer. Eur J Cell Biol 2022; 101:151209. [DOI: 10.1016/j.ejcb.2022.151209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/29/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
|
27
|
Lakshman Kumar P, Wilson AC, Rocco A, Cho MH, Wan E, Hobbs BD, Washko GR, Ortega VE, Christenson SA, Li X, Wells JM, Bhatt SP, DeMeo DL, Lutz SM, Rossiter H, Casaburi R, Rennard SI, Lomas DA, Labaki WW, Tal‐Singer R, Bowler RP, Hersh CP, Tiwari HK, Dransfield M, Thalacker‐Mercer A, Meyers DA, Silverman EK, McDonald MN. Genetic variation in genes regulating skeletal muscle regeneration and tissue remodelling associated with weight loss in chronic obstructive pulmonary disease. J Cachexia Sarcopenia Muscle 2021; 12:1803-1817. [PMID: 34523824 PMCID: PMC8718068 DOI: 10.1002/jcsm.12782] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/08/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is the third leading cause of death globally. COPD patients with cachexia or weight loss have increased risk of death independent of body mass index (BMI) and lung function. We tested the hypothesis genetic variation is associated with weight loss in COPD using a genome-wide association study approach. METHODS Participants with COPD (N = 4308) from three studies (COPDGene, ECLIPSE, and SPIROMICS) were analysed. Discovery analyses were performed in COPDGene with replication in SPIROMICS and ECLIPSE. In COPDGene, weight loss was defined as self-reported unintentional weight loss > 5% in the past year or low BMI (BMI < 20 kg/m2 ). In ECLIPSE and SPIROMICS, weight loss was calculated using available longitudinal visits. Stratified analyses were performed among African American (AA) and Non-Hispanic White (NHW) participants with COPD. Single variant and gene-based analyses were performed adjusting for confounders. Fine mapping was performed using a Bayesian approach integrating genetic association results with linkage disequilibrium and functional annotation. Significant gene networks were identified by integrating genetic regions associated with weight loss with skeletal muscle protein-protein interaction (PPI) data. RESULTS At the single variant level, only the rs35368512 variant, intergenic to GRXCR1 and LINC02383, was associated with weight loss (odds ratio = 3.6, 95% confidence interval = 2.3-5.6, P = 3.2 × 10-8 ) among AA COPD participants in COPDGene. At the gene level in COPDGene, EFNA2 and BAIAP2 were significantly associated with weight loss in AA and NHW COPD participants, respectively. The EFNA2 association replicated among AA from SPIROMICS (P = 0.0014), whereas the BAIAP2 association replicated in NHW from ECLIPSE (P = 0.025). The EFNA2 gene encodes the membrane-bound protein ephrin-A2 involved in the regulation of developmental processes and adult tissue homeostasis such as skeletal muscle. The BAIAP2 gene encodes the insulin-responsive protein of mass 53 kD (IRSp53), a negative regulator of myogenic differentiation. Integration of the gene-based findings participants with PPI data revealed networks of genes involved in pathways such as Rho and synapse signalling. CONCLUSIONS The EFNA2 and BAIAP2 genes were significantly associated with weight loss in COPD participants. Collectively, the integrative network analyses indicated genetic variation associated with weight loss in COPD may influence skeletal muscle regeneration and tissue remodelling.
Collapse
Affiliation(s)
- Preeti Lakshman Kumar
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of MedicineUniversity of Alabama at BirminghamBirminghamALUSA
| | - Ava C. Wilson
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of MedicineUniversity of Alabama at BirminghamBirminghamALUSA
- Department of EpidemiologyUniversity of Alabama at BirminghamBirminghamALUSA
| | - Alison Rocco
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of MedicineUniversity of Alabama at BirminghamBirminghamALUSA
- Department of EpidemiologyUniversity of Alabama at BirminghamBirminghamALUSA
| | - Michael H. Cho
- Channing Division of Network MedicineBrigham and Women's HospitalBostonMAUSA
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalBostonMAUSA
| | - Emily Wan
- Channing Division of Network MedicineBrigham and Women's HospitalBostonMAUSA
- Veterans Affairs Boston Health Care System, Jamaica PlainBostonMAUSA
| | - Brian D. Hobbs
- Channing Division of Network MedicineBrigham and Women's HospitalBostonMAUSA
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalBostonMAUSA
| | - George R. Washko
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalBostonMAUSA
| | - Victor E. Ortega
- Department of Internal Medicine, Section on Pulmonary, Critical Care, Allergy and Immunologic DiseasesWake Forest School of MedicineWinston‐SalemNCUSA
| | - Stephanie A. Christenson
- Division of Pulmonary, Critical Care, Allergy, & Sleep Medicine, Department of MedicineUniversity of California San FranciscoSan FranciscoCAUSA
| | - Xingnan Li
- Department of MedicineUniversity of Arizona College of MedicineTucsonAZUSA
| | - J. Michael Wells
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of MedicineUniversity of Alabama at BirminghamBirminghamALUSA
| | - Surya P. Bhatt
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of MedicineUniversity of Alabama at BirminghamBirminghamALUSA
| | - Dawn L. DeMeo
- Channing Division of Network MedicineBrigham and Women's HospitalBostonMAUSA
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalBostonMAUSA
| | - Sharon M. Lutz
- Department of Population MedicineHarvard Medical SchoolBostonMAUSA
| | - Harry Rossiter
- Rehabilitation Clinical Trials CenterLos Angeles Biomedical Research Institute at Harbor Harbor‐UCLA Medical CenterTorranceCAUSA
| | - Richard Casaburi
- Rehabilitation Clinical Trials CenterLos Angeles Biomedical Research Institute at Harbor Harbor‐UCLA Medical CenterTorranceCAUSA
| | | | | | - Wassim W. Labaki
- Division of Pulmonary and Critical Care MedicineUniversity of MichiganAnn ArborMIUSA
| | | | - Russel P. Bowler
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep MedicineNational Jewish HealthDenverCOUSA
| | - Craig P. Hersh
- Channing Division of Network MedicineBrigham and Women's HospitalBostonMAUSA
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalBostonMAUSA
| | - Hemant K. Tiwari
- Department of BiostatisticsUniversity of Alabama at BirminghamBirminghamALUSA
| | - Mark Dransfield
