1
|
Su X, Zhang M, Yang G, Cui X, Yuan X, Du L, Pei Y. Bioinformatics and machine learning approaches reveal key genes and underlying molecular mechanisms of atherosclerosis: A review. Medicine (Baltimore) 2024; 103:e38744. [PMID: 39093811 PMCID: PMC11296484 DOI: 10.1097/md.0000000000038744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/07/2024] [Indexed: 08/04/2024] Open
Abstract
Atherosclerosis (AS) causes thickening and hardening of the arterial wall due to accumulation of extracellular matrix, cholesterol, and cells. In this study, we used comprehensive bioinformatics tools and machine learning approaches to explore key genes and molecular network mechanisms underlying AS in multiple data sets. Next, we analyzed the correlation between AS and immune fine cell infiltration, and finally performed drug prediction for the disease. We downloaded GSE20129 and GSE90074 datasets from the Gene expression Omnibus database, then employed the Cell-type Identification By Estimating Relative Subsets Of RNA Transcripts algorithm to analyze 22 immune cells. To enrich for functional characteristics, the black module correlated most strongly with T cells was screened with weighted gene co-expression networks analysis. Functional enrichment analysis revealed that the genes were mainly enriched in cell adhesion and T-cell-related pathways, as well as NF-κ B signaling. We employed the Lasso regression and random forest algorithms to screen out 5 intersection genes (CCDC106, RASL11A, RIC3, SPON1, and TMEM144). Pathway analysis in gene set variation analysis and gene set enrichment analysis revealed that the key genes were mainly enriched in inflammation, and immunity, among others. The selected key genes were analyzed by single-cell RNA sequencing technology. We also analyzed differential expression between these 5 key genes and those involved in iron death. We found that ferroptosis genes ACSL4, CBS, FTH1 and TFRC were differentially expressed between AS and the control groups, RIC3 and FTH1 were significantly negatively correlated, whereas SPON1 and VDAC3 were significantly positively correlated. Finally, we used the Connectivity Map database for drug prediction. These results provide new insights into AS genetic regulation.
Collapse
Affiliation(s)
- Xiaoxue Su
- Vascular Surgery Department of Weifang Yidu Central Hospital, Weifang, Shandong, China
| | - Meng Zhang
- Vascular Surgery Department of Weifang Yidu Central Hospital, Weifang, Shandong, China
| | - Guinan Yang
- Department of Urology, People’s Hospital of Qingdao West Coast New Area, Qingdao, Shandong, China
| | - Xuebin Cui
- Vascular Surgery Department of Weifang Yidu Central Hospital, Weifang, Shandong, China
| | | | | | - Yuanmin Pei
- Vascular Surgery Department of Weifang Yidu Central Hospital, Weifang, Shandong, China
| |
Collapse
|
2
|
Shah AM, Myhre PL, Arthur V, Dorbala P, Rasheed H, Buckley LF, Claggett B, Liu G, Ma J, Nguyen NQ, Matsushita K, Ndumele C, Tin A, Hveem K, Jonasson C, Dalen H, Boerwinkle E, Hoogeveen RC, Ballantyne C, Coresh J, Omland T, Yu B. Large scale plasma proteomics identifies novel proteins and protein networks associated with heart failure development. Nat Commun 2024; 15:528. [PMID: 38225249 PMCID: PMC10789789 DOI: 10.1038/s41467-023-44680-3] [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: 03/02/2023] [Accepted: 12/21/2023] [Indexed: 01/17/2024] Open
Abstract
Heart failure (HF) causes substantial morbidity and mortality but its pathobiology is incompletely understood. The proteome is a promising intermediate phenotype for discovery of novel mechanisms. We measured 4877 plasma proteins in 13,900 HF-free individuals across three analysis sets with diverse age, geography, and HF ascertainment to identify circulating proteins and protein networks associated with HF development. Parallel analyses in Atherosclerosis Risk in Communities study participants in mid-life and late-life and in Trøndelag Health Study participants identified 37 proteins consistently associated with incident HF independent of traditional risk factors. Mendelian randomization supported causal effects of 10 on HF, HF risk factors, or left ventricular size and function, including matricellular (e.g. SPON1, MFAP4), senescence-associated (FSTL3, IGFBP7), and inflammatory (SVEP1, CCL15, ITIH3) proteins. Protein co-regulation network analyses identified 5 modules associated with HF risk, two of which were influenced by genetic variants that implicated trans hotspots within the VTN and CFH genes.
Collapse
Affiliation(s)
- Amil M Shah
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| | - Peder L Myhre
- Akershus University Hospital and K.G. Jebsen Center for Cardiac Biomarkers, University of Oslo, Oslo, Norway
| | - Victoria Arthur
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Pranav Dorbala
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Humaira Rasheed
- Akershus University Hospital and K.G. Jebsen Center for Cardiac Biomarkers, University of Oslo, Oslo, Norway
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Public Health and Nursing, HUNT Research Center, Norwegian University of Science and Technology, Trondheim, Norway
| | - Leo F Buckley
- Department of Pharmacy, Brigham and Women's Hospital, Boston, MA, USA
| | - Brian Claggett
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Guning Liu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Jianzhong Ma
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Ngoc Quynh Nguyen
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Chiadi Ndumele
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Adrienne Tin
- University of Mississippi Medical Center, Jackson, MS, USA
| | - Kristian Hveem
- Department of Public Health and Nursing, HUNT Research Center, Norwegian University of Science and Technology, Trondheim, Norway
| | - Christian Jonasson
- Department of Public Health and Nursing, HUNT Research Center, Norwegian University of Science and Technology, Trondheim, Norway
| | - Håvard Dalen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Cardiology, St Olavs University Hospital, Trondheim, Norway
- Department of Internal Medicine, Levanger Hospital, Levanger, Norway
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Ron C Hoogeveen
- Division of Cardiology, Baylor College of Medicine, Houston, TX, USA
| | | | - Josef Coresh
- Departments of Medicine and Population Health, NYU Langone Health, New York, NY, USA
| | - Torbjørn Omland
- Akershus University Hospital and K.G. Jebsen Center for Cardiac Biomarkers, University of Oslo, Oslo, Norway
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| |
Collapse
|
3
|
Srialluri N, Surapaneni A, Schlosser P, Chen TK, Schmidt IM, Rhee EP, Coresh J, Grams ME. Circulating Proteins and Mortality in CKD: A Proteomics Study of the AASK and ARIC Cohorts. Kidney Med 2023; 5:100714. [PMID: 37711886 PMCID: PMC10498294 DOI: 10.1016/j.xkme.2023.100714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023] Open
Abstract
Rationale & Objective Proteomics could provide pathophysiologic insight into the increased risk of mortality in patients with chronic kidney disease (CKD). This study aimed to investigate associations between the circulating proteome and all-cause mortality among patients with CKD. Study Design Observational cohort study. Setting & Participants Primary analysis in 703 participants in the African American Study of Kidney Disease and Hypertension (AASK) and validation in 1,628 participants with CKD in the Atherosclerosis Risk in Communities (ARIC) study who attended visit 5. Exposure Circulating proteins. Outcome All-cause mortality. Analytical Approach Among AASK participants, we evaluated the associations of 6,790 circulating proteins with all-cause mortality using multivariable Cox proportional hazards models. Proteins with significant associations were further studied in ARIC Visit 5 participants with CKD. Results In the AASK cohort, the mean age was 54.5 years, 271 (38.5%) were women, and the mean measured glomerular filtration rate (GFR) was 46 mL/min/1.73 m2. The median follow-up was 9.6 years, and 7 distinct proteins were associated with all-cause mortality at the Bonferroni-level threshold (P < 0.05 of the 6,790) after adjustment for demographics and clinical factors, including baseline measured estimated GFR and proteinuria. In the ARIC visit 5 cohort, the mean age was 77.2 years, 903 (55.5%) were women, the mean estimated GFR was 54 mL/min/1.73 m2 and median follow-up was 6.9 years. Of the 7 proteins found in AASK, 3 (β2-microglobulin, spondin-1, and N-terminal pro-brain natriuretic peptide) were available in the ARIC data, with all 3 significantly associated with death in ARIC. Limitations Possibility of unmeasured confounding. Cause of death was not known. Conclusions Using large-scale proteomic analysis, proteins were reproducibly associated with mortality in 2 cohorts of participants with CKD. Plain-Language Summary Patients with chronic kidney disease (CKD) have a high risk of premature death, with various pathophysiological processes contributing to this increased risk of mortality. This observational cohort study aimed to investigate the associations between circulating proteins and all-cause mortality in patients with CKD using large-scale proteomic analysis. The study analyzed data from the African American Study of Kidney Disease and Hypertension (AASK) study and validated the findings in the Atherosclerosis Risk in Communities (ARIC) Study. A total of 6,790 circulating proteins were evaluated in AASK, and 7 proteins were significantly associated with all-cause mortality. Three of these proteins (β2-microglobulin, spondin-1, and N-terminal pro-brain natriuretic peptide (BNP)) were also measured in ARIC and were significantly associated with death. Additional studies assessing biomarkers associated with mortality among patients with CKD are needed to evaluate their use in clinical practice.
