1
|
Rippe C, Bastrup JA, Holmberg J, Kawka K, Arévalo Martinez M, Albinsson S, Jepps TA, Swärd K. Declining activity of serum response factor in aging aorta in relation to aneurysm progression. J Biol Chem 2025; 301:108400. [PMID: 40081573 PMCID: PMC12002835 DOI: 10.1016/j.jbc.2025.108400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025] Open
Abstract
Age is a critical determinant of arterial disease, including aneurysm formation. Here, to understand the impact of aging on the arterial transcriptome, we leveraged RNA-sequencing data to define transcripts that change with advancing age in human arteries. Among the most repressed transcripts in aged individuals were those that are relevant for actomyosin structure and organization, including both myosin light chain kinase (MYLK) and smooth muscle γ-actin (ACTG2). This was associated with a reduction of serum response factor (SRF), which controls these transcripts via defined promoter elements. To determine the consequences of isolated Srf depletion, we conditionally deleted Srf in vascular smooth muscle of young mice (i8-SRF-KO mice). This led to a reduction of the SRF regulon, including Mylk and Actg2, and impaired arterial contractility, but left endothelial-dependent dilatation unaffected. Srf-depletion also increased aortic diameter and Alcian blue staining of the aortic media, which are cardinal features of aortopathy, such as aortic aneurysmal disease. Despite this, i8-SRF-KO mice were protected from aortic lesions elicited by angiotensin II (AngII). Proteomics demonstrated that Srf-depletion mimicked a protein signature of AngII treatment involving increases of the mechanoresponsive transcriptional coactivators YAP and TAZ and reduction of the Hippo kinase Lats2. Protection from aortopathy could be overcome by changing the order of KO induction and AngII administration resulting in advanced aneurysms in both i8-SRF-KO and control mice. Our work provides important insights into the molecular underpinnings of age-dependent changes in aortic function and mechanisms of adaptation in hypertension.
Collapse
Affiliation(s)
- Catarina Rippe
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Joakim Armstrong Bastrup
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Johan Holmberg
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Katarzyna Kawka
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Sebastian Albinsson
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Thomas A Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Karl Swärd
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| |
Collapse
|
2
|
Shi J, Liang Z, Liu Z, Pan L, Hu X, Tian Y, Jin H, Liu Y, Cheng Y, Zhang M. Identification of Novel Proteins Mediating Causal Association Between Smoking and Essential Hypertension: A Mendelian Randomization Study. J Am Heart Assoc 2024:e036202. [PMID: 39604029 DOI: 10.1161/jaha.124.036202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 09/06/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Smoking is a factor for hypertension. We aim to reveal novel plasma proteins mediating the relationship of smoking with hypertension and identify potential drug targets for hypertension on the basis of Mendelian randomization design. METHODS AND RESULTS Data for smoking were selected from the largest genome-wide association study meta-analysis performed by the Genome-Wide Association Study and Sequencing Consortium of Alcohol and Nicotine Use. Data for plasma proteins were selected from the deCODE Health study and the UK Biobank Pharma Proteomics Project. Data for hypertension were extracted from the FinnGen Study. Moreover, proteome-wide Mendelian randomization and colocalization analyses, 2-step Mendelian randomization, and gene function and network prediction, as well as druggability assessment were performed. We finally identified 8 proteins (ANXA4 [annexin A4], DLK1 [protein delta homolog 1], KLB [β-klotho], MMP8 [matrix metallopeptidase 8], PLAT [tissue-type plasminogen activator], POSTN [periostin], SAT2 [thialysine N-ε-acetyltransferase], and IFNLR1 [interferon λ receptor 1]) mediating association of smoking with hypertension. PLAT and IFNLR1 were identified to be involved in the complement and coagulation cascades and the Janus kinase/signal transducer and activator of transcription signaling pathway. ANXA4, KLB, MMP8, PLAT, and IFNLR1 had druggability. Moreover, IFNLR1 had strong evidence of genetic colocalization, because the posterior probability for H4 of IFNLR1 was 91.3%. CONCLUSIONS This study identified the 8 proteins that mediate causal association between smoking and essential hypertension. Interferon λ receptor agonist targeting IFNLR1 may open a new avenue for treating hypertension. Our discoveries provide new insights into protein pathogenesis of hypertension and to better guide hypertension prevention and treatment among smokers.
Collapse
Affiliation(s)
- Jikang Shi
- Department of Clinical Nutrition Peking University Shenzhen Hospital Shenzhen China
| | - Zhuoshuai Liang
- Department of Epidemiology and Biostatistics School of Public Health of Jilin University Changchun China
| | - Zhantong Liu
- Department of Epidemiology and Biostatistics School of Public Health of Jilin University Changchun China
| | - Lingfeng Pan
- Clinic for Plastic, Reconstructive and Hand Surgery, Klinikum Rechts der Isar Technical University of Munich Munich Germany
| | - Xinmeng Hu
- Department of Epidemiology and Biostatistics School of Public Health of Jilin University Changchun China
| | - Yuyang Tian
- Department of Epidemiology and Biostatistics School of Public Health of Jilin University Changchun China
| | - Huizhen Jin
- Department of Epidemiology and Biostatistics School of Public Health of Jilin University Changchun China
| | - Yawen Liu
- Department of Epidemiology and Biostatistics School of Public Health of Jilin University Changchun China
| | - Yi Cheng
- The Cardiovascular Center The First Hospital of Jilin University Changchun Jilin China
| | - Ming Zhang
- Department of Clinical Nutrition Peking University Shenzhen Hospital Shenzhen China
| |
Collapse
|
3
|
Sukumaran R, Nair AS, Banerjee M. Ethnic and region-specific genetic risk variants of stroke and its comorbid conditions can define the variations in the burden of stroke and its phenotypic traits. eLife 2024; 13:RP94088. [PMID: 39268810 PMCID: PMC11398864 DOI: 10.7554/elife.94088] [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: 09/15/2024] Open
Abstract
Burden of stroke differs by region, which could be attributed to differences in comorbid conditions and ethnicity. Genomewide variation acts as a proxy marker for ethnicity, and comorbid conditions. We present an integrated approach to understand this variation by considering prevalence and mortality rates of stroke and its comorbid risk for 204 countries from 2009 to 2019, and Genome-wide association studies (GWAS) risk variant for all these conditions. Global and regional trend analysis of rates using linear regression, correlation, and proportion analysis, signifies ethnogeographic differences. Interestingly, the comorbid conditions that act as risk drivers for stroke differed by regions, with more of metabolic risk in America and Europe, in contrast to high systolic blood pressure in Asian and African regions. GWAS risk loci of stroke and its comorbid conditions indicate distinct population stratification for each of these conditions, signifying for population-specific risk. Unique and shared genetic risk variants for stroke, and its comorbid and followed up with ethnic-specific variation can help in determining regional risk drivers for stroke. Unique ethnic-specific risk variants and their distinct patterns of linkage disequilibrium further uncover the drivers for phenotypic variation. Therefore, identifying population- and comorbidity-specific risk variants might help in defining the threshold for risk, and aid in developing population-specific prevention strategies for stroke.
Collapse
Affiliation(s)
- Rashmi Sukumaran
- Human Molecular Genetics Laboratory, Rajiv Gandhi Centre for BiotechnologyThiruvananthapuramIndia
- Department of Computational Biology and Bioinformatics, University of KeralaThiruvananthapuramIndia
| | - Achuthsankar S Nair
- Department of Computational Biology and Bioinformatics, University of KeralaThiruvananthapuramIndia
| | - Moinak Banerjee
- Human Molecular Genetics Laboratory, Rajiv Gandhi Centre for BiotechnologyThiruvananthapuramIndia
| |
Collapse
|
4
|
Olivera S, Graham D. Sex differences in preclinical models of hypertension. J Hum Hypertens 2023; 37:619-625. [PMID: 36335169 PMCID: PMC10403342 DOI: 10.1038/s41371-022-00770-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 10/03/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022]
Abstract
Hypertension remains the primary contributor in the development of cardiovascular disease which is rapidly increasing worldwide. High blood pressure affects men and women differently and understanding these sex differences is the ultimate unmet need for researchers in this field. Due to the inherent differences in hypertension prevalence, control and outcomes between men and women, novel research needs to be carried out to tackle these disparities and improve targeted treatment. Animal models of hypertension have provided valuable insights into the sexual dimorphism of blood pressure mechanisms. The availability of genetic and non-genetic hypertensive strains allows the opportunity to study diverse environmental and genetic factors that affect blood pressure, therefore presenting a valuable tool for researchers. Sex differences are present before birth and throughout life, which presents a challenge for the study of disease development in humans, but these complexities can be resolved with the use of in vivo models that display similarities to human disease. The aim of the present review is to provide an overview of the different available animal models of hypertension that present sexual dimorphisms and to discuss their relevance to humans.
Collapse
Affiliation(s)
- Sol Olivera
- School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK
| | - Delyth Graham
- School of Cardiovascular and Metabolic Health, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK.
| |
Collapse
|
5
|
Adua E. Decoding the mechanism of hypertension through multiomics profiling. J Hum Hypertens 2023; 37:253-264. [PMID: 36329155 PMCID: PMC10063442 DOI: 10.1038/s41371-022-00769-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 08/24/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Hypertension, characterised by a constant high blood pressure, is the primary risk factor for multiple cardiovascular events and a major cause of death in adults. Excitingly, innovations in high-throughput technologies have enabled the global exploration of the whole genome (genomics), revealing dysregulated genes that are linked to hypertension. Moreover, post-genomic biomarkers, from the emerging fields of transcriptomics, proteomics, glycomics and lipidomics, have provided new insights into the molecular underpinnings of hypertension. In this paper, we review the pathophysiology of hypertension, and highlight the multi-omics approaches for hypertension prediction and diagnosis.
Collapse
Affiliation(s)
- Eric Adua
- School of Clinical Medicine, Medicine & Health, Rural Clinical Campus, University of New South Wales, Wagga Wagga, NSW, Australia.
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.
| |
Collapse
|
6
|
Genetics of Neurogenic Orthostatic Hypotension in Parkinson’s Disease, Results from a Cross-Sectional In Silico Study. Brain Sci 2023; 13:brainsci13030506. [PMID: 36979316 PMCID: PMC10046202 DOI: 10.3390/brainsci13030506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/09/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
The genetic basis of Neurogenic Orthostatic Hypotension (NOH) in Parkinson’s disease (PD) has been inadequately explored. In a cross-sectional study, we examined the association between NOH and PD-related single-nucleotide polymorphisms (SNPs) and mapped their effects on gene expression and metabolic and signaling pathways. Patients with PD, free from pathological conditions associated with OH, and not taking OH-associated medications were included. NOH was defined as per international guidelines. Logistic regression was used to relate SNPs to NOH. Linkage-disequilibrium analysis, expression quantitative trait loci, and enrichment analysis were used to assess the effects on gene expression and metabolic/signaling pathways. We included 304 PD patients in the study, 35 of whom had NOH (11.5%). NOH was more frequent in patients with SNPs in SNCA, TMEM175, FAM47E-STBD1, CCDC62, SCN3A, MIR4696, SH3GL2, and LZTS3/DDRGK1 and less frequent in those with SNPs in ITGA8, IP6K2, SIPA1L2, NDUFAF2. These SNPs affected gene expression associated with the significant hierarchical central structures of the autonomic nervous system. They influenced several metabolic/signaling pathways, most notably IP3/Ca++ signaling, the PKA-CREB pathway, and the metabolism of fatty acids. These findings provide new insights into the pathophysiology of NOH in PD and may provide targets for future therapies.
