51
|
Oliveros W, Delfosse K, Lato DF, Kiriakopulos K, Mokhtaridoost M, Said A, McMurray BJ, Browning JW, Mattioli K, Meng G, Ellis J, Mital S, Melé M, Maass PG. Systematic characterization of regulatory variants of blood pressure genes. CELL GENOMICS 2023; 3:100330. [PMID: 37492106 PMCID: PMC10363820 DOI: 10.1016/j.xgen.2023.100330] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/29/2023] [Accepted: 04/28/2023] [Indexed: 07/27/2023]
Abstract
High blood pressure (BP) is the major risk factor for cardiovascular disease. Genome-wide association studies have identified genetic variants for BP, but functional insights into causality and related molecular mechanisms lag behind. We functionally characterize 4,608 genetic variants in linkage with 135 BP loci in vascular smooth muscle cells and cardiomyocytes by massively parallel reporter assays. High densities of regulatory variants at BP loci (i.e., ULK4, MAP4, CFDP1, PDE5A) indicate that multiple variants drive genetic association. Regulatory variants are enriched in repeats, alter cardiovascular-related transcription factor motifs, and spatially converge with genes controlling specific cardiovascular pathways. Using heuristic scoring, we define likely causal variants, and CRISPR prime editing finally determines causal variants for KCNK9, SFXN2, and PCGF6, which are candidates for developing high BP. Our systems-level approach provides a catalog of functionally relevant variants and their genomic architecture in two trait-relevant cell lines for a better understanding of BP gene regulation.
Collapse
Affiliation(s)
- Winona Oliveros
- Life Sciences Department, Barcelona Supercomputing Center, 08034 Barcelona, Catalonia, Spain
| | - Kate Delfosse
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Daniella F. Lato
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Katerina Kiriakopulos
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Milad Mokhtaridoost
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Abdelrahman Said
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Brandon J. McMurray
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Jared W.L. Browning
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Kaia Mattioli
- Division of Genetics, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Guoliang Meng
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - James Ellis
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Seema Mital
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Ted Rogers Centre for Heart Research, Toronto, ON M5G 1X8, Canada
- Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 0A4, Canada
| | - Marta Melé
- Life Sciences Department, Barcelona Supercomputing Center, 08034 Barcelona, Catalonia, Spain
| | - Philipp G. Maass
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
52
|
Wu TT, Zheng YY, Ma X, Xiu WJ, Yang HT, Hou XG, Yang Y, Chen Y, Ma YT, Xie X. Mutated CYP17A1 promotes atherosclerosis and early-onset coronary artery disease. Cell Commun Signal 2023; 21:155. [PMID: 37370070 PMCID: PMC10294473 DOI: 10.1186/s12964-023-01061-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/29/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Coronary artery disease (CAD) is a multi-factor complex trait and is heritable, especially in early-onset families. However, the genetic factors affecting the susceptibility of early-onset CAD are not fully characterized. METHODS In the present study, we identified a rare nonsense variant in the CYP17A1 gene from a Chinese Han family with CAD. To validate the effect of this variation on atherosclerosis and early-onset coronary artery disease, we conducted studies on population, cells, and mice. RESULTS The mutation precisely congregated with the clinical syndrome in all the affected family members and was absent in unaffected family members and unrelated controls. Similar to the human phenotype, the CYP17A1-deficient mice present the phenotype of metabolic syndrome with hypertension, increased serum glucose concentration, and presentation of central obesity and fatty liver. Furthermore, CYP17A1 knockout mice or CYP17A1 + ApoE double knockout mice developed more atherosclerotic lesions than wild type (WT) with high fat diary. In cell models, CYP17A1 was found to be involved in glucose metabolism by increasing glucose intake and utilization, through activating IGF1/mTOR/HIF1-α signaling way, which was consistent in CYP17A1 knockout mice with impaired glucose tolerance and insulin resistance. CONCLUSIONS Through our study of cells, mice and humans, we identified CYP17A1 as a key protein participating in the pathophysiology of the atherosclerotic process and the possible mechanism of CYP17A1 C987X mutation induced atherosclerosis and early-onset CAD involving glucose homeostasis regulation was revealed. Video Abstract.
Collapse
Affiliation(s)
- Ting-Ting Wu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China
| | - Ying-Ying Zheng
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China
| | - Xiang Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China
| | - Wen-Juan Xiu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China
| | - Hai-Tao Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China
| | - Xian-Geng Hou
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China
| | - Yi Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China
| | - You Chen
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China
| | - Yi-Tong Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China.
| | - Xiang Xie
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Urumqi, 830011, People's Republic of China.
| |
Collapse
|
53
|
De Beer D, Mels CMC, Schutte AE, Delles C, Mary S, Mullen W, Latosinska A, Mischak H, Kruger R. Identifying a urinary peptidomics profile for hypertension in young adults: The African-PREDICT study: Urinary peptidomics and hypertension: Urinary peptidomics and hypertension. Proteomics 2023; 23:e2200444. [PMID: 36943111 DOI: 10.1002/pmic.202200444] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/27/2023] [Accepted: 03/02/2023] [Indexed: 03/23/2023]
Abstract
Hypertension is one of the most important and complex risk factors for cardiovascular diseases (CVDs). By using urinary peptidomics analyses, we aimed to identify peptides associated with hypertension, building a framework for future research towards improved prediction and prevention of premature development of CVD. We included 78 hypertensive and 79 normotensive participants from the African-PREDICT study (aged 20-30 years), matched for sex (51% male) and ethnicity (49% black and 51% white). Urinary peptidomics data were acquired using capillary-electrophoresis-time-of-flight-mass-spectrometry. Hypertension-associated peptides were identified and combined into a support vector machine-based multidimensional classifier. When comparing the peptide data between the normotensive and hypertensive groups, 129 peptides were nominally differentially abundant (Wilcoxon p < 0.05). Nonetheless, only three peptides, all derived from collagen alpha-1(III), remained significantly different after rigorous adjustments for multiple comparisons. The 37 most significant peptides (all p ≤ 0.001) served as basis for the development of a classifier, with 20 peptides being combined into a unifying score, resulting in an AUC of 0.85 in the ROC analysis (p < 0.001), with 83% sensitivity at 80% specificity. Our study suggests potential value of urinary peptides in the classification of hypertension, which could enable earlier diagnosis and better understanding of the pathophysiology of hypertension and premature cardiovascular disease development.
Collapse
Affiliation(s)
- Dalene De Beer
- Hypertension in Africa Research Team (HART), North-West University (Potchefstroom Campus), Potchefstroom, South Africa
| | - Catharina M C Mels
- Hypertension in Africa Research Team (HART), North-West University (Potchefstroom Campus), Potchefstroom, South Africa
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Aletta E Schutte
- Hypertension in Africa Research Team (HART), North-West University (Potchefstroom Campus), Potchefstroom, South Africa
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
- School of Population Health, The George Institute for Global Health, University of New South Wales, Sydney, Australia
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Sheon Mary
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - William Mullen
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | | | | | - Ruan Kruger
- Hypertension in Africa Research Team (HART), North-West University (Potchefstroom Campus), Potchefstroom, South Africa
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| |
Collapse
|
54
|
Shang L, Zhao W, Wang YZ, Li Z, Choi JJ, Kho M, Mosley TH, Kardia SLR, Smith JA, Zhou X. meQTL mapping in the GENOA study reveals genetic determinants of DNA methylation in African Americans. Nat Commun 2023; 14:2711. [PMID: 37169753 PMCID: PMC10175543 DOI: 10.1038/s41467-023-37961-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 04/07/2023] [Indexed: 05/13/2023] Open
Abstract
Identifying genetic variants that are associated with variation in DNA methylation, an analysis commonly referred to as methylation quantitative trait locus (meQTL) mapping, is an important first step towards understanding the genetic architecture underlying epigenetic variation. Most existing meQTL mapping studies have focused on individuals of European ancestry and are underrepresented in other populations, with a particular absence of large studies in populations with African ancestry. We fill this critical knowledge gap by performing a large-scale cis-meQTL mapping study in 961 African Americans from the Genetic Epidemiology Network of Arteriopathy (GENOA) study. We identify a total of 4,565,687 cis-acting meQTLs in 320,965 meCpGs. We find that 45% of meCpGs harbor multiple independent meQTLs, suggesting potential polygenic genetic architecture underlying methylation variation. A large percentage of the cis-meQTLs also colocalize with cis-expression QTLs (eQTLs) in the same population. Importantly, the identified cis-meQTLs explain a substantial proportion (median = 24.6%) of methylation variation. In addition, the cis-meQTL associated CpG sites mediate a substantial proportion (median = 24.9%) of SNP effects underlying gene expression. Overall, our results represent an important step toward revealing the co-regulation of methylation and gene expression, facilitating the functional interpretation of epigenetic and gene regulation underlying common diseases in African Americans.
Collapse
Affiliation(s)
- Lulu Shang
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yi Zhe Wang
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Zheng Li
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jerome J Choi
- Population Health Sciences, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, 53726, USA
| | - Minjung Kho
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Thomas H Mosley
- Memory Impairment and Neurodegenerative Dementia (MIND) Center, University of Mississippi Medical Center, Jackson, MS, 39126, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Xiang Zhou
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
55
|
Clavero E, Sanchez-Maldonado JM, Macauda A, Ter Horst R, Sampaio-Marques B, Jurczyszyn A, Clay-Gilmour A, Stein A, Hildebrandt MAT, Weinhold N, Buda G, García-Sanz R, Tomczak W, Vogel U, Jerez A, Zawirska D, Wątek M, Hofmann JN, Landi S, Spinelli JJ, Butrym A, Kumar A, Martínez-López J, Galimberti S, Sarasquete ME, Subocz E, Iskierka-Jażdżewska E, Giles GG, Rybicka-Ramos M, Kruszewski M, Abildgaard N, Verdejo FG, Sánchez Rovira P, da Silva Filho MI, Kadar K, Razny M, Cozen W, Pelosini M, Jurado M, Bhatti P, Dudzinski M, Druzd-Sitek A, Orciuolo E, Li Y, Norman AD, Zaucha JM, Reis RM, Markiewicz M, Rodríguez Sevilla JJ, Andersen V, Jamroziak K, Hemminki K, Berndt SI, Rajkumar V, Mazur G, Kumar SK, Ludovico P, Nagler A, Chanock SJ, Dumontet C, Machiela MJ, Varkonyi J, Camp NJ, Ziv E, Vangsted AJ, Brown EE, Campa D, Vachon CM, Netea MG, Canzian F, Försti A, Sainz J. Polymorphisms within Autophagy-Related Genes as Susceptibility Biomarkers for Multiple Myeloma: A Meta-Analysis of Three Large Cohorts and Functional Characterization. Int J Mol Sci 2023; 24:8500. [PMID: 37239846 PMCID: PMC10218542 DOI: 10.3390/ijms24108500] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/10/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Multiple myeloma (MM) arises following malignant proliferation of plasma cells in the bone marrow, that secrete high amounts of specific monoclonal immunoglobulins or light chains, resulting in the massive production of unfolded or misfolded proteins. Autophagy can have a dual role in tumorigenesis, by eliminating these abnormal proteins to avoid cancer development, but also ensuring MM cell survival and promoting resistance to treatments. To date no studies have determined the impact of genetic variation in autophagy-related genes on MM risk. We performed meta-analysis of germline genetic data on 234 autophagy-related genes from three independent study populations including 13,387 subjects of European ancestry (6863 MM patients and 6524 controls) and examined correlations of statistically significant single nucleotide polymorphisms (SNPs; p < 1 × 10-9) with immune responses in whole blood, peripheral blood mononuclear cells (PBMCs), and monocyte-derived macrophages (MDM) from a large population of healthy donors from the Human Functional Genomic Project (HFGP). We identified SNPs in six loci, CD46, IKBKE, PARK2, ULK4, ATG5, and CDKN2A associated with MM risk (p = 4.47 × 10-4-5.79 × 10-14). Mechanistically, we found that the ULK4rs6599175 SNP correlated with circulating concentrations of vitamin D3 (p = 4.0 × 10-4), whereas the IKBKErs17433804 SNP correlated with the number of transitional CD24+CD38+ B cells (p = 4.8 × 10-4) and circulating serum concentrations of Monocyte Chemoattractant Protein (MCP)-2 (p = 3.6 × 10-4). We also found that the CD46rs1142469 SNP correlated with numbers of CD19+ B cells, CD19+CD3- B cells, CD5+IgD- cells, IgM- cells, IgD-IgM- cells, and CD4-CD8- PBMCs (p = 4.9 × 10-4-8.6 × 10-4) and circulating concentrations of interleukin (IL)-20 (p = 0.00082). Finally, we observed that the CDKN2Ars2811710 SNP correlated with levels of CD4+EMCD45RO+CD27- cells (p = 9.3 × 10-4). These results suggest that genetic variants within these six loci influence MM risk through the modulation of specific subsets of immune cells, as well as vitamin D3-, MCP-2-, and IL20-dependent pathways.