Collapse
Affiliation(s)
- Nityasree Srialluri
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, Maryland
| | - Aditya Surapaneni
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
- Division of Precision Medicine, Department of Medicine, New York University, New York, New York
| | - Pascal Schlosser
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Teresa K. Chen
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
- Kidney Health Research Collaborative; Division of Nephrology, Department of Medicine, University of California San Francisco and San Francisco VA Health Care System, San Francisco, California
| | - Insa M. Schmidt
- Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
| | - Eugene P. Rhee
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts
| | - Josef Coresh
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, Maryland
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Morgan E. Grams
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
- Division of Precision Medicine, Department of Medicine, New York University, New York, New York
| |
Collapse
|
4
|
Huo Y, Yang J, Zheng J, Xu D, Yang M, Tao L, Yao H, Fu X, Yang J, Liu D, Hua R, Zhang J, Sun Y, Hu L, Liu W. Increased SPON1 promotes pancreatic ductal adenocarcinoma progression by enhancing IL-6 trans-signalling. Cell Prolif 2022; 55:e13237. [PMID: 35487760 PMCID: PMC9136514 DOI: 10.1111/cpr.13237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 03/15/2022] [Accepted: 04/07/2022] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVES This study investigated the specific molecular mechanism and the roles of extracellular matrix protein Spondin 1 (SPON1) in the development of pancreatic ductal adenocarcinoma (PDAC). MATERIALS AND METHODS The expression pattern and clinical relevance of SPON1 was determined in GEO, Ren Ji and TCGA datasets, further validated by immunohistochemical staining and Kaplan-Meier analysis. Loss and gain of function experiments were employed to investigate the cellular function of SPON1 in vitro. Gene set enrichment analysis, luciferase assay, immunofluorescence and Western blot and immunoprecipitation were applied to reveal the underlying molecular mechanisms. Subcutaneous xenograft model was used to test the role of SPON1 in tumour growth and maintenance in vivo. RESULTS SPON1 is significantly upregulated in PDAC tumour tissues and correlated with progression of PDAC. Loss and gain of function experiments showed that SPON1 promotes the growth and colony formation ability of pancreatic cancer cells. Combining bioinformatics assays and experimental signalling evidences, we found that SPON1 can enhance the IL-6/JAK/STAT3 signalling. Mechanistically, SPON1 exerts its oncogenic roles in pancreatic cancer by maintaining IL-6R trans-signalling through stabilizing the interaction of soluble IL-6R (sIL-6R) and glycoprotein-130 (gp130) in PDAC cells. Furthermore, SPON1 depletion greatly reduced the tumour burden, exerted positive effect with gemcitabine, prolonging PDAC mice overall survival. CONCLUSIONS Our data indicate that SPON1 expression is dramatically increased in PDAC and that SPON1 promotes tumorigenicity by activating the sIL-6R/gp130/STAT3 axis. Collectively, our current work suggests SPON1 may be a potential therapy target for PDAC patient.
Collapse
Affiliation(s)
- Yanmiao Huo
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Jian Yang
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Jiahao Zheng
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Dapeng Xu
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Minwei Yang
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Lingye Tao
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Hongfei Yao
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Xueliang Fu
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Jianyu Yang
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Dejun Liu
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Rong Hua
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Junfeng Zhang
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Yongwei Sun
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Lipeng Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| | - Wei Liu
- Department of Biliary‐Pancreatic Surgery, Ren Ji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiPeople's Republic of China
| |
Collapse
|
5
|
Wallentin L, Eriksson N, Olszowka M, Grammer TB, Hagström E, Held C, Kleber ME, Koenig W, März W, Stewart RAH, White HD, Åberg M, Siegbahn A. Plasma proteins associated with cardiovascular death in patients with chronic coronary heart disease: A retrospective study. PLoS Med 2021; 18:e1003513. [PMID: 33439866 PMCID: PMC7817029 DOI: 10.1371/journal.pmed.1003513] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 01/20/2021] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Circulating biomarkers are associated with the development of coronary heart disease (CHD) and its complications by reflecting pathophysiological pathways and/or organ dysfunction. We explored the associations between 157 cardiovascular (CV) and inflammatory biomarkers and CV death using proximity extension assays (PEA) in patients with chronic CHD. METHODS AND FINDINGS The derivation cohort consisted of 605 cases with CV death and 2,788 randomly selected non-cases during 3-5 years follow-up included in the STabilization of Atherosclerotic plaque By Initiation of darapLadIb TherapY (STABILITY) trial between 2008 and 2010. The replication cohort consisted of 245 cases and 1,042 non-cases during 12 years follow-up included in the Ludwigshafen Risk and Cardiovascular Health (LURIC) study between 1997 and 2000. Biomarker levels were measured with conventional immunoassays and/or with the OLINK PEA panels CVD I and Inflammation. Associations with CV death were evaluated by Random Survival Forest (RF) and Cox regression analyses. Both cohorts had the same median age (65 years) and 20% smokers, while there were slight differences in male sex (82% and 76%), hypertension (70% and 78%), and diabetes (39% and 30%) in the respective STABILITY and LURIC cohorts. The analyses identified 18 biomarkers with confirmed independent association with CV death by Boruta analyses and statistical significance (all p < 0.0001) by Cox regression when adjusted for clinical characteristics in both cohorts. Most prognostic information was carried by N-terminal prohormone of brain natriuretic peptide (NTproBNP), hazard ratio (HR for 1 standard deviation [SD] increase of the log scale of the distribution of the biomarker in the replication cohort) 2.079 (95% confidence interval [CI] 1.799-2.402), and high-sensitivity troponin T (cTnT-hs) HR 1.715 (95% CI 1.491-1.973). The other proteins with independent associations were growth differentiation factor 15 (GDF-15) HR 1.728 (95% CI 1.527-1.955), transmembrane immunoglobulin and mucin domain protein (TIM-1) HR 1.555 (95% CI 1.362-1.775), renin HR 1.501 (95% CI 1.305-1.727), osteoprotegerin (OPG) HR 1.488 (95% CI 1.297-1.708), soluble suppression of tumorigenesis 2 protein (sST2) HR 1.478 (95% CI 1.307-1.672), cystatin-C (Cys-C) HR 1.370 (95% CI 1.243-1.510), tumor necrosis factor-related apoptosis-inducing ligand receptor 2 (TRAIL-R2) HR 1.205 (95% CI 1.131-1.285), carbohydrate antigen 125 (CA-125) HR 1.347 (95% CI 1.226-1.479), brain natriuretic peptide (BNP) HR 1.399 (95% CI 1.255-1.561), interleukin 6 (IL-6) HR 1.478 (95% CI 1.316-1.659), hepatocyte growth factor (HGF) HR 1.259 (95% CI 1.134-1.396), spondin-1 HR 1.295 (95% CI 1.156-1.450), fibroblast growth factor 23 (FGF-23) HR 1.349 (95% CI 1.237-1.472), chitinase-3 like protein 1 (CHI3L1) HR 1.284 (95% CI 1.129-1.461), tumor necrosis factor receptor 1 (TNF-R1) HR 1.486 (95% CI 1.307-1.689), and adrenomedullin (AM) HR 1.750 (95% CI 1.490-2.056). The study is limited by the differences in design, size, and length of follow-up of the 2 studies and the lack of results from coronary angiograms and follow-up of nonfatal events. CONCLUSIONS Profiles of levels of multiple plasma proteins might be useful for the identification of different pathophysiological pathways associated with an increased risk of CV death in patients with chronic CHD. TRIAL REGISTRATION ClinicalTrials.gov NCT00799903.