Collapse
|
7
|
Olczak KJ, Taylor-Bateman V, Nicholls HL, Traylor M, Cabrera CP, Munroe PB. Hypertension genetics past, present and future applications. J Intern Med 2021; 290:1130-1152. [PMID: 34166551 DOI: 10.1111/joim.13352] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Essential hypertension is a complex trait where the underlying aetiology is not completely understood. Left untreated it increases the risk of severe health complications including cardiovascular and renal disease. It is almost 15 years since the first genome-wide association study for hypertension, and after a slow start there are now over 1000 blood pressure (BP) loci explaining ∼6% of the single nucleotide polymorphism-based heritability. Success in discovery of hypertension genes has provided new pathological insights and drug discovery opportunities and translated to the development of BP genetic risk scores (GRSs), facilitating population disease risk stratification. Comparing highest and lowest risk groups shows differences of 12.9 mm Hg in systolic-BP with significant differences in risk of hypertension, stroke, cardiovascular disease and myocardial infarction. GRSs are also being trialled in antihypertensive drug responses. Drug targets identified include NPR1, for which an agonist drug is currently in clinical trials. Identification of variants at the PHACTR1 locus provided insights into regulation of EDN1 in the endothelin pathway, which is aiding the development of endothelin receptor EDNRA antagonists. Drug re-purposing opportunities, including SLC5A1 and canagliflozin (a type-2 diabetes drug), are also being identified. In this review, we present key studies from the past, highlight current avenues of research and look to the future focusing on gene discovery, epigenetics, gene-environment interactions, GRSs and drug discovery. We evaluate limitations affecting BP genetics, including ancestry bias and discuss streamlining of drug target discovery and applications for treating and preventing hypertension, which will contribute to tailored precision medicine for patients.
Collapse
Affiliation(s)
- Kaya J Olczak
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Victoria Taylor-Bateman
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Hannah L Nicholls
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Centre for Translational Bioinformatics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Matthew Traylor
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Claudia P Cabrera
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Centre for Translational Bioinformatics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,NIHR Barts Biomedical Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Patricia B Munroe
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,NIHR Barts Biomedical Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
8
|
Paterson MR, Jackson KL, Dona MSI, Farrugia GE, Visniauskas B, Watson AMD, Johnson C, Prieto MC, Evans RG, Charchar F, Pinto AR, Marques FZ, Head GA. Deficiency of MicroRNA-181a Results in Transcriptome-Wide Cell-Specific Changes in the Kidney and Increases Blood Pressure. Hypertension 2021; 78:1322-1334. [PMID: 34538100 PMCID: PMC8573069 DOI: 10.1161/hypertensionaha.121.17384] [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: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Madeleine R. Paterson
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Australia; Monash University, Melbourne, Australia
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Kristy L. Jackson
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University Parkville, Australia
| | - Malathi S. I. Dona
- Cardiac Cellular Systems Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Gabriella E. Farrugia
- Cardiac Cellular Systems Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Bruna Visniauskas
- Department of Physiology, School of Medicine, Tulane University, New Orleans, the USA
| | - Anna M. D. Watson
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| | - Chad Johnson
- Monash Micro Imaging, Monash University, Melbourne, Australia
| | - Minolfa C. Prieto
- Department of Physiology, School of Medicine, Tulane University, New Orleans, the USA
| | - Roger G. Evans
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Australia
| | - Fadi Charchar
- Health Innovation and Transformation Centre, Federation University, Ballarat, Australia
- Department of Physiology, University of Melbourne, Melbourne, Australia
| | - Alexander R. Pinto
- Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University Parkville, Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Australia
| | - Francine Z. Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Australia; Monash University, Melbourne, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Geoffrey A. Head
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Pharmacology, Monash University, Melbourne, Australia
| |
Collapse
|
9
|
Zhou Y, Yang B, Wang J, Zhu J, Tian G. A scaling-free minimum enclosing ball method to detect differentially expressed genes for RNA-seq data. BMC Genomics 2021; 22:479. [PMID: 34174824 PMCID: PMC8234728 DOI: 10.1186/s12864-021-07790-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 06/10/2021] [Indexed: 12/13/2022] Open
Abstract
Background Identifying differentially expressed genes between the same or different species is an urgent demand for biological and medical research. For RNA-seq data, systematic technical effects and different sequencing depths are usually encountered when conducting experiments. Normalization is regarded as an essential step in the discovery of biologically important changes in expression. The present methods usually involve normalization of the data with a scaling factor, followed by detection of significant genes. However, more than one scaling factor may exist because of the complexity of real data. Consequently, methods that normalize data by a single scaling factor may deliver suboptimal performance or may not even work.The development of modern machine learning techniques has provided a new perspective regarding discrimination between differentially expressed (DE) and non-DE genes. However, in reality, the non-DE genes comprise only a small set and may contain housekeeping genes (in same species) or conserved orthologous genes (in different species). Therefore, the process of detecting DE genes can be formulated as a one-class classification problem, where only non-DE genes are observed, while DE genes are completely absent from the training data. Results In this study, we transform the problem to an outlier detection problem by treating DE genes as outliers, and we propose a scaling-free minimum enclosing ball (SFMEB) method to construct a smallest possible ball to contain the known non-DE genes in a feature space. The genes outside the minimum enclosing ball can then be naturally considered to be DE genes. Compared with the existing methods, the proposed SFMEB method does not require data normalization, which is particularly attractive when the RNA-seq data include more than one scaling factor. Furthermore, the SFMEB method could be easily extended to different species without normalization. Conclusions Simulation studies demonstrate that the SFMEB method works well in a wide range of settings, especially when the data are heterogeneous or biological replicates. Analysis of the real data also supports the conclusion that the SFMEB method outperforms other existing competitors. The R package of the proposed method is available at https://bioconductor.org/packages/MEB. Supplementary Information The online version contains supplementary material available at (10.1186/s12864-021-07790-0).
Collapse
Affiliation(s)
- Yan Zhou
- College of Mathematics and Statistics, Institute of Statistical Sciences, Shenzhen Key Laboratory of Advanced Machine Learning and Applications, Shenzhen University, Shenzhen, China
| | - Bin Yang
- College of Mathematics and Statistics, Institute of Statistical Sciences, Shenzhen Key Laboratory of Advanced Machine Learning and Applications, Shenzhen University, Shenzhen, China
| | - Junhui Wang
- School of Data Science, City University of Hong Kong, Hong Kong
| | - Jiadi Zhu
- College of Mathematics and Statistics, Institute of Statistical Sciences, Shenzhen Key Laboratory of Advanced Machine Learning and Applications, Shenzhen University, Shenzhen, China.
| | - Guoliang Tian
- Department of Statistics and Data Science, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
10
|
Transport of L-Arginine Related Cardiovascular Risk Markers. J Clin Med 2020; 9:jcm9123975. [PMID: 33302555 PMCID: PMC7764698 DOI: 10.3390/jcm9123975] [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: 10/28/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
L-arginine and its derivatives, asymmetric and symmetric dimethylarginine (ADMA and SDMA) and L-homoarginine, have emerged as cardiovascular biomarkers linked to cardiovascular outcomes and various metabolic and functional pathways such as NO-mediated endothelial function. Cellular uptake and efflux of L-arginine and its derivatives are facilitated by transport proteins. In this respect the cationic amino acid transporters CAT1 and CAT2 (SLC7A1 and SLC7A2) and the system y+L amino acid transporters (SLC7A6 and SLC7A7) have been most extensively investigated, so far, but the number of transporters shown to mediate the transport of L-arginine and its derivatives is constantly increasing. In the present review we assess the growing body of evidence regarding the function, expression, and clinical relevance of these transporters and their possible relation to cardiovascular diseases.
Collapse
|
11
|
Checchi M, Bertacchini J, Cavani F, Magarò MS, Reggiani Bonetti L, Pugliese GR, Tamma R, Ribatti D, Maurel DB, Palumbo C. Scleral ossicles: angiogenic scaffolds, a novel biomaterial for regenerative medicine applications. Biomater Sci 2019; 8:413-425. [PMID: 31738355 DOI: 10.1039/c9bm01234f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Given the current prolonged life expectancy, various pathologies affect increasingly the aging subjects. Regarding the musculoskeletal apparatus, bone fragility induces more susceptibility to fractures, often not accompanied by good ability of self-repairing, in particular when critical-size defects (CSD) occur. Currently orthopedic surgery makes use of allografting and autografting which, however, have limitations due to the scarce amount of tissue that can be taken from the donor, the possibility of disease transmission and donor site morbidity. The need to develop new solutions has pushed the field of tissue engineering (TE) research to study new scaffolds to be functionalized in order to obtain constructs capable of promoting tissue regeneration and achieve stable bone recovery over time. This investigation focuses on the most important aspect related to bone tissue regeneration: the angiogenic properties of the scaffold to be used. As an innovative solution, scleral ossicles (SOs), previously characterized as natural, biocompatible and spontaneously decellularized scaffolds used for bone repair, were tested for angiogenic potential and biocompatibility. To reach this purpose, in ovo Chorioallantoic Membrane Assay (CAM) was firstly used to test the angiogenic potential; secondly, in vivo subcutaneous implantation of SOs (in a rat model) was performed in order to assess the biocompatibility and the inflammatory response. Finally, thanks to the analysis of mass spectrometry (LCMSQE), the putative proteins responsible for the SO angiogenic properties were identified. Thus, a novel natural biomaterial is proposed, which is (i) able to induce an angiogenic response in vivo by subcutaneous implantation in a non-immunodeficient animal model, (ii) which does not induce any inflammatory response, and (iii) is useful for regenerative medicine application for the healing of bone CSD.
Collapse
Affiliation(s)
- Marta Checchi
- Department of Biomedical, Metabolic Science and Neuroscience, University of Modena and Reggio Emilia, 41125 Modena, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Zhu J, Wang Z, Chen F, Liu C. Identification of genes and functional coexpression modules closely related to ulcerative colitis by gene datasets analysis. PeerJ 2019; 7:e8061. [PMID: 31741804 PMCID: PMC6858811 DOI: 10.7717/peerj.8061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 10/20/2019] [Indexed: 02/06/2023] Open
Abstract
Background Ulcerative colitis is a type of inflammatory bowel disease posing a great threat to the public health worldwide. Previously, gene expression studies of mucosal colonic biopsies have provided some insight into the pathophysiological mechanisms in ulcerative colitis; however, the exact pathogenesis is unclear. The purpose of this study is to identify the most related genes and pathways of UC by bioinformatics, so as to reveal the core of the pathogenesis. Methods Genome-wide gene expression datasets involving ulcerative colitis patients were collected from gene expression omnibus database. To identify most close genes, an integrated analysis of gene expression signature was performed by employing robust rank aggregation method. We used weighted gene co-expression network analysis to explore the functional modules involved in ulcerative colitis pathogenesis. Besides, biological process and pathways analysis of co-expression modules were figured out by gene ontology enrichment analysis using Metascape. Results A total of 328 ulcerative colitis patients and 138 healthy controls were from 14 datasets. The 150 most significant differentially expressed genes are likely to include causative genes of disease, and further studies are needed to demonstrate this. Seven main functional modules were identified, which pathway enrichment analysis indicated were associated with many biological processes. Pathways such as ‘extracellular matrix, immune inflammatory response, cell cycle, material metabolism’ are consistent with the core mechanism of ulcerative colitis. However, ‘defense response to virus’ and ‘herpes simplex infection’ suggest that viral infection is one of the aetiological agents. Besides, ‘Signaling by Receptor Tyrosine Kinases’ and ‘pathway in cancer’ provide new clues for the study of the risk and process of ulcerative colitis cancerization.