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of MedicineUniversity of Alabama at BirminghamBirminghamALUSA
| | - Anna Thalacker‐Mercer
- Department of Cell Development and Integrative BiologyUniversity of Alabama at BirminghamBirminghamALUSA
| | - Deborah A. Meyers
- Department of MedicineUniversity of Arizona College of MedicineTucsonAZUSA
| | - Edwin K. Silverman
- Channing Division of Network MedicineBrigham and Women's HospitalBostonMAUSA
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalBostonMAUSA
| | - Merry‐Lynn N. McDonald
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of MedicineUniversity of Alabama at BirminghamBirminghamALUSA
- Department of EpidemiologyUniversity of Alabama at BirminghamBirminghamALUSA
- Department of GeneticsUniversity of Alabama at BirminghamBirminghamALUSA
| | | |
Collapse
|
28
|
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: 3.0] [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
|
29
|
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
|
30
|
van der Molen MC, Hartman JE, Vermeulen CJ, van den Berge M, Faiz A, Kerstjens HAM, Charbonnier JP, Vanfleteren LEGW, Slebos DJ. Determinants of Lung Fissure Completeness. Am J Respir Crit Care Med 2021; 204:807-816. [PMID: 34126038 DOI: 10.1164/rccm.202102-0260oc] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/09/2021] [Indexed: 11/16/2022] Open
Abstract
Rationale: New advanced bronchoscopic treatment options for patients with severe chronic obstructive pulmonary disease (COPD) have led to increased interest for COPD phenotyping, including fissure completeness. Objectives: We investigated clinical, environmental, and genetic factors contributing to fissure completeness in patients with and without COPD. Methods: We used data from 9,926 participants of the COPDGene study who underwent chest computed tomographic (CT) scans. Fissure completeness was calculated from CT scans after quantitative CT analysis at baseline and 5-year follow-up. Clinical and environmental factors, including sex, race, smoking, COPD, emphysema, maternal smoking during pregnancy and maternal COPD, were tested for impact on fissure completeness. Genome-wide association analyses were performed separately in non-Hispanic White subjects and African American subjects. Measurements and Main Results: African American subjects had significantly higher fissure completeness than non-Hispanic White subjects for all three fissures (P < 0.001). There was no change in fissure completeness between baseline and 5-year follow-up. For all fissures, no clinically relevant differences in fissure completeness were found for other clinical or environmental factors, including COPD severity. Rs2173623, rs264866, rs2407284, rs7310342, rs4904145, rs6504172, and rs7209556 showed genome-wide significant associations with fissure completeness in non-Hispanic White subjects. In African American subjects, rs264866, rs4904145 and rs6504172 were identified as significant associations. Rs2173623, rs6504172, and rs7209556 lead to WNT5A and HOXB antisense RNA expression, which play an important role during embryogenesis. Conclusions: Fissure completeness is genetically determined and not dependent on age, sex, smoking status, the presence and severity of COPD (including exacerbation frequency), maternal smoking during pregnancy, or maternal COPD.
Collapse
Affiliation(s)
- Marieke C van der Molen
- Department of Pulmonary Diseases
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jorine E Hartman
- Department of Pulmonary Diseases
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Cornelis J Vermeulen
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Maarten van den Berge
- Department of Pulmonary Diseases
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Alen Faiz
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Respiratory Bioinformatics and Molecular Biology, University of Technology Sydney, Sydney, Australia
| | - Huib A M Kerstjens
- Department of Pulmonary Diseases
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Lowie E G W Vanfleteren
- COPD Centre, Institute of Medicine, and
- Department of Respiratory Medicine and Allergology, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Dirk-Jan Slebos
- Department of Pulmonary Diseases
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
31
|
Okyere DO, Bui DS, Washko GR, Lodge CJ, Lowe AJ, Cassim R, Perret JL, Abramson MJ, Walters EH, Waidyatillake NT, Dharmage SC. Predictors of lung function trajectories in population-based studies: A systematic review. Respirology 2021; 26:938-959. [PMID: 34490723 DOI: 10.1111/resp.14142] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/08/2021] [Accepted: 08/11/2021] [Indexed: 01/11/2023]
Abstract
Despite the growing body of evidence on lung function trajectories over the life course and their risk factors, the literature has not been systematically synthesized. Publications related to lung function trajectories were identified from PubMed, EMBASE and CINAHL databases. Two authors independently identified publications for inclusion according to predefined selection criteria. Studies that modelled lung function trajectories and reported associated exposures were included. Meta-analyses could not be conducted due to heterogeneity in the exposures and methods used to model lung function trajectories. Nine publications were eligible for inclusion of which four used group-based trajectory modelling to model lung function trajectories, while five used latent profile analysis. Studies with repeated lung function measurements over the life course identified more trajectories than others. Only one study spanning from childhood to middle age reported catch-up trajectory. The following childhood risk factors for subnormal lung function trajectories were observed in at least across two studies: low birth weight, early wheezing, asthma, allergic sensitization, eczema, allergic rhinitis, lower respiratory tract infections, family history of asthma and second-hand smoke exposure. Adult active asthma and personal cigarette smoking were observed to be associated with accelerated decline lung trajectories. Our review identified 10 risk factors associated with the growth, catch-up, reduced plateau and decline trajectories of lung function. Intervention directed at childhood asthma and infections, and tobacco smoke exposure at all ages would help promote lung health and prevent subnormal lung function trajectories.
