1
|
Sarı-Tunel F, Demirkan A, Vural B, Yıldız CE, Komurcu-Bayrak E. Omics Data Integration Uncovers mRNA-miRNA Interaction Regions in Genes Associated with Chronic Venous Insufficiency. Genes (Basel) 2024; 16:40. [PMID: 39858587 PMCID: PMC11765502 DOI: 10.3390/genes16010040] [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: 11/15/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Chronic venous insufficiency (CVI), a chronic vascular dysfunction, is a common health problem that causes serious complications such as painful varicose veins and even skin ulcers. Identifying the underlying genetic and epigenetic factors is important for improving the quality of life of individuals with CVI. In the literature, many genes, variants, and miRNAs associated with CVI have been identified through genomic and transcriptomic studies. Despite molecular pathogenesis studies, how the genes associated with CVI are regulated by miRNAs and the effect of variants in binding regions on expression levels are still not fully understood. In this study, previously identified genes, variants, and miRNAs associated with CVI, common variants in the mRNA-miRNA binding regions, were investigated using in silico analyses. Methods: For this purpose, miRNA research tools, MBS (miRNA binding site) database, genome browsers, and the eQTL Calculator in the GTEx portal were used. Results: We identified SNVs associated with CVI that may play a direct role in the miRNA-mediated regulation of the ZNF664, COL1A2, HFE, MDN, MTHFR, SRPX, TDRD5, TSPYL4, VEGFA, and APOE genes. In addition, when the common SNVs in the mRNA binding region of 75 unique CVI related-miRNAs in five candidate genes associated with CVI were examined, seven miRNAs associated with the expression profiles of ABCA1, PIEZO1, and CASZ1 genes were identified. Conclusions: In conclusion, the relationship between genetic markers identified in the literature that play a role in the pathogenesis of the CVI and the expression profiles was evaluated for the first time in the mRNA-miRNA interaction axis.
Collapse
Affiliation(s)
- Fatma Sarı-Tunel
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, 34093 Istanbul, Turkey; (F.S.-T.); (B.V.)
- Graduate School Institute of Health Sciences, Istanbul University, 34093 Istanbul, Turkey
| | - Ayse Demirkan
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, School of Biosciences and Medicine and People-Centred AI Institute, University of Surrey, Guildford GU2 7XH, UK
| | - Burcak Vural
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, 34093 Istanbul, Turkey; (F.S.-T.); (B.V.)
| | - Cenk Eray Yıldız
- Department of Cardiovascular Surgery, Institute of Cardiology, Istanbul University-Cerrahpasa, 34098 Istanbul, Turkey;
| | - Evrim Komurcu-Bayrak
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, 34093 Istanbul, Turkey;
| |
Collapse
|
2
|
Mol MO, van Ham TJ, Bannink N, Bruggenwirth HT, Escher JC, Kros JM, Renkens JJM, van Unen L, Verdijk RM, Vlot J, Verhoeven VJM, Demirdas S. Biallelic and monoallelic variants in EFEMP1 can cause a severe and distinct subtype of heritable connective tissue disorder. Eur J Hum Genet 2024; 32:1567-1573. [PMID: 39367272 DOI: 10.1038/s41431-024-01692-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/15/2024] [Accepted: 09/10/2024] [Indexed: 10/06/2024] Open
Abstract
Variants in EFEMP1, encoding Fibulin-3, were previously reported as a rare cause of heritable connective tissue disorder (HCTD) with recurrent hernias and joint hypermobility. We report three new cases with biallelic or monoallelic EFEMP1 variants and severe hernia phenotypes. Two male siblings of 10 and 13 years old presented with marfanoid habitus, recurrent inguinal and umbilical hernias, generalized joint hypermobility, and scoliosis. Parents and halfsiblings reported joint hypermobility and umbilical hernias. The eldest boy died at age 16 from incarcerated gastrointestinal herniation complicated by gastric and bowel necrosis with perforation. Autopsy revealed widespread intestinal diverticula. Immunohistochemistry of skin and fascia tissue did not reveal any abnormalities, including normal staining of elastic fibers. Both siblings harbored compound heterozygous likely pathogenic EFEMP1 variants (c.1320 + 2T > A, p.? and c.698G > A, p.Gly233Asp). An unrelated 58-year-old male had marfanoid features, high myopia, recurrent diaphragmatic and inguinal hernias, and chronic gastrointestinal dilatation with severe malabsorption. Both his dizygotic twin-brother and mother had recurrent hernias and high myopia. This man died at 59 years of age, and autopsy showed extensive diaphragmatic herniation, bowel diverticula, and pulmonary emphysema. A heterozygous EFEMP1 splice-variant (c.81 + 1G > A, p.?) was identified, causing exon skipping leading to a start-loss. Targeted genome reanalysis nor RNA-sequencing revealed a second variant at the other allele. The reported individuals expand the clinical and pathological phenotypes of EFEMP1-related disease, a distinct entity within the spectrum of HCTD. The severe and recurrent hernias, gastrointestinal dilatation, and diverticulosis result in an increased risk for life-threatening complications, demanding early recognition and close monitoring.
Collapse
Affiliation(s)
- M O Mol
- Department of Clinical Genetics, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - T J van Ham
- Department of Clinical Genetics, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - N Bannink
- Department of Pediatrics, Franciscus Gasthuis and Vlietland, Rotterdam and Schiedam, The Netherlands
| | - H T Bruggenwirth
- Department of Clinical Genetics, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J C Escher
- Department of Pediatric Gastroenterology, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J M Kros
- Department of Pathology, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J J M Renkens
- Department of Orthopedics and Sportsmedicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - L van Unen
- Department of Clinical Genetics, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - R M Verdijk
- Department of Pathology, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J Vlot
- Department of Pediatric Surgery, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - V J M Verhoeven
- Department of Clinical Genetics, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Ophthalmology, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - S Demirdas
- Department of Clinical Genetics, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
3
|
Diaz JA, Gianesini S, Khalil RA. Glycocalyx disruption, endothelial dysfunction and vascular remodeling as underlying mechanisms and treatment targets of chronic venous disease. INT ANGIOL 2024; 43:563-590. [PMID: 39873224 PMCID: PMC11839207 DOI: 10.23736/s0392-9590.24.05339-2] [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] [Indexed: 01/30/2025]
Abstract
The glycocalyx is an essential structural and functional component of endothelial cells. Extensive hemodynamic changes cause endothelial glycocalyx disruption and vascular dysfunction, leading to multiple arterial and venous disorders. Chronic venous disease (CVD) is a common disorder of the lower extremities with major health and socio-economic implications, but complex pathophysiology. Genetic aberrations accentuated by environmental factors, behavioral tendencies, and hormonal disturbances promote venous reflux, valve incompetence, and venous blood stasis. Increased venous hydrostatic pressure and changes in shear-stress cause glycocalyx injury, endothelial dysfunction, secretion of adhesion molecules, leukocyte recruitment/activation, and release of cytokines, chemokines, and hypoxia-inducible factor, causing smooth muscle cell switch from contractile to synthetic proliferative phenotype, imbalance in matrix metalloproteinases (MMPs), degradation of collagen and elastin, and venous tissue remodeling, leading to venous dilation and varicose veins. In the advanced stages of CVD, leukocyte infiltration of the vein wall causes progressive inflammation, fibrosis, disruption of junctional proteins, accumulation of tissue metabolites and reactive oxygen and nitrogen species, and iron deposition, leading to skin changes and venous leg ulcer (VLU). CVD management includes compression stockings, venotonics, and surgical intervention. In addition to its antithrombotic and fibrinolytic properties, literature suggests sulodexide benefits in reducing inflammation, promoting VLU healing, improving endothelial function, exhibiting venotonic properties, and inhibiting MMP-9. Understanding the role of glycocalyx, endothelial dysfunction, and vascular remodeling should help delineate the underlying mechanisms and develop improved biomarkers and targeted therapy for CVD and VLU.
Collapse
Affiliation(s)
- Jose A. Diaz
- Division of Surgical Research, Light Surgical Research and Training Laboratory, Surgical Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sergio Gianesini
- Vascular Diseases Center, Translational Surgery Unit, University of Ferrara, Ferrara, Italy, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD
| | - Raouf A. Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
4
|
Chang R, Wang C, Kong X, Li W, Wu J. Retracted article: The role of second generation sequencing technology and nanomedicine in the monitoring and treatment of lower extremity deep vein thrombosis susceptibility genes. Bioengineered 2024; 15:2003926. [PMID: 34787073 PMCID: PMC10826625 DOI: 10.1080/21655979.2021.2003926] [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: 08/26/2021] [Revised: 11/03/2021] [Accepted: 11/03/2021] [Indexed: 10/19/2022] Open
Abstract
Rong Chang, Chunsheng Wang, Xiangqi Kong, Wenhui Li and Jinchun Wu. The role of second generation sequencing technology and nanomedicine in the monitoring and treatment of lower extremity deep vein thrombosis susceptibility genes. Bioengineered. 2021 Nov. doi: 10.1080/21655979.2021.2003926.Since publication, significant concerns have been raised about the compliance with ethical policies for human research and the integrity of the data reported in the article.When approached for an explanation, the authors provided some original data but were not able to provide all the necessary supporting information. As verifying the validity of published work is core to the scholarly record's integrity, we are retracting the article. All authors listed in this publication have been informed.We have been informed in our decision-making by our editorial policies and the COPE guidelines.The retracted article will remain online to maintain the scholarly record, but it will be digitally watermarked on each page as 'Retracted.'
Collapse
Affiliation(s)
- Rong Chang
- Department of Cardiovascular Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong Province, China
- Department of Cardiovascular Medicine, Longhua Hospital Affiliated to Guangdong Medical University, Shenzhen, Guangdong Province, China
| | - Chunsheng Wang
- Department of Cardiovascular Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong Province, China
- Department of Cardiovascular Medicine, Longhua Hospital Affiliated to Guangdong Medical University, Shenzhen, Guangdong Province, China
| | - Xiangqi Kong
- Department of Cardiovascular Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong Province, China
- Department of Cardiovascular Medicine, Longhua Hospital Affiliated to Guangdong Medical University, Shenzhen, Guangdong Province, China
| | - Wenhui Li
- Department of Cardiovascular Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong Province, China
- Department of Cardiovascular Medicine, Longhua Hospital Affiliated to Guangdong Medical University, Shenzhen, Guangdong Province, China
| | - Jinchun Wu
- Department of Cardiovascular Medicine, Qinghai Provincial People’s Hospital, Xining, Qinghai Province, China
| |
Collapse
|
5
|
Shen K, Zhu F, Cheng C, Yu H. Exploring the causal relationship between plasma lipids and varicose veins of lower extremity: A comprehensive two-sample Mendelian randomization study. Medicine (Baltimore) 2024; 103:e39514. [PMID: 39252276 PMCID: PMC11383461 DOI: 10.1097/md.0000000000039514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 08/09/2024] [Indexed: 09/11/2024] Open
Abstract
Varicose veins of the lower extremities (VVs) is a common chronic vascular disease, with high prevalence rates in some countries; however, their pathogenesis remains unclear. Some studies have identified associations between changes in specific plasma lipid molecules, such as phosphatidylethanolamine (PE), phosphatidylcholine (PC), and sphingomyelin (SM), and the onset of VVs, but due to confounders and reverse causality, the causal relationship remains unclear. Meanwhile, studies on the potential link between other plasma lipids beyond PE, PC, and SM and the risk of VVs in the lower extremities are lacking. This study aimed to explore the potential causal relationship between VVs and plasma lipid levels to provide theoretical insights into the interrelation of plasma lipids and VVs in their occurrence and progression. We conducted a two-sample Mendelian randomization (MR) analysis to assess the potential connection between genetically predicted levels of individual plasma lipids and the risk of developing VVs. We utilized data from a large-scale genome-wide association study involving 7174 Finnish individuals for 179 plasma lipidomes along with VVs genome-wide association study data from 408,455 UK individuals. MR analysis employed methods, such as inverse-variance weighting, weighted median, Bayesian Weighted Mendelian Randomization, and MR-Egger regression. The inverse-variance weighting method was primarily used to assess causality. The validity of the results was demonstrated through sensitivity analysis. In total, 12 lipids were found to have their plasma levels associated with an increased risk of VVs. This includes 3 types of PE, 7 types of PC, and 2 types of phosphatidylinositol. However, no significant causal relationship was found between the plasma levels of 11 types of SM and VVs. These results support the existence of a potential causal relationship between specific types of lipid levels and the risk of VVs, which can provide clues for further studies on biological mechanisms and the exploration of potential therapeutic targets.