Collapse
Affiliation(s)
- Jie Zhu
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Zheng Wang
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Fengzhe Chen
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Changhong Liu
- Department of Gastroenterology, Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
13
|
Zhu J, Wang Z, Chen F. Association of Key Genes and Pathways with Atopic Dermatitis by Bioinformatics Analysis. Med Sci Monit 2019; 25:4353-4361. [PMID: 31184315 PMCID: PMC6582687 DOI: 10.12659/msm.916525] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Atopic dermatitis is a chronic inflammatory disease of the skin. It has a high prevalence worldwide and affected persons are prone to recurrent attacks, seriously affecting the physical and mental of patients. The exact etiology of the disease is still unclear. Material/Methods There are 7 datasets on atopic dermatitis in the Gene Expression Omnibus database, including 142 lesional and 134 non-lesional skin biopsy samples. Differential analysis was performed after datasets were integrated by robust multi-array average method. Functional modules of GSE99802 were explored by weighted gene co-expression network analysis. The 4 most important modules were enriched into the pathways by Metascape. Results Significantly differentially expressed genes included 41 upregulated and 10 downregulated genes. The following 5 of the most important upregulated genes had the strongest association with atopic dermatitis. SERPINB3&4 promote inflammation and impaired skin barrier function in the early stage of atopic dermatitis. S100A9 aggravates the inflammatory response by inducing the activation of toll-like receptor 4, neutrophil chemotaxis, neutrophilic inflammation, and the amplification of interleukin-8. MMP1 is the key protease of skin collagen degradation, keeping the extracellular matrix in dynamic balance. MMP12 induces the aggregation of various inflammatory cells into inflammatory tissue. The enriched pathways of each module mainly include Cellular responses to external stimuli, Metabolism of RNA and Translation, and Infectious disease. Conclusions The associated pathways and genes not only help us understand the molecular mechanism of the disease, but also provide research directions or targets for accurate diagnosis and treatment.
Collapse
Affiliation(s)
- Jie Zhu
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Zheng Wang
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Fengzhe Chen
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
14
|
A statistical normalization method and differential expression analysis for RNA-seq data between different species. BMC Bioinformatics 2019; 20:163. [PMID: 30925894 PMCID: PMC6441199 DOI: 10.1186/s12859-019-2745-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 03/18/2019] [Indexed: 02/06/2023] Open
Abstract
Background High-throughput techniques bring novel tools and also statistical challenges to genomic research. Identifying genes with differential expression between different species is an effective way to discover evolutionarily conserved transcriptional responses. To remove systematic variation between different species for a fair comparison, normalization serves as a crucial pre-processing step that adjusts for the varying sample sequencing depths and other confounding technical effects. Results In this paper, we propose a scale based normalization (SCBN) method by taking into account the available knowledge of conserved orthologous genes and by using the hypothesis testing framework. Considering the different gene lengths and unmapped genes between different species, we formulate the problem from the perspective of hypothesis testing and search for the optimal scaling factor that minimizes the deviation between the empirical and nominal type I errors. Conclusions Simulation studies show that the proposed method performs significantly better than the existing competitor in a wide range of settings. An RNA-seq dataset of different species is also analyzed and it coincides with the conclusion that the proposed method outperforms the existing method. For practical applications, we have also developed an R package named “SCBN”, which is freely available at http://www.bioconductor.org/packages/devel/bioc/html/SCBN.html. Electronic supplementary material The online version of this article (10.1186/s12859-019-2745-1) contains supplementary material, which is available to authorized users.
Collapse
|
15
|
Shi J, Liu Y, Liu Y, Li Y, Qiu S, Bai Y, Gu Y, Luo J, Cui H, Li Y, Zhao Q, Zhang K, Cheng Y. Association between ApoE polymorphism and hypertension: A meta-analysis of 28 studies including 5898 cases and 7518 controls. Gene 2018; 675:197-207. [PMID: 30180966 DOI: 10.1016/j.gene.2018.06.097] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 06/23/2018] [Accepted: 06/27/2018] [Indexed: 02/06/2023]
Abstract
Hypertension is one of the most common chronic diseases, constituting an independent risk factor for many diseases. Our study aimed to evaluate the association between apolipoprotein E (ApoE) genetic polymorphism and hypertension, and to provide evidence for the etiology of hypertension. Case-control studies of ApoE polymorphism and hypertension, which were included in PubMed, Embase, Web of Science, Medline, WanFang, Vip, and CNKI information databases, were selected and evaluated according to criteria of inclusion and exclusion. Eligible data were extracted and pooled, and were analyzed and assessed using Stata 12.0. Random-effect models were used when heterogeneity existed in between-study, and fixed-effect models were applied otherwise. A total of 28 studies that consisted of 5898 cases with hypertension and 7518 controls were selected. Alleles and genotypes of ApoE between cases and controls were compared. For ApoE alleles, we observed the contrast of ApoE ε2 versus ε3 allele yielded a pooled OR of 0.99 (95% CI: 0.87-1.11; P = 0.823), whereas the contrast of ε4 versus ε3 allele yielded a pooled OR of 1.95 (95% CI: 1.50-2.54; P < 0.001). For ApoE genotypes, compared with ε3/ε3 genotype, genotypes (ε2/ε2 and ε2/ε3) showed a possible association with hypertension (OR = 0.88; 95% CI: 0.79-0.99; P = 0.033), and genotypes (ε3/ε4 and ε4/ε4) had a 2.08-fold risk of developing hypertension (OR = 2.08; 95% CI: 1.58-2.74; P < 0.001). There is the association between ApoE polymorphism and hypertension: the genotypes carrying ε2 allele may be a protective factor, and the ApoE ε4 allele and the genotypes carrying ε4 allele may be risk factors for hypertension.
Collapse
Affiliation(s)
- Jikang Shi
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun 130021, China
| | - Yawen Liu
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun 130021, China
| | - Yunkai Liu
- The Cardiovascular Center, the First Hospital of Jilin University, Changchun 130021, China
| | - Yong Li
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun 130021, China
| | - Shuang Qiu
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun 130021, China
| | - Ye Bai
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun 130021, China
| | - Yulu Gu
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun 130021, China
| | - Jingjing Luo
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun 130021, China
| | - Heran Cui
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun 130021, China
| | - Yan Li
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun 130021, China
| | - Qian Zhao
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun 130021, China
| | - Kaixin Zhang
- Department of Epidemiology and Biostatistics, School of Public Health of Jilin University, Changchun 130021, China
| | - Yi Cheng
- The Cardiovascular Center, the First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
16
|
Interactions of Genes and Sodium Intake on the Development of Hypertension: A Cohort-Based Case-Control Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:ijerph15061110. [PMID: 29848945 PMCID: PMC6025596 DOI: 10.3390/ijerph15061110] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 05/17/2018] [Accepted: 05/24/2018] [Indexed: 11/16/2022]
Abstract
There have been few studies investigating interactions of G-protein beta3 subunit (GNB3) C825T (rs5443) and dietary sodium intake on the risk of hypertension, i.e., BP salt sensitivity. The study aims to evaluate joint effects of GNB3 polymorphisms and sodium consumption on the development of hypertension. A cohort-based case-control study was conducted in 2014. There are 233 participants with newly diagnosed hypertension in the case group and 699 participants in the gender-matched control group. The primary outcome is the development of hypertension over a 10-year period. The determinants of hypertension were three genotypes of SNP in GNB3 (TT; CT; and CC) and two dietary salt categories on the basis of the level of sodium consumption representing high (>4800 mg/day) and low-sodium (<2400 mg/day) diets. The development of hypertension increased with participants carrying TT genotype and high-sodium diets comparing with those carrying TC or CC genotype with low-sodium diets (adjusted OR 3.23, 95% CI 1.52–6.83) (Rothman synergy index = 3.79). The study suggests that GNB3 C825T polymorphism may influence the response of the renin-angiotensin system to high-sodium diet. It implies that GNB3 can be served as an easy, inexpensive, and early genetic marker of salt sensitivity to blood pressure. Salt-sensitive individuals should pay more attention to salt intake to reduce cardiovascular morbidity or mortality.
Collapse
|
17
|
Yan S, Wang W, Gao G, Cheng M, Wang X, Wang Z, Ma X, Chai C, Xu D. Key genes and functional coexpression modules involved in the pathogenesis of systemic lupus erythematosus. J Cell Physiol 2018; 233:8815-8825. [PMID: 29806703 DOI: 10.1002/jcp.26795] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 04/30/2018] [Indexed: 02/06/2023]
Abstract
We performed a systematic review of genome-wide gene expression datasets to identify key genes and functional modules involved in the pathogenesis of systemic lupus erythematosus (SLE) at a systems level. Genome-wide gene expression datasets involving SLE patients were searched in Gene Expression Omnibus and ArrayExpress databases. Robust rank aggregation (RRA) analysis was used to integrate those public datasets and identify key genes associated with SLE. The weighted gene coexpression network analysis (WGCNA) was adapted to identify functional modules involved in SLE pathogenesis, and the gene ontology enrichment analysis was utilized to explore their functions. The aberrant expressions of several randomly selected key genes were further validated in SLE patients through quantitative real-time polymerase chain reaction. Fifteen genome-wide gene expression datasets were finally included, which involved a total of 1,778 SLE patients and 408 healthy controls. A large number of significantly upregulated or downregulated genes were identified through RRA analysis, and some of those genes were novel SLE gene signatures and their molecular roles in etiology of SLE remained vague. WGCNA further successfully identified six main functional modules involved in the pathogenesis of SLE. The most important functional module involved in SLE included 182 genes and mainly enriched in biological processes, including defense response to virus, interferon signaling pathway, and cytokine-mediated signaling pathway. This study identifies a number of key genes and functional coexpression modules involved in SLE, which provides deepening insights into the molecular mechanism of SLE at a systems level and also provides some promising therapeutic targets.