Collapse
Affiliation(s)
- Daniel O Okyere
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Dinh S Bui
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - George R Washko
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, Massachusetts, USA.,Laboratory of Mathematics in Imaging, Harvard Medical School, Boston, Massachusetts, USA
| | - Caroline J Lodge
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia.,Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Adrian J Lowe
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia.,Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Raisa Cassim
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jennifer L Perret
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Michael J Abramson
- School of Public Health & Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - E Haydn Walters
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia.,School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Nilakshi T Waidyatillake
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Shyamali C Dharmage
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia.,Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| |
Collapse
|
32
|
Guzmán-Vargas J, Ambrocio-Ortiz E, Pérez-Rubio G, Ponce-Gallegos MA, Hernández-Zenteno RDJ, Mejía M, Ramírez-Venegas A, Buendia-Roldan I, Falfán-Valencia R. Differential Genomic Profile in TERT, DSP, and FAM13A Between COPD Patients With Emphysema, IPF, and CPFE Syndrome. Front Med (Lausanne) 2021; 8:725144. [PMID: 34490311 PMCID: PMC8416604 DOI: 10.3389/fmed.2021.725144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/22/2021] [Indexed: 12/31/2022] Open
Abstract
Background: Genetic association studies have identified single nucleotide polymorphisms (SNPs) associated with lasting lung diseases such as Chronic Obstructive Pulmonary Disease (COPD) and Idiopathic Pulmonary Fibrosis (IPF), as well as the simultaneous presentation, known as Combined Pulmonary Fibrosis and Emphysema (CPFE) Syndrome. It is unknown if these diseases share genetic variants previously described in an independent way. This study aims to identify common or differential variants between COPD, IPF, and CPFE. Materials and methods: The association analysis was carried out through a case-control design in a Mexican mestizo population (n = 828); three patients' groups were included: COPD smokers (COPD-S, n = 178), IPF patients (n = 93), and CPFE patients (n = 16). Also, two comparison groups were analyzed: smokers without COPD (SWOC, n = 367) and healthy subjects belonging to the Mexican Pulmonary Aging Cohort (PAC, n = 174). Five SNPs in four genes previously associated to interstitial and obstructive diseases were selected: rs2609255 (FAM13A), rs2736100 (TERT), rs2076295 (DSP) rs5743890, and rs111521887 (TOLLIP). Genotyping was performed by qPCR using predesigned Taqman probes. Results: In comparing IPF vs. PAC, significant differences were found in the frequency of the rs260955 G allele associated with the IPF risk (OR = 1.68, p = 0.01). Also, the genotypes, GG of rs260955 (OR = 2.86, p = 0.01) and TT of rs2076295 (OR = 1.79, p = 0.03) were associated with an increased risk of IPF; after adjusting by covariables, only the rs260955 G allele remain significant (p = 0.01). For the CPFE vs. PAC comparison, an increased CPFE risk was identified since there is a difference in the rs2736100 C allele (OR = 4.02, p < 0.01; adjusted p < 0.01). For COPD-S, the rs2609255 TG genotype was associated with increased COPD risk after adjusting by covariables. Conclusion: The rs2736100 C allele is associated with decreased IPF risk and confers an increased risk for CPFE. Also, the rs2076295 TT genotype is associated with increased IPF risk, while the GG genotype is associated with CFPE susceptibility. The rs2609255 G allele and GG genotype are associated with IPF susceptibility, while the TG genotype is present in patients with emphysema.
Collapse
Affiliation(s)
- Javier Guzmán-Vargas
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Enrique Ambrocio-Ortiz
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Gloria Pérez-Rubio
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | | | | | - Mayra Mejía
- Interstitial Pulmonary Diseases and Rheumatology Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Alejandra Ramírez-Venegas
- Tobacco Smoking and COPD Research Department, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Ivette Buendia-Roldan
- Translational Research Laboratory on Aging and Pulmonary Fibrosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Ramcés Falfán-Valencia
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| |
Collapse
|
33
|
Lin X, Li Y, Gong L, Yun JH, Xu S, Tesfaigzi Y, Qiao D, Zhou X. Tempo-spatial regulation of the Wnt pathway by FAM13A modulates the stemness of alveolar epithelial progenitors. EBioMedicine 2021; 69:103463. [PMID: 34224973 PMCID: PMC8264115 DOI: 10.1016/j.ebiom.2021.103463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/06/2021] [Accepted: 06/11/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Family with Sequence Similarity 13, Member A (FAM13A) gene has been consistently associated with COPD by Genome-wide association studies (GWAS). Our previous study demonstrated that FAM13A was mainly expressed in the lung epithelial progenitors including Club cells and alveolar type II epithelial (ATII) cells. Fam13a-/- mice were resistant to cigarette smoke (CS)-induced emphysema through promoting β-catenin/Wnt activation. Given the important roles of β-catenin/Wnt activation in alveolar regeneration during injury, it is unclear when and where FAM13A regulates the Wnt pathway, the requisite pathway for alveolar epithelial repair, in vivo during CS exposure in lung epithelial progenitors. METHODS Fam13a+/+ or Fam13a-/- mice were crossed with TCF/Lef:H2B-GFP Wnt-signaling reporter mouse line to indicate β-catenin/Wnt-activated cells labeled with GFP followed by acute (1 month) or chronic (7 months) CS exposure. Fluorescence-activated flow cytometry analysis, immunofluorescence and organoid culture system were performed to identify the β-catenin/Wnt-activated cells in Fam13a+/+ or Fam13a-/- mice exposed to CS. Fam13a;SftpcCreERT2;Rosa26RmTmG mouse line, where GFP labels ATII cells, was generated for alveolar organoid culture followed by analyses of organoid number, immunofluorescence and gene expression. Single cell RNA-seq data from COPD ever smokers and nonsmoker control lungs were further analyzed. FINDINGS We found that FAM13A-deficiency significantly increased Wnt activation mainly in lung epithelial cells. Consistently, after long-term CS exposure in vivo, FAM13A deficiency bestows alveolar epithelial progenitor cells with enhanced proliferation and differentiation in the ex vivo organoid model. Importantly, expression of FAM13A is significantly increased in human COPD-derived ATII cells compared to healthy ATII cells as suggested by single cell RNA-sequencing data. INTERPRETATION Our findings suggest that FAM13A-deficiency promotes the Wnt pathway-mediated ATII cell repair/regeneration, and thereby possibly mitigating CS-induced alveolar destruction. FUND: This project is funded by the National Institutes of Health of United States of America (NIH) grants R01HL127200, R01HL137927, R01HL148667 and R01HL147148 (XZ).