Collapse
Affiliation(s)
- Lars Wallentin
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
- Uppsala Clinical Research Center (UCR), Uppsala University, Uppsala, Sweden
- * E-mail: (LW); (AS)
| | - Niclas Eriksson
- Uppsala Clinical Research Center (UCR), Uppsala University, Uppsala, Sweden
| | - Maciej Olszowka
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
- Uppsala Clinical Research Center (UCR), Uppsala University, Uppsala, Sweden
| | - Tanja B. Grammer
- Mannheim Institute of Public Health, Social and Preventive Medicine, Mannheim Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Emil Hagström
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
- Uppsala Clinical Research Center (UCR), Uppsala University, Uppsala, Sweden
| | - Claes Held
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
- Uppsala Clinical Research Center (UCR), Uppsala University, Uppsala, Sweden
| | - Marcus E. Kleber
- Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
| | - Winfried März
- Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Mannheim and Augsburg, Germany
| | - Ralph A. H. Stewart
- Green Lane Cardiovascular Service, Auckland City Hospital and University of Auckland, Auckland, New Zealand
| | - Harvey D. White
- Green Lane Cardiovascular Service, Auckland City Hospital and University of Auckland, Auckland, New Zealand
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Agneta Siegbahn
- Uppsala Clinical Research Center (UCR), Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- * E-mail: (LW); (AS)
| |
Collapse
|
6
|
Klimczak-Tomaniak D, Bouwens E, Schuurman AS, Akkerhuis KM, Constantinescu A, Brugts J, Westenbrink BD, van Ramshorst J, Germans T, Pączek L, Umans V, Boersma E, Kardys I. Temporal patterns of macrophage- and neutrophil-related markers are associated with clinical outcome in heart failure patients. ESC Heart Fail 2020; 7:1190-1200. [PMID: 32196993 PMCID: PMC7261550 DOI: 10.1002/ehf2.12678] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 02/17/2020] [Accepted: 02/22/2020] [Indexed: 12/18/2022] Open
Abstract
AIMS Evidence on the association of macrophage- and neutrophil-related blood biomarkers with clinical outcome in heart failure patients is limited, and, with the exception of C-reactive protein, no data exist on their temporal evolution. We aimed to investigate whether temporal patterns of these biomarkers are related to clinical outcome in patients with stable chronic heart failure (CHF). METHODS AND RESULTS In 263 patients with CHF, we performed serial plasma measurements of scavenger receptor cysteine-rich type 1 protein M130 (CD163), tartrate-resistant acid phosphatase type 5 (TRAP), granulins (GRN), spondin-1 (SPON1), peptidoglycan recognition protein 1 (PGLYRP1), and tissue factor pathway inhibitor (TFPI). The Cardiovascular Panel III (Olink Proteomics AB, Uppsala, Sweden) was used. During 2.2 years of follow-up, we collected 1984 samples before the occurrence of the composite primary endpoint (PE) or censoring. For efficiency, we selected 567 samples for the measurements (all baseline samples, the last two samples preceding the PE, and the last sample before censoring in event-free patients). The relationship between repeatedly measured biomarker levels and the PE was evaluated by joint models. Mean (±standard deviation) age was 67 ± 13 years; 189 (72%) were men; left ventricular ejection fraction (%) was 32 ± 11. During follow-up, 70 (27%) patients experienced the PE. Serially measured biomarkers predicted the PE in a multivariable model adjusted for baseline clinical characteristics [hazard ratio (95% confidence interval) per 1-standard deviation change in biomarker]: CD163 [2.07(1.47-2.98), P < 0.001], TRAP [0.62 (0.43-0.90), P = 0.009], GRN [2.46 (1.64-3.84), P < 0.001], SPON1 [3.94 (2.50-6.50), P < 0.001], and PGLYRP1 [1.62 (1.14-2.31), P = 0.006]. CONCLUSIONS Changes in plasma levels of CD163, TRAP, GRN, SPON1, and PGLYRP1 precede adverse cardiovascular events in patients with CHF.
Collapse
Affiliation(s)
- Dominika Klimczak-Tomaniak
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Immunology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland.,Division of Heart Failure and Cardiac Rehabilitation, Medical University of Warsaw, Warsaw, Poland
| | - Elke Bouwens
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Anne-Sophie Schuurman
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - K Martijn Akkerhuis
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Alina Constantinescu
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jasper Brugts
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - B Daan Westenbrink
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan van Ramshorst
- Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | - Tjeerd Germans
- Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | - Leszek Pączek
- Department of Immunology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Victor Umans
- Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | - Eric Boersma
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Isabella Kardys
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
7
|
Wang G, Wu L, Chen Z, Sun J. Identification of crucial miRNAs and the targets in renal cortex of hypertensive patients by expression profiles. Ren Fail 2017; 39:92-99. [PMID: 27802793 PMCID: PMC6014400 DOI: 10.1080/0886022x.2016.1244083] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 09/04/2016] [Accepted: 09/16/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUNDS Defect in kidney is one major reason of hypertension. The study aimed ao uncovering the regulatory mechanisms of miRNAs and the targets in hypertensive kidney. METHODS Gene expression profile of GSE28345 and miRNA expression profile of GSE28283 were downloaded from GEO database. After data preprocessing, differently expressed genes (DEGs) and miRNAs (DE-miRs) were identified using limma package. Then targets of miRNAs were predicted according to information in relevant databases. Function and pathway enrichment analyses were performed for DEGs using DAVID software. Furthermore, protein-protein interaction (PPI) networks were constructed for up- and down-regulated genes, respectively, using the Cytoscape. Additionally, for down-regulated DEGs, the integrated regulatory network was established combining PPI network with the miRNA-mRNA interactions. RESULTS As a result, 285 DEGs were identified, including 177 up-regulated and 108 down-regulated genes. Combined with the predicted targets of miRNAs, 22 up-regulated DE-miRs were identified. In the integrated network for down-regulated DEGs, three crucial nodes were identified as ASPN, COL12A1, and SCN2A. ASPN was predicted as target of miR-21 and miR-374b, and COL12A1 was the target of miR-30e, miR-21, and miR-195, while SCN2A was the target of miR-30e, miR-374b, and miR-195. Notably, COL12A1 and ASPN were linked with each other in the network. CONCLUSION Three crucial genes were identified in hypertensive kidney, such as COL12A1, ASPN, and SCN2A. ASPN might co-function with COL12A1, and they both might be the targets of miR-21. SCN2A might be a novel target of miR-30e and miR-374b. However, more experiments are needed to validate these results.