Collapse
Affiliation(s)
- Esther Clavero
- Hematology Department, Virgen de las Nieves University Hospital, 18012 Granada, Spain; (E.C.); (M.J.)
| | - José Manuel Sanchez-Maldonado
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain;
- Instituto de Investigación Biosanataria IBs, Granada, 18014 Granada, Spain
| | - Angelica Macauda
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.M.); (A.S.); (F.C.)
| | - Rob Ter Horst
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (R.T.H.); (Y.L.); (M.G.N.)
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Belém Sampaio-Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (B.S.-M.); (P.L.)
| | - Artur Jurczyszyn
- Plasma Cell Dyscrasias Center, Department of Hematology, Jagiellonian University Medical College, 31-066 Kraków, Poland;
| | - Alyssa Clay-Gilmour
- Department of Biostatistics and Epidemiology, Arnold School of Public Health, University of South Carolina, Greenville, SC 29208, USA;
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55902, USA; (A.D.N.); (C.M.V.)
| | - Angelika Stein
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.M.); (A.S.); (F.C.)
| | - Michelle A. T. Hildebrandt
- Department of Lymphoma–Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Niels Weinhold
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
- Department of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
| | - Gabriele Buda
- Haematology Unit, Department of Clinical and Experimental Medicine, University of Pisa/AOUP, 56126 Pisa, Italy; (G.B.); (S.G.); (E.O.)
| | - Ramón García-Sanz
- Diagnostic Laboratory Unit in Hematology, University Hospital of Salamanca, IBSAL, CIBERONC, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (R.G.-S.); (M.E.S.)
| | - Waldemar Tomczak
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-059 Lublin, Poland;
| | - Ulla Vogel
- National Research Centre for the Working Environment, DK-2100 Copenhagen, Denmark;
| | - Andrés Jerez
- Department of Hematology, Experimental Hematology Unit, Vall d’Hebron Institute of Oncology (VHIO), University Hospital Vall d’Hebron, 08035 Barcelona, Spain;
| | - Daria Zawirska
- Department of Hematology, University Hospital, 30-688 Kraków, Poland;
| | - Marzena Wątek
- Holycross Medical Oncology Center, 25-735 Kielce, Poland;
- Institute of Hematology and Transfusion Medicine, 00-791 Warsaw, Poland
| | - Jonathan N. Hofmann
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (J.N.H.); (S.I.B.); (S.J.C.); (M.J.M.)
| | - Stefano Landi
- Department of Biology, University of Pisa, 56126 Pisa, Italy; (S.L.); (D.C.)
| | - John J. Spinelli
- Division of Population Oncology, BC Cancer, Vancouver, BC V5Z 4E6, Canada;
- School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Aleksandra Butrym
- Department of Cancer Prevention and Therapy, Wroclaw Medical University, 50-367 Wroclaw, Poland;
- Alfred Sokolowski Specialist Hospital in Walbrzych Oncology Support Centre for Clinical Trials, 58-309 Walbrzych, Poland
| | - Abhishek Kumar
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India;
- Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | | | - Sara Galimberti
- Haematology Unit, Department of Clinical and Experimental Medicine, University of Pisa/AOUP, 56126 Pisa, Italy; (G.B.); (S.G.); (E.O.)
| | - María Eugenia Sarasquete
- Diagnostic Laboratory Unit in Hematology, University Hospital of Salamanca, IBSAL, CIBERONC, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), 37007 Salamanca, Spain; (R.G.-S.); (M.E.S.)
| | - Edyta Subocz
- Department of Hematology, Military Institute of Medicine, 04-141 Warsaw, Poland;
| | | | - Graham G. Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC 3004, Australia;
- Centre for Epidemiology and Biostatistics, School of Population and Global Health, The University of Melbourne, Melbourne, VIC 3010, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC 3168, Australia
| | - Malwina Rybicka-Ramos
- Department of Hematology, Specialist Hospital No. 1 in Bytom, Academy of Silesia, Faculty of Medicine, 40-055 Katowice, Poland;
| | - Marcin Kruszewski
- Department of Hematology, University Hospital No. 2, 85-168 Bydgoszcz, Poland;
| | - Niels Abildgaard
- Department of Hematology, Odense University Hospital, DK-5000 Odense, Denmark;
| | - Francisco García Verdejo
- Department of Medical Oncology, Complejo Hospitalario de Jaén, 23007 Jaén, Spain; (F.G.V.); (P.S.R.)
| | - Pedro Sánchez Rovira
- Department of Medical Oncology, Complejo Hospitalario de Jaén, 23007 Jaén, Spain; (F.G.V.); (P.S.R.)
| | - Miguel Inacio da Silva Filho
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany;
| | | | - Małgorzata Razny
- Department of Hematology, Rydygier Hospital, 31-826 Cracow, Poland;
| | - Wendy Cozen
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, Department of Pathology, School of Medicine, Susan and Henry Samueli College of Health Sciences, Chao Family Comprehensive Cancer Center, University of California at Irvine, Irvine, CA 92697, USA;
| | - Matteo Pelosini
- U.O. Dipartimento di Ematologia, Azienda USL Toscana Nord Ovest, 57124 Livorno, Italy;
| | - Manuel Jurado
- Hematology Department, Virgen de las Nieves University Hospital, 18012 Granada, Spain; (E.C.); (M.J.)
- Instituto de Investigación Biosanataria IBs, Granada, 18014 Granada, Spain
- Department of Medicine, University of Granada, 18012 Granada, Spain
| | - Parveen Bhatti
- Cancer Control Research, BC Cancer, Vancouver, BC V5Z 4E6, Canada;
- Program in Epidemiology, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Marek Dudzinski
- Department of Hematology, Institute of Medical Sciences, College of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland; (M.D.); (M.M.)
| | - Agnieszka Druzd-Sitek
- Department of Lymphoproliferative Diseases, Maria Skłodowska Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| | - Enrico Orciuolo
- Haematology Unit, Department of Clinical and Experimental Medicine, University of Pisa/AOUP, 56126 Pisa, Italy; (G.B.); (S.G.); (E.O.)
| | - Yang Li
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (R.T.H.); (Y.L.); (M.G.N.)
- Centre for Individualised Infection Medicine (CiiM) & TWINCORE, Joint Ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), 30625 Hannover, Germany
| | - Aaron D. Norman
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55902, USA; (A.D.N.); (C.M.V.)
- Genetic Epidemiology and Risk Assessment Program, Mayo Clinic Comprehensive Cancer Center, Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55902, USA
| | - Jan Maciej Zaucha
- Department of Hematology and Transplantology, Medical University of Gdansk, 80-210 Gdansk, Poland;
| | - Rui Manuel Reis
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal and ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal;
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil
| | - Miroslaw Markiewicz
- Department of Hematology, Institute of Medical Sciences, College of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland; (M.D.); (M.M.)
| | | | - Vibeke Andersen
- Molecular Diagnostics and Clinical Research Unit, Institute of Regional Health Research, University Hospital of Southern Denmark, DK-6200 Aabenraa, Denmark;
| | - Krzysztof Jamroziak
- Department of Hematology, Transplantology and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Kari Hemminki
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany;
- Faculty of Medicine and Biomedical Center in Pilsen, Charles University in Prague, 30605 Pilsen, Czech Republic
| | - Sonja I. Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (J.N.H.); (S.I.B.); (S.J.C.); (M.J.M.)
| | - Vicent Rajkumar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA; (V.R.); (S.K.K.)
| | - Grzegorz Mazur
- Department of Internal Diseases, Occupational Medicine, Hypertension and Clinical Oncology, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Shaji K. Kumar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA; (V.R.); (S.K.K.)
| | - Paula Ludovico
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (B.S.-M.); (P.L.)
| | - Arnon Nagler
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer 52621, Israel;
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (J.N.H.); (S.I.B.); (S.J.C.); (M.J.M.)
| | - Charles Dumontet
- UMR INSERM 1052/CNRS 5286, University of Lyon, Hospices Civils de Lyon, 69008 Lyon, France;
| | - Mitchell J. Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (J.N.H.); (S.I.B.); (S.J.C.); (M.J.M.)
| | | | - Nicola J. Camp
- Division of Hematology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA;
| | - Elad Ziv
- Department of Medicine, University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94143, USA;
| | - Annette Juul Vangsted
- Department of Hematology, Rigshospitalet, Copenhagen University, DK-2100 Copenhagen, Denmark;
| | - Elizabeth E. Brown
- Department of Pathology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Daniele Campa
- Department of Biology, University of Pisa, 56126 Pisa, Italy; (S.L.); (D.C.)
| | - Celine M. Vachon
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55902, USA; (A.D.N.); (C.M.V.)
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (R.T.H.); (Y.L.); (M.G.N.)
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.M.); (A.S.); (F.C.)
| | - Asta Försti
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany;
- Hopp Children’s Cancer Center (KiTZ), 69120 Heidelberg, Germany
| | - Juan Sainz
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain;
- Instituto de Investigación Biosanataria IBs, Granada, 18014 Granada, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, 18071 Granada, Spain
| |
Collapse
|
56
|
Ivanova T, Churnosova M, Abramova M, Ponomarenko I, Reshetnikov E, Aristova I, Sorokina I, Churnosov M. Risk Effects of rs1799945 Polymorphism of the HFE Gene and Intergenic Interactions of GWAS-Significant Loci for Arterial Hypertension in the Caucasian Population of Central Russia. Int J Mol Sci 2023; 24:ijms24098309. [PMID: 37176017 PMCID: PMC10179076 DOI: 10.3390/ijms24098309] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
The aim of this case-control replicative study was to investigate the link between GWAS-impact for arterial hypertension (AH) and/or blood pressure (BP) gene polymorphisms and AH risk in Russian subjects (Caucasian population of Central Russia). AH (n = 939) and control (n = 466) cohorts were examined for ten GWAS AH/BP risk loci. The genotypes/alleles of these SNP and their combinations (SNP-SNP interactions) were tested for their association with the AH development using a logistic regression statistical procedure. The genotype GG of the SNP rs1799945 (C/G) HFE was strongly linked with an increased AH risk (ORrecGG = 2.53; 95%CIrecGG1.03-6.23; ppermGG = 0.045). The seven SNPs such as rs1173771 (G/A) AC026703.1, rs1799945 (C/G) HFE, rs805303 (G/A) BAG6, rs932764 (A/G) PLCE1, rs4387287 (C/A) OBFC1, rs7302981 (G/A) CERS5, rs167479 (T/G) RGL3, out of ten regarded loci, were related with AH within eight SNP-SNP interaction models (<0.001 ≤ pperm-interaction ≤ 0.047). Three polymorphisms such as rs8068318 (T/C) TBX2, rs633185 (C/G) ARHGAP42, and rs2681472 (A/G) ATP2B1 were not linked with AH. The pairwise rs805303 (G/A) BAG6-rs7302981 (G/A) CERS5 combination was a priority in determining the susceptibility to AH (included in six out of eight SNP-SNP interaction models [75%] and described 0.82% AH entropy). AH-associated variants are conjecturally functional for 101 genes involved in processes related to the immune system (major histocompatibility complex protein, processing/presentation of antigens, immune system process regulation, etc.). In conclusion, the rs1799945 polymorphism of the HFE gene and intergenic interactions of BAG6, CERS5, AC026703.1, HFE, PLCE1, OBFC1, RGL3 have been linked with AH risky in the Caucasian population of Central Russia.