Collapse
Affiliation(s)
- Xin Lin
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yujun Li
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - Lu Gong
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jeong H Yun
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; The Division of Pulmonary and Critical Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Shuang Xu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yohannes Tesfaigzi
- The Division of Pulmonary and Critical Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Dandi Qiao
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
34
|
Zhu J, Wang F, Feng X, Li B, Ma L, Zhang J. Family with sequence similarity 13 member A mediates TGF-β1-induced EMT in small airway epithelium of patients with chronic obstructive pulmonary disease. Respir Res 2021; 22:192. [PMID: 34210319 PMCID: PMC8247231 DOI: 10.1186/s12931-021-01783-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 06/24/2021] [Indexed: 12/28/2022] Open
Abstract
Background To explore the role of family with sequence similarity 13 member A (FAM13A) in TGF-β1-induced EMT in the small airway epithelium of patients with chronic obstructive pulmonary disease (COPD). Methods Small airway wall thickness and protein levels of airway remodeling markers, EMT markers, TGF-β1, and FAM13A were measured in lung tissue samples from COPD and non-COPD patients. The correlations of FAM13A expression with COPD severity and EMT marker expression were evaluated. Gain- and loss-of-function assays were performed to explore the functions of FAM13A in cell proliferation, motility, and TGF-β1-induced EMT marker alterations in human bronchial epithelial cell line BEAS-2B. Results Independent of smoking status, lung tissue samples from COPD patients exhibited significantly increased small airway thickness and collagen fiber deposition, along with enhanced protein levels of remodeling markers (collagen I, fibronectin, and MMP-9), mesenchymal markers (α-SMA, vimentin, and N-cadherin), TGF-β1, and FAM13A, compared with those from non-COPD patients. FAM13A expression negatively correlated with FEV1% and PO2 in COPD patients. In small airway epithelium, FAM13A expression negatively correlated with E-cadherin protein levels and positively correlated with vimentin protein levels. In BEAS-2B cells, TGF-β1 dose-dependently upregulated FAM13A protein levels. FAM13A overexpression significantly promoted cell proliferation and motility in BEAS-2B cells, whereas FAM13A silencing showed contrasting results. Furthermore, FAM13A knockdown partially reversed TGF-β1-induced EMT marker protein alterations in BEAS-2B cells. Conclusions FAM13A upregulation is associated with TGF-β1-induced EMT in the small airway epithelium of COPD patients independent of smoking status, serving as a potential therapeutic target for anti-EMT therapy in COPD. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-021-01783-z.
Collapse
Affiliation(s)
- Jinyuan Zhu
- Department of Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Faxuan Wang
- School of Public Health and Management, Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Xueyan Feng
- Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Beibei Li
- Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Liqiong Ma
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan, 750004, People's Republic of China
| | - Jin Zhang
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan, 750004, People's Republic of China.
| |
Collapse
|
35
|
Szalontai K, Gémes N, Furák J, Varga T, Neuperger P, Balog JÁ, Puskás LG, Szebeni GJ. Chronic Obstructive Pulmonary Disease: Epidemiology, Biomarkers, and Paving the Way to Lung Cancer. J Clin Med 2021; 10:jcm10132889. [PMID: 34209651 PMCID: PMC8268950 DOI: 10.3390/jcm10132889] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/24/2021] [Accepted: 06/24/2021] [Indexed: 12/16/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD), the frequently fatal pathology of the respiratory tract, accounts for half a billion cases globally. COPD manifests via chronic inflammatory response to irritants, frequently to tobacco smoke. The progression of COPD from early onset to advanced disease leads to the loss of the alveolar wall, pulmonary hypertension, and fibrosis of the respiratory epithelium. Here, we focus on the epidemiology, progression, and biomarkers of COPD with a particular connection to lung cancer. Dissecting the cellular and molecular players in the progression of the disease, we aim to shed light on the role of smoking, which is responsible for the disease, or at least for the more severe symptoms and worse patient outcomes. We summarize the inflammatory conditions, as well as the role of EMT and fibroblasts in establishing a cancer-prone microenvironment, i.e., the soil for ‘COPD-derived’ lung cancer. We highlight that the major health problem of COPD can be alleviated via smoking cessation, early diagnosis, and abandonment of the usage of biomass fuels on a global basis.
Collapse
Affiliation(s)
- Klára Szalontai
- Csongrád County Hospital of Chest Diseases, Alkotmány u. 36., H6772 Deszk, Hungary;
| | - Nikolett Gémes
- Laboratory of Functional Genomics, Biological Research Centre, Temesvári krt. 62., H6726 Szeged, Hungary; (N.G.); (T.V.); (P.N.); (J.Á.B.); (L.G.P.)
- PhD School in Biology, University of Szeged, H6726 Szeged, Hungary
| | - József Furák
- Department of Surgery, University of Szeged, Semmelweis u. 8., H6725 Szeged, Hungary;
| | - Tünde Varga
- Laboratory of Functional Genomics, Biological Research Centre, Temesvári krt. 62., H6726 Szeged, Hungary; (N.G.); (T.V.); (P.N.); (J.Á.B.); (L.G.P.)
| | - Patrícia Neuperger
- Laboratory of Functional Genomics, Biological Research Centre, Temesvári krt. 62., H6726 Szeged, Hungary; (N.G.); (T.V.); (P.N.); (J.Á.B.); (L.G.P.)
- PhD School in Biology, University of Szeged, H6726 Szeged, Hungary
| | - József Á. Balog
- Laboratory of Functional Genomics, Biological Research Centre, Temesvári krt. 62., H6726 Szeged, Hungary; (N.G.); (T.V.); (P.N.); (J.Á.B.); (L.G.P.)
- PhD School in Biology, University of Szeged, H6726 Szeged, Hungary
| | - László G. Puskás
- Laboratory of Functional Genomics, Biological Research Centre, Temesvári krt. 62., H6726 Szeged, Hungary; (N.G.); (T.V.); (P.N.); (J.Á.B.); (L.G.P.)
- Avicor Ltd. Alsó Kikötő sor 11/D, H6726 Szeged, Hungary
| | - Gábor J. Szebeni
- Laboratory of Functional Genomics, Biological Research Centre, Temesvári krt. 62., H6726 Szeged, Hungary; (N.G.); (T.V.); (P.N.); (J.Á.B.); (L.G.P.)