Collapse
Affiliation(s)
- Guohua Wang
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, China
| | - Lan Wu
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, China
| | - Zhi Chen
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Jinghui Sun
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
8
|
Stenemo M, Nowak C, Byberg L, Sundström J, Giedraitis V, Lind L, Ingelsson E, Fall T, Ärnlöv J. Circulating proteins as predictors of incident heart failure in the elderly. Eur J Heart Fail 2017; 20:55-62. [PMID: 28967680 DOI: 10.1002/ejhf.980] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/30/2017] [Accepted: 07/29/2017] [Indexed: 12/20/2022] Open
Abstract
AIMS To identify novel risk markers for incident heart failure using proteomic profiling of 80 proteins previously associated with cardiovascular pathology. METHODS AND RESULTS Proteomic profiling (proximity extension assay) was performed in two community-based prospective cohorts of elderly individuals without heart failure at baseline: the Prospective Investigation of the Vasculature in Uppsala Seniors [PIVUS, n = 901, median age 70.2 (interquartile range 70.0-70.3) years, 80 events]; and the Uppsala Longitudinal Study of Adult Men [ULSAM, n = 685, median age 77.8 (interquartile range 76.9-78.1) years, 90 events]. Twenty-nine proteins were associated with incident heart failure in the discovery cohort PIVUS after adjustment for age and sex, and correction for multiple testing. Eighteen associations replicated in ULSAM. In pooled analysis of both cohorts, higher levels of nine proteins were associated with incident heart failure after adjustment for established risk factors: growth differentiation factor 15 (GDF-15), T-cell immunoglobulin and mucin domain 1 (TIM-1), tumour necrosis factor-related apoptosis-inducing ligand receptor 2 (TRAIL-R2), spondin-1 (SPON1), matrix metalloproteinase-12 (MMP-12), follistatin (FS), urokinase-type plasminogen activator surface receptor (U-PAR), osteoprotegerin (OPG), and suppression of tumorigenicity 2 (ST2). Of these, GDF-15, U-PAR, MMP-12, TRAIL-R2, SPON1 and FS were associated with worsened echocardiographic left ventricular systolic function at baseline, while only TIM-1 was positively associated with worsened diastolic function (P < 0.02 for all). CONCLUSION Proteomic profiling identified several novel associations between proteins involved in apoptosis, inflammation, matrix remodelling, and fibrinolysis with incident heart failure in elderly individuals. Our results encourage additional studies investigating the underlying mechanisms and the clinical utility of our findings.
Collapse
Affiliation(s)
- Markus Stenemo
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Christoph Nowak
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Liisa Byberg
- Department of Surgical Sciences, Orthopedics, Uppsala University, Uppsala, Sweden
| | - Johan Sundström
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden.,Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Lars Lind
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Tove Fall
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Johan Ärnlöv
- School of Health and Social Studies, Dalarna University, Falun, Sweden.,Division of Family Medicine and Primary Care, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
9
|
Doris PA. Genetics of hypertension: an assessment of progress in the spontaneously hypertensive rat. Physiol Genomics 2017; 49:601-617. [PMID: 28916635 DOI: 10.1152/physiolgenomics.00065.2017] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The application of gene mapping methods to uncover the genetic basis of hypertension in the inbred spontaneously hypertensive rat (SHR) began over 25 yr ago. This animal provides a useful model of genetic high blood pressure, and some of its features are described. In particular, it appears to be a polygenic model of disease, and polygenes participate in human hypertension genetic risk. The SHR hypertension alleles were fixed rapidly by selective breeding in just a few generations and so are presumably common genetic variants present in the outbred Wistar strain from which SHR was created. This review provides a background to the origins and genesis of this rat line. It considers its usefulness as a model organism for a common cardiovascular disease. The progress and obstacles facing mapping are considered in depth, as are the emergence and application of other genome-wide genetic discovery approaches that have been applied to investigate this model. Candidate genes, their identification, and the evidence to support their potential role in blood pressure elevation are considered. The review assesses the progress that has arisen from this work has been limited. Consideration is given to some of the factors that have impeded progress, and prospects for advancing understanding of the genetic basis of hypertension in this model are discussed.
Collapse
Affiliation(s)
- Peter A Doris
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center, Houston, Texas
| |
Collapse
|
10
|
Deng AY, Chauvet C, Ménard A. Alterations in Fibronectin Type III Domain Containing 1 Protein Gene Are Associated with Hypertension. PLoS One 2016; 11:e0151399. [PMID: 27064407 PMCID: PMC4827815 DOI: 10.1371/journal.pone.0151399] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 02/26/2016] [Indexed: 12/02/2022] Open
Abstract
Multiple quantitative trait loci (QTLs) for blood pressure (BP) have been detected in rat models of human polygenic hypertension. Great challenges confronting us include molecular identifications of individual QTLs. We first defined the chromosome region harboring C1QTL1 to a segment of 1.9 megabases that carries 9 genes. Among them, we identified the gene encoding the fibronectin type III domain containing 1 protein (Fndc1)/activator of G protein signaling 8 (Ags8) to be the strongest candidate for C1QTL1, since numerous non-synonymous mutations are found. Moreover, the 5’ Fndc1/Ags8 putative promoter contains numerous mutations that can account for its differential expression in kidneys and the heart, prominent organs in modulating BP, although the Fndc1/Ags8 protein was not detectable in these organs under our experimental conditions. This work has provided the premier evidence that Fndc1/Ags8 is a novel and strongest candidate gene for C1QTL1 without completely excluding other 8 genes in the C1QTL1-residing interval. If proven true by future in vivo function studies such as single-gene Fndc1/Ags8 congenics, transgenesis or targeted-gene modifications, it might represent a part of the BP genetic architecture that operates in the upstream position distant from the end-phase physiology of BP control, since it activates a Gbetagamma component in a signaling pathway. Its functional role could validate the concept that a QTL in itself can influence BP ‘indirectly’ by regulating other genes downstream in a pathway. The elucidation of the mechanisms initiated by Fndc/Ags8 variations will reveal novel insights into the BP modulation via a regulatory hierarchy.
Collapse
Affiliation(s)
- Alan Y. Deng
- Research Centre, CRCHUM (Centre hospitalier de l’Université de Montréal), Department of Medicine, Université de Montréal, Montréal, Québec, Canada
- * E-mail:
| | - Cristina Chauvet
- Research Centre, CRCHUM (Centre hospitalier de l’Université de Montréal), Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Annie Ménard
- Research Centre, CRCHUM (Centre hospitalier de l’Université de Montréal), Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
11
|
Enroth S, Bosdotter Enroth S, Johansson Å, Gyllensten U. Effect of genetic and environmental factors on protein biomarkers for common non-communicable disease and use of personally normalized plasma protein profiles (PNPPP). Biomarkers 2015; 20:355-64. [PMID: 26551787 DOI: 10.3109/1354750x.2015.1093546] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To study the impact of genetic and lifestyle factors on protein biomarkers and develop personally normalized plasma protein profiles (PNPPP) controlling for non-disease-related variance. MATERIALS AND METHODS Proximity extension assays were used to measure 145 proteins in 632 controls and 344 cases with non-communicable diseases. RESULTS Genetic and lifestyle factors explained 20-88% of the variation in healthy controls. Adjusting for these factors reduced the number of candidate biomarkers by 63%. CONCLUSION PNPPP efficiently controls for non-disease-related variance, allowing both for efficient discovery of novel biomarkers and for covariate-independent linear cut-offs suitable for clinical use.