Collapse
Affiliation(s)
- Tatiana Ivanova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Maria Churnosova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Maria Abramova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Irina Ponomarenko
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Evgeny Reshetnikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Inna Aristova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Inna Sorokina
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| |
Collapse
|
57
|
Zucker R, Kovalerchik M, Linial M. Gene-based association study reveals a distinct female genetic signal in primary hypertension. Hum Genet 2023:10.1007/s00439-023-02567-9. [PMID: 37133573 DOI: 10.1007/s00439-023-02567-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/25/2023] [Indexed: 05/04/2023]
Abstract
Hypertension is a polygenic disease that affects over 1.2 billion adults aged 30-79 worldwide. It is a major risk factor for renal, cerebrovascular, and cardiovascular diseases. The heritability of hypertension is estimated to be high; nevertheless, our understanding of its underlying mechanisms remains scarce and incomplete. This study covered the entries from European ancestry from the UK-Biobank (UKB), with 74,090 cases diagnosed with essential (primary) hypertension and 200,734 controls. We compared the findings from large-scale genome-wide association studies (GWAS) to the gene-based method of proteome-wide association studies (PWAS). We focused on 70 statistically significant associated genes, most of which failed to reach significance in variant-based GWAS. A total of 30% of the PWAS-associated genes were validated against independent cohorts, including the Finnish Biobank. Furthermore, gene-based analyses that were performed on both sexes revealed sex-dependent genetics with a stronger genetic component associated with females. Analysis of systolic and diastolic blood pressure measurements confirms a strong genetic effect associated with females. We demonstrated that gene-based approaches provide insight into the underlying biology of hypertension. Specifically, the expression profiles of the identified genes exposed the enrichment of endothelial cells from multiple organs. Furthermore, females' top-ranked significant genes are involved in cellular immunity. We conclude that studying hypertension and blood pressure via gene-based association methods improves interpretability and exposes sex-dependent genetic effects, which enhances clinical utility.
Collapse
Affiliation(s)
- Roei Zucker
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| | - Michael Kovalerchik
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| | - Michal Linial
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel.
| |
Collapse
|
58
|
Ivanova T, Churnosova M, Abramova M, Plotnikov D, Ponomarenko I, Reshetnikov E, Aristova I, Sorokina I, Churnosov M. Sex-Specific Features of the Correlation between GWAS-Noticeable Polymorphisms and Hypertension in Europeans of Russia. Int J Mol Sci 2023; 24:ijms24097799. [PMID: 37175507 PMCID: PMC10178435 DOI: 10.3390/ijms24097799] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/13/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
The aim of the study was directed at studying the sex-specific features of the correlation between genome-wide association studies (GWAS)-noticeable polymorphisms and hypertension (HTN). In two groups of European subjects of Russia (n = 1405 in total), such as men (n = 821 in total: n = 564 HTN, n = 257 control) and women (n = 584 in total: n = 375 HTN, n = 209 control), the distribution of ten specially selected polymorphisms (they have confirmed associations of GWAS level with blood pressure (BP) parameters and/or HTN in Europeans) has been considered. The list of studied loci was as follows: (PLCE1) rs932764 A > G, (AC026703.1) rs1173771 G > A, (CERS5) rs7302981 G > A, (HFE) rs1799945 C > G, (OBFC1) rs4387287 C > A, (BAG6) rs805303 G > A, (RGL3) rs167479 T > G, (ARHGAP42) rs633185 C > G, (TBX2) rs8068318 T > C, and (ATP2B1) rs2681472 A > G. The contribution of individual loci and their inter-locus interactions to the HTN susceptibility with bioinformatic interpretation of associative links was evaluated separately in men's and women's cohorts. The men-women differences in involvement in the disease of the BP/HTN-associated GWAS SNPs were detected. Among women, the HTN risk has been associated with HFE rs1799945 C > G (genotype GG was risky; ORGG = 11.15 ppermGG = 0.014) and inter-locus interactions of all 10 examined SNPs as part of 26 intergenic interactions models. In men, the polymorphism BAG6 rs805303 G > A (genotype AA was protective; ORAA = 0.30 ppermAA = 0.0008) and inter-SNPs interactions of eight loci in only seven models have been founded as HTN-correlated. HTN-linked loci and strongly linked SNPs were characterized by pronounced polyvector functionality in both men and women, but at the same time, signaling pathways of HTN-linked genes/SNPs in women and men were similar and were represented mainly by immune mechanisms. As a result, the present study has demonstrated a more pronounced contribution of BP/HTN-associated GWAS SNPs to the HTN susceptibility (due to weightier intergenic interactions) in European women than in men.
Collapse
Affiliation(s)
- Tatiana Ivanova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Maria Churnosova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Maria Abramova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Denis Plotnikov
- Genetic Epidemiology Lab, Kazan State Medical University, 420012 Kazan, Russia
| | - Irina Ponomarenko
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Evgeny Reshetnikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Inna Aristova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Inna Sorokina
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| |
Collapse
|
59
|
Fehrenbach DJ, Nguyen B, Alexander MR, Madhur MS. Modulating T Cell Phenotype and Function to Treat Hypertension. KIDNEY360 2023; 4:e534-e543. [PMID: 36951464 PMCID: PMC10278787 DOI: 10.34067/kid.0000000000000090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 01/25/2023] [Indexed: 03/24/2023]
Abstract
Hypertension is the leading modifiable risk factor of worldwide morbidity and mortality because of its effects on cardiovascular and renal end-organ damage. Unfortunately, BP control is not sufficient to fully reduce the risks of hypertension, underscoring the need for novel therapies that address end-organ damage in hypertension. Over the past several decades, the link between immune activation and hypertension has been well established, but there are still no therapies for hypertension that specifically target the immune system. In this review, we describe the critical role played by T cells in hypertension and hypertensive end-organ damage and outline potential therapeutic targets to modulate T-cell phenotype and function in hypertension without causing global immunosuppression.
Collapse
Affiliation(s)
- Daniel J. Fehrenbach
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee
| | - Bianca Nguyen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
| | - Matthew R. Alexander
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, Tennessee
| | - Meena S. Madhur
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, Tennessee
| |
Collapse
|
60
|
Sajal IH, Biswas S. Bivariate quantitative Bayesian LASSO for detecting association of rare haplotypes with two correlated continuous phenotypes. Front Genet 2023; 14:1104727. [PMID: 36968609 PMCID: PMC10033866 DOI: 10.3389/fgene.2023.1104727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
In genetic association studies, the multivariate analysis of correlated phenotypes offers statistical and biological advantages compared to analyzing one phenotype at a time. The joint analysis utilizes additional information contained in the correlation and avoids multiple testing. It also provides an opportunity to investigate and understand shared genetic mechanisms of multiple phenotypes. Bivariate logistic Bayesian LASSO (LBL) was proposed earlier to detect rare haplotypes associated with two binary phenotypes or one binary and one continuous phenotype jointly. There is currently no haplotype association test available that can handle multiple continuous phenotypes. In this study, by employing the framework of bivariate LBL, we propose bivariate quantitative Bayesian LASSO (QBL) to detect rare haplotypes associated with two continuous phenotypes. Bivariate QBL removes unassociated haplotypes by regularizing the regression coefficients and utilizing a latent variable to model correlation between two phenotypes. We carry out extensive simulations to investigate the performance of bivariate QBL and compare it with that of a standard (univariate) haplotype association test, Haplo.score (applied twice to two phenotypes individually). Bivariate QBL performs better than Haplo.score in all simulations with varying degrees of power gain. We analyze Genetic Analysis Workshop 19 exome sequencing data on systolic and diastolic blood pressures and detect several rare haplotypes associated with the two phenotypes.
Collapse
Affiliation(s)
| | - Swati Biswas
- Department of Mathematical Sciences, University of Texas at Dallas, Richardson, TX, United States
| |
Collapse
|
61
|
Hodonsky CJ, Turner AW, Khan MD, Barrientos NB, Methorst R, Ma L, Lopez NG, Mosquera JV, Auguste G, Farber E, Ma WF, Wong D, Onengut-Gumuscu S, Kavousi M, Peyser PA, van der Laan SW, Leeper NJ, Kovacic JC, Björkegren JLM, Miller CL. Integrative multi-ancestry genetic analysis of gene regulation in coronary arteries prioritizes disease risk loci. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.09.23285622. [PMID: 36824883 PMCID: PMC9949190 DOI: 10.1101/2023.02.09.23285622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Genome-wide association studies (GWAS) have identified hundreds of genetic risk loci for coronary artery disease (CAD). However, non-European populations are underrepresented in GWAS and the causal gene-regulatory mechanisms of these risk loci during atherosclerosis remain unclear. We incorporated local ancestry and haplotype information to identify quantitative trait loci (QTL) for gene expression and splicing in coronary arteries obtained from 138 ancestrally diverse Americans. Of 2,132 eQTL-associated genes (eGenes), 47% were previously unreported in coronary arteries and 19% exhibited cell-type-specific expression. Colocalization analysis with GWAS identified subgroups of eGenes unique to CAD and blood pressure. Fine-mapping highlighted additional eGenes of interest, including TBX20 and IL5 . Splicing (s)QTLs for 1,690 genes were also identified, among which TOR1AIP1 and ULK3 sQTLs demonstrated the importance of evaluating splicing events to accurately identify disease-relevant gene expression. Our work provides the first human coronary artery eQTL resource from a patient sample and exemplifies the necessity of diverse study populations and multi-omic approaches to characterize gene regulation in critical disease processes. Study Design Overview
Collapse
|
62
|
Huang S, Wang J, Liu N, Li P, Wu S, Qi L, Xia L. A cross-tissue transcriptome association study identifies key genes in essential hypertension. Front Genet 2023; 14:1114174. [PMID: 36845374 PMCID: PMC9950398 DOI: 10.3389/fgene.2023.1114174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/30/2023] [Indexed: 02/12/2023] Open
Abstract
Genome-wide association study (GWAS) have identified over 1,000 loci associated with blood pressure. However, these loci only explain 6% of heritability. Transcriptome-wide association studies (TWAS) combine GWAS summary data with expression quantitative trait loci (eQTL) to provide a better approach to finding genes associated with complex traits. GWAS summary data (N = 450,584) for essential hypertension originating from European samples were subjected to Post-GWAS analysis using FUMA software and then combined with eQTL data from Genotype-Tissues Expression Project (GTEx) v8 for TWAS analysis using UTMOST, FUSION software, and then validated the results with SMR. FUMA identified 346 significant genes associated with hypertension, FUSION identified 461, and UTMOST cross-tissue analysis identified 34, of which 5 were common. SMR validation identified 3 key genes: ENPEP, USP38, and KCNK3. In previous GWAS studies on blood pressure regulation, the association of ENPEP and KCNK3 with hypertension has been established, and the association between USP38 and blood pressure regulation still needs further validation.
Collapse
Affiliation(s)
- Sihui Huang
- College of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China,Key Laboratory of Traditional Chinese Medicine Regimen and Health Industry Development, State Administration of TCM, Chengdu, China,Leshan Vocational and Technical College, Leshan, China
| | - Jie Wang
- College of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China,Key Laboratory of Traditional Chinese Medicine Regimen and Health Industry Development, State Administration of TCM, Chengdu, China
| | - Nannan Liu
- College of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China,Key Laboratory of Traditional Chinese Medicine Regimen and Health Industry Development, State Administration of TCM, Chengdu, China
| | - Ping Li
- College of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China,Key Laboratory of Traditional Chinese Medicine Regimen and Health Industry Development, State Administration of TCM, Chengdu, China
| | - Sha Wu
- College of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China,Key Laboratory of Traditional Chinese Medicine Regimen and Health Industry Development, State Administration of TCM, Chengdu, China
| | - Luming Qi
- College of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China,Key Laboratory of Traditional Chinese Medicine Regimen and Health Industry Development, State Administration of TCM, Chengdu, China,*Correspondence: Luming Qi, ; Lina Xia,
| | - Lina Xia
- College of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China,Key Laboratory of Traditional Chinese Medicine Regimen and Health Industry Development, State Administration of TCM, Chengdu, China,*Correspondence: Luming Qi, ; Lina Xia,
| |
Collapse
|
63
|
Yokota K, Shibata H, Kurihara I, Itoh H, Sone M. CASZ1: a promising factor modulating aldosterone biosynthesis and mineralocorticoid receptor activity. Hypertens Res 2023; 46:417-420. [PMID: 36522424 DOI: 10.1038/s41440-022-01131-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022]
Abstract
Hypertension is the definitive risk factor for cardiovascular disease. Primary aldosteronism (PA), a typical form of secondary hypertension, is responsible for treatment-resistant hypertension and carries an even higher risk of causing cardiovascular complications than essential hypertension. Several genes involved in the pathogenesis of hypertension have been identified recently using genome-wide association studies (GWASs). Among these, castor zinc finger 1(CASZ1) is considered to be involved in the pathophysiology of hypertension via modulation of aldosterone action. In 2021, using a biochemical approach with liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis, we identified CASZ1b, an isoform of CASZ1, as a novel mineralocorticoid receptor (MR) coregulator. Our further analysis revealed that CASZ1b is coexpressed with MR in MR targets such as kidney tubule cells, and that a decrease in CASZ1 protein levels promotes aldosterone-dependent transcriptional activity of MR. Further, a recent study of GWAS on PA identified CASZ1 to be a PA-related gene and demonstrated that overexpression of CASZ1 suppresses aldosterone biosynthesis in adrenal cells. These results suggest CASZ1 plays a pivotal role in the pathophysiology of hypertension and PA via dual mechanisms: aldosterone biosynthesis and transcriptional activity of MR.