Collapse
Affiliation(s)
- Shushan Yan
- Department of Gastrointestinal and Anal Diseases Surgery, The Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Weijie Wang
- Department of Neurosurgery, The Affiliated Huaian First Hospital of Nanjing Medical University, Huai'an, China
| | - Guohong Gao
- Department of Ophthalmology, The Affiliated Hospital of Weifang Medical University, Clinical Medical Institute, Weifang Medical University, Weifang, China
| | - Min Cheng
- Department of Physiology, Weifang Medical University, Weifang, China
| | - Xiaodong Wang
- Department of Rheumatology and Immunology, Affiliated Hospital of Weifang Medical University, Clinical Medical Institute, Weifang Medical University, Weifang, China
| | - Zengyan Wang
- Department of Surgery, Zhucheng People's Hospital, Weifang, China
| | - Xiufen Ma
- Department of Rheumatology and Immunology, Affiliated Hospital of Weifang Medical University, Clinical Medical Institute, Weifang Medical University, Weifang, China
| | - Chunxiang Chai
- Department of Rheumatology and Immunology, Affiliated Hospital of Weifang Medical University, Clinical Medical Institute, Weifang Medical University, Weifang, China
| | - Donghua Xu
- Department of Rheumatology and Immunology, Affiliated Hospital of Weifang Medical University, Clinical Medical Institute, Weifang Medical University, Weifang, China
| |
Collapse
|
18
|
Zhu W, Wang H, Wei J, Sartor GC, Bao MM, Pierce CT, Wahlestedt CR, Dykxhoorn DM, Dong C. Cocaine Exposure Increases Blood Pressure and Aortic Stiffness via the miR-30c-5p-Malic Enzyme 1-Reactive Oxygen Species Pathway. Hypertension 2018; 71:752-760. [PMID: 29483230 DOI: 10.1161/hypertensionaha.117.10213] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 08/28/2017] [Accepted: 12/14/2017] [Indexed: 02/07/2023]
Abstract
Cocaine abuse increases the risk of cardiovascular mortality and morbidity; however, the underlying molecular mechanisms remain elusive. By using a mouse model for cocaine abuse/use, we found that repeated cocaine injection led to increased blood pressure and aortic stiffness in mice associated with elevated levels of reactive oxygen species (ROS) in the aortas, a phenomenon similar to that observed in hypertensive humans. This ROS elevation was correlated with downregulation of Me1 (malic enzyme 1), an important redox molecule that counteracts ROS generation, and upregulation of microRNA (miR)-30c-5p that targets Me1 expression by directly binding to its 3'UTR (untranslated region). Remarkably, lentivirus-mediated overexpression of miR-30c-5p in aortic smooth muscle cells recapitulated the effect of cocaine on Me1 suppression, which in turn led to ROS elevation. Moreover, in vivo silencing of miR-30c-5p in smooth muscle cells resulted in Me1 upregulation, ROS reduction, and significantly suppressed cocaine-induced increases in blood pressure and aortic stiffness-a similar effect to that produced by treatment with the antioxidant N-acetyl cysteine. Discovery of this novel cocaine-↑miR-30c-5p-↓Me1-↑ROS pathway provides a potential new therapeutic avenue for treatment of cocaine abuse-related cardiovascular disease.
Collapse
Affiliation(s)
- Wei Zhu
- From the Interdisciplinary Stem Cell Institute (W.Z., H.W., J.W., M.M.B., C.T.P., C.D.), Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences (G.C.S., C.R.W.), and Department of Human Genetics, John P. Hussman Institute for Human Genomics (D.M.D.), University of Miami Miller School of Medicine, FL
| | - Huilan Wang
- From the Interdisciplinary Stem Cell Institute (W.Z., H.W., J.W., M.M.B., C.T.P., C.D.), Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences (G.C.S., C.R.W.), and Department of Human Genetics, John P. Hussman Institute for Human Genomics (D.M.D.), University of Miami Miller School of Medicine, FL
| | - Jianqin Wei
- From the Interdisciplinary Stem Cell Institute (W.Z., H.W., J.W., M.M.B., C.T.P., C.D.), Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences (G.C.S., C.R.W.), and Department of Human Genetics, John P. Hussman Institute for Human Genomics (D.M.D.), University of Miami Miller School of Medicine, FL
| | - Gregory C Sartor
- From the Interdisciplinary Stem Cell Institute (W.Z., H.W., J.W., M.M.B., C.T.P., C.D.), Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences (G.C.S., C.R.W.), and Department of Human Genetics, John P. Hussman Institute for Human Genomics (D.M.D.), University of Miami Miller School of Medicine, FL
| | - Michelle Meiqi Bao
- From the Interdisciplinary Stem Cell Institute (W.Z., H.W., J.W., M.M.B., C.T.P., C.D.), Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences (G.C.S., C.R.W.), and Department of Human Genetics, John P. Hussman Institute for Human Genomics (D.M.D.), University of Miami Miller School of Medicine, FL
| | - Clay T Pierce
- From the Interdisciplinary Stem Cell Institute (W.Z., H.W., J.W., M.M.B., C.T.P., C.D.), Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences (G.C.S., C.R.W.), and Department of Human Genetics, John P. Hussman Institute for Human Genomics (D.M.D.), University of Miami Miller School of Medicine, FL
| | - Claes R Wahlestedt
- From the Interdisciplinary Stem Cell Institute (W.Z., H.W., J.W., M.M.B., C.T.P., C.D.), Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences (G.C.S., C.R.W.), and Department of Human Genetics, John P. Hussman Institute for Human Genomics (D.M.D.), University of Miami Miller School of Medicine, FL
| | - Derek M Dykxhoorn
- From the Interdisciplinary Stem Cell Institute (W.Z., H.W., J.W., M.M.B., C.T.P., C.D.), Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences (G.C.S., C.R.W.), and Department of Human Genetics, John P. Hussman Institute for Human Genomics (D.M.D.), University of Miami Miller School of Medicine, FL
| | - Chunming Dong
- From the Interdisciplinary Stem Cell Institute (W.Z., H.W., J.W., M.M.B., C.T.P., C.D.), Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences (G.C.S., C.R.W.), and Department of Human Genetics, John P. Hussman Institute for Human Genomics (D.M.D.), University of Miami Miller School of Medicine, FL.
| |
Collapse
|
19
|
Rask-Andersen M, Martinsson D, Ahsan M, Enroth S, Ek WE, Gyllensten U, Johansson Å. Epigenome-wide association study reveals differential DNA methylation in individuals with a history of myocardial infarction. Hum Mol Genet 2018; 25:4739-4748. [PMID: 28172975 DOI: 10.1093/hmg/ddw302] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 08/24/2016] [Accepted: 08/26/2016] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading causes of death worldwide and represent a substantial economic burden on public health care systems. Epigenetic markers have potential as diagnostic markers before clinical symptoms have emerged, and as prognostic markers to inform the choice of clinical intervention. In this study, we performed an epigenome-wide association study (EWAS) for CVDs, to identify disease-specific alterations in DNA methylation. CpG methylation in blood samples from the northern Sweden population health study (NSPHS) (n = 729) was assayed on the Illumina Infinium HumanMethylation450 BeadChip. Individuals with a history of a CVD were identified in the cohort. It included individuals with hypertension (N = 147), myocardial infarction (MI) (N = 48), stroke (N = 27), thrombosis (N = 22) and cardiac arrhythmia (N = 5). Differential DNA methylation was observed at 211 CpG-sites in individuals with a history of MI (q <0.05). These sites represent 196 genes, of which 42 have been described in the scientific literature to be related to cardiac function, cardiovascular disease, cardiogenesis and recovery after ischemic injury. We have shown that individuals with a history of MI have a deviating pattern of DNA methylation at many genomic loci of which a large fraction has previously been linked to CVD. Our results highlight genes that might be important in the pathogenesis of MI or in recovery. In addition, the sites pointed out in this study can serve as candidates for further evaluation as potential biomarkers for MI.
Collapse
Affiliation(s)
- Mathias Rask-Andersen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - David Martinsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Muhammad Ahsan
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Stefan Enroth
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Weronica E Ek
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ulf Gyllensten
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Åsa Johansson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
20
|
Identification of susceptible genes for complex chronic diseases based on disease risk functional SNPs and interaction networks. J Biomed Inform 2017; 74:137-144. [DOI: 10.1016/j.jbi.2017.09.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 01/05/2023]
|
21
|
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
|
22
|
Prediction and Subtyping of Hypertension from Pan-Tissue Transcriptomic and Genetic Analyses. Genetics 2017; 207:1121-1134. [PMID: 28899996 DOI: 10.1534/genetics.117.300280] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 08/23/2017] [Indexed: 01/01/2023] Open
Abstract
Hypertension (HT) is a complex systemic disease involving transcriptional changes in multiple organs. Here we systematically investigate the pan-tissue transcriptional and genetic landscape of HT spanning dozens of tissues in hundreds of individuals. We find that in several tissues, previously identified HT-linked genes are dysregulated and the gene expression profile is predictive of HT. Importantly, many expression quantitative trait loci (eQTL) SNPs associated with the population variance of the dysregulated genes are linked with blood pressure in an independent genome-wide association study, suggesting that the functional effect of HT-associated SNPs may be mediated through tissue-specific transcriptional dysregulation. Analyses of pan-tissue transcriptional dysregulation profile, as well as eQTL SNPs underlying the dysregulated genes, reveals substantial heterogeneity among the HT patients, revealing two broad groupings - a Diffused group where several tissues exhibit HT-associated molecular alterations and a Localized group where such alterations are localized to very few tissues. These two patient subgroups differ in several clinical phenotypes including respiratory, cerebrovascular, diabetes, and heart disease. These findings suggest that the Diffused and Localized subgroups may be driven by different molecular mechanisms and have different genetic underpinning.
Collapse
|
23
|
Positive allosteric modulation of GABAA receptors attenuates high blood pressure in Schlager hypertensive mice. J Hypertens 2017; 35:546-557. [PMID: 28009705 DOI: 10.1097/hjh.0000000000001210] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Blood pressure high Schlager (BPH/2J) mice have neurogenic hypertension associated with differences in hypothalamic GABAA receptors compared with their normotensive counterparts (BPN/3J). Allopregnanolone is an endogenous neurosteroid reduced in chronic stress, and when administered, decreases anxiety by positive allosteric modulation of GABAA receptors. METHODS To determine if allopregnanolone could be a viable therapeutic for neurogenic hypertension, male BPH/2J (n = 6-7) and BPN/3J (n = 8-9) mice were equipped with radiotelemetry probes to compare cardiovascular variables before and after implantation of subcutaneous minipumps delivering allopregnanolone (5 mg/kg per day), or its vehicle, for a period of 2 weeks. In addition to baseline recordings, the response to stress and ganglionic blockade with pentolinium was recorded, before and 7-14 days after minipump implantation. Following treatment, brains were processed for c-Fos immunohistochemistry and quantitative real-time polymerase chain reaction. RESULTS Administration of allopregnanolone selectively reduced mean arterial pressure (-8.0 ± 2.7 mmHg; P = 0.02) and the depressor response to pentolinium (-15.3 ± 3.2 mmHg; P = 0.001) in BPH/2J mice, with minimal effects observed in BPN/3J mice. Following allopregnanolone treatment, the diminished expression of GABAA δ, α4 and β2 subunits in the hypothalamus (-1.6 to 4.8-fold; Pstrain < 0.05) was abolished. Furthermore, in BPH/2J mice, allopregnanolone treatment reduced the pressor response to dirty cage switch stress (-26.7 ± 4.5%; P < 0.001) and abolished the elevated c-Fos expression in pre-sympathetic nuclei. CONCLUSION The selective antihypertensive and stress inhibitory effects of allopregnanolone in BPH/2J mice suggest that allosteric modulation of GABAA receptors, in amygdalo-hypothalamic pathways, may contribute to the development of hypertension in this model and may offer a potential new therapeutic avenue.