Collapse
Affiliation(s)
- Stefan Enroth
- a Department of Immunology , Genetics, and Pathology, Biomedical Center, SciLifeLab Uppsala, Uppsala University , Uppsala , Sweden and
| | | | - Åsa Johansson
- a Department of Immunology , Genetics, and Pathology, Biomedical Center, SciLifeLab Uppsala, Uppsala University , Uppsala , Sweden and
| | - Ulf Gyllensten
- a Department of Immunology , Genetics, and Pathology, Biomedical Center, SciLifeLab Uppsala, Uppsala University , Uppsala , Sweden and
| |
Collapse
|
12
|
Fan Y, Wei C, Xiao W, Zhang W, Wang N, Chuang PY, He JC. Temporal profile of the renal transcriptome of HIV-1 transgenic mice during disease progression. PLoS One 2014; 9:e93019. [PMID: 24667548 PMCID: PMC3965528 DOI: 10.1371/journal.pone.0093019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 02/27/2014] [Indexed: 01/15/2023] Open
Abstract
Profiling of temporal changes of gene expression in the same kidney over the course of renal disease progression is challenging because repeat renal biopsies are rarely indicated in clinical practice. Here, we profiled the temporal change in renal transcriptome of HIV-1 transgenic mice (Tg26), an animal model for human HIV-associated nephropathy (HIVAN), and their littermates at three different time points (4, 8, and 12 weeks of age) representing early, middle, and late stages of renal disease by serial kidney biopsy. We analyzed both static levels of gene expression at three stages of disease and dynamic changes in gene expression between different stages. Analysis of static and dynamic changes in gene expression revealed that up-regulated genes at the early and middle stages are mostly involved in immune response and inflammation, whereas down-regulated genes mostly related to fatty acid and retinoid metabolisms. We validated the expression of a selected panel of genes that are up-regulated at the early stage (CCL2, CCL5, CXCL11, Ubd, Anxa1, and Spon1) by real-time PCR. Among these up-regulated genes, Spon1, which is a previously identified candidate gene for hypertension, was found to be up-regulated in kidney of human with diabetic nephropathy. Immunostaining of human biopsy samples demonstrated that protein expression of Spon1 was also markedly increased in kidneys of patients with both early and late HIVAN and diabetic nephropathy. Our studies suggest that analysis of both static and dynamic changes of gene expression profiles in disease progression avails another layer of information that could be utilized to gain a more comprehensive understanding of disease progression and identify potential biomarkers and drug targets.
Collapse
Affiliation(s)
- Ying Fan
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chengguo Wei
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Wenzhen Xiao
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Weijia Zhang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Niansong Wang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Peter Y. Chuang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
13
|
Dreger H, Ludwig A, Weller A, Stangl V, Baumann G, Meiners S, Stangl K. Epigenetic regulation of cell adhesion and communication by enhancer of zeste homolog 2 in human endothelial cells. Hypertension 2012; 60:1176-83. [PMID: 22966008 DOI: 10.1161/hypertensionaha.112.191098] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The histone methyltransferase enhancer of zeste homolog 2 (Ezh2) mediates trimethylation of lysine 27 in histone 3, which acts as a repressive epigenetic mark. Ezh2 is essential for maintaining pluripotency of stem cells, but information on its role in differentiated cells is sparse. Whole-genome mRNA expression arrays identified 964 genes that were regulated by >2-fold 72 hours after small interfering RNA-mediated silencing of Ezh2 in human umbilical vein endothelial cells. Among them, genes associated with the gene ontology terms cell communication and cell adhesion were significantly overrepresented, suggesting a functional role for Ezh2 in the regulation of angiogenesis. Indeed, adhesion, migration, and tube formation assays revealed significantly altered angiogenic properties of human umbilical vein endothelial cells after silencing of Ezh2. To identify direct target genes of Ezh2, we performed chromatin immunoprecipitation experiments followed by whole-genome promoter arrays (chromatin immunoprecipitation-on-chip) and identified 5585 genes associated with trimethylation of lysine 27 in histone 3. Comparative analysis with our mRNA expression data identified 276 genes that met our criteria for putative Ezh2 target genes, upregulation by >2-fold after Ezh2 silencing and association with trimethylation of lysine 27 in histone 3. Notably, we observed a striking overrepresentation of genes involved in wingless-type mouse mammary tumor virus integration site (WNT) signaling pathways. Epigenetic regulation of several of these genes by Ezh2 was specifically confirmed by polymerase chain reaction analysis of DNA enrichment after chromatin immunoprecipitation using an antibody specific for trimethylation of lysine 27 in histone 3. Combining mRNA expression arrays and chromatin immunoprecipitation-on-chip analysis, we identified 276 Ezh2 target genes in endothelial cells. Ezh2-dependent repression of genes involved in cell adhesion and communication contributes to the regulation of angiogenesis.
Collapse
Affiliation(s)
- Henryk Dreger
- Medizinische Klinik für Kardiologie und Angiologie, Charité - Universitätsmedizin Berlin, Charitéplatz 1, D-10117 Berlin, Germany.
| | | | | | | | | | | | | |
Collapse
|
14
|
Friese RS, Schmid-Schönbein GW, O'Connor DT. Systematic polymorphism discovery after genome-wide identification of potential susceptibility loci in a hereditary rodent model of human hypertension. Blood Press 2011; 20:222-31. [PMID: 21428728 DOI: 10.3109/08037051.2011.566012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Genetic strategies such as linkage analysis and quantitative trait locus (QTL) mapping have identified a multitude of loci implicated in the pathogenesis of hypertension in the spontaneously hypertensive rat (SHR). While several candidate genetic regions have been identified in the SHR and its control, the Wistar-Kyoto rat (WKY), systematic follow-up of candidate identification with polymorphism discovery has not been widespread. In the current report, we develop a data-mining strategy to identify candidate genes for hypertension in the SHR, and then sequence each gene in the SHR and WKY strains. We integrate blood pressure QTL data, microarray data and data-mining methods. First, we determined the set of genes differentially expressed in SHR and WKY adrenal glands. Next, the chromosomal position of all differentially expressed genes was compared with peak marker position of all reported SHR blood pressure QTLs. We also identified the set of differentially expressed genes with the most extreme fold-change. Finally, the QTL positional candidates and the genes with extreme differential expression were proposed as candidate genes if they had biologically plausible roles in hypertensive pathology. We identified seven candidate genes that merit resequencing (catechol-O-methyltransferase [Comt], chromogranin A [Chga], dopamine beta-hydroxylase [Dbh], electron transferring flavoprotein dehydrogenase [Etfdh], endothelin receptor type B [Ednrb], neuropeptide Y [Npy] and phenylethanolamine-N-methyltransferase [Pnmt]), and then discovered polymorphism in four of these seven candidate genes. Chga is proposed as the strongest candidate for additional functional investigation. Our method for candidate gene identification is portable and can be applied to microarray data from any tissue, in any disease model with a QTL database.
Collapse
Affiliation(s)
- Ryan S Friese
- Department of Bioengineering, University of California at San Diego, USA
| | | | | |
Collapse
|
15
|
Gopalakrishnan K, Kumarasamy S, Rapp JP, Joe B. Reply to “Letter to the Editor: ‘Mapping genes for hypertension using experimental models: a challenging and unanticipated very long journey’”. Physiol Genomics 2011. [DOI: 10.1152/physiolgenomics.00230.2010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- K. Gopalakrishnan
- Physiological Genomics Laboratory, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, Ohio
| | - S. Kumarasamy
- Physiological Genomics Laboratory, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, Ohio
| | - J. P. Rapp
- Physiological Genomics Laboratory, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, Ohio
| | - B. Joe
- Physiological Genomics Laboratory, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, Ohio
| |
Collapse
|
16
|
Kinoshita K, Ashenagar MS, Tabuchi M, Higashino H. Whole rat DNA array survey for candidate genes related to hypertension in kidneys from three spontaneously hypertensive rat substrains at two stages of age and with hypotensive induction caused by hydralazine hydrochloride. Exp Ther Med 2011; 2:201-212. [PMID: 22977489 DOI: 10.3892/etm.2011.193] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Accepted: 12/28/2010] [Indexed: 11/06/2022] Open
Abstract
Clarification of the genetic nature and more effective care for hypertension are required, given the high incidences of cardiovascular and cerebrovascular mortality. Thus, we surveyed candidate genes for hypertension with rat whole gene DNA microarrays using three novel methods. Gene expression analyses were conducted as follows: Method 1, three types of spontaneously hypertensive rat (SHR) substrains, SHR, stroke-prone SHR (SHRSP) and malignant type of SHRSP (M-SHRSP) were used and compared to normotensive Wistar Kyoto rats; Method 2, the expressed genes between rats of different ages were compared for different blood pressures; and Method 3, genes that were expressed in rats treated with or without an acute hypotensive stimulus, the antihypertensive hydralazine hydrochloride, were compared. This approach identified dozens of genes, including Dusp15, Cyp8b1, Armc 3, Gtpbp4, Mettl2, Mapk14, Prkar2b, frame 12, Anxa13, Ephx2, Myr8 and Pcdh9 by Method 1; Cyp2C and Atp12a by Method 2; and Kcnc3, Vnn1, TC560558 and Gabrq and a number of unknown genes by Methods 2 and 3, as probable candidate genes for hypertension in SHR substrains. Ephx2 was previously reported as a candidate gene in SHRs; however other genes were identified for the first time in this study. Since it was not always possible to completely demonstrate that these genes are responsible for hypertension in SHRs, further research into true candidate genes that participate in the genesis of hypertension in SHR substrains is warranted.