Collapse
Affiliation(s)
- Kenichi Yokota
- Division of Metabolism and Endocrinology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan.
| | - Hirotaka Shibata
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Oita, Japan
| | - Isao Kurihara
- Department of Medical Education, National Defense Medical College, Tokorozawa, Japan.,Division of Endocrinology, Metabolism and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Itoh
- Division of Endocrinology, Metabolism and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Masakatsu Sone
- Division of Metabolism and Endocrinology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
64
|
Udosen B, Soremekun O, Kamiza A, Machipisa T, Cheickna C, Omotuyi O, Soliman M, Wélé M, Nashiru O, Chikowore T, Fatumo S. Meta-Analysis and Multivariate GWAS Analyses in 80,950 Individuals of African Ancestry Identify Novel Variants Associated with Blood Pressure Traits. Int J Mol Sci 2023; 24:2164. [PMID: 36768488 PMCID: PMC9916484 DOI: 10.3390/ijms24032164] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/24/2022] [Accepted: 01/03/2023] [Indexed: 01/25/2023] Open
Abstract
High blood pressure (HBP) has been implicated as a major risk factor for cardiovascular diseases in several populations, including individuals of African ancestry. Despite the elevated burden of HBP-induced cardiovascular diseases in Africa and other populations of African descent, limited genetic studies have been carried out to explore the genetic mechanism driving this phenomenon. We performed genome-wide association univariate and multivariate analyses of both systolic (SBP) and diastolic blood pressure (DBP) traits in 80,950 individuals of African ancestry. We used summary statistics data from six independent cohorts, including the African Partnership for Chronic Disease Research (APCDR), the UK Biobank, and the Million Veteran Program (MVP). FUMA was used to annotate, prioritize, visualize, and interpret our findings to gain a better understanding of the molecular mechanism(s) underlying the genetics of BP traits. Finally, we undertook a Bayesian fine-mapping analysis to identify potential causal variants. Our meta-analysis identified 10 independent variants associated with SBP and 9 with DBP traits. Whilst our multivariate GWAS method identified 21 independent signals, 18 of these SNPs have been previously identified. SBP was linked to gene sets involved in biological processes such as synapse assembly and cell-cell adhesion via plasma membrane adhesion. Of the 19 independent SNPs identified in the BP meta-analysis, only 11 variants had posterior probability (PP) of > 50%, including one novel variant: rs562545 (MOBP, PP = 77%). To facilitate further research and fine-mapping of high-risk loci/variants in highly susceptible groups for cardiovascular disease and other related traits, large-scale genomic datasets are needed. Our findings highlight the importance of including ancestrally diverse populations in large GWASs and the need for diversity in genetic research.
Collapse
Affiliation(s)
- Brenda Udosen
- The African Computational Genomics (TACG) Research Group, MRC/UVRI and LSHTM, Entebbe 7545, Uganda; (B.U.); (O.S.); (A.K.)
- The African Center of Excellence in Bioinformatics of Bamako (ACE-B), University of Sciences, Techniques and Technologies of Bamako, Bamako 3206, Mali; (C.C.); (M.W.)
- H3Africa Bioinformatics Network (H3ABioNet) Node, Centre for Genomics Research and Innovation, NABDA/FMST, Abuja 901101, Nigeria;
| | - Opeyemi Soremekun
- The African Computational Genomics (TACG) Research Group, MRC/UVRI and LSHTM, Entebbe 7545, Uganda; (B.U.); (O.S.); (A.K.)
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, Westville Campus, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Abram Kamiza
- The African Computational Genomics (TACG) Research Group, MRC/UVRI and LSHTM, Entebbe 7545, Uganda; (B.U.); (O.S.); (A.K.)
- Malawi Epidemiology and Intervention Research Unit, Lilongwe P.O. Box 46, Malawi
| | - Tafadzwa Machipisa
- Hatter Institute for Cardiovascular Diseases Research in Africa (HICRA), Department of Medicine, University of Cape Town, Cape Town 7701, South Africa;
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, ON L8L 2X2, Canada
| | - Cisse Cheickna
- The African Center of Excellence in Bioinformatics of Bamako (ACE-B), University of Sciences, Techniques and Technologies of Bamako, Bamako 3206, Mali; (C.C.); (M.W.)
- Department of Biological Sciences, Faculty of Sciences and Techniques, University of Sciences, Techniques and Technologies of Bamako, Bamako 3206, Mali
| | - Olaposi Omotuyi
- Institute for Drug Research and Development, S.E. Bogoro Center, Afe Babalola University, Ado Ekiti 360101, Nigeria;
| | - Mahmoud Soliman
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, Westville Campus, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Mamadou Wélé
- The African Center of Excellence in Bioinformatics of Bamako (ACE-B), University of Sciences, Techniques and Technologies of Bamako, Bamako 3206, Mali; (C.C.); (M.W.)
- Department of Biological Sciences, Faculty of Sciences and Techniques, University of Sciences, Techniques and Technologies of Bamako, Bamako 3206, Mali
| | - Oyekanmi Nashiru
- H3Africa Bioinformatics Network (H3ABioNet) Node, Centre for Genomics Research and Innovation, NABDA/FMST, Abuja 901101, Nigeria;
| | - Tinashe Chikowore
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2050, South Africa;
- MRC/Wits Developmental Pathways for Health Research Unit, Department of Pediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Segun Fatumo
- The African Computational Genomics (TACG) Research Group, MRC/UVRI and LSHTM, Entebbe 7545, Uganda; (B.U.); (O.S.); (A.K.)
- H3Africa Bioinformatics Network (H3ABioNet) Node, Centre for Genomics Research and Innovation, NABDA/FMST, Abuja 901101, Nigeria;
- Segun Fatumo, Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| |
Collapse
|
65
|
mRNA Metabolism and Hypertension. Biomedicines 2023; 11:biomedicines11010118. [PMID: 36672629 PMCID: PMC9855994 DOI: 10.3390/biomedicines11010118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
Hypertension is the most frequent cardiovascular risk factor all over the world. It remains a leading contributor to the risk of cardiovascular events and death. In the year 2015, about 1.5 billion of adult people worldwide had hypertension (as defined by office systolic blood pressure ≥ 140 mmHg or office diastolic blood pressure ≥ 90 mmHg). Moreover, the number of hypertensive patients with age ranging from 30 to 79 years doubled in the last 30 years (from 317 million men and 331 million women in the year 1990 to 652 million men and 626 million women in 2019) despite stable age-standardized prevalence worldwide. Despite such impressive growth, the proportion of controlled hypertension is very low. A better understanding of the pathogenesis of hypertension may contribute to the development of innovative therapeutic strategies. In this context, alterations of the messenger RNA metabolism have been recently evaluated as contributors to the pathogenesis of hypertension, and pharmacological modulation of RNA metabolism is under investigation as potential and novel therapeutic armamentarium in hypertension.
Collapse
|
66
|
Cao Y, Li P, Zhang Y, Qiu M, Li J, Ma S, Yan Y, Li Y, Han Y. Association of systemic immune inflammatory index with all-cause and cause-specific mortality in hypertensive individuals: Results from NHANES. Front Immunol 2023; 14:1087345. [PMID: 36817427 PMCID: PMC9932782 DOI: 10.3389/fimmu.2023.1087345] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Background The relationship between the systemic immune inflammatory index (SII) and the prognosis of hypertensive patients is unclear. This study aims to explore the association of SII with all-cause and cause-specific mortality in patients with hypertension. Methods This study included 8524 adults with hypertension from the National Health and Nutritional Examination Surveys (NHANES) 2011-2018, and followed for survival through December 31, 2019. Cox proportional hazards models were used to investigate the associations between SII and mortality from all causes, cardiovascular disease (CVD), and cancer. Restricted cubic spline, piecewise linear regression, subgroup and sensitivity analyses were also used. Results During a median follow-up of 4.58 years, 872 all-cause deaths occurred. After adjusting for covariates, higher SII was significantly associated with an elevated risk of CVD mortality. There was a 102% increased risk of CVD mortality per one-unit increment in natural log-transformed SII (lnSII) (P < 0.001). Consistent results were also observed when SII was examined as categorical variable (quartiles). The associations of SII with all-cause and cancer mortality were detected as U-shaped with threshold values of 5.97 and 6.18 for lnSII respectively. Below thresholds, higher SII was significantly associated with lower all-cause mortality (HR=0.79, 95%CI=0.64-0.97) and cancer mortality (HR=0.73, 95%CI=0.53-1.00). Above thresholds, SII was significantly positive associated with all-cause mortality (HR=1.93, 95%CI=1.55-2.40) and cancer mortality (HR=1.93, 95%CI=1.22-3.05). The results were robust in subgroup and sensitivity analyses. Conclusion Higher SII (either as a continuous or categorical variable) were significantly associated with a higher risk of CVD mortality. The U-shaped associations were observed between SII and all-cause and cancer mortality.
Collapse
Affiliation(s)
- Yang Cao
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China.,The Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shanxi, China
| | - Pengxiao Li
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China.,The Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shanxi, China
| | - Yan Zhang
- The Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shanxi, China
| | - Miaohan Qiu
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Jing Li
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Sicong Ma
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Yudong Yan
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China.,The Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shanxi, China
| | - Yi Li
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Yaling Han
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| |
Collapse
|
67
|
Chew NWS, Loong SSE, Foo R. Progress in molecular biology and translational science: Epigenetics in cardiovascular health and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 197:105-134. [PMID: 37019589 DOI: 10.1016/bs.pmbts.2023.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Conrad Waddington's epigenetics landscape has provided a metaphorical framework for how cells progress from undifferentiated states to one of several discrete, distinct, differentiated cell fates. The understanding of epigenetics has evolved over time, with DNA methylation being the most studied epigenetic modification, followed by histone modifications and non-coding RNA. Cardiovascular diseases (CVD) are leading contributors to death worldwide, with the prevalence of CVDs increasing across the last couple of decades. Significant amount of resources being poured into researching key mechanisms and underpinnings of the various CVDs. These molecular studies looked at the genetics, epigenetics as well as the transcriptomics of various cardiovascular conditions, aiming to provide mechanistic insights. It has paved the way for therapeutics to be developed and in recent years, epi-drugs for the treatment of CVDs. This chapter aims to cover the various roles of epigenetics in the context of cardiovascular health and disease. The following will be examined in detail: the developments in basic experimental techniques used to study epigenetics, the role of epigenetics in various CVDs (hypertension, atrial fibrillation, atherosclerosis, and heart failure), and current advances in epi-therapeutics, providing a holistic view of the current concerted efforts in advancing the field of epigenetics in CVDs.