Collapse
|
24
|
IP 3 receptor signaling and endothelial barrier function. Cell Mol Life Sci 2017; 74:4189-4207. [PMID: 28803370 DOI: 10.1007/s00018-017-2624-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 07/18/2017] [Accepted: 08/08/2017] [Indexed: 12/14/2022]
Abstract
The endothelium, a monolayer of endothelial cells lining vessel walls, maintains tissue-fluid homeostasis by restricting the passage of the plasma proteins and blood cells into the interstitium. The ion Ca2+, a ubiquitous secondary messenger, initiates signal transduction events in endothelial cells that is critical to control of vascular tone and endothelial permeability. The ion Ca2+ is stored inside the intracellular organelles and released into the cytosol in response to environmental cues. The inositol 1,4,5-trisphosphate (IP3) messenger facilitates Ca2+ release through IP3 receptors which are Ca2+-selective intracellular channels located within the membrane of the endoplasmic reticulum. Binding of IP3 to the IP3Rs initiates assembly of IP3R clusters, a key event responsible for amplification of Ca2+ signals in endothelial cells. This review discusses emerging concepts related to architecture and dynamics of IP3R clusters, and their specific role in propagation of Ca2+ signals in endothelial cells.
Collapse
|
25
|
Wang S, Fisher VA, Chen Y, Dupuis J. Comparison of multiple single-nucleotide variant association tests in a meta-analysis of Genetic Analysis Workshop 19 family and unrelated data. BMC Proc 2016; 10:187-191. [PMID: 27980634 PMCID: PMC5133513 DOI: 10.1186/s12919-016-0028-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background Meta-analysis has been widely used in genetic association studies to increase sample size and to improve power, both in the context of single-variant analysis, as well as for gene-based tests. Meta-analysis approaches for haplotype analysis have not been extensively developed and used, and have not been compared with other ways of jointly analysing multiple genetic variants. Methods We propose a novel meta-analysis approach for a gene-based haplotype association test, and compare it with an existing meta-analysis approach of the sequence kernel association test (SKAT), using the unrelated samples and family samples of the Genetic Analysis Workshop 19 data sets. We performed association tests with diastolic blood pressure and restricted our analyses to all variants in exonic regions on all odd chromosomes. Results Meta-analysis of haplotype results and SKAT identified different genes. The most significantly associated gene identified by SKAT was the ALCAM gene on chromosome 3 with a p value of 7.0 × 10− 5. Two of the most associated genes identified by the haplotype method were FPGT (p = 6.7 × 10− 8) on chromosome 1 and SPARC (p = 3.3 × 10− 7) on chromosome 5. Both genes were previously implicated in blood pressure regulation and hypertension. Conclusion We compared two meta-analysis approaches to jointly analyze multiple variants: SKAT and haplotype tests. The difference in observed results may be because the haplotype method considered all observed haplotypes, whereas SKAT weighted variants inversely to their minor allele frequency, masking the effects of common variants. The two approaches identified different top genes, and appear to be complementary.
Collapse
|
26
|
Yuan Q, Yang J, Santulli G, Reiken SR, Wronska A, Kim MM, Osborne BW, Lacampagne A, Yin Y, Marks AR. Maintenance of normal blood pressure is dependent on IP3R1-mediated regulation of eNOS. Proc Natl Acad Sci U S A 2016; 113:8532-8537. [PMID: 27402766 PMCID: PMC4968706 DOI: 10.1073/pnas.1608859113] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Endothelial cells (ECs) are critical mediators of blood pressure (BP) regulation, primarily via the generation and release of vasorelaxants, including nitric oxide (NO). NO is produced in ECs by endothelial NO synthase (eNOS), which is activated by both calcium (Ca(2+))-dependent and independent pathways. Here, we report that intracellular Ca(2+) release from the endoplasmic reticulum (ER) via inositol 1,4,5-trisphosphate receptor (IP3R) is required for Ca(2+)-dependent eNOS activation. EC-specific type 1 1,4,5-trisphosphate receptor knockout (IP3R1(-/-)) mice are hypertensive and display blunted vasodilation in response to acetylcholine (ACh). Moreover, eNOS activity is reduced in both isolated IP3R1-deficient murine ECs and human ECs following IP3R1 knockdown. IP3R1 is upstream of calcineurin, a Ca(2+)/calmodulin-activated serine/threonine protein phosphatase. We show here that the calcineurin/nuclear factor of activated T cells (NFAT) pathway is less active and eNOS levels are decreased in IP3R1-deficient ECs. Furthermore, the calcineurin inhibitor cyclosporin A, whose use has been associated with the development of hypertension, reduces eNOS activity and vasodilation following ACh stimulation. Our results demonstrate that IP3R1 plays a crucial role in the EC-mediated vasorelaxation and the maintenance of normal BP.
Collapse
Affiliation(s)
- Qi Yuan
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; Wu Center for Molecular Cardiology, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032
| | - Jingyi Yang
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; Wu Center for Molecular Cardiology, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Gaetano Santulli
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; Wu Center for Molecular Cardiology, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032;
| | - Steven R Reiken
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; Wu Center for Molecular Cardiology, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032
| | - Anetta Wronska
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; Wu Center for Molecular Cardiology, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032
| | - Mindy M Kim
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032
| | - Brent W Osborne
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032
| | - Alain Lacampagne
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; INSERM U1046, CNRS UMR-9214, Université de Montpellier, 34295 Montpellier, France
| | - Yuxin Yin
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China;
| | - Andrew R Marks
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; Wu Center for Molecular Cardiology, College of Physicians and Surgeons of Columbia University Medical Center, New York, NY 10032; Department of Medicine, Columbia University, New York, NY 10032
| |
Collapse
|
27
|
LoVerso PR, Cui F. A Computational Pipeline for Cross-Species Analysis of RNA-seq Data Using R and Bioconductor. Bioinform Biol Insights 2015; 9:165-74. [PMID: 26692761 PMCID: PMC4668955 DOI: 10.4137/bbi.s30884] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 10/21/2015] [Accepted: 10/24/2015] [Indexed: 01/25/2023] Open
Abstract
RNA sequencing (RNA-seq) has revolutionized transcriptome analysis through profiling the expression of thousands of genes at the same time. Systematic analysis of orthologous transcripts across species is critical for understanding the evolution of gene expression and uncovering important information in animal models of human diseases. Several computational methods have been published for analyzing gene expression between species, but they often lack crucial details and therefore cannot serve as a practical guide. Here, we present the first step-by-step protocol for cross-species RNA-seq analysis with a concise workflow that is largely based on the free open-source R language and Bioconductor packages. This protocol covers the entire process from short-read mapping, gene expression quantification, differential expression analysis to pathway enrichment. Many useful utilities for data visualization are included. This complete and easy-to-follow protocol provides hands-on guidance for users who are new to cross-species gene expression analysis.
Collapse
Affiliation(s)
- Peter R LoVerso
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, One Lomb Memorial Drive, Rochester, NY, USA
| | - Feng Cui
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, One Lomb Memorial Drive, Rochester, NY, USA
| |
Collapse
|
28
|
Zhang J, Wang G, Feng J, Zhang L, Li J. Identifying ion channel genes related to cardiomyopathy using a novel decision forest strategy. MOLECULAR BIOSYSTEMS 2015; 10:2407-14. [PMID: 24977958 DOI: 10.1039/c4mb00193a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Ion channels play many crucial functions in life. Their dysfunction may lead to a number of diseases, such as arrhythmia and beta cell dysfunction. In this study, we firstly selected the ion channel gene expression profiles using a dimensionality reduction method. After that, we applied a novel decision forest strategy to mine cardiomyopathy related ion channel genes. The novel proposed Zi integrated the information of the decision trees' height and the frequency at which a gene was located in the tree. It achieved a much higher ability of feature selection. In the result, 26 cardiomyopathy related ion channel genes were identified. Their Zi were higher than the threshold Z*. Furthermore, most of these genes had been reported to have relationships with cardiomyopathies. In conclusion, our proposed decision forest strategy had a better classification performance. Our result can provide a theoretical basis for cardiovascular researchers.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Prevention, Tongji University School of Medicine, Shanghai, China.
| | | | | | | | | |
Collapse
|
29
|
Morris BJ, Dampney RAL. Brain-stem microRNAs implicated in hypertension. Physiol Genomics 2015; 47:386-7. [PMID: 26242934 DOI: 10.1152/physiolgenomics.00079.2015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Brian J Morris
- Discipline of Physiology, School of Medical Sciences and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Roger A L Dampney
- Discipline of Physiology, School of Medical Sciences and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
30
|
LaDisa JF, Bozdag S, Olson J, Ramchandran R, Kersten JR, Eddinger TJ. Gene Expression in Experimental Aortic Coarctation and Repair: Candidate Genes for Therapeutic Intervention? PLoS One 2015. [PMID: 26207811 PMCID: PMC4514739 DOI: 10.1371/journal.pone.0133356] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Coarctation of the aorta (CoA) is a constriction of the proximal descending thoracic aorta and is one of the most common congenital cardiovascular defects. Treatments for CoA improve life expectancy, but morbidity persists, particularly due to the development of chronic hypertension (HTN). Identifying the mechanisms of morbidity is difficult in humans due to confounding variables such as age at repair, follow-up duration, coarctation severity and concurrent anomalies. We previously developed an experimental model that replicates aortic pathology in humans with CoA without these confounding variables, and mimics correction at various times using dissolvable suture. Here we present the most comprehensive description of differentially expressed genes (DEGs) to date from the pathology of CoA, which were obtained using this model. Aortic samples (n=4/group) from the ascending aorta that experiences elevated blood pressure (BP) from induction of CoA, and restoration of normal BP after its correction, were analyzed by gene expression microarray, and enriched genes were converted to human orthologues. 51 DEGs with >6 fold-change (FC) were used to determine enriched Gene Ontology terms, altered pathways, and association with National Library of Medicine Medical Subject Headers (MeSH) IDs for HTN, cardiovascular disease (CVD) and CoA. The results generated 18 pathways, 4 of which (cell cycle, immune system, hemostasis and metabolism) were shared with MeSH ID’s for HTN and CVD, and individual genes were associated with the CoA MeSH ID. A thorough literature search further uncovered association with contractile, cytoskeletal and regulatory proteins related to excitation-contraction coupling and metabolism that may explain the structural and functional changes observed in our experimental model, and ultimately help to unravel the mechanisms responsible for persistent morbidity after treatment for CoA.