Collapse
Affiliation(s)
- Kailin Shen
- Department of Intervention, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Fangtao Zhu
- Department of Intervention, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Cunwei Cheng
- Department of Intervention, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Haibin Yu
- Department of Intervention, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| |
Collapse
|
6
|
Zhang DD, He XY, Yang L, Wu BS, Fu Y, Liu WS, Guo Y, Fei CJ, Kang JJ, Feng JF, Cheng W, Tan L, Yu JT. Exome sequencing identifies novel genetic variants associated with varicose veins. PLoS Genet 2024; 20:e1011339. [PMID: 38980841 PMCID: PMC11233024 DOI: 10.1371/journal.pgen.1011339] [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: 09/30/2023] [Accepted: 06/13/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Varicose veins (VV) are one of the common human diseases, but the role of genetics in its development is not fully understood. METHODS We conducted an exome-wide association study of VV using whole-exome sequencing data from the UK Biobank, and focused on common and rare variants using single-variant association analysis and gene-level collapsing analysis. FINDINGS A total of 13,823,269 autosomal genetic variants were obtained after quality control. We identified 36 VV-related independent common variants mapping to 34 genes by single-variant analysis and three rare variant genes (PIEZO1, ECE1, FBLN7) by collapsing analysis, and most associations between genes and VV were replicated in FinnGen. PIEZO1 was the closest gene associated with VV (P = 5.05 × 10-31), and it was found to reach exome-wide significance in both single-variant and collapsing analyses. Two novel rare variant genes (ECE1 and METTL21A) associated with VV were identified, of which METTL21A was associated only with females. The pleiotropic effects of VV-related genes suggested that body size, inflammation, and pulmonary function are strongly associated with the development of VV. CONCLUSIONS Our findings highlight the importance of causal genes for VV and provide new directions for treatment.
Collapse
Affiliation(s)
- Dan-Dan Zhang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xiao-Yu He
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Wei-Shi Liu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen-Jie Fei
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ju-Jiao Kang
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
- Department of Computer Science, University of Warwick, Coventry, United Kingdom
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
- Department of Computer Science, University of Warwick, Coventry, United Kingdom
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Zalewski D, Chmiel P, Kołodziej P, Kocki M, Feldo M, Kocki J, Bogucka-Kocka A. Key Regulators of Angiogenesis and Inflammation Are Dysregulated in Patients with Varicose Veins. Int J Mol Sci 2024; 25:6785. [PMID: 38928491 PMCID: PMC11204110 DOI: 10.3390/ijms25126785] [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: 05/22/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Varicose veins (VVs) are the most common manifestation of chronic venous disease (CVD) and appear as abnormally enlarged and tortuous superficial veins. VVs result from functional abnormalities in the venous circulation of the lower extremities, such as venous hypertension, venous valve incompetence, and venous reflux. Previous studies indicate that enhanced angiogenesis and inflammation contribute to the progression and onset of VVs; however, dysregulations in signaling pathways associated with these processes in VVs patients are poorly understood. Therefore, in our study, we aimed to identify key regulators of angiogenesis and inflammation that are dysregulated in patients with VVs. Expression levels of 18 genes were analyzed in peripheral blood mononuclear cells (PBMC) using real-time PCR, as well as plasma levels of 6 proteins were investigated using ELISA. Higher levels of CCL5, PDGFA, VEGFC, TGF-alpha, TGF-beta 1, and VEGF-A, as well as lower levels of VEGFB and VEGF-C, were found to be statistically significant in the VV group compared to the control subjects without VVs. None of the analyzed factors was associated with the venous localization of the varicosities. The presented study identified dysregulations in key angiogenesis- and inflammation-related factors in PBMC and plasma from VVs patients, providing new insight into molecular mechanisms that could contribute to the development of VVs and point out promising candidates for circulatory biomarkers of this disease.
Collapse
Affiliation(s)
- Daniel Zalewski
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland; (P.C.); (A.B.-K.)
| | - Paulina Chmiel
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland; (P.C.); (A.B.-K.)
| | - Przemysław Kołodziej
- Laboratory of Diagnostic Parasitology, Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland;
| | - Marcin Kocki
- Department of Neonatology and Neonatal Intensive Care, Independent Public Hospital No. 4 in Lublin, 8 Jaczewski St., 20-954 Lublin, Poland;
| | - Marcin Feldo
- Chair and Department of Vascular Surgery and Angiology, Medical University of Lublin, 11 Staszica St., 20-081 Lublin, Poland;
| | - Janusz Kocki
- Department of Clinical Genetics, Chair of Medical Genetics, Medical University of Lublin, 11 Radziwiłłowska St., 20-080 Lublin, Poland;
| | - Anna Bogucka-Kocka
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20-093 Lublin, Poland; (P.C.); (A.B.-K.)
| |
Collapse
|
8
|
Nadasy GL, Patai BB, Molnar AA, Hetthessy JR, Tokes AM, Varady Z, Dornyei G. Vicious Circle With Venous Hypertension, Irregular Flow, Pathological Venous Wall Remodeling, and Valve Destruction in Chronic Venous Disease: A Review. Angiology 2024:33197241256680. [PMID: 38839285 DOI: 10.1177/00033197241256680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Substantial advances occurred in phlebological practice in the last two decades. With the use of modern diagnostic equipment, the patients' venous hemodynamics can be examined in detail in everyday practice. Application of venous segments for arterial bypasses motivated studies on the effect of hemodynamic load on the venous wall. New animal models have been developed to study hemodynamic effects on the venous system. In vivo and in vitro studies revealed cellular phase transitions of venous endothelial, smooth muscle, and fibroblastic cells and changes in connective tissue composition, under hemodynamic load and at different locations of the chronically diseased venous system. This review is an attempt to integrate our knowledge from epidemiology, paleoanthropology and anthropology, clinical and experimental hemodynamic studies, histology, cell physiology, cell pathology, and molecular biology on the complex pathomechanism of this frequent disease. Our conclusion is that the disease is initiated by limited genetic adaptation of mankind not to bipedalism but to bipedalism in the unmoving standing or sitting position. In the course of the disease several pathologic vicious circles emerge, sustained venous hypertension inducing cellular phase transitions, chronic wall inflammation, apoptosis of cells, pathologic dilation, and valvular damage which, in turn, further aggravate the venous hypertension.
Collapse
Affiliation(s)
- Gyorgy L Nadasy
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | | | - Andrea A Molnar
- Department of Cardiology, Semmelweis University, Budapest, Hungary
| | | | - Anna-Maria Tokes
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | | | - Gabriella Dornyei
- Department of Morphology and Physiology, Health Science Faculty, Semmelweis University, Budapest, Hungary
| |
Collapse
|
9
|
Mehra R, Patra V, Dhillan R, Sharma A, Kashyap S, Rastogi G, Gupta L, Singh R, Chopra C, Sharma V. Replication study identified EFEMP1 association with varicose vein predisposition among Indians. Eur J Med Res 2024; 29:232. [PMID: 38609985 PMCID: PMC11015598 DOI: 10.1186/s40001-024-01786-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 03/11/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Varicose vein is a chronic condition that affects the lower extremities of the human body. Several factors have been implicated in the development of this disease, viz age, gender, weight, height and prolonged standing. Recently, genome-wide studies have identified genetic biomarkers that are associated with varicose veins in different ethnic groups. Such genetic studies are lacking in South Asians specifically in Indians where the prevalence of varicose veins is high, and it is important to replicate these variants in the stated population. The study aimed to replicate the association of genetic variants associated with varicose veins in this target population, which were found to be associated with the other ethnic groups. METHODOLOGY The studied cohort is of the Indian population comprising unrelated 104 varicose veins cases and 448 non-varicose vein controls. The samples were genotyped using the Illumina Global Screening Array. Using the genomic data from UK BioBank and 23andMe studied cohorts; eight genetic variants were selected to replicate in our dataset. The allelic association was performed to identify the effective allele and risk was estimated using odds ratio and p-value as level of significance. Multifactor Dimensionality Reduction was used to estimate the cumulative effect of variants in Indians. RESULT Variant rs3791679 of EFEMP1 was found to be associated with varicose veins in Indians. After observing the association of the EFEMP1 with varicose veins, we further ensued to identify all genetic variants within EFEMP1 to uncover the additional variants associated with this trait. Interestingly, we identified six new variants of EFEMP1 gene that have shown association. Moreover, the cumulative effect of all associated variations was estimated and the risk was 2.7 times higher in cases than controls whereas independently their effect ranges from 0.37-1.58. CONCLUSION This study identifies EFEMP1 as a potential gene related to the risk of varicose veins in Indians. It also highlights that evaluating the maximum number of variants of a gene rather than focusing solely on replicating single variations offers a more comprehensive and nuanced understanding of the genetic factors contributing to a complex trait like varicose veins.
Collapse
Affiliation(s)
- Rohit Mehra
- Department of Vascular and Endovascular Surgery, Command Hospital (Southern Command), Pune, India.
| | - Vikram Patra
- Department of Vascular and Endovascular Surgery, Command Hospital (Northern Command), Udhampur, India
| | - Rishi Dhillan
- Department of Vascular and Endovascular Surgery, Army Hospital (Research and Referral), Delhi, India
| | - Anuka Sharma
- NMC Genetics India Pvt. Ltd. Gurugram, Haryana, 122001, India
| | - Sonal Kashyap
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Garima Rastogi
- NMC Genetics India Pvt. Ltd. Gurugram, Haryana, 122001, India
| | - Love Gupta
- NMC Genetics India Pvt. Ltd. Gurugram, Haryana, 122001, India
| | - Reena Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Chirag Chopra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Varun Sharma
- NMC Genetics India Pvt. Ltd. Gurugram, Haryana, 122001, India.
| |
Collapse
|
10
|
Wilmink M, Spalinger MR. SKAP2-A Molecule at the Crossroads for Integrin Signalling and Immune Cell Migration and Function. Biomedicines 2023; 11:2788. [PMID: 37893161 PMCID: PMC10603853 DOI: 10.3390/biomedicines11102788] [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: 09/07/2023] [Revised: 09/28/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Src-kinase associated protein 2 (SKAP2) is an intracellular scaffolding protein that is broadly expressed in immune cells and is involved in various downstream signalling pathways, including, but not limited to, integrin signalling. SKAP2 has a wide range of binding partners and fine-tunes the rearrangement of the cytoskeleton, thereby regulating cell migration and immune cell function. Mutations in SKAP2 have been associated with several inflammatory disorders such as Type 1 Diabetes and Crohn's disease. Rodent studies showed that SKAP2 deficient immune cells have diminished pathogen clearance due to impaired ROS production and/or phagocytosis. However, there is currently no in-depth understanding of the functioning of SKAP2. Nevertheless, this review summarises the existing knowledge with a focus of its role in signalling cascades involved in cell migration, tissue infiltration and immune cell function.