Collapse
Affiliation(s)
- Kosho Kinoshita
- Department of Pharmacology, Kinki University School of Medicine, Osaka 589-8511, Japan
| | | | | | | |
Collapse
|
17
|
Gopalakrishnan K, Saikumar J, Peters CG, Kumarasamy S, Farms P, Yerga-Woolwine S, Toland EJ, Schnackel W, Giovannucci DR, Joe B. Defining a rat blood pressure quantitative trait locus to a <81.8 kb congenic segment: comprehensive sequencing and renal transcriptome analysis. Physiol Genomics 2010; 42A:153-61. [PMID: 20716646 PMCID: PMC2957796 DOI: 10.1152/physiolgenomics.00122.2010] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Accepted: 08/12/2010] [Indexed: 01/11/2023] Open
Abstract
Evidence from multiple linkage and genome-wide association studies suggest that human chromosome 2 (HSA2) contains alleles that influence blood pressure (BP). Homologous to a large segment of HSA2 is rat chromosome 9 (RNO9), to which a BP quantitative trait locus (QTL) was previously mapped. The objective of the current study was to further resolve this BP QTL. Eleven congenic strains with introgressed segments spanning <81.8 kb to <1.33 Mb were developed by introgressing genomic segments of RNO9 from the Dahl salt-resistant (R) rat onto the genome of the Dahl salt-sensitive (S) rat and tested for BP. The congenic strain with the shortest introgressed segment spanning <81.8 kb significantly lowered BP of the hypertensive S rat by 25 mmHg and significantly increased its mean survival by 45 days. In contrast, two other congenic strains had increased BP compared with the S. We focused on the <81.8 kb congenic strain, which represents the shortest genomic segment to which a BP QTL has been mapped to date in any species. Sequencing of this entire region in both S and R rats detected 563 variants. The region did not contain any known or predicted rat protein coding genes. Furthermore, a whole genome renal transcriptome analysis between S and the <81.8 kb S.R congenic strain revealed alterations in several critical genes implicated in renal homeostasis. Taken together, our results provide the basis for future studies to examine the relationship between the candidate variants within the QTL region and the renal differentially expressed genes as potential causal mechanisms for BP regulation.
Collapse
Affiliation(s)
- K. Gopalakrishnan
- Physiological Genomics Laboratory, Departments of Physiology and Pharmacology and
| | - J. Saikumar
- Physiological Genomics Laboratory, Departments of Physiology and Pharmacology and
| | - C. G. Peters
- Neurosciences, University of Toledo College of Medicine, Toledo, Ohio
| | - S. Kumarasamy
- Physiological Genomics Laboratory, Departments of Physiology and Pharmacology and
| | - P. Farms
- Physiological Genomics Laboratory, Departments of Physiology and Pharmacology and
| | - S. Yerga-Woolwine
- Physiological Genomics Laboratory, Departments of Physiology and Pharmacology and
| | - E. J. Toland
- Physiological Genomics Laboratory, Departments of Physiology and Pharmacology and
| | - W. Schnackel
- Physiological Genomics Laboratory, Departments of Physiology and Pharmacology and
| | - D. R. Giovannucci
- Neurosciences, University of Toledo College of Medicine, Toledo, Ohio
| | - B. Joe
- Physiological Genomics Laboratory, Departments of Physiology and Pharmacology and
| |
Collapse
|
18
|
Marques FZ, Campain AE, Yang YHJ, Morris BJ. Meta-analysis of genome-wide gene expression differences in onset and maintenance phases of genetic hypertension. Hypertension 2010; 56:319-24. [PMID: 20585107 DOI: 10.1161/hypertensionaha.110.155366] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Gene expression differences accompany both the onset and established phases of hypertension. By an integrated genome-transcriptome approach we performed a meta-analysis of data from 74 microarray experiments available on public databases to identify genes with altered expression in the kidney, adrenal, heart, and artery of spontaneously hypertensive and Lyon hypertensive rats. To identify genes responsible for the onset of hypertension we used a statistical approach that sought to eliminate expression differences that occur during maturation unrelated to hypertension. Based on this adjusted fold-difference statistic, we found 36 genes for which the expression differed between the prehypertensive phase and established hypertension. Genes having possible relevance to hypertension onset included Actn2, Ankrd1, ApoE, Cd36, Csrp3, Me1, Myl3, Nppa, Nppb, Pln, Postn, Spp1, Slc21a4, Slc22a2, Thbs4, and Tnni3. In established hypertension 102 genes exhibited altered expression after Bonferroni correction (P<0.05). These included Atp5o, Ech1, Fabp3, Gnb3, Ldhb, Myh6, Lpl, Pkkaca, Vegfb, Vcam1, and reduced nicotinamide-adenine dinucleotide dehydrogenases. Among the genes identified, there was an overrepresentation of gene ontology terms involved in energy production, fatty acid and lipid metabolism, oxidation, and transport. These could contribute to increases in reactive oxygen species. Our meta-analysis has revealed many new genes for which the expression is altered in hypertension, so pointing to novel potential causative, maintenance, and responsive mechanisms and pathways.
Collapse
Affiliation(s)
- Francine Z Marques
- Basic & Clinical Genomics Laboratory, School of Medical Sciences and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | | | | | | |
Collapse
|
19
|
Charchar FJ, Kaiser M, Bingham AJ, Fotinatos N, Ahmady F, Tomaszewski M, Samani NJ. Whole genome survey of copy number variation in the spontaneously hypertensive rat: relationship to quantitative trait loci, gene expression, and blood pressure. Hypertension 2010; 55:1231-8. [PMID: 20231529 PMCID: PMC5266550 DOI: 10.1161/hypertensionaha.109.141663] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Accepted: 02/15/2010] [Indexed: 11/16/2022]
Abstract
Copy number variation has emerged recently as an important genetic mechanism leading to phenotypic heterogeneity. The aim of our study was to determine whether copy number variants (CNVs) exist between the spontaneously hypertensive rat (SHR) and its control strain, the Wistar-Kyoto rat, whether these map to quantitative trait loci in the rat and whether CNVs associate with gene expression or blood pressure differences between the 2 strains. We performed a comparative genomic hybridization assay between SHR and Wistar-Kyoto strains using a whole-genome array. In total, 16 CNVs were identified and validated (6 because of a relative loss of copy number in the SHR and 10 because of a relative gain). CNVs were present on rat autosomes 1, 3, 4, 6, 7, 10, 14, and 17 and varied in size from 10.0 kb to 1.6 Mb. Most of these CNVs mapped to chromosomal regions within previously identified quantitative trait loci, including those for blood pressure in the SHR. Transcriptomic experiments confirmed differences in the renal expression of several genes (including Ms4a6a, Ndrg3, Egln1, Cd36, Sema3a, Ugt2b, and Idi21) located in some of the CNVs between SHR and Wistar-Kyoto rats. In F(2) animals derived from an SHRxWistar-Kyoto cross, we also found a significant increase in blood pressure associated with an increase in copy number in the Egln1 gene. Our findings suggest that CNVs may play a role in the susceptibility to hypertension and related traits in the SHR.