Collapse
Affiliation(s)
- John F. LaDisa
- Department of Biomedical Engineering, Marquette University, Milwaukee, Wisconsin, United States of America
- Department of Medicine, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Biotechnology and Bioengineering Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Herma Heart Center, Children’s Hospital of Wisconsin, Milwaukee, Wisconsin, United States of America
- * E-mail:
| | - Serdar Bozdag
- Department of Mathematics, Statistics, and Computer Science, Marquette University, Milwaukee, Wisconsin, United States of America
| | - Jessica Olson
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Ramani Ramchandran
- Departments of Pediatrics and Obstetrics and Gynecology, Medical College of Wisconsin and the Developmental Vascular Biology Program, Children’s Hospital of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Judy R. Kersten
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Thomas J. Eddinger
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, United States of America
| |
Collapse
|
31
|
Fetal-adult cardiac transcriptome analysis in rats with contrasting left ventricular mass reveals new candidates for cardiac hypertrophy. PLoS One 2015; 10:e0116807. [PMID: 25646840 PMCID: PMC4315412 DOI: 10.1371/journal.pone.0116807] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 12/15/2014] [Indexed: 01/20/2023] Open
Abstract
Reactivation of fetal gene expression patterns has been implicated in common cardiac diseases in adult life including left ventricular (LV) hypertrophy (LVH) in arterial hypertension. Thus, increased wall stress and neurohumoral activation are discussed to induce the return to expression of fetal genes after birth in LVH. We therefore aimed to identify novel potential candidates for LVH by analyzing fetal-adult cardiac gene expression in a genetic rat model of hypertension, i.e. the stroke-prone spontaneously hypertensive rat (SHRSP). To this end we performed genome-wide transcriptome analysis in SHRSP to identify differences in expression patterns between day 20 of fetal development (E20) and adult animals in week 14 in comparison to a normotensive rat strain with contrasting low LV mass, i.e. Fischer (F344). 15232 probes were detected as expressed in LV tissue obtained from rats at E20 and week 14 (p < 0.05) and subsequently screened for differential expression. We identified 24 genes with SHRSP specific up-regulation and 21 genes with down-regulation as compared to F344. Further bioinformatic analysis presented Efcab6 as a new candidate for LVH that showed only in the hypertensive SHRSP rat differential expression during development (logFC = 2.41, p < 0.001) and was significantly higher expressed in adult SHRSP rats compared with adult F344 (+ 76%) and adult normotensive Wistar-Kyoto rats (+ 82%). Thus, it represents an interesting new target for further functional analyses and the elucidation of mechanisms leading to LVH. Here we report a new approach to identify candidate genes for cardiac hypertrophy by combining the analysis of gene expression differences between strains with a contrasting cardiac phenotype with a comparison of fetal-adult cardiac expression patterns.
Collapse
|
32
|
Morris BJ. Renin, genes, microRNAs, and renal mechanisms involved in hypertension. Hypertension 2015; 65:956-62. [PMID: 25601934 DOI: 10.1161/hypertensionaha.114.04366] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 12/23/2014] [Indexed: 12/20/2022]
Affiliation(s)
- Brian J Morris
- From the Basic & Clinical Genomics Laboratory, School of Medical Sciences and Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
33
|
Briant LJB, Stalbovskiy AO, Nolan MF, Champneys AR, Pickering AE. Increased intrinsic excitability of muscle vasoconstrictor preganglionic neurons may contribute to the elevated sympathetic activity in hypertensive rats. J Neurophysiol 2014; 112:2756-78. [PMID: 25122704 PMCID: PMC4254885 DOI: 10.1152/jn.00350.2014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 08/06/2014] [Indexed: 02/07/2023] Open
Abstract
Hypertension is associated with pathologically increased sympathetic drive to the vasculature. This has been attributed to increased excitatory drive to sympathetic preganglionic neurons (SPN) from brainstem cardiovascular control centers. However, there is also evidence supporting increased intrinsic excitability of SPN. To test this hypothesis, we made whole cell recordings of muscle vasoconstrictor-like (MVClike) SPN in the working-heart brainstem preparation of spontaneously hypertensive (SH) and normotensive Wistar-Kyoto (WKY) rats. The MVClike SPN have a higher spontaneous firing frequency in the SH rat (3.85 ± 0.4 vs. 2.44 ± 0.4 Hz in WKY; P = 0.011) with greater respiratory modulation of their activity. The action potentials of SH SPN had smaller, shorter afterhyperpolarizations (AHPs) and showed diminished transient rectification indicating suppression of an A-type potassium conductance (IA). We developed mathematical models of the SPN to establish if changes in their intrinsic properties in SH rats could account for their altered firing. Reduction of the maximal conductance density of IA by 15-30% changed the excitability and output of the model from the WKY to a SH profile, with increased firing frequency, amplified respiratory modulation, and smaller AHPs. This change in output is predominantly a consequence of altered synaptic integration. Consistent with these in silico predictions, we found that intrathecal 4-aminopyridine (4-AP) increased sympathetic nerve activity, elevated perfusion pressure, and augmented Traube-Hering waves. Our findings indicate that IA acts as a powerful filter on incoming synaptic drive to SPN and that its diminution in the SH rat is potentially sufficient to account for the increased sympathetic output underlying hypertension.
Collapse
Affiliation(s)
- Linford J B Briant
- School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Department of Engineering Mathematics, University of Bristol, Bristol, United Kingdom; and
| | - Alexey O Stalbovskiy
- School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom
| | - Matthew F Nolan
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Alan R Champneys
- Department of Engineering Mathematics, University of Bristol, Bristol, United Kingdom; and
| | - Anthony E Pickering
- School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Department of Anaesthesia, University Hospitals Bristol, Bristol, United Kingdom;
| |
Collapse
|
34
|
Abstract
Multiple genes and pathways are involved in the pathogenesis of hypertension. Epigenomic studies of hypertension are beginning to emerge and hold great promise of providing novel insights into the mechanisms underlying hypertension. Epigenetic marks or mediators including DNA methylation, histone modifications, and noncoding RNA can be studied at a genome or near-genome scale using epigenomic approaches. At the single gene level, several studies have identified changes in epigenetic modifications in genes expressed in the kidney that correlate with the development of hypertension. Systematic analysis and integration of epigenetic marks at the genome-wide scale, demonstration of cellular and physiological roles of specific epigenetic modifications, and investigation of inheritance are among the major challenges and opportunities for future epigenomic and epigenetic studies of hypertension.
Collapse
Affiliation(s)
- Mingyu Liang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI.
| | | | | | | | | |
Collapse
|
35
|
Chen Q, Huang CQ, Hu XY, Li SB, Zhang XM. Functional CLOCK gene rs1554483 G/C polymorphism is associated with susceptibility to Alzheimer's disease in the Chinese population. J Int Med Res 2014; 41:340-6. [PMID: 23781009 DOI: 10.1177/0300060513476430] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE To examine the association between the circadian locomotor output cycles kaput (CLOCK) gene rs1554483 G/C polymorphism and susceptibility to Alzheimer's disease in Chinese people. METHODS This case-control study determined apolipoprotein E (APOE) and CLOCK rs1554483 G/C genotypes using polymerase chain reaction restriction fragment length polymorphism. RESULTS Unrelated patients with Alzheimer's disease (n = 130) and healthy controls (n = 188) were analysed for an association between the CLOCK gene rs1554483 G/C polymorphism and susceptibility to Alzheimer's disease. In the whole sample and in APOE ε4 isoform noncarriers, the prevalence of CLOCK gene rs1554483 G allele carriers was significantly higher in patients with Alzheimer's disease than in controls. Among APOE ε4 carriers, the prevalence of CLOCK rs1554483 G allele carriers was not significantly different between patients with Alzheimer's disease and controls. CONCLUSION Among APOE ε4 noncarriers, but not APOE ε4 carriers, the CLOCK rs1554483 G allele was associated with increased susceptibility to Alzheimer's disease.
Collapse
Affiliation(s)
- Qian Chen
- Department of Geriatrics, The West China Hospital of Sichuan University, Sichuan Province, China
| | | | | | | | | |
Collapse
|
36
|
Flister MJ, Tsaih SW, O'Meara CC, Endres B, Hoffman MJ, Geurts AM, Dwinell MR, Lazar J, Jacob HJ, Moreno C. Identifying multiple causative genes at a single GWAS locus. Genome Res 2013; 23:1996-2002. [PMID: 24006081 PMCID: PMC3847770 DOI: 10.1101/gr.160283.113] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Accepted: 09/03/2013] [Indexed: 01/11/2023]
Abstract
Genome-wide association studies (GWAS) are useful for nominating candidate genes, but typically are unable to establish disease causality or differentiate between the effects of variants in linkage disequilibrium (LD). Additionally, some GWAS loci might contain multiple causative variants or genes that contribute to the overall disease susceptibility at a single locus. However, the majority of current GWAS lack the statistical power to test whether multiple causative genes underlie the same locus, prompting us to adopt an alternative approach to testing multiple GWAS genes empirically. We used gene targeting in a disease-susceptible rat model of genetic hypertension to test all six genes at the Agtrap-Plod1 locus (Agtrap, Mthfr, Clcn6, Nppa, Nppb, and Plod1) for blood pressure (BP) and renal phenotypes. This revealed that the majority of genes at this locus (five out of six) can impact hypertension by modifying BP and renal phenotypes. Mutations of Nppa, Plod1, and Mthfr increased disease susceptibility, whereas Agtrap and Clcn6 mutations decreased hypertension risk. Reanalysis of the human AGTRAP-PLOD1 locus also implied that disease-associated haplotype blocks with polygenic effects were not only possible, but rather were highly plausible. Combined, these data demonstrate for the first time that multiple modifiers of hypertension can cosegregate at a single GWAS locus.
Collapse
Affiliation(s)
- Michael J. Flister
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Shirng-Wern Tsaih
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Caitlin C. O'Meara
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Bradley Endres
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Matthew J. Hoffman
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Aron M. Geurts
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Melinda R. Dwinell
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Jozef Lazar
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
- Department of Dermatology
| | - Howard J. Jacob
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Carol Moreno
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| |
Collapse
|
37
|
HIF isoforms in the skin differentially regulate systemic arterial pressure. Proc Natl Acad Sci U S A 2013; 110:17570-5. [PMID: 24101470 DOI: 10.1073/pnas.1306942110] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Vascular flow through tissues is regulated via a number of homeostatic mechanisms. Localized control of tissue blood flow, or autoregulation, is a key factor in regulating tissue perfusion and oxygenation. We show here that the net balance between two hypoxia-inducible factor (HIF) transcription factor isoforms, HIF-1α and HIF-2α, is an essential mechanism regulating both local and systemic blood flow in the skin of mice. We also show that balance of HIF isoforms in keratinocyte-specific mutant mice affects thermal adaptation, exercise capacity, and systemic arterial pressure. The two primary HIF isoforms achieve these effects in opposing ways that are associated with HIF isoform regulation of nitric oxide production. We also show that a correlation exists between altered levels of HIF isoforms in the skin and the degree of idiopathic hypertension in human subjects. Thus, the balance between HIF-1α and HIF-2α expression in keratinocytes is a control element of both tissue perfusion and systemic arterial pressure, with potential implications in human hypertension.
Collapse
|
38
|
Abstract
Arterial hypertension is a major cardiovascular risk factor that affects between 10 and 40% of the population in industrialized countries. Primary aldosteronism (PA) is the most common form of secondary hypertension with an estimated prevalence of around 10% in referral centers and 4% in a primary care setting. Despite its high prevalence until recently, the underlying genetic and molecular basis of this common disease had remained largely obscure. Over the past decade, a number of insights have been achieved that have relied on in vitro cellular systems, wild-type and genetically modified in vivo models, as well as clinical studies in well-characterized patient populations. This progress has been made possible by a number of independent technical developments including that of specific hormone assays that allow measurement in small sample volumes as well as genetic techniques that enable high-throughput sequencing of a large number of samples. Furthermore, animal models have provided important insights into the physiology of aldosterone regulation that have served as a starting point for investigation of mechanisms involved in autonomous aldosterone secretion. Finally, national and international networks that have built up registries and biobanks have been instrumental in fostering translational research endeavors in PA. Therefore, it is to be expected that in the near future, further pathophysiological mechanisms that result in autonomous aldosterone secretion will be unraveled.