Collapse
Affiliation(s)
| | - Marianne Rebecca Spalinger
- Department for Gastroenterology and Hepatology, University Hospital Zürich, Sternwartstrasse 14, 8091 Zürich, Switzerland;
| |
Collapse
|
11
|
Helkkula P, Hassan S, Saarentaus E, Vartiainen E, Ruotsalainen S, Leinonen JT, Palotie A, Karjalainen J, Kurki M, Ripatti S, Tukiainen T. Genome-wide association study of varicose veins identifies a protective missense variant in GJD3 enriched in the Finnish population. Commun Biol 2023; 6:71. [PMID: 36653477 PMCID: PMC9849365 DOI: 10.1038/s42003-022-04285-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 11/21/2022] [Indexed: 01/19/2023] Open
Abstract
Varicose veins is the most common manifestation of chronic venous disease that displays female-biased incidence. To identify protein-inactivating variants that could guide identification of drug target genes for varicose veins and genetic evidence for the disease prevalence difference between the sexes, we conducted a genome-wide association study of varicose veins in Finns using the FinnGen dataset with 17,027 cases and 190,028 controls. We identified 50 associated genetic loci (P < 5.0 × 10-8) of which 29 were novel including one near ERG with female-specificity (rs2836405-G, OR[95% CI] = 1.09[1.05-1.13], P = 3.1 × 10-8). These also include two X-chromosomal (ARHGAP6 and SRPX) and two autosomal novel loci (TGFB2 and GJD3) with protein-coding lead variants enriched above 56-fold in Finns over non-Finnish non-Estonian Europeans. A low-frequency missense variant in GJD3 (p.Pro59Thr) is exclusively associated with a lower risk for varicose veins (OR = 0.62 [0.55-0.70], P = 1.0 × 10-14) in a phenome-wide scan of the FinnGen data. The absence of observed pleiotropy and its membership of the connexin gene family underlines GJD3 as a potential connexin-modulating therapeutic strategy for varicose veins. Our results provide insights into varicose veins etiopathology and highlight the power of isolated populations, including Finns, to discover genetic variants that inform therapeutic development.
Collapse
Grants
- MC_PC_17228 Medical Research Council
- Academy of Finland (Suomen Akatemia)
- Sydäntutkimussäätiö (Finnish Foundation for Cardiovascular Research)
- Academy of Finland Center of Excellence in Complex Disease Genetics (Grant No 312062), Sigrid Juselius Foundation (S.Ri. and T.T.), University of Helsinki HiLIFE Fellow and Grand Challenge grants (S.Ri.), University of Helsinki three-year research project grant (T.T.), FIMM-EMBL PhD program doctoral funding (S.H.), Nylands Nation, University of Helsinki (P.H.) The FinnGen project is funded by two grants from Business Finland (HUS 4685/31/2016 and UH 4386/31/2016) and the following industry partners: AbbVie Inc., AstraZeneca UK Ltd, Biogen MA Inc., Bristol Myers Squibb (and Celgene Corporation & Celgene International II Sàrl), Genentech Inc., Merck Sharp & Dohme Corp, Pfizer Inc., GlaxoSmithKline Intellectual Property Development Ltd., Sanofi US Services Inc., Maze Therapeutics Inc., Janssen Biotech Inc, Novartis AG, and Boehringer Ingelheim. Following biobanks are acknowledged for delivering biobank samples to FinnGen: Auria Biobank (www.auria.fi/biopankki), THL Biobank (www.thl.fi/biobank), Helsinki Biobank (www.helsinginbiopankki.fi), Biobank Borealis of Northern Finland (https://www.ppshp.fi/Tutkimus-ja-opetus/Biopankki/Pages/Biobank-Borealis-briefly-in-English.aspx), Finnish Clinical Biobank Tampere (www.tays.fi/en-US/Research_and_development/Finnish_Clinical_Biobank_Tampere), Biobank of Eastern Finland (www.ita-suomenbiopankki.fi/en), Central Finland Biobank (www.ksshp.fi/fi-FI/Potilaalle/Biopankki), Finnish Red Cross Blood Service Biobank (www.veripalvelu.fi/verenluovutus/biopankkitoiminta) and Terveystalo Biobank (www.terveystalo.com/fi/Yritystietoa/Terveystalo-Biopankki/Biopankki/).
Collapse
Affiliation(s)
- Pyry Helkkula
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Shabbeer Hassan
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Elmo Saarentaus
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Emilia Vartiainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Sanni Ruotsalainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Jaakko T Leinonen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Juha Karjalainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Mitja Kurki
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Department of Public Health, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Taru Tukiainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.
| |
Collapse
|
12
|
Costa D, Andreucci M, Ielapi N, Serraino GF, Mastroroberto P, Bracale UM, Serra R. Molecular Determinants of Chronic Venous Disease: A Comprehensive Review. Int J Mol Sci 2023; 24:ijms24031928. [PMID: 36768250 PMCID: PMC9916309 DOI: 10.3390/ijms24031928] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Chronic Venous Disease (CVD) refers to several pathological and hemodynamic alterations of the veins of lower limbs causing a wide range of symptoms and signs with a high prevalence in the general population and with disabling consequences in the most severe forms. The etiology and pathophysiology of CVD is complex and multifactorial, involving genetic, proteomic, and cellular mechanisms that result in changes to the venous structure and functions. Expressions of several genes associated with angiogenesis, vascular development, and the regulation of veins are responsible for the susceptibility to CVD. Current evidence shows that several extracellular matrix alterations (ECM) could be identified and in some cases pharmacologically targeted. This review shows the most up to date information on molecular determinants of CVD in order to provide a complete overview of the current knowledge on this topic. In particular, the article explores the genetic influence, the hormonal influence, ECM imbalance, and histopathology of CVD and the role of endothelial dysfunction in CVD.
Collapse
Affiliation(s)
- Davide Costa
- Department of Law, Economics and Sociology, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Michele Andreucci
- Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Nicola Ielapi
- Department of Public Health and Infectious Disease, “Sapienza” University of Rome, 00185 Rome, Italy
| | - Giuseppe Filiberto Serraino
- Department of Experimental and Clinical Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Pasquale Mastroroberto
- Department of Experimental and Clinical Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | | | - Raffaele Serra
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
- Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
- Correspondence:
| |
Collapse
|
13
|
Levin MG, Huffman JE, Verma A, Sullivan KA, Rodriguez AA, Kainer D, Garvin MR, Lane M, Cashman M, Miller JI, Won H, Li B, Luo Y, Jarvik GP, Hakonarson H, Jasper EA, Bick AG, Tsao PS, Ritchie MD, Jacobson DA, Madduri RK, Damrauer SM. Genetics of varicose veins reveals polygenic architecture and genetic overlap with arterial and venous disease. NATURE CARDIOVASCULAR RESEARCH 2023; 2:44-57. [PMID: 39196206 DOI: 10.1038/s44161-022-00196-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 11/23/2022] [Indexed: 08/29/2024]
Abstract
Varicose veins represent a common cause of cardiovascular morbidity, with limited available medical therapies. Although varicose veins are heritable and epidemiologic studies have identified several candidate varicose vein risk factors, the molecular and genetic basis remains uncertain. Here we analyzed the contribution of common genetic variants to varicose veins using data from the Veterans Affairs Million Veteran Program and four other large biobanks. Among 49,765 individuals with varicose veins and 1,334,301 disease-free controls, we identified 139 risk loci. We identified genetic overlap between varicose veins, other vascular diseases and dozens of anthropometric factors. Using Mendelian randomization, we prioritized therapeutic targets via integration of proteomic and transcriptomic data. Finally, topological enrichment analyses confirmed the biologic roles of endothelial shear flow disruption, inflammation, vascular remodeling and angiogenesis. These findings may facilitate future efforts to develop nonsurgical therapies for varicose veins.
Collapse
Affiliation(s)
- Michael G Levin
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Jennifer E Huffman
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | - Anurag Verma
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kyle A Sullivan
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Alexis A Rodriguez
- Data Science and Learning Division, Argonne National Laboratory, Lemont, IL, USA
| | - David Kainer
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Michael R Garvin
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Matthew Lane
- Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee, Knoxville, TN, USA
| | - Mikaela Cashman
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - J Izaak Miller
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Hyejung Won
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Binglan Li
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Yuan Luo
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Gail P Jarvik
- Departments of Medicine (Division of Medical Genetics) and Genome Sciences, University of Washington Medical Center, Seattle, WA, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elizabeth A Jasper
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexander G Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Philip S Tsao
- VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Marylyn D Ritchie
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel A Jacobson
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Ravi K Madduri
- Data Science and Learning Division, Argonne National Laboratory, Lemont, IL, USA
| | - Scott M Damrauer
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA.
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
14
|
Sharma P, Beck D, Murtha LA, Figtree G, Boyle A, Gentile C. Fibulin-3 Deficiency Protects Against Myocardial Injury Following Ischaemia/ Reperfusion in in vitro Cardiac Spheroids. Front Cardiovasc Med 2022; 9:913156. [PMID: 35795376 PMCID: PMC9251181 DOI: 10.3389/fcvm.2022.913156] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/30/2022] [Indexed: 12/02/2022] Open
Abstract
Myocardial infarction (MI, or heart attack) is a leading cause of death worldwide. Myocardial ischaemia reperfusion (I/R) injury typical of MI events is also associated with the development of cardiac fibrosis and heart failure in patients. Fibulin-3 is an extracellular matrix component that plays a role in regulating MI response in the heart. In this study, we generated and compared in vitro cardiac spheroids (CSs) from wild type (WT) and fibulin-3 knockout (Fib-3 KO) mice. These were then exposed to pathophysiological changes in oxygen (O2) concentrations to mimic an MI event. We finally measured changes in contractile function, cell death, and mRNA expression levels of cardiovascular disease genes between WT and Fib-3 KO CSs. Our results demonstrated that there are significant differences in growth kinetics and endothelial network formation between WT and Fib-3 KO CSs, however, they respond similarly to changes in O2 concentrations. Fib-3 deficiency resulted in an increase in viability of cells and improvement in contraction frequency and fractional shortening compared to WT I/R CSs. Gene expression analyses demonstrated that Fib-3 deficiency inhibits I/R injury and cardiac fibrosis and promotes angiogenesis in CSs. Altogether, our findings suggest that Fib-3 deficiency makes CSs resistant to I/R injury and associated cardiac fibrosis and helps to improve the vascular network in CSs.
Collapse
Affiliation(s)
- Poonam Sharma
- College of Health Medicine and Wellbeing, The University of Newcastle, Callaghan, NSW, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
- Faculty of Medicine and Health, Northern Clinical School, The University of Sydney, Sydney, NSW, Australia
- Faculty of Engineering and IT, School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - Dominik Beck
- Faculty of Engineering and IT, School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - Lucy A. Murtha
- College of Health Medicine and Wellbeing, The University of Newcastle, Callaghan, NSW, Australia
| | - Gemma Figtree
- Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
- Faculty of Medicine and Health, Northern Clinical School, The University of Sydney, Sydney, NSW, Australia
| | - Andrew Boyle
- College of Health Medicine and Wellbeing, The University of Newcastle, Callaghan, NSW, Australia
| | - Carmine Gentile
- Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
- Faculty of Medicine and Health, Northern Clinical School, The University of Sydney, Sydney, NSW, Australia
- Faculty of Engineering and IT, School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
- *Correspondence: Carmine Gentile
| |
Collapse
|
15
|
Ahmed WUR, Kleeman S, Ng M, Wang W, Auton A, Lee R, Handa A, Zondervan KT, Wiberg A, Furniss D. Genome-wide association analysis and replication in 810,625 individuals with varicose veins. Nat Commun 2022; 13:3065. [PMID: 35654884 PMCID: PMC9163161 DOI: 10.1038/s41467-022-30765-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 05/17/2022] [Indexed: 12/30/2022] Open
Abstract
Varicose veins affect one-third of Western society, with a significant subset of patients developing venous ulceration, costing $14.9 billion annually in the USA. Current management consists of either compression stockings, or surgical ablation for more advanced disease. Most varicose veins patients report a positive family history, and heritability is ~17%. We describe the largest two-stage genome-wide association study of varicose veins in 401,656 individuals from UK Biobank, and replication in 408,969 individuals from 23andMe (total 135,514 cases and 675,111 controls). Forty-nine signals at 46 susceptibility loci were discovered. We map 237 genes to these loci, several of which are biologically plausible and tractable to therapeutic targeting. Pathway analysis identified enrichment in extracellular matrix biology, inflammation, (lymph)angiogenesis, vascular smooth muscle cell migration, and apoptosis. Using a polygenic risk score (PRS) derived in an independent cohort, we demonstrate its predictive utility and correlation with varicose veins surgery.