Collapse
Affiliation(s)
- Nicholas W S Chew
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore, Singapore.
| | - Shaun S E Loong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Roger Foo
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
68
|
Yao J, Mao X, Sun Q, Wu B, Yu W, Huang Y, Luo S, Zeng J, Lin J. TBX5 Variants are Associated with Susceptibility to and the Incidence of Liver Cirrhosis and Hepatocellular Carcinoma in the Chinese Population: A Multicenter and Follow-Up Study. Infect Drug Resist 2023; 16:2653-2665. [PMID: 37159827 PMCID: PMC10163879 DOI: 10.2147/idr.s410151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/19/2023] [Indexed: 05/11/2023] Open
Abstract
Purpose Liver cirrhosis (LC) and hepatocellular carcinoma (HCC) are progressions affected by genetic predispositions, and persistent hepatitis B virus infection also demonstrates genetic susceptibility. All HBV-related outcomes have been compared in parallel to identify risk polymorphism in HBV progression. Methods The multiple-stage association study filtered and validated the risk SNPs for HBV progression and explored their association with persistent infection, with a total of 8906 subjects in China from three sites. Cox proportional hazards models and Kaplan-Meier Log rank tests were used to determine the time to the progressive event in relation to the risk SNPs. Results Rs3825214 in TBX5 replicated a specific association with LC and HCC in 4 progression cohorts and was not related to persistent infection, naivety to HBV infection and natural clearance in 3 persistent cohorts. In combined samples, rs3825214 was associated with an increased risk of LC (P<0.001; OR = 1.98) and HCC (P<0.001; OR = 1.68). The results of bioinformatics analysis indicated that rs3825214 genotypes change RNA structure and intron excision ratio. In the follow-up of 571 hospital-based persistent HBV infection patients, ninety-three (16.29%) developed LC, and seventy-four (12.96%) progressed to HCC at a median follow-up of 5.1 years. Rs3825214 was associated with HCC and LC events in Cox proportional hazards models (P<0.001). Conclusion We identified and confirmed that genetic variants in TBX5 are significantly associated with susceptibility to and the incidence of LC and HCC.
Collapse
Affiliation(s)
- JinJian Yao
- Department of Emergency, Hainan General Hospital, Haikou, Hainan, People’s Republic of China
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Correspondence: JinJian Yao; Jusheng Lin, Email ;
| | - Xiaochun Mao
- Department of Ophthalmology, Xiangyang Central Hospital Affiliated to Hubei University of Arts and Science, Xiangyang, Hubei, People’s Republic of China
| | - Qigang Sun
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Haikou, Hainan, People’s Republic of China
| | - Biao Wu
- Infectious Disease, Hainan General Hospital, Haikou, Hainan, People’s Republic of China
| | - Weiling Yu
- Oncology Department, Haikou City People’s Hospital, Haikou, Hainan, People’s Republic of China
| | - Yanjing Huang
- Oncology Department of Hainan General Hospital, Haikou, Hainan, People’s Republic of China
| | - Shuai Luo
- Department of Emergency, Hainan General Hospital, Haikou, Hainan, People’s Republic of China
| | - Jia Zeng
- Department of Emergency, Hainan General Hospital, Haikou, Hainan, People’s Republic of China
| | - Jusheng Lin
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
69
|
Ou H, Liu D, Zhao G, Gong C, Li Y, Zhao Q. Association between AT1 receptor gene polymorphism and left ventricular hypertrophy and arterial stiffness in essential hypertension patients: a prospective cohort study. BMC Cardiovasc Disord 2022; 22:571. [PMID: 36577936 PMCID: PMC9795750 DOI: 10.1186/s12872-022-03024-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AT1 receptor gene (AGTR1) is related to essential hypertension (EH), and left ventricular hypertrophy (LVH) and arterial stiffness are common complications of EH. This study aimed to explore the association between AGTR1 genotype and LVH and arterial stiffness in EH patients. METHODS A total of 179 EH patients were recruited in this study. Oral exfoliated cells were collected from each patient, and the genetic polymorphism of AGTR1(rs4524238) was assessed using a gene sequencing platform. The outcomes were LVH and arterial stiffness. RESULTS Among 179 patients, 114 were with AGTR1 genotype of GG (57 males, aged 59.54 ± 13.49 years) and 65 were with AGTR1 genotype of GA or AA (36 males, aged 61.28 ± 12.79 years). Patients with AGTR1 genotype of GG were more likely to have LVH (47 [41.23%] vs. 14 [21.54%], P = 0.006) and arterial stiffness (30 [26.32%] vs. 8 [12.31%], P = 0.036). The AGTR1 polymorphism frequency was in accordance with Hardy-Weinberg equilibrium (P = 0.291). The multivariate logistic regression showed that AGTR1 genotype of GA or AA was independently associated with lower risk of LVH (OR = 0.344, 95%CI 160~0.696, P = 0.003) and arterial stiffness (OR = 0.371, 95%CI 0.155~0.885, P = 0.025) after adjusting for gender, age, and diabetes. CONCLUSION EH patients with the AGTR1 genotype of GA or AA were at lower risk for LVH and arterial stiffness than those with the GG genotype.
Collapse
Affiliation(s)
- Hangjun Ou
- grid.452244.1Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou China
| | - Danan Liu
- grid.452244.1Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou China
| | - Guangjian Zhao
- grid.452244.1Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou China
| | - Caiwei Gong
- grid.452244.1Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou China
| | - Yunyun Li
- grid.452244.1Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou China
| | - Quanwei Zhao
- grid.452244.1Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou China
| |
Collapse
|
70
|
Abramova M, Churnosova M, Efremova O, Aristova I, Reshetnikov E, Polonikov A, Churnosov M, Ponomarenko I. Effects of Pre-Pregnancy Overweight/Obesity on the Pattern of Association of Hypertension Susceptibility Genes with Preeclampsia. Life (Basel) 2022; 12:2018. [PMID: 36556383 PMCID: PMC9784908 DOI: 10.3390/life12122018] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/21/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to explore the effects of pre-pregnancy overweight/obesity on the pattern of association of hypertension susceptibility genes with preeclampsia (PE). Ten single-nucleotide polymorphisms (SNPs) of the 10 genome-wide association studies (GWAS)-significant hypertension/blood pressure (BP) candidate genes were genotyped in 950 pregnant women divided into two cohorts according to their pre-pregnancy body mass index (preBMI): preBMI ≥ 25 (162 with PE and 159 control) and preBMI < 25 (290 with PE and 339 control). The PLINK software package was utilized to study the association (analyzed four genetic models using logistic regression). The functionality of PE-correlated loci was analyzed by performing an in silico database analysis. Two SNP hypertension/BP genes, rs805303 BAG6 (OR: 0.36−0.66) and rs167479 RGL3 (OR: 1.86), in subjects with preBMI ≥ 25 were associated with PE. No association between the studied SNPs and PE in the preBMI < 25 group was determined. Further analysis showed that two PE-associated SNPs are functional (have weighty eQTL, sQTL, regulatory, and missense values) and could be potentially implicated in PE development. In conclusion, this study was the first to discover the modifying influence of overweight/obesity on the pattern of association of GWAS-significant hypertension/BP susceptibility genes with PE: these genes are linked with PE in preBMI ≥ 25 pregnant women and are not PE-involved in the preBMI < 25 group.
Collapse
Affiliation(s)
- Maria Abramova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Maria Churnosova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Olesya Efremova
- Department of Medical Genetics, Kharkiv National Medical University, 61022 Kharkov, Ukraine
- Grishchenko Clinic of Reproductive Medicine, 61052 Kharkov, Ukraine
| | - Inna Aristova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Evgeny Reshetnikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Alexey Polonikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
- Department of Biology, Medical Genetics and Ecology and Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Irina Ponomarenko
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| |
Collapse
|
71
|
Identifying susceptibility genes for essential hypertension by transcriptome-wide association study. Biochem Biophys Rep 2022; 32:101387. [DOI: 10.1016/j.bbrep.2022.101387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/29/2022] [Accepted: 11/12/2022] [Indexed: 11/22/2022] Open
|
72
|
Zumaraga MPP, Rodriguez MP, Aman AYC, Deguit CDT, Biwang JH, Melegrito JB, Duante CA, Madrid ML, Concepcion MAR, Nevado JB. Nutritional and genetic determinants of essential hypertension among adult respondents of the 2013 national nutrition survey, Philippines: a preliminary observational study. J Nutr Biochem 2022; 110:109152. [PMID: 36245136 DOI: 10.1016/j.jnutbio.2022.109152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 08/11/2022] [Accepted: 08/19/2022] [Indexed: 01/13/2023]
Abstract
Preventive strategies for hypertension and its sequelae require an understanding of their predisposing conditions and recognition of at-risk individuals. Several factors, both genetic and nongenetic, are influential, and likely vary in their effects across ethnicities. This study aimed to identify dietary, lifestyle-related differences and genetic variants associated with hypertension in Filipinos. The study included 147 adult Filipino respondents of the 2013 Philippine National Nutrition Survey living in the National Capital Region. Data on the socio-demographic profile and selected lifestyle factors were obtained via face-to-face interviews. Blood pressure, anthropometric and biochemical indicators of health were determined using standard procedures. Hypertension incidence was determined following American College of Cardiology/American Heart Association guideline. Genotyping utilized the customized Illumina Golden Gate genotyping array, with subsequent allele and genotypic association analytics. Genetic variant effects were adjusted to clinical parameters via logistic regression. Between those with and without hypertension, there was relatively higher intake of dietary protein, fat but not carbohydrates in the latter (P<.05). Of note, other established risk factors for hypertension, such as high lipid levels and fasting blood sugar, were consistently frequently seen among hypertensive respondents. Of the gene markers, 3 SNPs (rs10492602 of APOC [3' UTR], rs12721054 of CYP2C19 [exon] and rs4244285 [intergenic between PCDH17-DIAPH3 locus]) remained significant after multivariable logistic regression. The study highlights that both nutrition and genetic information may contribute to hypertension among Filipinos. This could guide public health initiatives to identify Filipinos susceptible to hypertension and recommend control strategies in lowering its morbidity rate.
Collapse
Affiliation(s)
- Mark Pretzel P Zumaraga
- Department of Science and Technology-Food and Nutrition Research Institute, Bicutan, Taguig City, Philippines.
| | - Marietta P Rodriguez
- Department of Science and Technology-Food and Nutrition Research Institute, Bicutan, Taguig City, Philippines
| | - Aimee Yvonne Criselle Aman
- Microarray Core Laboratory, Institute of Human Genetics, National Institutes of Health, University of the Philippines Manila, Philippines; Department of Science and Technology-Philippine Council for Health Research and Development, Bicutan, Taguig City, Philippines
| | - Christian Deo T Deguit
- Microarray Core Laboratory, Institute of Human Genetics, National Institutes of Health, University of the Philippines Manila, Philippines
| | - Jessica H Biwang
- Microarray Core Laboratory, Institute of Human Genetics, National Institutes of Health, University of the Philippines Manila, Philippines
| | - Jodelyn B Melegrito
- Microarray Core Laboratory, Institute of Human Genetics, National Institutes of Health, University of the Philippines Manila, Philippines
| | - Charmaine A Duante
- Department of Science and Technology-Food and Nutrition Research Institute, Bicutan, Taguig City, Philippines
| | - Marilou L Madrid
- Department of Science and Technology-Food and Nutrition Research Institute, Bicutan, Taguig City, Philippines
| | - Mae Anne R Concepcion
- Department of Science and Technology-Food and Nutrition Research Institute, Bicutan, Taguig City, Philippines
| | - Jose B Nevado
- Microarray Core Laboratory, Institute of Human Genetics, National Institutes of Health, University of the Philippines Manila, Philippines
| |
Collapse
|
73
|
Morris GE, Denniff MJ, Karamanavi E, Andrews SA, Kostogrys RB, Bountziouka V, Ghaderi‐Najafabadi M, Shamkhi N, McConnell G, Kaiser MA, Carleton L, Schofield C, Kessler T, Rainbow RD, Samani NJ, Webb TR. The integrin ligand SVEP1 regulates GPCR-mediated vasoconstriction via integrins α9β1 and α4β1. Br J Pharmacol 2022; 179:4958-4973. [PMID: 35802072 PMCID: PMC9805129 DOI: 10.1111/bph.15921] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 06/10/2022] [Accepted: 06/27/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE Vascular tone is regulated by the relative contractile state of vascular smooth muscle cells (VSMCs). Several integrins directly modulate VSMC contraction by regulating calcium influx through L-type voltage-gated Ca2+ channels (VGCCs). Genetic variants in ITGA9, which encodes the α9 subunit of integrin α9β1, and SVEP1, a ligand for integrin α9β1, associate with elevated blood pressure; however, neither SVEP1 nor integrin α9β1 has reported roles in vasoregulation. We determined whether SVEP1 and integrin α9β1 can regulate VSMC contraction. EXPERIMENTAL APPROACH SVEP1 and integrin binding were confirmed by immunoprecipitation and cell binding assays. Human induced pluripotent stem cell-derived VSMCs were used in in vitro [Ca2+ ]i studies, and aortas from a Svep1+/- knockout mouse model were used in wire myography to measure vessel contraction. KEY RESULTS We confirmed the ligation of SVEP1 to integrin α9β1 and additionally found SVEP1 to directly bind to integrin α4β1. Inhibition of SVEP1, integrin α4β1 or α9β1 significantly enhanced [Ca2+ ]i levels in isolated VSMCs to Gαq/11 -vasoconstrictors. This response was confirmed in whole vessels where a greater contraction to U46619 was seen in vessels from Svep1+/- mice compared to littermate controls or when integrin α4β1 or α9β1 was inhibited. Inhibition studies suggested that this effect was mediated via VGCCs, PKC and Rho A/Rho kinase dependent mechanisms. CONCLUSIONS AND IMPLICATIONS Our studies reveal a novel role for SVEP1 and the integrins α4β1 and α9β1 in reducing VSMC contractility. This could provide an explanation for the genetic associations with blood pressure risk at the SVEP1 and ITGA9 loci.