Collapse
Affiliation(s)
- Felix Beuschlein
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ziemssenstrasse 1, D-80336 Munich, Germany.
| |
Collapse
|
39
|
Marques FZ, Morris BJ. Neurogenic hypertension: revelations from genome-wide gene expression profiling. Curr Hypertens Rep 2013; 14:485-91. [PMID: 22639016 DOI: 10.1007/s11906-012-0282-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There is now good evidence for a role of the sympathetic nervous system in the etiology of essential hypertension in humans. Although genetic variation is expected to underlie the elevated sympathetic outflow in this complex polygenic condition, only limited information has emerged from classic molecular genetic studies. Recently, progress has been made in understanding neurogenic aspects by determination of global alterations in gene expression in key brain regions of animal models of neurogenic hypertension. Such genome-wide expression studies in the hypothalamus and brainstem support roles for factors such as neuronal nitric oxide synthase, inflammation and reactive oxygen species. A role for non-coding RNAs such as microRNAs, and epigenetic alterations await exploration. Ongoing novel approaches should provide a better understanding of the processes responsible for the increased sympathetic outflow in animal models, as well as essential hypertension in humans. Such information may lead to better therapies for neurogenic hypertension in humans.
Collapse
|
40
|
Hernández-Abreu O, Torres-Piedra M, García-Jiménez S, Ibarra-Barajas M, Villalobos-Molina R, Montes S, Rembao D, Estrada-Soto S. Dose-dependent antihypertensive determination and toxicological studies of tilianin isolated from Agastache mexicana. JOURNAL OF ETHNOPHARMACOLOGY 2013; 146:187-91. [PMID: 23276782 DOI: 10.1016/j.jep.2012.12.029] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 12/03/2012] [Accepted: 12/20/2012] [Indexed: 05/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Agastache mexicana is used in Mexican traditional medicine for the treatment of hypertension, anxiety and related diseases. AIM OF THE STUDY Current work was developed to establish pharmacological/toxicological parameters of tilianin, a flavone extracted from Agastache mexicana in order to propose it for clinical trials. MATERIALS AND METHODS Acute and sub-acute toxicology studies in Imprinting Control Region (ICR) mice and median effective dose (ED50) determination in conscious spontaneously hypertensive rats (SHR) were done. RESULTS A median lethal dose (LD50) of 6624 mg/kg (6201, 7076) in mice and significant antihypertensive effect (ED50=53.51 mg/kg) in SHR were determined. Moreover, sub-acute oral administration of tilianin did not alter body weight, clinical chemistry parameters (alanine amino-transferase, aspartate amino-transferase, total cholesterol, high density lipoprotein, low density lipoprotein, triglycerides, glucose and insulin), and also did not induce any toxic or adverse effects on kidney, heart, liver, and lung functions. CONCLUSIONS We have shown that tilianin, isolated from Agastache mexicana, was not toxic for rodents. Also, its antihypertensive effect was dose-dependent and ED50 (53.51 mg/kg) calculated was lesser than LD50 determined (6624 mg/kg), which suggest a wide range of pharmacology-toxicology patterns. Results support the hypothesis that tilianin must be investigated and developed for clinical trials as antihypertensive drug.
Collapse
Affiliation(s)
- Oswaldo Hernández-Abreu
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001 Col. Chamilpa, 62209 Cuernavaca, Morelos, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Kristiansson E, Österlund T, Gunnarsson L, Arne G, Larsson DGJ, Nerman O. A novel method for cross-species gene expression analysis. BMC Bioinformatics 2013; 14:70. [PMID: 23444967 PMCID: PMC3679856 DOI: 10.1186/1471-2105-14-70] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 02/13/2013] [Indexed: 12/27/2022] Open
Abstract
Background Analysis of gene expression from different species is a powerful way to identify evolutionarily conserved transcriptional responses. However, due to evolutionary events such as gene duplication, there is no one-to-one correspondence between genes from different species which makes comparison of their expression profiles complex. Results In this paper we describe a new method for cross-species meta-analysis of gene expression. The method takes the homology structure between compared species into account and can therefore compare expression data from genes with any number of orthologs and paralogs. A simulation study shows that the proposed method results in a substantial increase in statistical power compared to previously suggested procedures. As a proof of concept, we analyzed microarray data from heat stress experiments performed in eight species and identified several well-known evolutionarily conserved transcriptional responses. The method was also applied to gene expression profiles from five studies of estrogen exposed fish and both known and potentially novel responses were identified. Conclusions The method described in this paper will further increase the potential and reliability of meta-analysis of gene expression profiles from evolutionarily distant species. The method has been implemented in R and is freely available at
http://bioinformatics.math.chalmers.se/Xspecies/.
Collapse
Affiliation(s)
- Erik Kristiansson
- Department of Mathematical Statistics, Chalmers University of Technology/University of Gothenburg, Gothenburg, Sweden.
| | | | | | | | | | | |
Collapse
|
42
|
Bubb KJ, Khambata RS, Ahluwalia A. Sexual dimorphism in rodent models of hypertension and atherosclerosis. Br J Pharmacol 2013; 167:298-312. [PMID: 22582712 DOI: 10.1111/j.1476-5381.2012.02036.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Approximately one third of all deaths are attributed to cardiovascular disease (CVD), making it the biggest killer worldwide. Despite a number of therapeutic options available, the burden of CVD morbidity continues to grow indicating the need for continued research to address this unmet need. In this respect, investigation of the mechanisms underlying the protection that premenopausal females enjoy from cardiovascular-related disease and mortality is of interest. In this review, we discuss the essential role that rodent animal models play in enabling this field of research. In particular, we focus our discussion on models of hypertension and atherosclerosis.
Collapse
Affiliation(s)
- Kristen J Bubb
- William Harvey Research Institute, Clinical Pharmacology, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
| | | | | |
Collapse
|
43
|
Yang C, Stingo FC, Ahn KW, Liu P, Vannucci M, Laud PW, Skelton M, O'Connor P, Kurth T, Ryan RP, Moreno C, Tsaih SW, Patone G, Hummel O, Jacob HJ, Liang M, Cowley AW. Increased proliferative cells in the medullary thick ascending limb of the loop of Henle in the Dahl salt-sensitive rat. Hypertension 2012. [PMID: 23184381 DOI: 10.1161/hypertensionaha.112.199380] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Studies of transcriptome profiles have provided new insights into mechanisms underlying the development of hypertension. Cell type heterogeneity in tissue samples, however, has been a significant hindrance in these studies. We performed a transcriptome analysis in medullary thick ascending limbs of the loop of Henle isolated from Dahl salt-sensitive rats. Genes differentially expressed between Dahl salt-sensitive rats and salt-insensitive consomic SS.13(BN) rats on either 0.4% or 7 days of 8.0% NaCl diet (n=4) were highly enriched for genes located on chromosome 13, the chromosome substituted in the SS.13(BN) rat. A pathway involving cell proliferation and cell cycle regulation was identified as one of the most highly ranked pathways based on differentially expressed genes and by a Bayesian model analysis. Immunofluorescent analysis indicated that just 1 week of a high-salt diet resulted in a severalfold increase in proliferative medullary thick ascending limb cells in both rat strains, and that Dahl salt-sensitive rats exhibited a significantly greater proportion of medullary thick ascending limb cells in a proliferative state than in SS.13(BN) rats (15.0±1.4% versus 10.1±0.6%; n=7-9; P<0.05). The total number of cells per medullary thick ascending limb section analyzed was not different between the 2 strains. The study revealed alterations in regulatory pathways in Dahl salt-sensitive rats in tissues highly enriched for a single cell type, leading to the unexpected finding of a greater increase in the number of proliferative medullary thick ascending limb cells in Dahl salt-sensitive rats on a high-salt diet.
Collapse
Affiliation(s)
- Chun Yang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Cordwell SJ, Edwards AVG, Liddy KA, Moshkanbaryans L, Solis N, Parker BL, Yong ASC, Wong C, Kritharides L, Hambly BD, White MY. Release of tissue-specific proteins into coronary perfusate as a model for biomarker discovery in myocardial ischemia/reperfusion injury. J Proteome Res 2012; 11:2114-26. [PMID: 22250753 DOI: 10.1021/pr2006928] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Diagnosis of acute coronary syndromes is based on protein biomarkers, such as the cardiac troponins (cTnI/cTnT) and creatine kinase (CK-MB) that are released into the circulation. Biomarker discovery is focused on identifying very low abundance tissue-derived analytes from within albumin-rich plasma, in which the wide dynamic range of the native protein complement hinders classical proteomic investigations. We employed an ex vivo rabbit model of myocardial ischemia/reperfusion (I/R) injury using Langendorff buffer perfusion. Nonrecirculating perfusate was collected over a temporal profile of 60 min reperfusion following brief, reversible ischemia (15 min; 15I/60R) for comparison with irreversible I/R (60I/60R). Perfusate proteins were separated using two-dimensional gel electrophoresis (2-DE) and identified by mass spectrometry (MS), revealing 26 tissue-specific proteins released during reperfusion post-15I. Proteins released during irreversible I/R (60I/60R) were profiled using gel-based (2-DE and one-dimensional gel electrophoresis coupled to liquid chromatography and tandem mass spectrometry; geLC-MS) and gel-free (LC-MS/MS) methods. A total of 192 tissue-specific proteins were identified during reperfusion post-60I. Identified proteins included those previously associated with I/R (myoglobin, CK-MB, cTnI, and cTnT), in addition to examples currently under investigation in large cohort studies (heart-type fatty acid binding protein; FABPH). The postischemic release profile of a novel cardiac-specific protein, cysteine and glycine-rich protein 3 (Csrp3; cardiac LIM domain protein) was validated by Western blot analysis. We also identified Csrp3 in serum from 6 of 8 patients postreperfusion following acute myocardial infarction. These studies indicate that animal modeling of biomarker release using ex vivo buffer perfused tissue to limit the presence of obfuscating plasma proteins may identify candidates for further study in humans.
Collapse
Affiliation(s)
- Stuart J Cordwell
- School of Molecular Bioscience, The University of Sydney, and Department of Cardiology, Concord Repatriation General Hospital, New South Wales, Australia 2006.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Asghar M, Tayebati SK, Lokhandwala MF, Hussain T. Potential dopamine-1 receptor stimulation in hypertension management. Curr Hypertens Rep 2011; 13:294-302. [PMID: 21633929 DOI: 10.1007/s11906-011-0211-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The role of dopamine receptors in blood pressure regulation is well established. Genetic ablation of both dopamine D1-like receptor subtypes (D1, D5) and D2-like receptor subtypes (D2, D3, D4) results in a hypertensive phenotype in mice. This review focuses on the dopamine D1-like receptor subtypes D1 and D5 (especially D1 receptors), as they play a major role in regulating sodium homeostasis and blood pressure. Studies mostly describing the role of renal dopamine D1-like receptors are included, as the kidneys play a pivotal role in the maintenance of sodium homeostasis and the long-term regulation of blood pressure. We also attempt to describe the interaction between D1-like receptors and other proteins, especially angiotensin II type 1 and type 2 receptors, which are involved in the maintenance of sodium homeostasis and blood pressure. Finally, we discuss a new concept of renal D1 receptor regulation in hypertension that involves oxidative stress mechanisms.