Collapse
Affiliation(s)
- Waheed-Ul-Rahman Ahmed
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK
| | - Sam Kleeman
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Michael Ng
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK
| | - Wei Wang
- 23andMe, Inc., Sunnyvale, CA, USA
| | | | | | - Regent Lee
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Ashok Handa
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Krina T Zondervan
- Nuffield Department of Women's & Reproductive Health, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK.,Wellcome Centre for Human Genetics, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Akira Wiberg
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK.,Department of Plastic and Reconstructive Surgery, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Dominic Furniss
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK. .,Department of Plastic and Reconstructive Surgery, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
| |
Collapse
|
16
|
Lee ML, Liang C, Chuang CH, Lee PS, Chen TH, Sun S, Liao KW, Huang HD. A genome-wide association study for varicose veins. Phlebology 2022; 37:267-278. [DOI: 10.1177/02683555211069248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background The aim was to compare the genetic information of varicose vein patients with that of a healthy population attempting to identify certain significant genetic associations. Method Patients’ clinical characteristics and demographics were collected, and their genetic samples were examined. The results were compared to the genetic information of one thousand sex-matched healthy controls from Taiwan Biobank database. The Clinical-Etiology-Anatomy-Pathophysiology classification was applied for further subgroup analysis. Results After comparison of genetic information of ninety-six patients to that of healthy controls, two significant single nucleotide polymorphisms (SNPs) were identified. One was in DPYSL2 gene, and the other was in VSTM2L gene. A further comparison between C2-3 patient subgroup and C4-6 subgroup identified another four significant SNPs, which were located in ZNF664-FAM101A, PHF2, ACOT11, and TOM1L1 genes. Conclusion Our preliminary result identified six significant SNPs located in six different genes. All of them and their genetic products may warrant further investigations.
Collapse
Affiliation(s)
- Meng-Lin Lee
- Division of Cardiovascular Surgery, Department of Surgery, Cathay General Hospital, Taipei, Republic of China
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Republic of China
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Republic of China
| | - Chao Liang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Republic of China
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Republic of China
| | - Cheng-Hsun Chuang
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Republic of China
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu, Republic of China
| | - Pei-Shyuan Lee
- Department of Family Medicine, Cathay General Hospital, Taipei, Republic of China
| | - Thay-Hsiung Chen
- Division of Cardiovascular Surgery, Department of Surgery, Cathay General Hospital, Taipei, Republic of China
| | - Shen Sun
- Division of Cardiovascular Surgery, Department of Surgery, Mackay Memorial Hospital, Taipei, Republic of China
| | - Kuang-Wen Liao
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Republic of China
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu, Republic of China
| | - Hsien-Da Huang
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Republic of China
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Republic of China
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong Province, China
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Guangdong Province, China
| |
Collapse
|
17
|
Pannier F, Noppeney T, Alm J, Breu FX, Bruning G, Flessenkämper I, Gerlach H, Hartmann K, Kahle B, Kluess H, Mendoza E, Mühlberger D, Mumme A, Nüllen H, Rass K, Reich-Schupke S, Stenger D, Stücker M, Schmedt CG, Schwarz T, Tesmann J, Teßarek J, Werth S, Valesky E. S2k guidelines: diagnosis and treatment of varicose veins. DER HAUTARZT 2022; 73:1-44. [PMID: 35438355 PMCID: PMC9358954 DOI: 10.1007/s00105-022-04977-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 02/23/2022] [Indexed: 11/29/2022]
Affiliation(s)
- F Pannier
- Praxis für Dermatologie und Phlebologie, Helmholtzstr. 4-6, 53123, Bonn, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Ma Z, Hu X, Ding HF, Zhang M, Huo Y, Dong Z. Single-Nucleus Transcriptional Profiling of Chronic Kidney Disease after Cisplatin Nephrotoxicity. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:613-628. [PMID: 35092726 PMCID: PMC8978211 DOI: 10.1016/j.ajpath.2021.12.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/12/2021] [Accepted: 12/16/2021] [Indexed: 01/09/2023]
Abstract
Cisplatin induces both acute and chronic nephrotoxicity during chemotherapy in patients with cancer. Presented here is the first study of single-nucleus RNA sequencing (snRNA-seq) of cisplatin-induced nephrotoxicity. Repeated low-dose cisplatin treatment (RLDC) led to decreases in renal function and kidney weight in mice at 9 weeks. The kidneys of these mice showed tubular degeneration and dilation. snRNA-seq identified 16 cell types and 17 cell clusters in these kidneys. Cluster-by-cluster comparison demonstrated cell type-specific changes in gene expression and identified a unique proximal tubule (PT) injury/repair cluster that co-expressed the injury marker kidney injury molecule-1 (Kim1) and the proliferation marker Ki-67. Compared with control, post-RLDC kidneys had 424 differentially expressed genes in PT cells, including tubular transporters and cytochrome P450 enzymes involved in lipid metabolism. snRNA-seq also revealed transcriptional changes in potential PT injury markers (Krt222, Eda2r, Ltbp2, and Masp1) and repair marker (Bex4). RLDC induced inflammation and proinflammatory cytokines (RelB, TNF-α, Il7, Ccl2, and Cxcl2) and the expression of fibrosis markers (fibronectin, collagen I, connective tissue growth factor, vimentin, and α-smooth muscle actin). Together, these results provide new insights into RLDC-induced transcriptional changes at the single-cell level that may contribute to the development of chronic kidney problems in patients with cancer after cisplatin chemotherapy.
Collapse
Affiliation(s)
- Zhengwei Ma
- Department of Cellular Biology and Anatomy, Vascular Biology Center, Augusta, Georgia.
| | - Xiaoru Hu
- Department of Cellular Biology and Anatomy, Vascular Biology Center, Augusta, Georgia; Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Han-Fei Ding
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ming Zhang
- Department of Cellular Biology and Anatomy, Vascular Biology Center, Augusta, Georgia
| | - Yuqing Huo
- Department of Cellular Biology and Anatomy, Vascular Biology Center, Augusta, Georgia; Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Vascular Biology Center, Augusta, Georgia; Charlie Norwood VA Medical Center, Augusta, Georgia.
| |
Collapse
|
19
|
He R, Cai H, Jiang Y, Liu R, Zhou Y, Qin Y, Yao C, Wang S, Hu Z. Integrative analysis prioritizes the relevant genes and risk factors for chronic venous disease. J Vasc Surg Venous Lymphat Disord 2022; 10:738-748.e5. [PMID: 35218958 DOI: 10.1016/j.jvsv.2022.02.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 02/03/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Chronic venous disease (CVD) refers to a range of symptoms resulting from long-term morphological and functional abnormalities of the venous system. However, the mechanism of CVD development remains largely unknown. Here we aim to provide more information on CVD pathogenesis, prevention strategies and therapy development through the integrative analysis of large-scale genetic data. METHODS Genetic data were obtained from publicly accessible databases. We utilized different approaches, including FUMA, DEPICT, Sherlock, SMR/HEIDIS, DEPICT and NetWAS to identify possible causal genes for CVD. Candidate genes were prioritized to further literature-based review. The differential expression of prioritized genes was validated by microarray from the Gene Expression Omnibus (GEO), a public genomics data repository" and Real-time quantitative PCR (qPCR) of varicose veins (VVs) specimens. The causal relationships between risk factors and CVD were assessed using the Two-sample Mendelian randomization (MR) approach. RESULTS We identified 46 lead single-nucleotide polymorphisms (SNPs) and 26 plausible causal genes for CVD. Microarray data indicated differential expression of possible causal genes in CVD when compared to controls. The expression levels of WDR92, RSPO3, LIMA, ABCB10, DNAJC7, C1S, CXCL1 were significantly down-regulated (P<0.05). PHLDA1 and SERPINE1 were significantly upregulated (P<0.05). Dysregulated expression of WDR92, RSPO3 and CASZ1 was also found in varicose vein specimens by qPCR. Two-sample MR suggested causative effects of BMI (OR, 1.008, 95%CI, 1.005-1.010), standing height (OR, 1.009, 95%CI, 1.007-1.011), college degree (OR, 0.983, 95%CI, 0.991-0.976), insulin (OR, 0.858, 95%CI, 0.794-0.928) and metformin (OR, 0.944, 95%CI, 0.904-0.985) on CVD. CONCLUSIONS Our study integrates genetic and gene expression data to make an effective risk gene prediction and etiological inferences for CVD. Prioritized candidate genes provide more insights into CVD pathogenesis, and the causative effects of risk factors on CVD that deserve further investigation.
Collapse
Affiliation(s)
- Rongzhou He
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Vascular Disease Treatment, Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Huoying Cai
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Vascular Disease Treatment, Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Yu Jiang
- Department of Ophthalmology, the First People's Hospital of Guangzhou City, Guangzhou, China; Zhongshan ophthalmic center, Sun Yat-sen University, Guangzhou, China
| | - Ruiming Liu
- National-Guangdong Joint Engineering Laboratory for Vascular Disease Treatment, Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China; Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu Zhou
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Vascular Disease Treatment, Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Yuansen Qin
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Vascular Disease Treatment, Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Chen Yao
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Vascular Disease Treatment, Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Shenming Wang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Vascular Disease Treatment, Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| | - Zuojun Hu
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Vascular Disease Treatment, Guangdong Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China.
| |
Collapse
|
20
|
Abstract
Chronic venous disease is a worldwide problem associated with significant morbidity and is expected to increase in prevalence as the current population ages. This is a comprehensive review of the anatomy, pathophysiology, genomics, clinical classification, and treatment modalities of chronic venous disease.
Collapse
Affiliation(s)
- Tom Alsaigh
- Division of Vascular Surgery, Vascular Medicine Section, Stanford University, 780 Welch Road, Suite CJ 350, Palo Alto, CA 94304, USA
| | - Eri Fukaya
- Division of Vascular Surgery, Vascular Medicine Section, Stanford University, 780 Welch Road, Suite CJ 350, Palo Alto, CA 94304, USA.
| |
Collapse
|
21
|
Ortega MA, Fraile-Martínez O, García-Montero C, Álvarez-Mon MA, Chaowen C, Ruiz-Grande F, Pekarek L, Monserrat J, Asúnsolo A, García-Honduvilla N, Álvarez-Mon M, Bujan J. Understanding Chronic Venous Disease: A Critical Overview of Its Pathophysiology and Medical Management. J Clin Med 2021; 10:3239. [PMID: 34362022 PMCID: PMC8348673 DOI: 10.3390/jcm10153239] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 01/19/2023] Open
Abstract
Chronic venous disease (CVD) is a multifactorial condition affecting an important percentage of the global population. It ranges from mild clinical signs, such as telangiectasias or reticular veins, to severe manifestations, such as venous ulcerations. However, varicose veins (VVs) are the most common manifestation of CVD. The explicit mechanisms of the disease are not well-understood. It seems that genetics and a plethora of environmental agents play an important role in the development and progression of CVD. The exposure to these factors leads to altered hemodynamics of the venous system, described as ambulatory venous hypertension, therefore promoting microcirculatory changes, inflammatory responses, hypoxia, venous wall remodeling, and epigenetic variations, even with important systemic implications. Thus, a proper clinical management of patients with CVD is essential to prevent potential harms of the disease, which also entails a significant loss of the quality of life in these individuals. Hence, the aim of the present review is to collect the current knowledge of CVD, including its epidemiology, etiology, and risk factors, but emphasizing the pathophysiology and medical care of these patients, including clinical manifestations, diagnosis, and treatments. Furthermore, future directions will also be covered in this work in order to provide potential fields to explore in the context of CVD.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, 28806 Alcalá de Henares, Spain
| | - Oscar Fraile-Martínez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
| | - Miguel A. Álvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
| | - Chen Chaowen
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
| | - Fernando Ruiz-Grande
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain;
- Department of Vascular Surgery, Príncipe de Asturias Hospital, 28801 Alcalá de Henares, Spain
| | - Leonel Pekarek
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
| | - Jorge Monserrat
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
| | - Angel Asúnsolo
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain;
- Department of Epidemiology and Biostatistics, Graduate School of Public Health and Health Policy, The City University of New York, New York, NY 10027, USA
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
- Immune System Diseases—Rheumatology and Internal Medicine Service, University Hospital Príncipe de Asturias, (CIBEREHD), 28806 Alcalá de Henares, Spain
| | - Julia Bujan
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (M.A.O.); (O.F.-M.); (C.G.-M.); (C.C.); (L.P.); (J.M.); (N.G.-H.); (M.Á.-M.); (J.B.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
| |
Collapse
|
22
|
Abstract
Venous disease is a term that broadly covers both venous thromboembolic disease and chronic venous disease. The basic pathophysiology of venous thromboembolism and chronic venous disease differ as venous thromboembolism results from an imbalance of hemostasis and thrombosis while chronic venous disease occurs in the setting of tissue damage because of prolonged venous hypertension. Both diseases are common and account for significant mortality and morbidity, respectively, and collectively make up a large health care burden. Despite both diseases having well-characterized environmental components, it has been known for decades that family history is an important risk factor, implicating a genetic element to a patient's risk. Our understanding of the pathogenesis of these diseases has greatly benefited from an expansion of population genetic studies from pioneering familial studies to large genome-wide association studies; we now have multiple risk loci for each venous disease. In this review, we will highlight the current state of knowledge on the epidemiology and genetics of venous thromboembolism and chronic venous disease and directions for future research.