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, H6726 Szeged, Hungary
- CS-Smartlab Devices Ltd., Ady E. u. 14., H7761 Kozármisleny, Hungary
- Correspondence:
| |
Collapse
|
36
|
Zhang PD, Zhang XR, Zhang A, Li ZH, Liu D, Zhang YJ, Mao C. Associations of genetic risk and smoking with incident chronic obstructive pulmonary disease. Eur Respir J 2021; 59:13993003.01320-2021. [PMID: 34172472 DOI: 10.1183/13993003.01320-2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 06/14/2021] [Indexed: 11/05/2022]
Abstract
BACKGROUND Genetic and smoking contribute to chronic obstructive pulmonary disease (COPD), but whether a combined polygenic risk score (PRS) is associated with incident COPD and whether it has a synergistic effect on the smoking remains unclear. We aimed to investigate the association of PRS with COPD and explore whether smoking behaviors could modify such association. METHODS Multivariable Cox proportional models were used to estimate hazard ratios (HRs) and 95% confidence intervals (95% CIs) for the association of the PRS and smoking with COPD. RESULTS The study included 439 255 participants (mean age 56.5; 53.9% female), with a median follow-up of 9.0 years. The PRSlasso containing 2.5 million variants showed better discrimination and a stronger association for incident COPD than the PRS279 containing 279 genome-wide significance variants. Compared with the low genetic risk, the HRs of the medium and high genetic risk were 1.39 (95% CI, 1.31-1.48) and 2.40 (95% CI, 2.24-2.56), respectively. The HR of high genetic risk and current smoking was 11.62 (95% CI, 10.31-13.10) times of low genetic risk and never smoking. There were significant interactions between the PRSlasso and smoking status for incident COPD (p for interaction<0.001). From low genetic risk to high genetic risk, the HRs of current smoking increased from 4.32 (95% CI, 3.69-5.06) to 6.89 (95% CI, 6.21-7.64), and the population-attributable risks of smoking increased from 42.7% to 61.1%. CONCLUSION PRS constructed from millions of variants below genome-wide significance showed significant associations with incident COPD. Participants with a high genetic risk may be more susceptible to developing COPD when exposed to smoking.
Collapse
Affiliation(s)
- Pei-Dong Zhang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Pei-Dong Zhang and Xi-Ru Zhang contributed to the work equally
| | - Xi-Ru Zhang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.,Pei-Dong Zhang and Xi-Ru Zhang contributed to the work equally
| | - Ao Zhang
- State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| | - Zhi-Hao Li
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Dan Liu
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu-Jie Zhang
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Chen Mao
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China .,Department of Laboratory Medicine, Microbiome Medicine Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
37
|
Huo X, Jin S, Wang Y, Ma L. DNA methylation in chronic obstructive pulmonary disease. Epigenomics 2021; 13:1145-1155. [PMID: 34142873 DOI: 10.2217/epi-2021-0111] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD), a complex disease with polygenetic tendency, is one of the most important health problems in the world. Recently, in the study of the pathogenesis of the COPD, epigenetic changes caused by environmental factors, such as DNA methylation, started to attract more attention than genetic factors. In this review, we discuss the main features of DNA methylation, such as DNA methyltransferases and the methylation sites that modulate the DNA methylation level, and their roles in COPD progression. Finally, to promote new ideas for the prevention and treatment of COPD, we focus on the potential of DNA methylation as a COPD therapeutic target.
Collapse
Affiliation(s)
- XinXin Huo
- School of Public Health, Lanzhou University, Lanzhou, China
| | - SiHui Jin
- School of Public Health, Lanzhou University, Lanzhou, China
| | - YiGe Wang
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Li Ma
- School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|
38
|
Tam A, Leclair P, Li LV, Yang CX, Li X, Witzigmann D, Kulkarni JA, Hackett TL, Dorscheid DR, Singhera GK, Hogg JC, Cullis PR, Sin DD, Lim CJ. FAM13A as potential therapeutic target in modulating TGF-β-induced airway tissue remodeling in COPD. Am J Physiol Lung Cell Mol Physiol 2021; 321:L377-L391. [PMID: 34105356 DOI: 10.1152/ajplung.00477.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Genome-wide association studies have shown that a gene variant in the Family with sequence similarity 13, member A (FAM13A) is strongly associated with reduced lung function and the appearance of respiratory symptoms in patients with chronic obstructive pulmonary disease (COPD). A key player in smoking-induced tissue injury and airway remodeling is the transforming growth factor-β1 (TGF-β1). To determine the role of FAM13A in TGF-β1 signaling, FAM13A-/- airway epithelial cells were generated using CRISPR-Cas9, whereas overexpression of FAM13A was achieved using lipid nanoparticles. Wild-type (WT) and FAM13A-/- cells were treated with TGF-β1, followed by gene and/or protein expression analyses. FAM13A-/- cells augmented TGF-β1-induced increase in collagen type 1 (COL1A1), matrix metalloproteinase 2 (MMP2), expression compared with WT cells. This effect was mediated by an increase in β-catenin (CTNNB1) expression in FAM13A-/- cells compared with WT cells after TGF-β1 treatment. FAM13A overexpression was partially protective from TGF-β1-induced COL1A1 expression. Finally, we showed that airway epithelial-specific FAM13A protein expression is significantly increased in patients with severe COPD compared with control nonsmokers, and negatively correlated with lung function. In contrast, β-catenin (CTNNB1), which has previously been linked to be regulated by FAM13A, is decreased in the airway epithelium of smokers with COPD compared with non-COPD subjects. Together, our data showed that FAM13A may be protective from TGF-β1-induced fibrotic response in the airway epithelium via sequestering CTNNB1 from its regulation on downstream targets. Therapeutic increase in FAM13A expression in the airway epithelium of smokers at risk for COPD, and those with mild COPD, may reduce the extent of airway tissue remodeling.