Collapse
Affiliation(s)
- Gavin E. Morris
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Matthew J. Denniff
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Elisavet Karamanavi
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Sarah A. Andrews
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Renata B. Kostogrys
- Department of Human Nutrition, Faculty of Food TechnologyUniversity of Agriculture in KrakowKrakowPoland
| | - Vasiliki Bountziouka
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Maryam Ghaderi‐Najafabadi
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Noor Shamkhi
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - George McConnell
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Michael A. Kaiser
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | | | | | - Thorsten Kessler
- Department of Cardiology, German Heart Centre MunichTechnical University of MunichMunichGermany,German Centre of Cardiovascular Research (DZHK e. V.), Partner Site Munich Heart AllianceMunichGermany
| | - Richard D. Rainbow
- Department of Cardiovascular and Metabolic Medicine & Liverpool Centre for Cardiovascular ScienceUniversity of LiverpoolLiverpoolUK
| | - Nilesh J. Samani
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Thomas R. Webb
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| |
Collapse
|
74
|
Alexander MR, Hank S, Dale BL, Himmel L, Zhong X, Smart CD, Fehrenbach DJ, Chen Y, Prabakaran N, Tirado B, Centrella M, Ao M, Du L, Shyr Y, Levy D, Madhur MS. A Single Nucleotide Polymorphism in SH2B3/LNK Promotes Hypertension Development and Renal Damage. Circ Res 2022; 131:731-747. [PMID: 36169218 PMCID: PMC9588739 DOI: 10.1161/circresaha.121.320625] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 09/15/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND SH2B3 (SH2B adaptor protein 3) is an adaptor protein that negatively regulates cytokine signaling and cell proliferation. A common missense single nucleotide polymorphism in SH2B3 (rs3184504) results in substitution of tryptophan (Trp) for arginine (Arg) at amino acid 262 and is a top association signal for hypertension in human genome-wide association studies. Whether this variant is causal for hypertension, and if so, the mechanism by which it impacts pathogenesis is unknown. METHODS We used CRISPR-Cas9 technology to create mice homozygous for the major (Arg/Arg) and minor (Trp/Trp) alleles of this SH2B3 polymorphism. Mice underwent angiotensin II (Ang II) infusion to evaluate differences in blood pressure (BP) elevation and end-organ damage including albuminuria and renal fibrosis. Cytokine production and Stat4 phosphorylation was also assessed in Arg/Arg and Trp/Trp T cells. RESULTS Trp/Trp mice exhibit 10 mmHg higher systolic BP during chronic Ang II infusion compared to Arg/Arg controls. Renal injury and perivascular fibrosis are exacerbated in Trp/Trp mice compared to Arg/Arg controls following Ang II infusion. Renal and ex vivo stimulated splenic CD8+ T cells from Ang II-infused Trp/Trp mice produce significantly more interferon gamma (IFNg) compared to Arg/Arg controls. Interleukin-12 (IL-12)-induced IFNg production is greater in Trp/Trp compared to Arg/Arg CD8+ T cells. In addition, IL-12 enhances Stat4 phosphorylation to a greater degree in Trp/Trp compared to Arg/Arg CD8+ T cells, suggesting that Trp-encoding SH2B3 exhibits less negative regulation of IL-12 signaling to promote IFNg production. Finally, we demonstrated that a multi-SNP model genetically predicting increased SH2B3 expression in lymphocytes is inversely associated with hypertension and hypertensive chronic kidney disease in humans.. CONCLUSIONS Taken together, these results suggest that the Trp encoding allele of rs3184504 is causal for BP elevation and renal dysfunction, in part through loss of SH2B3-mediated repression of T cell IL-12 signaling leading to enhanced IFNg production.
Collapse
Affiliation(s)
- Matthew R. Alexander
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Medicine, Division of Cardiovascular Medicine, VUMC, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Samuel Hank
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Bethany L. Dale
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Lauren Himmel
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA
| | - Xue Zhong
- Department of Medicine, Division of Genetic Medicine, VUMC, Nashville, TN, USA
| | - Charles D. Smart
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Daniel J. Fehrenbach
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Yuhan Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Cardiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China
| | | | | | - Megan Centrella
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Mingfang Ao
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Liping Du
- Department of Biostatistics, VUMC, Nashville, TN
| | - Yu Shyr
- Department of Biostatistics, VUMC, Nashville, TN
| | - Daniel Levy
- Framingham Heart Study, Framingham, MA and Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Meena S. Madhur
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Medicine, Division of Cardiovascular Medicine, VUMC, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
75
|
Portilla-Fernandez E, Klarin D, Hwang SJ, Biggs ML, Bis JC, Weiss S, Rospleszcz S, Natarajan P, Hoffmann U, Rogers IS, Truong QA, Völker U, Dörr M, Bülow R, Criqui MH, Allison M, Ganesh SK, Yao J, Waldenberger M, Bamberg F, Rice KM, Essers J, Kapteijn DMC, van der Laan SW, de Knegt RJ, Ghanbari M, Felix JF, Ikram MA, Kavousi M, Uitterlinden AG, Roks AJM, Danser AHJ, Tsao PS, Damrauer SM, Guo X, Rotter JI, Psaty BM, Kathiresan S, Völzke H, Peters A, Johnson C, Strauch K, Meitinger T, O’Donnell CJ, Dehghan A. Genetic and clinical determinants of abdominal aortic diameter: genome-wide association studies, exome array data and Mendelian randomization study. Hum Mol Genet 2022; 31:3566-3579. [PMID: 35234888 PMCID: PMC9558840 DOI: 10.1093/hmg/ddac051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
Progressive dilation of the infrarenal aortic diameter is a consequence of the ageing process and is considered the main determinant of abdominal aortic aneurysm (AAA). We aimed to investigate the genetic and clinical determinants of abdominal aortic diameter (AAD). We conducted a meta-analysis of genome-wide association studies in 10 cohorts (n = 13 542) imputed to the 1000 Genome Project reference panel including 12 815 subjects in the discovery phase and 727 subjects [Partners Biobank cohort 1 (PBIO)] as replication. Maximum anterior-posterior diameter of the infrarenal aorta was used as AAD. We also included exome array data (n = 14 480) from seven epidemiologic studies. Single-variant and gene-based associations were done using SeqMeta package. A Mendelian randomization analysis was applied to investigate the causal effect of a number of clinical risk factors on AAD. In genome-wide association study (GWAS) on AAD, rs74448815 in the intronic region of LDLRAD4 reached genome-wide significance (beta = -0.02, SE = 0.004, P-value = 2.10 × 10-8). The association replicated in the PBIO1 cohort (P-value = 8.19 × 10-4). In exome-array single-variant analysis (P-value threshold = 9 × 10-7), the lowest P-value was found for rs239259 located in SLC22A20 (beta = 0.007, P-value = 1.2 × 10-5). In the gene-based analysis (P-value threshold = 1.85 × 10-6), PCSK5 showed an association with AAD (P-value = 8.03 × 10-7). Furthermore, in Mendelian randomization analyses, we found evidence for genetic association of pulse pressure (beta = -0.003, P-value = 0.02), triglycerides (beta = -0.16, P-value = 0.008) and height (beta = 0.03, P-value < 0.0001), known risk factors for AAA, consistent with a causal association with AAD. Our findings point to new biology as well as highlighting gene regions in mechanisms that have previously been implicated in the genetics of other vascular diseases.
Collapse
Affiliation(s)
- Eliana Portilla-Fernandez
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Derek Klarin
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Shih-Jen Hwang
- Population Sciences Branch, Division of Intramural Research, NHLBI/NIH, Bethesda MD, USA
- National Heart Lung and Blood Institute's Intramural Research Program's Framingham Heart Study, Framingham, MA, USA
| | - Mary L Biggs
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Stefan Weiss
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Susanne Rospleszcz
- Institute of Epidemiology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Pradeep Natarajan
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Udo Hoffmann
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Ian S Rogers
- Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| | - Quynh A Truong
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Marcus Dörr
- Department of Internal Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Robin Bülow
- Department of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Michael H Criqui
- Department of Family Medicine, University of California, San Diego, CA, USA
| | - Matthew Allison
- Department of Family Medicine, University of California, San Diego, CA, USA
| | - Santhi K Ganesh
- Department of Internal Medicine and Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Melanie Waldenberger
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Fabian Bamberg
- Department of Diagnostic and Interventional Radiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kenneth M Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Jeroen Essers
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiation Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Daniek M C Kapteijn
- Laboratory of Experimental Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Sander W van der Laan
- Laboratory of Clinical Chemistry & Hematology, Division Laboratories, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Rob J de Knegt
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Janine F Felix
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Andre G Uitterlinden
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Anton J M Roks
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A H Jan Danser
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Philip S Tsao
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- School of Medicine, Stanford University, Stanford, CA, USA
| | - Scott M Damrauer
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Services, University of Washington, Seattle, WA, USA
| | - Sekar Kathiresan
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Henry Völzke
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Epidemiology, Institute for Medical Information Processing, Biometry, and Epidemiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Craig Johnson
- Collaborative Health Studies Coordinating Center, Department of Biostatistics in the School of Public Health, University of Washington, Seattle, WA, USA
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Genetic Epidemiology, Institute for Medical Information Processing, Biometry, and Epidemiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Thomas Meitinger
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Institute of Human Genetics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, München, Germany
| | - Christopher J O’Donnell
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | | |
Collapse
|
76
|
Jaago M, Rähni A, Pupina N, Pihlak A, Sadam H, Tuvikene J, Avarlaid A, Planken A, Planken M, Haring L, Vasar E, Baćević M, Lambert F, Kalso E, Pussinen P, Tienari PJ, Vaheri A, Lindholm D, Timmusk T, Ghaemmaghami AM, Palm K. Differential patterns of cross-reactive antibody response against SARS-CoV-2 spike protein detected for chronically ill and healthy COVID-19 naïve individuals. Sci Rep 2022; 12:16817. [PMID: 36207326 PMCID: PMC9540097 DOI: 10.1038/s41598-022-20849-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
Immunity to previously encountered viruses can alter response to unrelated pathogens. We reasoned that similar mechanism may also involve SARS-CoV-2 and thereby affect the specificity and the quality of the immune response against the virus. Here, we employed high-throughput next generation phage display method to explore the link between antibody immune response to previously encountered antigens and spike (S) glycoprotein. By profiling the antibody response in COVID-19 naïve individuals with a diverse clinical history (including cardiovascular, neurological, or oncological diseases), we identified 15 highly antigenic epitopes on spike protein that showed cross-reactivity with antigens of seasonal, persistent, latent or chronic infections from common human viruses. We observed varying degrees of cross-reactivity of different viral antigens with S in an epitope-specific manner. The data show that pre-existing SARS-CoV-2 S1 and S2 cross-reactive serum antibody is readily detectable in pre-pandemic cohort. In the severe COVID-19 cases, we found differential antibody response to the 15 defined antigenic and cross-reactive epitopes on spike. We also noted that despite the high mutation rates of Omicron (B.1.1.529) variants of SARS-CoV-2, some of the epitopes overlapped with the described mutations. Finally, we propose that the resolved epitopes on spike if targeted by re-called antibody response from SARS-CoV-2 infections or vaccinations can function in chronically ill COVID-19 naïve/unvaccinated individuals as immunogenic targets to boost antibodies augmenting the chronic conditions. Understanding the relationships between prior antigen exposure at the antibody epitope level and the immune response to subsequent infections with viruses from a different strain is paramount to guiding strategies to exit the COVID-19 pandemic.