Collapse
Affiliation(s)
- Richard A. Baylis
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, CA
| | - Nicholas L. Smith
- Department of Epidemiology, University of Washington, Seattle WA 98195, USA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle WA 98101, USA
- Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle WA 98108, USA
| | - Derek Klarin
- Division of Vascular Surgery, University of Florida College of Medicine, Gainesville, FL
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eri Fukaya
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, CA
| |
Collapse
|
23
|
Genetic Polymorphisms Related to VO2max Adaptation Are Associated With Elite Rugby Union Status and Competitive Marathon Performance. Int J Sports Physiol Perform 2021; 16:1858-1864. [PMID: 34088882 DOI: 10.1123/ijspp.2020-0856] [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: 10/28/2020] [Revised: 02/19/2021] [Accepted: 03/23/2021] [Indexed: 11/18/2022]
Abstract
PURPOSE Genetic polymorphisms have been associated with the adaptation to training in maximal oxygen uptake (V˙O2max). However, the genotype distribution of selected polymorphisms in athletic cohorts is unknown, with their influence on performance characteristics also undetermined. This study investigated whether the genotype distributions of 3 polymorphisms previously associated with V˙O2max training adaptation are associated with elite athlete status and performance characteristics in runners and rugby athletes, competitors for whom aerobic metabolism is important. METHODS Genomic DNA was collected from 732 men including 165 long-distance runners, 212 elite rugby union athletes, and 355 nonathletes. Genotype and allele frequencies of PRDM1 rs10499043 C/T, GRIN3A rs1535628 G/A, and KCNH8 rs4973706 T/C were compared between athletes and nonathletes. Personal-best marathon times in runners, as well as in-game performance variables and playing position, of rugby athletes were analyzed according to genotype. RESULTS Runners with PRDM1 T alleles recorded marathon times ∼3 minutes faster than CC homozygotes (02:27:55 [00:07:32] h vs 02:31:03 [00:08:24] h, P = .023). Rugby athletes had 1.57 times greater odds of possessing the KCNH8 TT genotype than nonathletes (65.5% vs 54.7%, χ2 = 6.494, P = .013). No other associations were identified. CONCLUSIONS This study is the first to demonstrate that polymorphisms previously associated with V˙O2max training adaptations in nonathletes are also associated with marathon performance (PRDM1) and elite rugby union status (KCNH8). The genotypes and alleles previously associated with superior endurance-training adaptation appear to be advantageous in long-distance running and achieving elite status in rugby union.
Collapse
|
24
|
Joshi H, Vastrad B, Joshi N, Vastrad C, Tengli A, Kotturshetti I. Identification of Key Pathways and Genes in Obesity Using Bioinformatics Analysis and Molecular Docking Studies. Front Endocrinol (Lausanne) 2021; 12:628907. [PMID: 34248836 PMCID: PMC8264660 DOI: 10.3389/fendo.2021.628907] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 05/19/2021] [Indexed: 01/01/2023] Open
Abstract
Obesity is an excess accumulation of body fat. Its progression rate has remained high in recent years. Therefore, the aim of this study was to diagnose important differentially expressed genes (DEGs) associated in its development, which may be used as novel biomarkers or potential therapeutic targets for obesity. The gene expression profile of E-MTAB-6728 was downloaded from the database. After screening DEGs in each ArrayExpress dataset, we further used the robust rank aggregation method to diagnose 876 significant DEGs including 438 up regulated and 438 down regulated genes. Functional enrichment analysis was performed. These DEGs were shown to be significantly enriched in different obesity related pathways and GO functions. Then protein-protein interaction network, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. The module analysis was performed based on the whole PPI network. We finally filtered out STAT3, CORO1C, SERPINH1, MVP, ITGB5, PCM1, SIRT1, EEF1G, PTEN and RPS2 hub genes. Hub genes were validated by ICH analysis, receiver operating curve (ROC) analysis and RT-PCR. Finally a molecular docking study was performed to find small drug molecules. The robust DEGs linked with the development of obesity were screened through the expression profile, and integrated bioinformatics analysis was conducted. Our study provides reliable molecular biomarkers for screening and diagnosis, prognosis as well as novel therapeutic targets for obesity.
Collapse
Affiliation(s)
- Harish Joshi
- Department of Endocrinology, Endocrine and Diabetes Care Center, Hubbali, India
| | - Basavaraj Vastrad
- Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, India
| | - Nidhi Joshi
- Department of Medicine, Dr. D. Y. Patil Medical College, Kolhapur, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad, India
- *Correspondence: Chanabasayya Vastrad,
| | - Anandkumar Tengli
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru and JSS Academy of Higher Education & Research, Mysuru, India
| | - Iranna Kotturshetti
- Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, India
| |
Collapse
|
25
|
Mazuchová J, Halašová E, Mazuch J, Šarlinová M, Valentová V, Franeková M, Zelník Š, Krkošková K, Javorka K, Péč M, Grendár M. Investigation of association between genetic polymorphisms of MMP2, MMP8, MMP9 and TIMP2 and development of varicose veins in the Slovak Population - pilot study. Physiol Res 2020; 69:S443-S454. [PMID: 33471544 DOI: 10.33549/physiolres.934597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent metalloendopeptidases that degrades extracellular matrix (ECM) components. MMPs are associated with venous wall remodelling, proliferation, migration, phenotypic and functional transformation of vascular smooth muscle cells and ECM organization under the physiological and pathophysiological conditions. We investigated possible association of genetic promoter polymorphisms of MMP2 (rs243866), MMP8 (rs11225395), MMP9 (rs3918242) and TIMP2 (rs8179090) to varicose veins development in the Slovak population. Genomic DNA from 276 Slovak individuals (138 cases, 138 controls) was genotyped for selected SNPs (rs243866, rs11225395, rs3918242 and rs8179090) using the PCR-RFLP analysis. The data were analysed by chi-squared (chi2) test, logistic regression, and Mann-Whitney test. The risk of varicose veins development was evaluated in dominant, codominant and recessive genetic models. The statistical evaluation of selected polymorphisms in patients in all three genetic models has not shown a significant risk of varicose veins development. Our study has not shown the association between selected polymorphisms and increased risk of varicose veins development in Slovak population. More evidence with broaden sample size is needed.
Collapse
Affiliation(s)
- J Mazuchová
- Department of Medical Biology, Jessenius Faculty of Medicine, Martin, Slovakia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Zhang C, Yu C, Li W, Zhu Y, Ye Y, Wang Z, Lin Z. Fibulin-3 affects vascular endothelial function and is regulated by angiotensin II. Microvasc Res 2020; 132:104043. [PMID: 32707048 DOI: 10.1016/j.mvr.2020.104043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 07/15/2020] [Accepted: 07/15/2020] [Indexed: 11/11/2022]
Abstract
OBJECTIVE The aim of the present study was to investigate the effect of fibulin-3 on vascular endothelial function, and to explore the relevant underlying mechanism with regard to the involvement of angiotensin II (AngII). METHODS One hundred and eight patients with essential hypertension (EH) and 31 controls were included to measure the flow-mediated dilatation (FMD). Serum fibulin-3 and AngII were examined using enzyme-linked immunosorbent assay (ELISA) and radioimmunoassay. Stable transfection of fibulin-3 was conducted on human umbilical vein endothelial cells (HUVECs) and SV40T-transformed HUVECs (PUMC-HUVEC-T1 cells). Cell counting kit-8 assay, cell cycle assay, wound healing assay, Transwell assay, apoptosis assay, and tube formation assay were subsequently performed. The expression of angiogenesis-associated genes [endothelial nitric oxide synthase (eNOS) and vascular endothelial growth factor A (VEGFA)] were measured by western blot analysis. HUVECs and PUMC-HUVEC-T1 cells were treated with AngII, and with or without an inhibitor of nuclear factor κB (NF-κB), BAY 11-7082. Pro-inflammatory cytokines [interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α)] were detected by ELISA. The expression levels of fibulin-3 and p65 were then measured by western blotting. RESULTS Lower levels of serum fibulin-3 were accompanied by poorer FMD and higher levels of serum AngII in patients with EH. Fibulin-3 overexpression promoted cell proliferation, migration, and angiogenesis, but led to an inhibition of apoptosis. By contrast, fibulin-3 downregulation inhibited cell proliferation, migration and angiogenesis, but promoted apoptosis. AngII induced inflammation and inhibited the expression of fibulin-3. BAY 11-7082 eliminated the inhibitory effect of AngII on fibulin-3. CONCLUSIONS Taken together, the results of the present study have shown that serum fibulin-3 may be a predictor of vascular endothelial function in patients with EH. Fibulin-3 gene may also have a beneficial role in repairing the vascular endothelium. Furthermore, the results also suggested that fibulin-3 gene was suppressed by AngII via the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Chiming Zhang
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Chan Yu
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Wenlei Li
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Yaoyao Zhu
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Yuling Ye
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Zhuo Wang
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China.
| | - Zhongwei Lin
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, PR China.
| |
Collapse
|
27
|
Livingstone I, Uversky VN, Furniss D, Wiberg A. The Pathophysiological Significance of Fibulin-3. Biomolecules 2020; 10:E1294. [PMID: 32911658 PMCID: PMC7563619 DOI: 10.3390/biom10091294] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 02/07/2023] Open
Abstract
Fibulin-3 (also known as EGF-containing fibulin extracellular matrix protein 1 (EFEMP1)) is a secreted extracellular matrix glycoprotein, encoded by the EFEMP1 gene that belongs to the eight-membered fibulin protein family. It has emerged as a functionally unique member of this family, with a diverse array of pathophysiological associations predominantly centered on its role as a modulator of extracellular matrix (ECM) biology. Fibulin-3 is widely expressed in the human body, especially in elastic-fibre-rich tissues and ocular structures, and interacts with enzymatic ECM regulators, including tissue inhibitor of metalloproteinase-3 (TIMP-3). A point mutation in EFEMP1 causes an inherited early-onset form of macular degeneration called Malattia Leventinese/Doyne honeycomb retinal dystrophy (ML/DHRD). EFEMP1 genetic variants have also been associated in genome-wide association studies with numerous complex inherited phenotypes, both physiological (namely, developmental anthropometric traits) and pathological (many of which involve abnormalities of connective tissue function). Furthermore, EFEMP1 expression changes are implicated in the progression of numerous types of cancer, an area in which fibulin-3 has putative significance as a therapeutic target. Here we discuss the potential mechanistic roles of fibulin-3 in these pathologies and highlight how it may contribute to the development, structural integrity, and emergent functionality of the ECM and connective tissues across a range of anatomical locations. Its myriad of aetiological roles positions fibulin-3 as a molecule of interest across numerous research fields and may inform our future understanding and therapeutic approach to many human diseases in clinical settings.