Collapse
Affiliation(s)
- Fadi J Charchar
- School of Science and Engineering, University of Ballarat, University Dr, Mt Helen, Ballarat, Australia 3350.
| | | | | | | | | | | | | |
Collapse
|
20
|
Chen Q, Gruber H, Swist E, Coville K, Pakenham C, Ratnayake WM, Scoggan KA. Dietary phytosterols and phytostanols decrease cholesterol levels but increase blood pressure in WKY inbred rats in the absence of salt-loading. Nutr Metab (Lond) 2010; 7:11. [PMID: 20637058 PMCID: PMC2843689 DOI: 10.1186/1743-7075-7-11] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Accepted: 02/12/2010] [Indexed: 12/04/2022] Open
Abstract
Background There are safety concerns regarding widespread consumption of phytosterol and phytostanol supplemented food products. The aim of this study was to determine, in the absence of excess dietary salt, the individual effects of excess accumulation of dietary phytosterols and phytostanols on blood pressure in Wistar Kyoto (WKY) inbred rats that have a mutation in the Abcg5 gene and thus over absorb phytosterols and phytostanols. Methods Thirty 35-day old male WKY inbred rats (10/group) were fed a control diet or a diet containing phytosterols or phytostanols (2.0 g/kg diet) for 5 weeks. The sterol composition of the diets, plasma and tissues were analysed by gas chromatography. Blood pressure was measured by the tail cuff method. mRNA levels of several renal blood pressure regulatory genes were measured by real-time quantitative PCR. Results Compared to the control diet, the phytosterol diet resulted in 3- to 4-fold increases in the levels of phytosterols in plasma, red blood cells, liver, aorta and kidney of WKY inbred rats (P < 0.05). The phytostanol diet dramatically increased (> 9-fold) the levels of phytostanols in plasma, red blood cells, liver, aorta and kidney of these rats (P < 0.05). The phytosterol diet decreased cholesterol levels by 40%, 31%, and 19% in liver, aorta and kidney, respectively (P < 0.05). The phytostanol diet decreased cholesterol levels by 15%, 16%, 20% and 14% in plasma, liver, aorta and kidney, respectively (P < 0.05). The phytostanol diet also decreased phytosterol levels by 29% to 54% in plasma and tissues (P < 0.05). Both the phytosterol and phytostanol diets produced significant decreases in the ratios of cholesterol to phytosterols and phytostanols in plasma, red blood cells, liver, aorta and kidney. Rats that consumed the phytosterol or phytostanol diets displayed significant increases in systolic and diastolic blood pressure compared to rats that consumed the control diet (P < 0.05). The phytosterol diet increased renal angiotensinogen mRNA levels of these rats. Conclusion These data suggest that excessive accumulation of dietary phytosterols and phytostanols in plasma and tissues may contribute to the increased blood pressure in WKY inbred rats in the absence of excess dietary salt. Therefore, even though phytosterols and phytostanols lower cholesterol levels, prospective clinical studies testing the net beneficial effects of dietary phytosterols and phytostanols on cardiovascular events for subgroups of individuals that have an increased incorporation of these substances are needed.
Collapse
Affiliation(s)
- Qixuan Chen
- Nutrition Research Division, Food Directorate, Health Products and Food Branch, Health Canada, Banting Research Centre, Ottawa, Ontario K1A 0K9, Canada.
| | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Diurnal rhythms influence cardiovascular physiology such as heart rate and blood pressure and the incidence of adverse cardiac events such as heart attack and stroke. For example, shift workers and patients with sleep disturbances, such as obstructive sleep apnea, have an increased risk of heart attack, stroke, and sudden death. Diurnal variation is also evident at the molecular level, as gene expression in the heart and blood vessels is remarkably different in the day as compared to the night. Much of the evidence presented here indicates that growth and renewal (structural remodeling) are highly dependent on processes that occur during the subjective night. Myocardial metabolism is also dynamic with substrate preference also differing day from night. The risk/benefit ratio of some therapeutic strategies and the appearance of biomarkers also vary across the 24-hour diurnal cycle. Synchrony between external and internal diurnal rhythms and harmony among the molecular rhythms within the cell is essential for normal organ biology. Cell physiology is 4 dimensional; the substrate and enzymatic components of a given metabolic pathway must be present not only in the right compartmental space within the cell but also at the right time. As a corollary, we show disrupting this integral relationship has devastating effects on cardiovascular, renal and possibly other organ systems. Harmony between our biology and our environment is vital to good health.
Collapse
Affiliation(s)
- Tami A Martino
- Department of Biomedical Sciences, OVC, University of Guelph, Guelph, ON, Canada, N1G2W1.
| | | |
Collapse
|
22
|
Abstract
Hypertension represents a global public health burden. In addition to the rarer Mendelian forms of hypertension, classic genetic studies have documented a significant heritable component to the most common form, essential hypertension (EH). Extensive efforts are under way to elucidate the genetic basis of this disease. Recently, a new form of Mendelian hypertension has been identified, pharmacogenetic association studies in hypertensive patients have identified novel gene-by-drug interactions, and the first genome-wide association studies of EH have been published. New findings in consomic and congenic rat models also offer new clues to the genetic architecture of this complex phenotype. In this review, the authors summarize and evaluate the most recent findings related to hypertension gene identification.
Collapse
|
23
|
Influence of dietary phytosterols and phytostanols on diastolic blood pressure and the expression of blood pressure regulatory genes in SHRSP and WKY inbred rats. Br J Nutr 2008; 102:93-101. [DOI: 10.1017/s0007114508137904] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The aim of the present study was to determine the impact of increased consumption of phytosterols or phytostanols on blood pressure and renal blood pressure regulatory gene expression in stroke-prone spontaneously hypertensive (SHRSP) and normotensive Wistar–Kyoto (WKY) inbred rats. SHRSP and WKY inbred rats (10/group) were fed a control diet or a diet supplemented with phytosterols or phytostanols (2·0 g/kg diet). After 5 weeks, SHRSP rats demonstrated higher systolic and diastolic blood pressures than WKY inbred rats. SHRSP rats that consumed the phytosterol or phytostanol supplemental diets displayed a 2- or 3-fold respective increase in the diastolic blood pressure than those that consumed the control diet. Angiotensinogen (Agt), angiotensin I-converting enzyme 1 (Ace1), nitric oxide synthase (Nos) 1,Nos3, cyclooxygenase 2 (Cox2) and THUMP domain containing 1 were expressed at higher levels in SHRSP compared with WKY inbred rats.Reninand angiotensin II receptor type 1a were expressed at lower levels in SHRSP than WKY inbred rats. Phytostanol supplementation up-regulated the expression ofAce1andNos3in SHRSP rats. Phytosterol supplementation increased the mRNA levels ofNos1and spondin 1 (Spon1) in SHRSP and WKY inbred rats.Cox2mRNA levels were elevated in both phytosterol- and phytostanol-supplemented SHRSP and WKY inbred rats. Therefore, the increased blood pressure in SHRSP rats may be partly due to altered renal expression of blood pressure regulatory genes. Specifically, up-regulation ofAce1,Nos1,Nos3,Cox2andSpon1were associated with the increased diastolic blood pressure observed in phytosterol- or phytostanol-supplemented SHRSP rats.