Collapse
Affiliation(s)
- Anthony Tam
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.,Center for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Pascal Leclair
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ling Vicky Li
- Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Chen X Yang
- Center for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Xuan Li
- Center for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Dominik Witzigmann
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada.,NanoMedicines Innovation Network, Vancouver, British Columbia, Canada
| | - Jayesh A Kulkarni
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada.,NanoMedicines Innovation Network, Vancouver, British Columbia, Canada
| | - Tillie-Louise Hackett
- Center for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Delbert R Dorscheid
- Center for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Gurpreet K Singhera
- Center for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - James C Hogg
- Center for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada.,NanoMedicines Innovation Network, Vancouver, British Columbia, Canada
| | - Don D Sin
- Center for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Chinten James Lim
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
39
|
Samaha E, Vierlinger K, Weinhappel W, Godnic-Cvar J, Nöhammer C, Koczan D, Thiesen HJ, Yanai H, Fraifeld VE, Ziesche R. Expression Profiling Suggests Loss of Surface Integrity and Failure of Regenerative Repair as Major Driving Forces for Chronic Obstructive Pulmonary Disease Progression. Am J Respir Cell Mol Biol 2021; 64:441-452. [PMID: 33524306 DOI: 10.1165/rcmb.2020-0270oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) poses a major risk for public health, yet remarkably little is known about its detailed pathophysiology. Definition of COPD as nonreversible pulmonary obstruction revealing more about spatial orientation than about mechanisms of pathology may be a major reason for this. We conducted a controlled observational study allowing for simultaneous assessment of clinical and biological development in COPD. Sixteen healthy control subjects and 104 subjects with chronic bronchitis, with or without pulmonary obstruction at baseline, were investigated. Using both the extent of and change in bronchial obstruction as main scoring criteria for the analysis of gene expression in lung tissue, we identified 410 genes significantly associated with progression of COPD. One hundred ten of these genes demonstrated a distinctive expression pattern, with their functional annotations indicating participation in the regulation of cellular coherence, membrane integrity, growth, and differentiation, as well as inflammation and fibroproliferative repair. The regulatory pattern indicates a sequentially unfolding pathology that centers on a two-step failure of surface integrity commencing with a loss of epithelial coherence as early as chronic bronchitis. Decline of regenerative repair starting in Global Initiative for Chronic Obstructive Lung Disease stage I then activates degradation of extracellular-matrix hyaluronan, causing structural failure of the bronchial wall that is only resolved by scar formation. Although they require independent confirmation, our findings provide the first tangible pathophysiological concept of COPD to be further explored.Clinical trial registered with www.clinicaltrials.gov (NCT00618137).
Collapse
Affiliation(s)
- Eslam Samaha
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Klemens Vierlinger
- Department of Health and Environment, Austrian Institute of Technology, Vienna, Austria
| | - Wolfgang Weinhappel
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Jasminka Godnic-Cvar
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Christa Nöhammer
- Department of Health and Environment, Austrian Institute of Technology, Vienna, Austria
| | - Dirk Koczan
- Department of Immunology, University of Rostock, Rostock, Germany; and
| | | | - Hagai Yanai
- Faculty of Health Sciences, Beer-Sheva Campus, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Vadim E Fraifeld
- Faculty of Health Sciences, Beer-Sheva Campus, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Rolf Ziesche
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
40
|
Ziółkowska-Suchanek I, Podralska M, Żurawek M, Łaczmańska J, Iżykowska K, Dzikiewicz-Krawczyk A, Rozwadowska N. Hypoxia-Induced FAM13A Regulates the Proliferation and Metastasis of Non-Small Cell Lung Cancer Cells. Int J Mol Sci 2021; 22:ijms22094302. [PMID: 33919074 PMCID: PMC8122400 DOI: 10.3390/ijms22094302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/13/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022] Open
Abstract
Hypoxia in non-small cell lung cancer (NSCLC) affects cancer progression, metastasis and metabolism. We previously showed that FAM13A was induced by hypoxia in NSCLC but the biological function of this gene has not been fully elucidated. This study aimed to investigate the role of hypoxia-induced FAM13A in NSCLC progression and metastasis. Lentiviral shRNAs were used for FAM13A gene silencing in NSCLC cell lines (A549, CORL-105). MTS assay, cell tracking VPD540 dye, wound healing assay, invasion assay, BrdU assay and APC Annexin V staining assays were performed to examine cell proliferation ability, migration, invasion and apoptosis rate in NSCLC cells. The results of VPD540 dye and MTS assays showed a significant reduction in cell proliferation after FAM13A knockdown in A549 cells cultured under normal and hypoxia (1% O2) conditions (p < 0.05), while the effect of FAM13A downregulation on CORL-105 cells was observed after 96 h exposition to hypoxia. Moreover, FAM13A inhibition induced S phase cell cycle arrest in A549 cells under hypoxia conditions. Silencing of FAM13A significantly suppressed migration of A549 and CORL-105 cells in both oxygen conditions, especially after 72 and 96 h (p < 0.001 in normoxia, p < 0.01 after hypoxia). It was showed that FAM13A reduction resulted in disruption of the F-actin cytoskeleton altering A549 cell migration. Cell invasion rates were significantly decreased in A549 FAM13A depleted cells compared to controls (p < 0.05), mostly under hypoxia. FAM13A silencing had no effect on apoptosis induction in NSCLC cells. In the present study, we found that FAM13A silencing has a negative effect on proliferation, migration and invasion activity in NSCLC cells in normal and hypoxic conditions. Our data demonstrated that FAM13A depleted post-hypoxic cells have a decreased cell proliferation ability and metastatic potential, which indicates FAM13A as a potential therapeutic target in lung cancer.
Collapse
|
41
|
DeMeo DL. Sex and Gender Omic Biomarkers in Men and Women With COPD: Considerations for Precision Medicine. Chest 2021; 160:104-113. [PMID: 33745988 DOI: 10.1016/j.chest.2021.03.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/25/2021] [Accepted: 03/08/2021] [Indexed: 11/17/2022] Open
Abstract
Sex and gender differences in lung health and disease are imperative to consider and study if precision pulmonary medicine is to be achieved. The development of reliable COPD biomarkers has been elusive, and the translation of biomarkers to clinical care has been limited. Useful and effective biomarkers must be developed with attention to clinical heterogeneity of COPD; inherent heterogeneity exists related to grouping women and men together in the studies of COPD. Considering sex and gender differences and influences related to -omics may represent progress in susceptibility, diagnostic, prognostic, and therapeutic biomarker development and clinical innovation to improve the lung health of men and women.