Collapse
Affiliation(s)
- Mohammad Asghar
- Heart and Kidney Institute, College of Pharmacy, University of Houston, Houston, TX 77204, USA.
| | | | | | | |
Collapse
|
46
|
Wang F, Song Y, Jiang Y, Yang C, Ding Z. Associations among periostin gene polymorphisms, clinical parameters and heart failure: a case-control study in 1104 Chinese individuals. J Cardiovasc Med (Hagerstown) 2011; 12:469-74. [PMID: 21558869 DOI: 10.2459/jcm.0b013e328347e48c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AIMS To determine the associations among periostin gene polymorphisms, clinical parameters and heart failure in a Chinese population. METHODS In total, 464 patients with heart failure and 640 control individuals were included in this study. rs3829365 and rs1028728 were genotyped through PCR and restriction fragment length polymorphism. Multivariate logistic regression was employed to analyze the independent strength of association among clinical parameters, genotypes and heart failure. RESULTS rs3829365 was associated with heart failure (P = 0.043), whereas rs1028728 was not (P = 0.188). After adjusting for age, sex, hypertension, diabetes mellitus, smoking and hypertriglyceridemia in multivariate logistic regression, we found that CG or GG of rs3829365 [P = 0.015, odds ratio (OR) = 1.88] was an independent risk factor for heart failure and that CG or GG of rs3829365 (P = 0.039, OR = 0.94) and AT or TT of rs1028728 (P = 0.011, OR = 0.68) were significantly protective factors for heart failure in patients who smoked and in patients with hypertriglyceridemia, respectively. Moreover, heart failure in patients with CG or GG genotype of rs3829365 tended to be more severe than in those with CC genotype. CONCLUSION These findings suggest that rs3829365 of the periostin gene may be helpful to determine the susceptibility to, and severity of, heart failure. The interactions between rs3829365 and smoking and between rs1028728 and hypertriglyceridemia warrant further investigations for underlying mechanisms.
Collapse
Affiliation(s)
- Fangfang Wang
- Department of Cardiovascular Disease, Changzhou No. 2 People's Hospital, Affiliated Nanjing Medical University, Changzhou, Jiangsu Province, China
| | | | | | | | | |
Collapse
|
47
|
Dummler S, Eichhorn S, Tesche C, Schreiber U, Voss B, Deutsch MA, Hauner H, Lahm H, Lange R, Krane M. Pulsatile ex vivo perfusion of human saphenous vein grafts under controlled pressure conditions increases MMP-2 expression. Biomed Eng Online 2011; 10:62. [PMID: 21777461 PMCID: PMC3148203 DOI: 10.1186/1475-925x-10-62] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 07/21/2011] [Indexed: 11/16/2022] Open
Abstract
Background The use of human saphenous vein grafts (HSVGs) as a bypass conduit is a standard procedure in the treatment of coronary artery disease while their early occlusion remains a major problem. Methods We have developed an ex vivo perfusion system, which uses standardized and strictly controlled hemodynamic parameters for the pulsatile and non-static perfusion of HSVGs to guarantee a reliable analysis of molecular parameters under different pressure conditions. Cell viability of HSVGs (n = 12) was determined by the metabolic conversion of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) into a purple formazan dye. Results Under physiological flow rates (10 mmHg) HSVGs remained viable for two weeks. Their exposure to arterial conditions (100 mmHg) was possible for one week without important reduction in viability. Baseline expression of matrix metalloproteinase-2 (MMP-2) after venous perfusion (2.2 ± 0.5, n = 5) was strongly up-regulated after exposure to arterial conditions for three days (19.8 ± 4.3) or five days (23.9 ± 6.1, p < 0.05). Zymographic analyses confirmed this increase on the protein level. Our results suggest that expression and activity of MMP-2 are strongly increased after exposure of HSVGs to arterial hemodynamic conditions compared to physiological conditions. Conclusion Therefore, our system might be helpful to more precisely understand the molecular mechanisms leading to an early failure of HSVGs.
Collapse
Affiliation(s)
- Sara Dummler
- German Heart Center Munich at the Technische Universität München, Department of Cardiovascular Surgery, Lazarettstrasse 36, D-80636 Munich, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Marques FZ, Campain AE, Davern PJ, Yang YHJ, Head GA, Morris BJ. Global identification of the genes and pathways differentially expressed in hypothalamus in early and established neurogenic hypertension. Physiol Genomics 2011; 43:766-71. [DOI: 10.1152/physiolgenomics.00009.2011] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The hypothalamus has an important etiological role in the onset and maintenance of hypertension and stress responses in the Schlager high blood pressure (BP) (BPH/2J) mouse, a genetic model of neurogenic hypertension. Using Affymetrix GeneChip Mouse Gene 1.0 ST Arrays we identified 1,019 hypothalamic genes whose expression differed between 6 wk old BPH/2J and normal BP (BPN/3J) strains, and 466 for 26 wk old mice. Of these, 459 were in 21 mouse BP quantitative trait loci. We validated 46 genes by qPCR. Gene changes that would increase sympathetic outflow at both ages were: Dynll1 encoding dynein light chain LC8-type 1, which physically destabilizes neuronal nitric oxide synthase, decreasing neuronal nitric oxide, and Hcrt encoding hypocretin and Npsr1 encoding neuropeptide S receptor 1, each involved in sympathetic response to stress. At both ages we identified genes for inflammation, such as CC-chemokine ligand 19 ( Ccl19), and oxidative stress. Via reactive oxygen species generation, these could contribute to oxidative damage. Other genes identified could be responding to such perturbations. Atp2b1, the major gene from genome-wide association studies of BP variation, was underexpressed in the early phase. Comparison of profiles of young and adult BPH/2J mice, after adjusting for maturation genes, pointed to the proopiomelanocortin-α gene ( Pomc) and neuropeptide Y gene ( Npy), among others, as potentially causative. The present study has identified a diversity of genes and possible mechanisms involved in hypertension etiology and maintenance in the hypothalamus of BPH/2J mice, highlighting both common and divergent processes in each phase of the condition.
Collapse
Affiliation(s)
- Francine Z. Marques
- Basic & Clinical Genomics Laboratory, School of Medical Sciences and Bosch Institute, and
| | - Anna E. Campain
- School of Mathematics and Statistics, The University of Sydney, Sydney, New South Wales; and
| | - Pamela J. Davern
- the Neuropharmacology Laboratory, Baker IDI Heart Research Institute, Melbourne, Victoria, Australia
| | - Yee Hwa J. Yang
- School of Mathematics and Statistics, The University of Sydney, Sydney, New South Wales; and
| | - Geoffrey A. Head
- the Neuropharmacology Laboratory, Baker IDI Heart Research Institute, Melbourne, Victoria, Australia
| | - Brian J. Morris
- Basic & Clinical Genomics Laboratory, School of Medical Sciences and Bosch Institute, and
| |
Collapse
|
49
|
Abstract
The assessment of salt sensitivity of blood pressure is difficult because of the lack of universal consensus on definition. Regardless of the variability in the definition of salt sensitivity, increased salt intake, independent of the actual level of blood pressure, is also a risk factor for cardiovascular morbidity and mortality and kidney disease. A modest reduction in salt intake results in an immediate decrease in blood pressure, with long-term beneficial consequences. However, some have suggested that dietary sodium restriction may not be beneficial to everyone. Thus, there is a need to distinguish salt-sensitive from salt-resistant individuals, but it has been difficult to do so with phenotypic studies. Therefore, there is a need to determine the genes that are involved in salt sensitivity. This review focuses on genes associated with salt sensitivity, with emphasis on the variants associated with salt sensitivity in humans that are not due to monogenic causes. Special emphasis is given to gene variants associated with salt sensitivity whose protein products interfere with cell function and increase blood pressure in transgenic mice.
Collapse
Affiliation(s)
- Hironobu Sanada
- Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan.
| | | | | |
Collapse
|
50
|
Marques FZ, Campain AE, Davern PJ, Yang YHJ, Head GA, Morris BJ. Genes influencing circadian differences in blood pressure in hypertensive mice. PLoS One 2011; 6:e19203. [PMID: 21541337 PMCID: PMC3082552 DOI: 10.1371/journal.pone.0019203] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 03/29/2011] [Indexed: 01/11/2023] Open
Abstract
Essential hypertension is a common multifactorial heritable condition in which increased sympathetic outflow from the central nervous system is involved in the elevation in blood pressure (BP), as well as the exaggerated morning surge in BP that is a risk factor for myocardial infarction and stroke in hypertensive patients. The Schlager BPH/2J mouse is a genetic model of hypertension in which increased sympathetic outflow from the hypothalamus has an important etiological role in the elevation of BP. Schlager hypertensive mice exhibit a large variation in BP between the active and inactive periods of the day, and also show a morning surge in BP. To investigate the genes responsible for the circadian variation in BP in hypertension, hypothalamic tissue was collected from BPH/2J and normotensive BPN/3J mice at the ‘peak’ (n = 12) and ‘trough’ (n = 6) of diurnal BP. Using Affymetrix GeneChip® Mouse Gene 1.0 ST Arrays, validation by quantitative real-time PCR and a statistical method that adjusted for clock genes, we identified 212 hypothalamic genes whose expression differed between ‘peak’ and ‘trough’ BP in the hypertensive strain. These included genes with known roles in BP regulation, such as vasopressin, oxytocin and thyrotropin releasing hormone, as well as genes not recognized previously as regulators of BP, including chemokine (C-C motif) ligand 19, hypocretin and zinc finger and BTB domain containing 16. Gene ontology analysis showed an enrichment of terms for inflammatory response, mitochondrial proton-transporting ATP synthase complex, structural constituent of ribosome, amongst others. In conclusion, we have identified genes whose expression differs between the peak and trough of 24-hour circadian BP in BPH/2J mice, pointing to mechanisms responsible for diurnal variation in BP. The findings may assist in the elucidation of the mechanism for the morning surge in BP in essential hypertension.
Collapse
Affiliation(s)
- Francine Z. Marques
- Basic and Clinical Genomics Laboratory, School of Medical Sciences and Bosch Institute, The University of Sydney, Sydney, Australia
| | - Anna E. Campain
- School of Mathematics and Statistics, The University of Sydney, Sydney, Australia
| | - Pamela J. Davern
- Neuropharmacology Laboratory, Baker IDI Heart Research Institute, Melbourne, Australia
| | - Yee Hwa J. Yang
- School of Mathematics and Statistics, The University of Sydney, Sydney, Australia
| | - Geoffrey A. Head
- Neuropharmacology Laboratory, Baker IDI Heart Research Institute, Melbourne, Australia
| | - Brian J. Morris
- Basic and Clinical Genomics Laboratory, School of Medical Sciences and Bosch Institute, The University of Sydney, Sydney, Australia
- * E-mail:
| |
Collapse
|