Collapse
Affiliation(s)
- Mariliis Jaago
- Protobios LLC, Tallinn, Estonia
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Annika Rähni
- Protobios LLC, Tallinn, Estonia
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | | | | | - Helle Sadam
- Protobios LLC, Tallinn, Estonia
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Jürgen Tuvikene
- Protobios LLC, Tallinn, Estonia
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
- DXLabs LLC, Tallinn, Estonia
| | - Annela Avarlaid
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Anu Planken
- North Estonia Medical Centre Foundation, Tallinn, Estonia
| | - Margus Planken
- North Estonia Medical Centre Foundation, Tallinn, Estonia
| | - Liina Haring
- Institute of Clinical Medicine, Psychiatry Clinic of Tartu University Hospital, University of Tartu, Tartu, Estonia
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
- Center of Excellence for Genomics and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Miljana Baćević
- Dental Biomaterial Research Unit (d-BRU), Faculty of Medicine, University of Liege, Liege, Belgium
| | - France Lambert
- Department of Periodontology and Oral Surgery, Faculty of Medicine, University of Liege, Liege, Belgium
| | - Eija Kalso
- Department of Anaesthesiology, Intensive Care and Pain Medicine, Helsinki University Hospital, Helsinki, Finland
- SleepWell Research Programme, Department of Pharmacology, University of Helsinki, Helsinki, Finland
| | - Pirkko Pussinen
- Oral and Maxillofacial Diseases, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Pentti J Tienari
- Translational Immunology Research Program, Department of Neurology, Neurocenter, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Antti Vaheri
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Dan Lindholm
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Tõnis Timmusk
- Protobios LLC, Tallinn, Estonia
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Amir M Ghaemmaghami
- Immunology and Immuno-Bioengineering Group, School of Life Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | | |
Collapse
|
77
|
Zu T, Lian H, Green B, Yu Y. Ultra-high Dimensional Quantile Regression for Longitudinal Data: an Application to Blood Pressure Analysis. J Am Stat Assoc 2022. [DOI: 10.1080/01621459.2022.2128806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Tianhai Zu
- Department of Operations, Business Analytics, & Information Systems, University of Cincinnati, Cincinnati, Ohio, USA
| | - Heng Lian
- Department of Mathematics, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong Hong Kong, China
| | - Brittany Green
- Department of Information Systems, Analytics, and Operations, University of Louisville, Louisville, Kentucky, USA
| | - Yan Yu
- Department of Operations, Business Analytics, & Information Systems, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
78
|
Huang L, Tang J, Lin L, Wang R, Chen F, Wei Y, Si Y, Fu W. Association of genetic variants in ULK4 with the age of first onset of type B aortic dissection. Front Genet 2022; 13:956866. [PMID: 36118886 PMCID: PMC9478570 DOI: 10.3389/fgene.2022.956866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/28/2022] [Indexed: 11/30/2022] Open
Abstract
Background: The association between autophagy, structural alterations of the aortic wall, and endothelial dysfunction in humans has yet to be fully elucidated. The family of ULK (UNC51-like) enzymes plays critical roles in autophagy and development. This study aimed to evaluate the association between ULK gene family members and patient age of first type B aortic dissection (TBAD) onset. Methods: The genotype data in a TBAD cohort from China and the related summary-level datasets were analyzed. We applied the sequence kernel association test (SKAT) to test the association between single-nucleotide polymorphisms (SNPs) and age of first onset of TBAD controlling for gender, hypertension, and renal function. Next, we performed a 2-sample Mendelian randomization (MR) to explore the potential causal relationship between ULK4 and early onset of TBAD at the level of gene expression coupled with DNA methylation with genetic variants as instrumental variables. Results: A total of 159 TBAD patients with 1,180,097 SNPs were included. Concerning the association between the ULK gene family and the age of first onset of the TBAD, only ULK4 was found to be significant according to SKAT analysis (q-FDR = 0.0088). From 2-sample MR, the high level of ULK4 gene expression was related to a later age of first onset of TBAD (β = 4.58, p = 0.0214). Conclusion: This is the first study of the ULK gene family in TBAD, regarding the association with the first onset age. We demonstrated that the ULK4 gene is associated with the time of onset of TBAD based on both the SKAT and 2-sample MR analyses.
Collapse
Affiliation(s)
- Lihong Huang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Biostatistics, Zhongshan Hospital, Fudan University, Shanghai, China
- Clinical Research Unit, Institute of Clinical Science, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Jiaqi Tang
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lijuan Lin
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ruihan Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Feng Chen
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yongyue Wei
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yi Si
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- *Correspondence: Yi Si, ; Weiguo Fu,
| | - Weiguo Fu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- *Correspondence: Yi Si, ; Weiguo Fu,
| |
Collapse
|
79
|
Tomaszewski M, Morris AP, Howson JMM, Franceschini N, Eales JM, Xu X, Dikalov S, Guzik TJ, Humphreys BD, Harrap S, Charchar FJ. Kidney omics in hypertension: from statistical associations to biological mechanisms and clinical applications. Kidney Int 2022; 102:492-505. [PMID: 35690124 PMCID: PMC9886011 DOI: 10.1016/j.kint.2022.04.045] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/10/2022] [Accepted: 04/22/2022] [Indexed: 02/06/2023]
Abstract
Hypertension is a major cardiovascular disease risk factor and contributor to premature death globally. Family-based investigations confirmed a significant heritable component of blood pressure (BP), whereas genome-wide association studies revealed >1000 common and rare genetic variants associated with BP and/or hypertension. The kidney is not only an organ of key relevance to BP regulation and the development of hypertension, but it also acts as the tissue mediator of genetic predisposition to hypertension. The identity of kidney genes, pathways, and related mechanisms underlying the genetic associations with BP has started to emerge through integration of genomics with kidney transcriptomics, epigenomics, and other omics as well as through applications of causal inference, such as Mendelian randomization. Single-cell methods further enabled mapping of BP-associated kidney genes to cell types, and in conjunction with other omics, started to illuminate the biological mechanisms underpinning associations of BP-associated genetic variants and kidney genes. Polygenic risk scores derived from genome-wide association studies and refined on kidney omics hold the promise of enhanced diagnostic prediction, whereas kidney omics-informed drug discovery is likely to contribute new therapeutic opportunities for hypertension and hypertension-mediated kidney damage.
Collapse
Affiliation(s)
- Maciej Tomaszewski
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK; Manchester Heart Centre and Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester, UK.
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, University of Manchester, Manchester, UK
| | - Joanna M M Howson
- Department of Genetics, Novo Nordisk Research Centre Oxford, Novo Nordisk Ltd, Oxford, UK
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
| | - James M Eales
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Xiaoguang Xu
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Sergey Dikalov
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK; Department of Internal and Agricultural Medicine, Jagiellonian University College of Medicine, Kraków, Poland
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Stephen Harrap
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Fadi J Charchar
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia; Health Innovation and Transformation Centre, School of Science, Psychology and Sport, Federation University Australia, Ballarat, Victoria, Australia; Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| |
Collapse
|
80
|
Ohara H, Nabika T. Genetic Modifications to Alter Blood Pressure Level. Biomedicines 2022; 10:biomedicines10081855. [PMID: 36009402 PMCID: PMC9405136 DOI: 10.3390/biomedicines10081855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 12/04/2022] Open
Abstract
Genetic manipulation is one of the indispensable techniques to examine gene functions both in vitro and in vivo. In particular, cardiovascular phenotypes such as blood pressure cannot be evaluated in vitro system, necessitating the creation of transgenic or gene-targeted knock-out and knock-in experimental animals to understand the pathophysiological roles of specific genes on the disease conditions. Although genome-wide association studies (GWAS) in various human populations have identified multiple genetic variations associated with increased risk for hypertension and/or its complications, the causal links remain unresolved. Genome-editing technologies can be applied to many different types of cells and organisms for creation of knock-out/knock-in models. In the post-GWAS era, it may be more worthwhile to validate pathophysiological implications of the risk variants and/or candidate genes by creating genome-edited organisms.
Collapse
|
81
|
Fujii R, Hishida A, Nakatochi M, Tsuboi Y, Suzuki K, Kondo T, Ikezaki H, Hara M, Okada R, Tamura T, Shimoshikiryo I, Suzuki S, Koyama T, Kuriki K, Takashima N, Arisawa K, Momozawa Y, Kubo M, Takeuchi K, Wakai K, Matsuo K, Tanaka K, Miura K, Kita Y, Takezaki T, Nagase H, Mikami H, Uehara R, Narimatsu H. Associations of Genome-Wide Polygenic Risk Score and Risk Factors With Hypertension in a Japanese Population. Circ Genom Precis Med 2022; 15:e003612. [DOI: 10.1161/circgen.121.003612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background:
Although many polygenic risk scores (PRS) for cardiovascular traits have been developed in European populations, it is an urgent task to construct a PRS and to evaluate its ability in non-European populations. We developed a genome-wide PRS for blood pressure in a Japanese population and examined the associations between this PRS and hypertension prevalence.
Methods:
We performed a cross-sectional study in 11 252 Japanese individuals who participated in the J-MICC (Japan Multi-Institutional Collaborative Cohort) study. Using publicly available GWAS summary statistics from Biobank Japan, we developed the PRS in the target data (n=7876). With >30 000 single nucleotide polymorphisms, we evaluated PRS performance in the test data (n=3376). Hypertension was defined as systolic blood pressure of 130 mm Hg or more, or diastolic blood pressure of 85 mm Hg or more, or taking an antihypertensive drug.
Results:
Compared with the middle PRS quintile, the prevalence of hypertension at the top PRS quintile was higher independently from traditional risk factors (odds ratio, 1.73 [95% CI, 1.32–2.27]). The difference of mean systolic blood pressure and diastolic blood pressure between the middle and the top PRS quintile was 4.55 (95% CI, 2.26–6.85) and 2.32 (95% CI, 0.86–3.78) mm Hg, respectively. Subgroups reflecting combinations of Japanese PRS and modifiable lifestyles and factors (smoking, alcohol intake, sedentary time, and obesity) were associated with the prevalence of hypertension. A European-derived PRS was not associated with hypertension in our participants.
Conclusions:
A PRS for blood pressure was significantly associated with hypertension and BP traits in a general Japanese population. Our findings also highlighted the importance of a combination of PRS and risk factors for identifying high-risk subgroups.
Collapse
Affiliation(s)
- Ryosuke Fujii
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, Toyoake, Japan (R.F., Y.T., K.S.), Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of interactive Medical & Healthcare Systems, Department of Integrated Health Sciences (R.F., T. Kondo), Nagoya University Graduate School of Medicine, Nagoya, Japan
- Institute for Biomedicine, Eurac Research (affiliated to the University of Lübeck), Bolzano/Bozen, Italy (R.F.)
| | - Asahi Hishida
- Department of Preventive Medicine (A.H., R.O., T.T., K.T., K.W.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences (M.N.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiki Tsuboi
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, Toyoake, Japan (R.F., Y.T., K.S.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Koji Suzuki
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, Toyoake, Japan (R.F., Y.T., K.S.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takaaki Kondo
- Division of interactive Medical & Healthcare Systems, Department of Integrated Health Sciences (R.F., T. Kondo), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Ikezaki
- Department of Comprehensive General Internal Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (H.I.)
| | - Megumi Hara
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan (M.H.)
| | - Rieko Okada
- Department of Preventive Medicine (A.H., R.O., T.T., K.T., K.W.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Tamura
- Department of Preventive Medicine (A.H., R.O., T.T., K.T., K.W.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ippei Shimoshikiryo
- Department of International Island & Community Medicine, Kagoshima University Graduate School of Medical & Dental Sciences, Kagoshima, Japan (I.S.)
| | - Sadao Suzuki
- Department of Public Health, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan (S.S.)
| | - Teruhide Koyama
- Department of Epidemiology for Community Health & Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan (T. Koyama)
| | - Kiyonori Kuriki
- Laboratory of Public Health, Division of Nutritional Sciences, School of Food & Nutritional Sciences, University of Shizuoka, Shizuoka, Shizuoka (K.K.)
| | - Naoyuki Takashima
- Department of Public Health, Shiga University of Medical Science, Otsu, Japan (N.T.)