Collapse
Affiliation(s)
- Imogen Livingstone
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford OX3 7LD, UK; (I.L.); (D.F.)
| | - Vladimir N. Uversky
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation, Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Pushchino 142290, Moscow Region, Russia;
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Dominic Furniss
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford OX3 7LD, UK; (I.L.); (D.F.)
- Department of Plastic and Reconstructive Surgery, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Akira Wiberg
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford OX3 7LD, UK; (I.L.); (D.F.)
- Department of Plastic and Reconstructive Surgery, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford OX3 9DU, UK
| |
Collapse
|
28
|
Kakkos SK, Guex JJ, Lugli M, Nicolaides AN. CEAP clinical classes C0S-C4: differences, similarities and role of Ruscus + HMC + vitamin C in patients with chronic venous disease. INT ANGIOL 2020; 39:118-124. [PMID: 32052951 DOI: 10.23736/s0392-9590.20.04341-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Since the publication of the CEAP classification, new research has enriched our knowledge; notably on the heritability of CVD and the genetic and environmental factors involved in this condition, as well as the symptoms apparent within the spectrum of the CEAP clinical classes and the benefits of medical treatment. Using the CEAP classification as a special theme, a symposium with the same title as the present paper was held at the annual meeting of the 2019 European Venous Forum. The lectures presented much valuable information, from which some key points can be extracted. The influence of environmental factors was demonstrated, and the fact that a large amount of information can be obtained from comprehensive history taking. There is robust evidence for heritability. Many candidate genes/loci have been identified, potentially offering new targets for treatment. More research is needed, notably using genome-wide association studies and also on microbiota, which may play a role in CVD through the inflammation pathway. Ruscus + HMC + vitamin C acts by increasing venous and lymphatic tone, protecting microcirculation, and reducing inflammation. It improves quality of life in C0S to C3 CVD patients, while a review of clinical studies and a meta-analysis have confirmed its clinical efficacy across a wide spectrum of CVD clinical classes: C0S, C1S, C2, C3 and C4. It has been awarded a Grade 1A recommendation by the international guidelines.
Collapse
Affiliation(s)
- Stavros K Kakkos
- Department of Vascular Surgery, University Hospital of Patras, Patras, Greece -
| | | | - Marzia Lugli
- National UEMS Reference Training Center in Phlebology, Department of Cardiovascular Surgery, Hesperia Hospital, Modena, Italy
| | - Andrew N Nicolaides
- Department of Surgery, University of Nicosia Medical School, Nicosia, Cyprus
| |
Collapse
|
29
|
Serra R, Ssempijja L, Provenzano M, Andreucci M. Genetic biomarkers in chronic venous disease. Biomark Med 2020; 14:75-80. [PMID: 32053001 DOI: 10.2217/bmm-2019-0408] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Raffaele Serra
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology at the Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Calabria, Italy.,Department of Medical & Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Calabria, Italy
| | - Lwanga Ssempijja
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology at the Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Calabria, Italy.,Department of Medical & Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Calabria, Italy
| | - Michele Provenzano
- Department of Health Sciences, University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Calabria, Italy
| | - Michele Andreucci
- Department of Health Sciences, University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Calabria, Italy
| |
Collapse
|
30
|
A variant of the castor zinc finger 1 (CASZ1) gene is differentially associated with the clinical classification of chronic venous disease. Sci Rep 2019; 9:14011. [PMID: 31570750 PMCID: PMC6769056 DOI: 10.1038/s41598-019-50586-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 09/16/2019] [Indexed: 01/25/2023] Open
Abstract
Recent reports have suggested a reproducible association between the rs11121615 SNP, located within an intron of the castor zinc finger 1 (CASZ1) gene, and varicose veins. This study aimed to determine if this variant is also differentially associated with the various clinical classifications of chronic venous disease (CVD). The rs11121615 SNP was genotyped in two independent cohorts from New Zealand (n = 1876 controls /1606 CVD cases) and the Netherlands (n = 1626/2966). Participants were clinically assessed using well-established CVD criteria. The association between the rs11121615 C-allele and varicose veins was validated in both cohorts. This was strongest in those with higher clinical severity classes and was not significant in those with non-varicose vein CVD. Functional analysis of the rs11121615 variant demonstrated that the risk allele was associated with increased enhancer activity. This study demonstrates that the CASZ1 gene associated C-allele of rs11121615 has a significant, reproducible, association with CVD (CEAP C ≥ 2 meta-odds ratio 1.31, 95% CI 1.27–1.34, P = 1 × 10−98, PHet = 0.25), but not with non-varicose vein (CEAP C1, telangiectasia or reticular veins) forms of venous disease. The effect size of this association therefore appears to be susceptible to influence by phenotypic heterogeneity, particularly if a cohort includes a large number of cases with lower severity CVD.
Collapse
|
31
|
Affiliation(s)
- Quinn S Wells
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
32
|
Next-Generation Sequencing Profiles of the Methylome and Transcriptome in Peripheral Blood Mononuclear Cells of Rheumatoid Arthritis. J Clin Med 2019; 8:jcm8091284. [PMID: 31443559 PMCID: PMC6780767 DOI: 10.3390/jcm8091284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/14/2019] [Accepted: 08/19/2019] [Indexed: 02/06/2023] Open
Abstract
Using next-generation sequencing to decipher methylome and transcriptome and underlying molecular mechanisms contributing to rheumatoid arthritis (RA) for improving future therapies, we performed methyl-seq and RNA-seq on peripheral blood mononuclear cells (PBMCs) from RA subjects and normal donors. Principal component analysis and hierarchical clustering revealed distinct methylation signatures in RA with methylation aberrations noted across chromosomes. Methylation alterations varied with CpG features and genic characteristics. Typically, CpG islands and CpG shores were hypermethylated and displayed the greatest methylation variance. Promoters were hypermethylated and enhancers/gene bodies were hypomethylated, with methylation variance associated with expression variance. RA genetically associated genes preferentially displayed differential methylation and differential expression or interacted with differentially methylated and differentially expressed genes. These differentially methylated and differentially expressed genes were enriched with several signaling pathways and disease categories. 10 genes (CD86, RAB20, XAF1, FOLR3, LTBR, KCNH8, DOK7, PDGFA, PITPNM2, CELSR1) with concomitantly differential methylation in enhancers/promoters/gene bodies and differential expression in B cells were validated. This integrated analysis of methylome and transcriptome identified novel epigenetic signatures associated with RA and highlighted the interaction between genetics and epigenetics in RA. These findings help our understanding of the pathogenesis of RA and advance epigenetic studies in regards to the disease.
Collapse
|
33
|
Schafmayer C, Harrison JW, Buch S, Lange C, Reichert MC, Hofer P, Cossais F, Kupcinskas J, von Schönfels W, Schniewind B, Kruis W, Tepel J, Zobel M, Rosendahl J, Jacobi T, Walther-Berends A, Schroeder M, Vogel I, Sergeev P, Boedeker H, Hinrichsen H, Volk A, Erk JU, Burmeister G, Hendricks A, Hinz S, Wolff S, Böttner M, Wood AR, Tyrrell J, Beaumont RN, Langheinrich M, Kucharzik T, Brezina S, Huber-Schönauer U, Pietsch L, Noack LS, Brosch M, Herrmann A, Thangapandi RV, Schimming HW, Zeissig S, Palm S, Focke G, Andreasson A, Schmidt PT, Weitz J, Krawczak M, Völzke H, Leeb G, Michl P, Lieb W, Grützmann R, Franke A, Lammert F, Becker T, Kupcinskas L, D'Amato M, Wedel T, Datz C, Gsur A, Weedon MN, Hampe J. Genome-wide association analysis of diverticular disease points towards neuromuscular, connective tissue and epithelial pathomechanisms. Gut 2019; 68:854-865. [PMID: 30661054 DOI: 10.1136/gutjnl-2018-317619] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 01/03/2019] [Accepted: 01/05/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Diverticular disease is a common complex disorder characterised by mucosal outpouchings of the colonic wall that manifests through complications such as diverticulitis, perforation and bleeding. We report the to date largest genome-wide association study (GWAS) to identify genetic risk factors for diverticular disease. DESIGN Discovery GWAS analysis was performed on UK Biobank imputed genotypes using 31 964 cases and 419 135 controls of European descent. Associations were replicated in a European sample of 3893 cases and 2829 diverticula-free controls and evaluated for risk contribution to diverticulitis and uncomplicated diverticulosis. Transcripts at top 20 replicating loci were analysed by real-time quatitative PCR in preparations of the mucosal, submucosal and muscular layer of colon. The localisation of expressed protein at selected loci was investigated by immunohistochemistry. RESULTS We discovered 48 risk loci, of which 12 are novel, with genome-wide significance and consistent OR in the replication sample. Nominal replication (p<0.05) was observed for 27 loci, and additional 8 in meta-analysis with a population-based cohort. The most significant novel risk variant rs9960286 is located near CTAGE1 with a p value of 2.3×10-10 and 0.002 (ORallelic=1.14 (95% CI 1.05 to 1.24)) in the replication analysis. Four loci showed stronger effects for diverticulitis, PHGR1 (OR 1.32, 95% CI 1.12 to 1.56), FAM155A-2 (OR 1.21, 95% CI 1.04 to 1.42), CALCB (OR 1.17, 95% CI 1.03 to 1.33) and S100A10 (OR 1.17, 95% CI 1.03 to 1.33). CONCLUSION In silico analyses point to diverticulosis primarily as a disorder of intestinal neuromuscular function and of impaired connective fibre support, while an additional diverticulitis risk might be conferred by epithelial dysfunction.