Collapse
Affiliation(s)
- Dawn L DeMeo
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
42
|
Hu Y, Ciminieri C, Hu Q, Lehmann M, Königshoff M, Gosens R. WNT Signalling in Lung Physiology and Pathology. Handb Exp Pharmacol 2021; 269:305-336. [PMID: 34463851 DOI: 10.1007/164_2021_521] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The main physiological function of the lung is gas exchange, mediated at the interface between the alveoli and the pulmonary microcapillary network and facilitated by conducting airway structures that regulate the transport of these gases from and to the alveoli. Exposure to microbial and environmental factors such as allergens, viruses, air pollution, and smoke contributes to the development of chronic lung diseases such as asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), and lung cancer. Respiratory diseases as a cluster are the commonest cause of chronic disease and of hospitalization in children and are among the three most common causes of morbidity and mortality in the adult population worldwide. Many of these chronic respiratory diseases are associated with inflammation and structural remodelling of the airways and/or alveolar tissues. They can often only be treated symptomatically with no disease-modifying therapies that normalize the pathological tissue destruction driven by inflammation and remodelling. In search for novel therapeutic strategies for these diseases, several lines of evidence revealed the WNT pathway as an emerging target for regenerative strategies in the lung. WNT proteins, their receptors, and signalling effectors have central regulatory roles under (patho)physiological conditions underpinning lung function and (chronic) lung diseases and we summarize these roles and discuss how pharmacological targeting of the WNT pathway may be utilized for the treatment of chronic lung diseases.
Collapse
Affiliation(s)
- Yan Hu
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, CO, USA
| | - Chiara Ciminieri
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Aurora, CO, USA.,Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands
| | - Qianjiang Hu
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Munich, Germany
| | - Mareike Lehmann
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Munich, Germany
| | - Melanie Königshoff
- Lung Repair and Regeneration Unit, Helmholtz-Zentrum Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, Munich, Germany. .,Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
43
|
Lee YJ, Choi S, Kwon SY, Lee Y, Lee JK, Heo EY, Chung HS, Kim DK. A Genome-Wide Association Study in Early COPD: Identification of One Major Susceptibility Loci. Int J Chron Obstruct Pulmon Dis 2020; 15:2967-2975. [PMID: 33235445 PMCID: PMC7680157 DOI: 10.2147/copd.s269263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/23/2020] [Indexed: 12/20/2022] Open
Abstract
Background Identifying the genetic basis of airflow limitation is one of the most interesting issues for understanding chronic obstructive pulmonary disease (COPD) pathophysiology. Several studies have shown that some genetic variants associated with COPD have been identified in genome-wide association study (GWAS), especially in patients with moderate to severe COPD; genetic susceptibility for airflow limitation in the early COPD phase has not been widely studied. Objective We investigated the genetic variants in early COPD. Methods The present study analyzed Gene-environment interaction and phenotype (GENIE) cohort that included participants who received health screening examination. The association between single nucleotide polymorphism (SNP) and susceptibility to early COPD (FEV1 predicted ≥50% and FEV1/FVC <0.7) was tested. Results A total of 130 patients with early COPD and 3478 controls (1700 ever smokers and 1778 never smokers) were recruited. When compared with the total controls, certain SNPs (rs2818103, rs875033, rs9354627, rs34552148) on chromosome 6 were included at the top of our list (p= 5.6 × 10–7 ~9.6 × 10–6) although they did not reach genome-wide significance. When compared with the never smoker controls, two SNPs (rs2857210, rs2621419) of the HLA-DQB2 gene class were persistently associated with susceptibility to early COPD. Conclusion Certain SNPs located on chromosome 6 or the HLA-DQB2 gene were the top-scoring SNPs for the association with susceptibility to early COPD in the Korean GENIE cohort.
Collapse
Affiliation(s)
- Ye-Jin Lee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Hallym University Kangdong Sacred Heart Hospital, Seoul, Korea
| | - SeungHo Choi
- Department of Internal Medicine, Healthcare Research Institute, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul 135-984 Korea
| | - Sung-Youn Kwon
- Department of Internal Medicine, Healthcare Research Institute, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul 135-984 Korea
| | - Yunhwan Lee
- Department of Internal Medicine, Healthcare Research Institute, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul 135-984 Korea
| | - Jung Kyu Lee
- Division of Pulmonary and Critical Care Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea
| | - Eun Young Heo
- Division of Pulmonary and Critical Care Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea
| | - Hee Soon Chung
- Division of Pulmonary and Critical Care Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Deog Kyeom Kim
- Division of Pulmonary and Critical Care Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
44
|
Montesi SB, Fisher JH, Martinez FJ, Selman M, Pardo A, Johannson KA. Update in Interstitial Lung Disease 2019. Am J Respir Crit Care Med 2020; 202:500-507. [PMID: 32412784 DOI: 10.1164/rccm.202002-0360up] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Sydney B Montesi
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jolene H Fisher
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Moisés Selman
- Instituto Nacional de Enfermedades Respiratorias, Ismael Cosío Villegas, Mexico City, Mexico
| | - Annie Pardo
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico; and
| | - Kerri A Johannson
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
45
|
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: 29] [Impact Index Per Article: 7.3] [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
|
46
|
Ahn HJ. Anesthetic management of patients with chronic obstructive pulmonary disease. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2020. [DOI: 10.5124/jkma.2020.63.9.532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the third most common cause of death worldwide. It has a prevalence of 14% among Koreans aged above 40 years and a prevalence of 31% among those aged above 65 years. However, only 6% of the COPD patients receive treatment. Most of the patients do not seek medical attention, as they think that dyspnea, cough, and productive sputum, which are the common symptoms of COPD, are normal aging phenomena. Smoking is a major risk factor for COPD, but environmental hazards and genetic susceptibility are also involved. With aging, lung injuries due to these risk factors accumulate, leading to increased prevalence of COPD. The major concerns regarding perioperative management of COPD patients include preoperative evaluation of cardiopulmonary risks, optimization of lung function, and evaluation of COPD-related physiological functions that are easily aggravated during anesthesia. These include respiratory muscle dysfunction, dynamic hyperinflation and auto-positive end-expiratory pressure, hypoxia-hypercarbia, and pulmonary hypertension-associated heart failure. Therefore, anesthesia for COPD patients should focus on preoperative evaluation, risk reduction measures, and prevention of postoperative pulmonary complications.