Collapse
|
24
|
Blood pressure and proteinuria effects of multiple quantitative trait loci on rat chromosome 9 that differentiate the spontaneously hypertensive rat from the Dahl salt-sensitive rat. J Hypertens 2008; 26:2134-41. [PMID: 18854752 DOI: 10.1097/hjh.0b013e32830ef95c] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A blood pressure (BP) quantitative trait locus (QTL) was previously located within 117 kb on rat chromosome 9 (RNO9) using hypertensive Dahl salt-sensitive and normotensive Dahl salt-resistant rats. An independent study between two hypertensive rat strains, the Dahl salt-sensitive rat and the spontaneously hypertensive rat (SHR), also detected a QTL encompassing this 117 kb region. Dahl salt-sensitive alleles in both of these studies were associated with increased BP. To map SHR alleles that decrease BP in the Dahl salt-sensitive rat, a panel of eight congenic strains introgressing SHR alleles onto the Dahl salt-sensitive genetic background were constructed and characterized. S.SHR(9)x3B, S.SHR(9)x3A and S.SHR(9)x2B, the congenic regions of which span a portion or all of the 1 logarithm of odds (LOD) interval identified by linkage analysis, did not significantly alter BP. However, S.SHR(9), S.SHR(9)x4A, S.SHR(9)x7A, S.SHR(9)x8A and S.SHR(9)x10A, the introgressed segments of which extend distal to the 1 LOD interval, significantly reduced BP. The shortest genomic segment, BP QTL1, to which this BP-lowering effect can be traced is the differential segment of S.SHR(9)x4A and S.SHR(9)x2B, to which an urinary protein excretion QTL also maps. However, the introgressed segment of S.SHR(9)x10A, located outside of this QTL1 region, represented a second BP QTL (BP QTL2) having no detectable effects on urinary protein excretion. In summary, the data suggest that there are multiple RNO9 alleles of the SHR that lower BP of the Dahl salt-sensitive rat with or without detectable effects on urinary protein excretion and that only one of these BP QTLs, QTL1, overlaps with the 117 kb BP QTL region identified using Dahl salt-sensitive and Dahl salt-resistant rats.
Collapse
|
25
|
Wang T, Takabatake T, Kobayashi Y, Nabika T. Sympathetic regulation of renal function in stroke-prone spontaneously hypertensive rats congenic for chromosome 1 blood pressure quantitative trait loci. Clin Exp Pharmacol Physiol 2008; 35:1365-70. [PMID: 18565192 DOI: 10.1111/j.1440-1681.2008.04990.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
1. Two reciprocal congenic strains, WKYpch1.0 and SHRSPwch1.0, were constructed, respectively, by introgressing the stroke-prone spontaneously hypertensive rat (SHRSP)-derived fragment for the chromosome 1 blood pressure (BP) quantitative trait locus (QTL) into Wistar-Kyoto (WKY) rats and vice versa. 2. Under basal conditions with intact renal sympathetic nerves, the renal noradrenaline content and renal vascular resistance (RVR) were decreased in the order of SHRSP, SHRSPwch1.0, WKYpch1.0 and WKY, exhibiting reciprocal changes in the congenic strains according to the genotype of the chromosome 1 QTL. 3. Renal denervation resulted in significant effects on RVR and the fractional excretion of sodium only in SHRSP and WKYpch1.0, both of which harboured the SHRSP-derived fragment of chromosome 1 QTL. 4. Thus, chromosome 1 QTL may influence both renal sympathetic nervous activity and the regulatory role of the sympathetic nervous system in vascular and tubular functions. The reciprocal congenic strains are thereby unique models that may help in the search for intermediate phenotypes and empower functional deduction of candidate genes.
Collapse
Affiliation(s)
- Tao Wang
- Department of Functional Pathology, Shimane University School of Medicine, Izumo, Japan
| | | | | | | |
Collapse
|
26
|
Toland EJ, Saad Y, Yerga-Woolwine S, Ummel S, Farms P, Ramdath R, Frank BC, Lee NH, Joe B. Closely linked non-additive blood pressure quantitative trait loci. Mamm Genome 2008; 19:209-18. [PMID: 18324438 DOI: 10.1007/s00335-008-9093-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2007] [Accepted: 01/04/2008] [Indexed: 11/30/2022]
Abstract
There is enough evidence through linkage and substitution mapping to indicate that rat chromosome 1 harbors multiple blood pressure (BP) quantitative trait loci (QTLs). Of these, BP QTL1b was previously reported from our laboratory using congenic strains derived by introgressing normotensive alleles from the LEW rat onto the genetic background of the hypertensive Dahl salt-sensitive (S) rat. The region spanned by QTL1b is quite large (20.92 Mb), thus requiring further mapping with improved resolution so as to facilitate systematic identification of the underlying genetic determinant(s). Using congenic strains containing the LEW rat chromosomal segments on the Dahl salt-sensitive (S) rat background, further iterations of congenic substrains were constructed and characterized. Collective data obtained from this new iteration of congenic substrains provided evidence for further fragmentation of QTL1b with improved resolution. At least two separate genetic determinants of blood pressure underlie QTL1b. These are within 7.40 Mb and 7.31 Mb and are known as the QTL1b1 region and the QTL1b2 region, respectively. A genetic interaction was detected between the two BP QTLs. Interestingly, five of the previously reported differentially expressed genes located within the newly mapped QTL1b1 region remained differentially expressed. The congenic strain S.LEW(D1Mco36-D1Mco101), which harbors the QTL1b1 region alone but not the QTL1b2 region, serves as a genetic tool for further dissection of the QTL1b1 region and validation of Nr2f2 as a positional candidate gene. Overall, this study represents an intermediary yet obligatory progression towards the identification of genetic elements controlling BP.
Collapse
Affiliation(s)
- Edward J Toland
- Physiological Genomics Laboratory, Department of Physiology and Pharmacology, University of Toledo College of Medicine, 3035 Arlington Avenue, Toledo, OH 43614, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Woon PY, Kaisaki PJ, Bragança J, Bihoreau MT, Levy JC, Farrall M, Gauguier D. Aryl hydrocarbon receptor nuclear translocator-like (BMAL1) is associated with susceptibility to hypertension and type 2 diabetes. Proc Natl Acad Sci U S A 2007; 104:14412-7. [PMID: 17728404 PMCID: PMC1958818 DOI: 10.1073/pnas.0703247104] [Citation(s) in RCA: 298] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2007] [Indexed: 12/22/2022] Open
Abstract
Many aspects of physiology and behavior follow a circadian rhythm. Brain and muscle Arnt-like protein-1 (BMAL1) is a key component of the mammalian molecular clock, which controls circadian oscillations. In the rat, the gene encoding Bmal1 is located within hypertension susceptibility loci. We analyzed the SNP distribution pattern in a congenic interval associated with hypertension in the spontaneously hypertensive rat (SHR), and we show that Bmal1 maps close to a region genetically divergent between SHR and its normotensive (Wistar-Kyoto) counterpart. Bmal1 sequencing in rat strains identified 19 polymorphisms, including an SHR promoter variant that significantly affects Gata-4 activation of transcription in transient transfection experiments. A genetic association study designed to test the relevance of these findings in 1,304 individuals from 424 families primarily selected for type 2 diabetes showed that two BMAL1 haplotypes are associated with type 2 diabetes and hypertension. This comparative genetics finding translated from mouse and rat models to human provides evidence of a causative role of Bmal1 variants in pathological components of the metabolic syndrome.
Collapse
Affiliation(s)
| | | | - José Bragança
- *Wellcome Trust Centre for Human Genetics and
- Department of Cardiovascular Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom; and
| | | | - Jonathan C. Levy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LJ, United Kingdom
| | - Martin Farrall
- *Wellcome Trust Centre for Human Genetics and
- Department of Cardiovascular Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom; and
| | | |
Collapse
|
28
|
|