Collapse
Affiliation(s)
- Clemens Schafmayer
- Department of Visceral and Thoracic Surgery, Kiel University, Kiel, Germany
| | | | - Stephan Buch
- Medical Department 1, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden (TU Dresden), Dresden, Germany
| | | | - Matthias C Reichert
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Philipp Hofer
- Institute of Cancer Research, Department of Medicine I, Medical University Vienna, Vienna, Austria
| | | | - Juozas Kupcinskas
- Department of Gastroenterology and Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | | | - Wolfgang Kruis
- Department of Internal Medicine, Gastroenterology and Pulmonology, Evangelic Hospital Köln-Kalk, Cologne, Germany
| | - Jürgen Tepel
- Department of General and Thoracic Surgery, Hospital Osnabrück, Osnabrück, Germany
| | - Myrko Zobel
- Department of Gastroenterology, Helios Hospital Weißeritztal, Freital, Germany
| | - Jonas Rosendahl
- Medical Department 1, University Hospital Halle, Martin-Luther Universität Halle-Wittenberg, Halle, Germany
| | | | | | | | - Ilka Vogel
- Department of Surgery, Community Hospital Kiel, Kiel, Germany
| | - Petr Sergeev
- Department of Internal Medicine II, Hospital Riesa, Kiel, Germany
| | - Hans Boedeker
- Department of Internal Medicine, Hospital Freiberg, Freiberg, Germany
| | | | - Andreas Volk
- Department of Visceral, Thoracic and Vascular Surgery, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Jens-Uwe Erk
- Medical Department 1, University Hospital Halle, Martin-Luther Universität Halle-Wittenberg, Halle, Germany
| | - Greta Burmeister
- Department of Visceral and Thoracic Surgery, Kiel University, Kiel, Germany
| | | | - Sebastian Hinz
- Department of Visceral and Thoracic Surgery, Kiel University, Kiel, Germany
| | - Sebastian Wolff
- Department of Internal Medicine, Gastroenterology and Pulmonology, Evangelic Hospital Köln-Kalk, Cologne, Germany
| | | | - Andrew R Wood
- University of Exeter Medical School, University of Exeter, United Kingdom, Exeter, UK
| | - Jessica Tyrrell
- University of Exeter Medical School, University of Exeter, United Kingdom, Exeter, UK
| | - Robin N Beaumont
- University of Exeter Medical School, University of Exeter, United Kingdom, Exeter, UK
| | | | | | - Stefanie Brezina
- Institute of Cancer Research, Department of Medicine I, Medical University Vienna, Vienna, Austria
| | - Ursula Huber-Schönauer
- Department of Internal Medicine, Hospital Oberndorf, Teaching Hospital of the Paracelsus Private Medical University of Salzburg, Oberndorf, Austria
| | - Leonora Pietsch
- Medical Department 1, University Hospital Halle, Martin-Luther Universität Halle-Wittenberg, Halle, Germany
| | - Laura Sophie Noack
- Medical Department 1, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Mario Brosch
- Medical Department 1, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Alexander Herrmann
- Medical Department 1, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Raghavan Veera Thangapandi
- Medical Department 1, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | | | - Sebastian Zeissig
- Medical Department 1, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Stefan Palm
- Outpatient Center for Gastroenterology, Dippoldiswalde, Germany
| | - Gerd Focke
- Outpatient Center for Gastroenterology Dresden-Blasewitz, Dresden, Germany
| | - Anna Andreasson
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Stress Research Institute, Stockholm University, Stockholm, Sweden
| | - Peter T Schmidt
- Department of Medicine Solna and Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Juergen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Michael Krawczak
- Institute of Medical Informatics and Statistics, Kiel University, Kiel, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Gernot Leeb
- Department of Gastroenterology, Hospital Oberpullendorf, Oberpullendorf, Austria
| | - Patrick Michl
- Medical Department 1, University Hospital Halle, Martin-Luther Universität Halle-Wittenberg, Halle, Germany
| | - Wolfgang Lieb
- Institute of Epidemiology & Popgen Biobank, Kiel University, Kiel, Germany
| | - Robert Grützmann
- Department of Surgery, University Hospital Erlangen, Erlangen, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Frank Lammert
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Thomas Becker
- Department of Visceral and Thoracic Surgery, Kiel University, Kiel, Germany
| | - Limas Kupcinskas
- Department of Gastroenterology and Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Mauro D'Amato
- Department of Medicine Solna and Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Thilo Wedel
- Institute of Anatomy, Kiel University, Kiel, Germany
| | - Christian Datz
- Department of Internal Medicine, Hospital Oberndorf, Teaching Hospital of the Paracelsus Private Medical University of Salzburg, Oberndorf, Austria
| | - Andrea Gsur
- Institute of Cancer Research, Department of Medicine I, Medical University Vienna, Vienna, Austria
| | - Michael N Weedon
- University of Exeter Medical School, University of Exeter, United Kingdom, Exeter, UK
| | - Jochen Hampe
- Medical Department 1, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden (TU Dresden), Dresden, Germany
| |
Collapse
|
34
|
Varicose veins of lower extremities: Insights from the first large-scale genetic study. PLoS Genet 2019; 15:e1008110. [PMID: 30998689 PMCID: PMC6490943 DOI: 10.1371/journal.pgen.1008110] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 04/30/2019] [Accepted: 03/25/2019] [Indexed: 11/19/2022] Open
Abstract
Varicose veins of lower extremities (VVs) are a common multifactorial vascular disease. Genetic factors underlying VVs development remain largely unknown. Here we report the first large-scale study of VVs performed on a freely available genetic data of 408,455 European-ancestry individuals. We identified the 12 reliably associated loci that explain 13% of the SNP-based heritability, and prioritized the most likely causal genes CASZ1, PIEZO1, PPP3R1, EBF1, STIM2, HFE, GATA2, NFATC2, and SOX9. VVs-associated variants within these loci exhibited pleiotropic effects on several phenotypes including blood pressure/hypertension and blood cell traits. Gene set enrichment analysis revealed gene categories related to abnormal vasculogenesis. Genetic correlation analysis confirmed known epidemiological associations between VVs and deep venous thrombosis, weight, rough labor, and standing job, and found a genetic overlap with multiple traits that have not been previously suspected to share common genetic background with VVs. These traits included educational attainment, fluid intelligence and prospective memory scores, walking pace (negative correlation with VVs), smoking, height, number of operations, pain, and gonarthrosis (positive correlation with VVs). Finally, Mendelian randomization analysis provided evidence for causal effects of plasma levels of MICB and CD209 proteins, and anthropometric traits such as waist and hip circumference, height, weight, and both fat and fat-free mass. Our results provide novel insight into both VVs genetics and etiology. The revealed genes and proteins can be considered as good candidates for follow-up functional studies and might be of interest as potential drug targets. Varicose veins of lower extremities (VVs) affect about 30% of adults in developed countries and cause both cosmetic and health problems. A strong body of evidence indicates that heredity plays an important role in the etiology of this condition. However, genetic basis of VVs remains poorly understood. Here, we present the results of the first large-scale genetic study for VVs. We identified genes which are the most likely involved in VVs pathogenesis. We show that VVs are correlated at a genetic level with numerous traits and phenotypes, including those already known from prior epidemiological studies (deep venous thrombosis, body mass index, standing job, etc.) as well as with those that have not been suspected to share common genetic background with VVs (fluid intelligence and prospective memory scores, smoking, walking pace, pain all over the body, and other traits). Finally, using genetic variants as instruments, we demonstrate direct causal effects of the traits related to anthropometry, such as height and weight, and plasma levels of immune-related proteins MICB and CD209. Our study provides novel insight into both VVs genetics and etiology. The revealed genes (CASZ1, PIEZO1, PPP3R1, EBF1, STIM2, HFE, GATA2, NFATC2, and SOX9) and proteins (MICB and CD209) can be considered as good candidates for follow-up functional studies and might be of interest as potential drug targets.
Collapse
|
35
|
Bagnati M, Moreno-Moral A, Ko JH, Nicod J, Harmston N, Imprialou M, Game L, Gil J, Petretto E, Behmoaras J. Systems genetics identifies a macrophage cholesterol network associated with physiological wound healing. JCI Insight 2019; 4:e125736. [PMID: 30674726 PMCID: PMC6413785 DOI: 10.1172/jci.insight.125736] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/18/2018] [Indexed: 01/18/2023] Open
Abstract
Among other cells, macrophages regulate the inflammatory and reparative phases during wound healing but genetic determinants and detailed molecular pathways that modulate these processes are not fully elucidated. Here, we took advantage of normal variation in wound healing in 1,378 genetically outbred mice, and carried out macrophage RNA-sequencing profiling of mice with extreme wound healing phenotypes (i.e., slow and fast healers, n = 146 in total). The resulting macrophage coexpression networks were genetically mapped and led to the identification of a unique module under strong trans-acting genetic control by the Runx2 locus. This macrophage-mediated healing network was specifically enriched for cholesterol and fatty acid biosynthetic processes. Pharmacological blockage of fatty acid synthesis with cerulenin resulted in delayed wound healing in vivo, and increased macrophage infiltration in the wounded skin, suggesting the persistence of an unresolved inflammation. We show how naturally occurring sequence variation controls transcriptional networks in macrophages, which in turn regulate specific metabolic pathways that could be targeted in wound healing.
Collapse
Affiliation(s)
- Marta Bagnati
- Centre for Inflammatory Disease, Imperial College London, Hammersmith Hospital, London, United Kingdom (UK)
| | | | - Jeong-Hun Ko
- Centre for Inflammatory Disease, Imperial College London, Hammersmith Hospital, London, United Kingdom (UK)
| | - Jérôme Nicod
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | - Martha Imprialou
- Centre for Inflammatory Disease, Imperial College London, Hammersmith Hospital, London, United Kingdom (UK)
| | - Laurence Game
- Genomics Laboratory, Medical Research Council (MRC) London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Jesus Gil
- Cell Proliferation Group, MRC London Institute of Medical Sciences (LMS), London, UK
| | - Enrico Petretto
- Duke-NUS Medical School, Singapore, Singapore
- MRC London Institute of Medical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Jacques Behmoaras
- Centre for Inflammatory Disease, Imperial College London, Hammersmith Hospital, London, United Kingdom (UK)
| |
Collapse
|
36
|
Tasaki M, Ueda M, Hoshii Y, Mizukami M, Matsumoto S, Nakamura M, Yamashita T, Ueda A, Misumi Y, Masuda T, Inoue Y, Torikai T, Nomura T, Tsuda Y, Kanenawa K, Isoguchi A, Okada M, Matsui H, Obayashi K, Ando Y. A novel age-related venous amyloidosis derived from EGF-containing fibulin-like extracellular matrix protein 1. J Pathol 2018; 247:444-455. [DOI: 10.1002/path.5203] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 09/28/2018] [Accepted: 11/16/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Masayoshi Tasaki
- Department of Neurology, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
- Department of Morphological and Physiological Sciences, Graduate School of Health Sciences; Kumamoto University; Kumamoto Japan
| | - Mitsuharu Ueda
- Department of Neurology, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
| | - Yoshinobu Hoshii
- Department of Pathology; Yamaguchi University Graduate School of Medicine; Ube Japan
| | - Mayumi Mizukami
- Department of Neurology, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
| | - Sayaka Matsumoto
- Department of Neurology, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
| | - Makoto Nakamura
- Department of Neurology, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
| | - Taro Yamashita
- Department of Neurology, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
| | - Akihiko Ueda
- Department of Neurology, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
| | - Yohei Misumi
- Department of Neurology, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
| | - Teruaki Masuda
- Department of Neurology, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
| | - Yasuteru Inoue
- Department of Neurology, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
| | - Tessei Torikai
- Department of Neurology, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
| | - Toshiya Nomura
- Department of Neurology, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
| | - Yukimoto Tsuda
- Department of Neurology, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
| | - Kyosuke Kanenawa
- Department of Neurology, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
| | - Aito Isoguchi
- Department of Neurology, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
| | - Masamitsu Okada
- Department of Neurology, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
| | - Hirotaka Matsui
- Department of Molecular Laboratory Medicine, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
| | - Konen Obayashi
- Department of Morphological and Physiological Sciences, Graduate School of Health Sciences; Kumamoto University; Kumamoto Japan
| | - Yukio Ando
- Department of Neurology, Graduate School of Medical Sciences; Kumamoto University; Kumamoto Japan
| |
Collapse
|
37
|
Roles of short fibulins, a family of matricellular proteins, in lung matrix assembly and disease. Matrix Biol 2018; 73:21-33. [DOI: 10.1016/j.matbio.2018.02.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/26/2017] [Accepted: 02/01/2018] [Indexed: 12/19/2022]
|
38
|
Serra R, Ielapi N, Barbetta A, Andreucci M, de Franciscis S. Novel biomarkers for cardiovascular risk. Biomark Med 2018; 12:1015-1024. [PMID: 30126290 DOI: 10.2217/bmm-2018-0056] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular disease refers to different diseases involving the heart and/or the arteries and/or the veins. Cardiovascular disease, overall considered, is a notable source of morbidity and mortality worldwide. Therefore, several research studies are dedicated to explore, by means of biomarkers, the possiblity to calculate the cardiovascular risk both for the onset and for the complications of the related clinical manifestations such as coronary artery disease, carotid artery stenosis, peripheral artery disease, arterial aneurysm, chronic venous disease and venous thromboembolism. This review discusses the most updated information in the area of the novel biomarkers related to omics, imaging techniques and clinical data, that may help physicians in order to improve the knowledge and the management of the cardiovascular risk.