Collapse
|
47
|
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: 3.5] [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
|
48
|
He LX, Tang ZH, Huang QS, Li WH. DNA Methylation: A Potential Biomarker of Chronic Obstructive Pulmonary Disease. Front Cell Dev Biol 2020; 8:585. [PMID: 32733890 PMCID: PMC7358425 DOI: 10.3389/fcell.2020.00585] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a serious public health concern worldwide. By 2040, 4.41 million people are estimated to expire annually due to COPD. However, till date, it has remained difficult to alter the activity or progress of the disease through treatment. In order to address this issue, the best way would be to find biomarkers and new therapeutic targets for COPD. DNA methylation (DNAm) may be a potential biomarker for disease prevention, diagnosis, and prognosis, and its reversibility further makes it a potential drug design target in COPD. In this review, we aimed to explore the role of DNAm as biomarkers and disease mediators in different tissue samples from patients with COPD.
Collapse
Affiliation(s)
- Lin-Xi He
- School of Basic Medicine Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhao-Hui Tang
- School of Basic Medicine Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qing-Song Huang
- Department of Respiratory, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei-Hong Li
- School of Basic Medicine Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
49
|
Wu Y, Guan S, Ge Y, Yang Y, Cao Y, Zhou J. Cigarette smoke promotes chronic obstructive pulmonary disease (COPD) through the miR-130a/Wnt1 axis. Toxicol In Vitro 2020; 65:104770. [PMID: 31935487 DOI: 10.1016/j.tiv.2020.104770] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 12/18/2019] [Accepted: 01/09/2020] [Indexed: 12/15/2022]
Abstract
Cigarette smoke (CS) is a crucial factor in chronic obstructive pulmonary disease (COPD). Wnt/β-catenin signaling deregulation may further contribute to COPD progression. The deregulation and dysfunction of miRNAs in COPD have been reported. Investigating the deregulated miRNAs and their potential role in COPD progression may provide novel strategies for COPD treatment. In the present study, we analyzed significantly differentially-expressed miRNAs in COPD according to GSE44531 and miR-130a was selected. We revealed the upregulation of miR-130a in COPD, both in cigarette smoke extract (CSE)-treated BEAS-2B cells and CS-exposed mice. MiR-130a negatively regulated three critical factors in Wnt/β-catenin signaling, Wnt1, β-Catenin, and LEF1. MiR-130a inhibition rescued CSE-blocked activation of Wnt/β-catenin signaling in vitro. MiR-130a targets WNT1 3'UTR to inhibit its expression. Moreover, in CSE-stimulated BEAS-2B cells, miR-130a overexpression aggravated, while miR-130a inhibition partially attenuated CSE-caused suppression on cell migration and proliferation. MiR-130a aggravates CSE-induced cellular injury in BEAS-2B cells by targeting Wnt signaling. In summary, miR-130a has a pathogenetic role in CS-induced COPD and regulates Wnt/β-catenin signaling via targeting Wnt1. Our findings indicate that miR-130a is a potential therapeutic target for the treatment of CS-induced COPD.
Collapse
Affiliation(s)
- Yudi Wu
- Department of Respiratory, The Third Affiliation Hospital of Soochow University, Changzhou, Jiangsu 213000, China
| | - Shuhong Guan
- Department of Respiratory, The Third Affiliation Hospital of Soochow University, Changzhou, Jiangsu 213000, China
| | - Yunqi Ge
- Department of Respiratory, The Third Affiliation Hospital of Soochow University, Changzhou, Jiangsu 213000, China
| | - Yun Yang
- Department of Respiratory, The Third Affiliation Hospital of Soochow University, Changzhou, Jiangsu 213000, China
| | - Yi Cao
- Department of Respiratory, The Third Affiliation Hospital of Soochow University, Changzhou, Jiangsu 213000, China
| | - Jun Zhou
- Department of Respiratory, The Third Affiliation Hospital of Soochow University, Changzhou, Jiangsu 213000, China.
| |
Collapse
|
50
|
Gim J, An J, Sung J, Silverman EK, Cho MH, Won S. A Between Ethnicities Comparison of Chronic Obstructive Pulmonary Disease Genetic Risk. Front Genet 2020; 11:329. [PMID: 32373161 PMCID: PMC7187688 DOI: 10.3389/fgene.2020.00329] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 03/20/2020] [Indexed: 12/13/2022] Open
Abstract
Heterogeneity of lung function levels and risk for developing chronic obstructive pulmonary disease (COPD) among people exposed to the same environmental risk factors, such as cigarette smoking, suggest an important role of genetic factors in COPD susceptibility. To investigate the possible role of different genetic factors in COPD susceptibility across ethnicities. We used a population-stratified analysis for: (i) identifying ethnic-specific genetic susceptibility loci, (ii) developing ethnic-specific polygenic risk prediction models using those SNPs, and (iii) validating the models with an independent dataset. We elucidated substantial differences in SNP heritability and susceptibility loci for the disease across ethnicities. Furthermore, the application of three ethnic-specific prediction models to an independent dataset showed that the best performance is achieved when the prediction model is applied to a dataset with the matched ethnic sample. Our study validates the necessity of considering ethnic differences in COPD risk; understanding these differences might help in preventing COPD and developing therapeutic strategies.
Collapse
Affiliation(s)
- Jungsoo Gim
- Department of Biomedical Science, Chosun University, Gwangju, South Korea
| | - Jaehoon An
- Graduate School of Public Health, Seoul National University, Seoul, South Korea
| | - Joohon Sung
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, South Korea.,Interdisciplinary Program of Bioinformatics, Seoul National University, Seoul, South Korea.,Institute of Health and Environment, Seoul National University, Seoul, South Korea
| | - Edwin K Silverman
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Michael H Cho
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Sungho Won
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, South Korea.,Interdisciplinary Program of Bioinformatics, Seoul National University, Seoul, South Korea.,Institute of Health and Environment, Seoul National University, Seoul, South Korea
| |
Collapse
|