Collapse
Affiliation(s)
- Raffaele Serra
- Interuniversity Center of Phlebolymphology (CIFL). International Research & Educational Program in Clinical & Experimental Biotechnology' at the Department of Surgical & Medical Sciences University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Italy.,Department of Surgical & Medical Sciences University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Italy
| | - Nicola Ielapi
- Interuniversity Center of Phlebolymphology (CIFL). International Research & Educational Program in Clinical & Experimental Biotechnology' at the Department of Surgical & Medical Sciences University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Italy
| | - Andrea Barbetta
- Interuniversity Center of Phlebolymphology (CIFL). International Research & Educational Program in Clinical & Experimental Biotechnology' at the Department of Surgical & Medical Sciences University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Italy
| | - Michele Andreucci
- Department of Health Sciences University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Italy
| | - Stefano de Franciscis
- Interuniversity Center of Phlebolymphology (CIFL). International Research & Educational Program in Clinical & Experimental Biotechnology' at the Department of Surgical & Medical Sciences University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Italy.,Department of Surgical & Medical Sciences University Magna Graecia of Catanzaro, Viale Europa 88100 Catanzaro, Italy
| |
Collapse
|
39
|
Chen Y, Gilbert MA, Grochowski CM, McEldrew D, Llewellyn J, Waisbourd-Zinman O, Hakonarson H, Bailey-Wilson JE, Russo P, Wells RG, Loomes KM, Spinner NB, Devoto M. A genome-wide association study identifies a susceptibility locus for biliary atresia on 2p16.1 within the gene EFEMP1. PLoS Genet 2018; 14:e1007532. [PMID: 30102696 PMCID: PMC6107291 DOI: 10.1371/journal.pgen.1007532] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 08/23/2018] [Accepted: 07/04/2018] [Indexed: 02/06/2023] Open
Abstract
Biliary atresia (BA) is a rare pediatric cholangiopathy characterized by fibrosclerosing obliteration of the extrahepatic bile ducts, leading to cholestasis, fibrosis, cirrhosis, and eventual liver failure. The etiology of BA remains unknown, although environmental, inflammatory, infectious, and genetic risk factors have been proposed. We performed a genome-wide association study (GWAS) in a European-American cohort of 343 isolated BA patients and 1716 controls to identify genetic loci associated with BA. A second GWAS was performed in an independent European-American cohort of 156 patients with BA and other extrahepatic anomalies and 212 controls to confirm the identified candidate BA-associated SNPs. Meta-analysis revealed three genome-wide significant BA-associated SNPs on 2p16.1 (rs10865291, rs6761893, and rs727878; P < 5 ×10-8), located within the fifth intron of the EFEMP1 gene, which encodes a secreted extracellular protein implicated in extracellular matrix remodeling, cell proliferation, and organogenesis. RNA expression analysis showed an increase in EFEMP1 transcripts from human liver specimens isolated from patients with either BA or other cholestatic diseases when compared to normal control liver samples. Immunohistochemistry demonstrated that EFEMP1 is expressed in cholangiocytes and vascular smooth muscle cells in liver specimens from patients with BA and other cholestatic diseases, but it is absent from cholangiocytes in normal control liver samples. Efemp1 transcripts had higher expression in cholangiocytes and portal fibroblasts as compared with other cell types in normal rat liver. The identification of a novel BA-associated locus, and implication of EFEMP1 as a new BA candidate susceptibility gene, could provide new insights to understanding the mechanisms underlying this severe pediatric disorder.
Collapse
Affiliation(s)
- Ying Chen
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Division of Human Genetics, Department of Pediatrics, at The Children's Hospital of Philadelphia, and The Perelman School of Medicine at The University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Melissa A. Gilbert
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, and The Perelman School of Medicine at The University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Christopher M. Grochowski
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, and The Perelman School of Medicine at The University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Deborah McEldrew
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, and The Perelman School of Medicine at The University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jessica Llewellyn
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Orith Waisbourd-Zinman
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics at The Children's Hospital of Philadelphia, and The Perelman School of Medicine at The University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Schneider Children's Medical Center of Israel, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Hakon Hakonarson
- Division of Human Genetics, Department of Pediatrics, at The Children's Hospital of Philadelphia, and The Perelman School of Medicine at The University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Joan E. Bailey-Wilson
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Pierre Russo
- Division of Anatomic Pathology, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Rebecca G. Wells
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kathleen M. Loomes
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics at The Children's Hospital of Philadelphia, and The Perelman School of Medicine at The University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Nancy B. Spinner
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, and The Perelman School of Medicine at The University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Marcella Devoto
- Division of Human Genetics, Department of Pediatrics, at The Children's Hospital of Philadelphia, and The Perelman School of Medicine at The University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| |
Collapse
|
40
|
Shadrina A, Tsepilov Y, Sokolova E, Smetanina M, Voronina E, Pakhomov E, Sevost'ianova K, Shevela A, Ilyukhin E, Seliverstov E, Zolotukhin I, Filipenko M. Genome-wide association study in ethnic Russians suggests an association of the MHC class III genomic region with the risk of primary varicose veins. Gene 2018; 659:93-99. [DOI: 10.1016/j.gene.2018.03.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 02/16/2018] [Accepted: 03/14/2018] [Indexed: 10/17/2022]
|
41
|
Shadrina A, Tsepilov Y, Smetanina M, Voronina E, Seliverstov E, Ilyukhin E, Kirienko A, Zolotukhin I, Filipenko M. Polymorphisms of genes involved in inflammation and blood vessel development influence the risk of varicose veins. Clin Genet 2018; 94:191-199. [PMID: 29660117 DOI: 10.1111/cge.13362] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/06/2018] [Accepted: 04/09/2018] [Indexed: 12/29/2022]
Abstract
Heredity plays an important role in the etiology of varicose veins (VVs). However, the genetic basis underlying this condition remains poorly understood. Our aim was to replicate top association signals from genome-wide association studies (GWASs) for VVs of lower extremities using 2 independent datasets-our sample of ethnic Russian individuals (709 cases and 278 controls) and a large cohort of British residents from UK Biobank (10 861 cases and 397 594 controls). Associations of polymorphisms rs11121615, rs6712038, rs507666, rs966562, rs7111987, rs6062618, and rs6905288 were validated in the UK Biobank individuals at a Bonferroni-corrected significance level. In Russian cohort, only rs11121615 reached a nominal significance level of P < .05. Results of original GWAS and replication studies were combined by a meta-analysis, and polymorphisms listed above as well as rs111434909 and rs4463578 passed a genome-wide significant threshold. Notably, the majority of these polymorphisms were located within or near genes involved in vascular development and remodeling, and regulation of inflammatory response. Our results confirm the role of these polymorphisms in genetic susceptibility to VVs and indicate the revealed genomic regions as good candidates for further fine-mapping studies and functional analysis. Moreover, our findings implicate inflammation and abnormal vascular architecture in VVs pathogenesis.
Collapse
Affiliation(s)
- A Shadrina
- Laboratory of Pharmacogenomics, Institute of Chemical Biology and Fundamental Medicine, Theoretical and Applied Functional Genomics Laboratory, Novosibirsk State University, Novosibirsk, Russia
| | - Y Tsepilov
- Laboratory of Recombination and Segregation Analysis, Institute of Cytology and Genetics, Theoretical and Applied Functional Genomics Laboratory, Novosibirsk State University, Novosibirsk, Russia
| | - M Smetanina
- Laboratory of Pharmacogenomics, Institute of Chemical Biology and Fundamental Medicine, Department of Fundamental Medicine, Novosibirsk State University, Novosibirsk, Russia
| | - E Voronina
- Laboratory of Pharmacogenomics, Institute of Chemical Biology and Fundamental Medicine, Department of Natural Sciences, Novosibirsk State University, Moscow, Russia
| | - E Seliverstov
- Department of Faculty Surgery, Pirogov Russian National Research Medical University, Moscow, Russia
| | - E Ilyukhin
- Private Surgery Center "Medalp", Saint Petersburg, Russia
| | - A Kirienko
- Department of Faculty Surgery, Pirogov Russian National Research Medical University, Moscow, Russia
| | - I Zolotukhin
- Department of Faculty Surgery, Pirogov Russian National Research Medical University, Laboratory of Pharmacogenomics, Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
| | - M Filipenko
- Laboratory of Pharmacogenomics, Institute of Chemical Biology and Fundamental Medicine, Department of Natural Sciences, Novosibirsk State University, Moscow, Russia
| |
Collapse
|
42
|
Yamada Y, Horibe H, Oguri M, Sakuma J, Takeuchi I, Yasukochi Y, Kato K, Sawabe M. Identification of novel hyper- or hypomethylated CpG sites and genes associated with atherosclerotic plaque using an epigenome-wide association study. Int J Mol Med 2018; 41:2724-2732. [PMID: 29436575 PMCID: PMC5846673 DOI: 10.3892/ijmm.2018.3453] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/23/2018] [Indexed: 12/12/2022] Open
Abstract
DNA methylation is an important epigenetic modification that has been implicated in the pathogenesis of atherosclerosis. Although previous studies have identified various CpG sites and genes whose methylation is associated with atherosclerosis in populations with European or Mexican ancestry, the genome‑wide pattern of DNA methylation in the atherosclerotic human aorta is yet to be elucidated in Japanese individuals. In the present study, a genome‑wide analysis of DNA methylation at ~853,000 CpG sites was performed using 128 postmortem aortic intima specimens obtained from 64 Japanese patients. To avoid the effects of interindividual variation, intraindividual paired comparisons were performed between atheromatous plaque lesions and corresponding plaque‑free tissue for each patient. Bisulfite‑modified genomic DNA was analyzed using a specific microarray for DNA methylation. DNA methylation at each CpG site was calculated as the β value, where β = (intensity of the methylated allele)/(intensity of the methylated allele + intensity of the unmethylated allele + 100). Bonferroni's correction for statistical significance of association was applied to compensate for multiple comparisons. The methylation of 2,679 CpG sites differed significantly (P<5.86x10‑8) between atheromatous plaque lesions and the corresponding plaque‑free intima, with 2,272 and 407 CpG sites in atheromatous plaques being hyper‑ or hypomethylated, respectively. A total of 5 hypermethylated CpG sites in atheromatous plaques were demonstrated to have a difference in β value of >0.15 (plaque lesion‑plaque‑free intima) and 11 had a β ratio of >1.50 (plaque/plaque‑free intima). A further 15 and 17 hypomethylated CpG sites in atheromatous plaques were observed to have a difference in β value of <‑0.15 or a β ratio of <0.67, respectively. According to these limits, a total of 16 novel genes that were significantly hyper‑ or hypomethylated in atheromatous plaque lesions compared with the plaque‑free intima were identified in the present study. The results of the present study suggest that the methylation of these genes may contribute to the pathogenesis of atherosclerosis in the Japanese population.
Collapse
Affiliation(s)
- Yoshiji Yamada
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, Tsu 514-8507
- CREST, Japan Science and Technology Agency, Kawaguchi 332-0012
| | - Hideki Horibe
- Department of Cardiovascular Medicine, Gifu Prefectural Tajimi Hospital, Tajimi 507-8522
| | - Mitsutoshi Oguri
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, Tsu 514-8507
- Department of Cardiology, Kasugai Municipal Hospital, Kasugai 486-8510
| | - Jun Sakuma
- CREST, Japan Science and Technology Agency, Kawaguchi 332-0012
- Computer Science Department, College of Information Science, University of Tsukuba, Tsukuba 305-8573
- RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027
| | - Ichiro Takeuchi
- CREST, Japan Science and Technology Agency, Kawaguchi 332-0012
- RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027
- Department of Computer Science, Nagoya Institute of Technology, Nagoya 466-8555
| | - Yoshiki Yasukochi
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, Tsu 514-8507
- CREST, Japan Science and Technology Agency, Kawaguchi 332-0012
| | - Kimihiko Kato
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, Tsu 514-8507
- Department of Internal Medicine, Meitoh Hospital, Nagoya 465-0025
| | - Motoji Sawabe
- Section of Molecular Pathology, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| |
Collapse
|
43
|
|