- Department of Public Health, Kindai University Faculty of Medicine, Osaka, Japan (N.T.)
| | - Kokichi Arisawa
- Department of Preventive Medicine, Tokushima University Graduate School of Biomedical Scinces, Tokushima, Japan (K.A.)
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, Center for Integrative Medical Sciences, RIKEN, Kanagawa, Japan (Y.M., M.K.)
| | - Michiaki Kubo
- Laboratory for Genotyping Development, Center for Integrative Medical Sciences, RIKEN, Kanagawa, Japan (Y.M., M.K.)
| | - Kenji Takeuchi
- Department of Preventive Medicine (A.H., R.O., T.T., K.T., K.W.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenji Wakai
- Department of Preventive Medicine (A.H., R.O., T.T., K.T., K.W.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Weng Z, Liu Q, Yan Q, Liang J, Zhang X, Xu J, Li W, Xu C, Gu A. Associations of genetic risk factors and air pollution with incident hypertension among participants in the UK Biobank study. CHEMOSPHERE 2022; 299:134398. [PMID: 35339527 DOI: 10.1016/j.chemosphere.2022.134398] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 03/19/2022] [Accepted: 03/19/2022] [Indexed: 06/14/2023]
Abstract
The purposes of this study were to quantify the association of the combination of air pollution and genetic risk factors with hypertension and explore the interactions between air pollution and genetic risk. This study included 391,366 participants of European ancestry initially free from pre-existing hypertension in the UK Biobank. Exposure to ambient air pollutants, including particulate matter (PM2.5 PM2.5-10, and PM10), nitrogen dioxide (NO2) and nitrogen oxides (NOX), was estimated through land use regression modelling, and the associations between air pollutants and the incidence of hypertension were investigated using a Cox proportional hazards model adjusted for covariates. Furthermore, we established a polygenic risk score for hypertension and assessed the combined effect of genetic susceptibility and air pollution on incident hypertension. The results showed significant associations between the risk of hypertension and exposure to PM2.5 (hazard ratio [HR]: 1.41, 95% confidence interval [CI]: 1.29-1.53; per 10 μg/m3), PM10 (1.05, 1.00-1.09; per 10 μg/m3), and NOX (1.01, 1.01-1.02 per 10 μg/m3). Additive effects of PM2.5 and NOX exposure and genetic risk were observed. Compared to individuals with a low genetic risk and low air pollution exposure, participants with high air pollution exposure and a high genetic risk had a significantly increased risk of hypertension (PM2.5: 71% (66%-76%), PM10: 59% (55%-64%), NOX: 65% (60%-70%)). Our results indicate that long-term exposure to air pollution is associated with an increased risk of hypertension, especially in individuals with a high genetic risk.
Collapse
Affiliation(s)
- Zhenkun Weng
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Qian Liu
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Qing Yan
- Department of Neurosurgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jingjia Liang
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Xin Zhang
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Jin Xu
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China; Department of Maternal, Child, and Adolescent Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Wenxiang Li
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Cheng Xu
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China.
| | - Aihua Gu
- State Key Laboratory of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
83
|
Berillo O, Huo KG, Richer C, Fraulob-Aquino JC, Briet M, Lipman ML, Sinnett D, Paradis P, Schiffrin EL. Distinct transcriptomic profile of small arteries of hypertensive patients with chronic kidney disease identified miR-338-3p targeting GPX3 and PTPRS. J Hypertens 2022; 40:1394-1405. [PMID: 35703228 DOI: 10.1097/hjh.0000000000003160] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Hypertension is associated with vascular injury, which contributes to end-organ damage. MicroRNAs regulating mRNAs have been shown to play a role in vascular injury in hypertensive mice. We aimed to identify differentially expressed microRNAs and their mRNA targets in small arteries of hypertensive patients with/without chronic kidney disease (CKD) to shed light on the pathophysiological molecular mechanisms of vascular remodeling. METHODS AND RESULTS Normotensive individuals and hypertensive patients with/without CKD were recruited ( n = 15-16 per group). Differentially expressed microRNAs and mRNAs were identified uniquely associated with hypertension (microRNAs: 10, mRNAs: 68) or CKD (microRNAs: 68, mRNAs: 395), and in both groups (microRNAs: 2, mRNAs: 32) with a P less than 0.05 and a fold change less than or greater than 1.3 in subcutaneous small arteries ( n = 14-15). One of the top three differentially expressed microRNAs, miR-338-3p that was down-regulated in CKD, presented the best correlation between RNA sequencing and reverse transcription-quantitative PCR (RT-qPCR, R2 = 0.328, P < 0.001). Profiling of human aortic vascular cells showed that miR-338-3p was mostly expressed in endothelial cells. Two of the selected top nine up-regulated miR-338-3p predicted targets, glutathione peroxidase 3 ( GPX3 ) and protein tyrosine phosphatase receptor type S ( PTPRS ), were validated with mimics by RT-qPCR in human aortic endothelial cells ( P < 0.05) and by a luciferase assay in HEK293T cells ( P < 0.05). CONCLUSION A distinct transcriptomic profile was observed in gluteal subcutaneous small arteries of hypertensive patients with CKD. Down-regulated miR-338-3p could contribute to GPX3 and PTPRS up-regulation via the canonical microRNA targeting machinery in hypertensive patients with CKD. http://links.lww.com/HJH/C27.
Collapse
Affiliation(s)
- Olga Berillo
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research
| | - Ku-Geng Huo
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research
| | - Chantal Richer
- Division of Hematology-Oncology, Research Center, CHU Ste-Justine
| | | | - Marie Briet
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research
- INSERM U1083, CNRS UMR 6214, Centre Hospitalo-Universitaire d'Angers, Université d'Angers, Angers, France
| | - Mark L Lipman
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research
- Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University
| | - Daniel Sinnett
- Division of Hematology-Oncology, Research Center, CHU Ste-Justine
- Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Pierre Paradis
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research
| | - Ernesto L Schiffrin
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research
- Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University
| |
Collapse
|
84
|
Transcriptomics of angiotensin II-induced long noncoding and coding RNAs in endothelial cells. J Hypertens 2022; 40:1303-1313. [PMID: 35762471 DOI: 10.1097/hjh.0000000000003140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Angiotensin II (Ang II)-induced endothelial dysfunction plays an important role in the pathogenesis of cardiovascular diseases such as systemic hypertension, cardiac hypertrophy and atherosclerosis. Recently, long noncoding RNAs (lncRNAs) have been shown to play an essential role in the pathobiology of cardiovascular diseases; however, the effect of Ang II on lncRNAs and coding RNAs expression in endothelial cells has not been evaluated. Accordingly, we sought to evaluate the expression profiles of lncRNAs and coding RNAs in endothelial cells following treatment with Ang II. METHODS Human umbilical vein endothelial cells (HUVECs) were cultured and treated with Ang II (10-6 mol/l) for 24 h. The cells were then profiled for the expression of lncRNAs and mRNAs using the Arraystar Human lncRNA Expression Microarray V3.0. RESULTS In HUVECs following Ang II treatment, from a total of 30 584 lncRNA targets screened, 25 targets were significantly upregulated, while 69 were downregulated. In the same HUVECs samples, from 26 106 mRNA targets screened, 28 targets were significantly upregulated and 67 were downregulated. Of the differentially expressed lncRNAs, RP11-354P11.2 and RP11-360F5.1 were the most upregulated (11-fold) and downregulated (three-fold) lncRNAs, respectively. Assigning the differentially regulated genes into functional groups using bioinformatics reveals numerous genes involved in the nucleotide excision repair and ECM-receptor interaction. CONCLUSION This is the first study to profile the Ang II-induced differentially expressed lncRNAs and mRNAs in human endothelial cells. Our results reveal novel targets and substantially extend the list of potential candidate genes involved in Ang II-induced endothelial dysfunction and cardiovascular diseases.
Collapse
|
85
|
Parcha V, Irvin MR, Lange LA, Armstrong ND, Pampana A, Meyer M, Judd SE, Arora G, Arora P. Corin Missense Variants, Blood Pressure, and Hypertension in 11 322 Black Individuals: Insights From REGARDS and the Jackson Heart Study. J Am Heart Assoc 2022; 11:e025582. [PMID: 35699180 PMCID: PMC9238660 DOI: 10.1161/jaha.121.025582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/27/2022] [Indexed: 11/18/2022]
Abstract
Background Corin enzyme contributes to the processing of inactive natriuretic peptides to bioactive hormones. In Black individuals, Corin gene variants (rs111253292 [Q568P] and rs75770792 [T555I]) have been previously reported to have a modest association with blood pressure (BP) and hypertension. Methods and Results We evaluated the association of Corin genotype with BP traits, prevalent hypertension, and incident hypertension among self-identified 11 322 Black Americans in the REGARDS (Reasons for Geographic and Racial Differences in Stroke) study and the JHS (Jackson Heart Study) using multivariable-adjusted regression modeling. Multivariable-adjusted genotype-stratified differences in NT-proBNP (N-terminal pro-B-type natriuretic peptide) and BNP (B-type natriuretic peptide) levels were assessed. Genotype-stratified NPPA and NPPB expression differences in healthy organ donor left atrial and left ventricular heart tissue (N=15) were also examined. The rs111253292 genotype was not associated with systolic BP (β±SE, 0.42±0.58; -1.24±0.82), diastolic BP (0.51±0.33; -0.41±0.46), mean arterial pressure (0.48±0.38; -0.68±0.51), and prevalent hypertension (odds ratio [OR], 0.93 [95% CI, 0.80-1.09]; OR, 0.79 [95% CI, 0.61-1.01]) in both REGARDS and JHS, respectively. The rs75770792 genotype was not associated with systolic BP (0.48±0.58; -1.26±0.81), diastolic BP (0.52±0.33; -0.33±0.45), mean arterial pressure (0.50±0.38; -0.63±0.50), and prevalent hypertension (OR, 1.02 [95% CI, 0.84-1.23]; OR, 0.87 [95% CI, 0.67-1.13]) in both cohorts, respectively. The Corin genotype was also not associated with incident hypertension (OR, 1.35 [95% CI, 0.94-1.93]; OR, 0.95 [95% CI, 0.64-1.39]) in the study cohorts. The NT-proBNP levels in REGARDS and BNP levels in JHS were similar between the Corin genotype groups. In heart tissue, the NPPA and NPPB expression was similar between the genotype groups. Conclusions Corin gene variants observed more commonly in Black individuals are not associated with differences in NP expression, circulating NP levels, and BP or hypertension as previously reported in candidate gene studies. Understanding the genetic determinants of complex cardiovascular traits in underrepresented populations requires further evaluation.
Collapse
Affiliation(s)
- Vibhu Parcha
- Division of Cardiovascular DiseaseUniversity of Alabama at BirminghamBirminghamAL
| | | | - Leslie A. Lange
- Division of Biomedical Informatics and Personalized MedicineDepartment of MedicineUniversity of Colorado School of MedicineAuroraCO
- Department of EpidemiologyUniversity of Colorado School of Public HealthAuroraCO
| | | | - Akhil Pampana
- Division of Cardiovascular DiseaseUniversity of Alabama at BirminghamBirminghamAL
| | - Mariah Meyer
- Division of Biomedical Informatics and Personalized MedicineDepartment of MedicineUniversity of Colorado School of MedicineAuroraCO
| | - Suzanne E. Judd
- Department of BiostatisticsUniversity of Alabama at BirminghamBirminghamAL
| | - Garima Arora
- Division of Cardiovascular DiseaseUniversity of Alabama at BirminghamBirminghamAL
| | - Pankaj Arora
- Division of Cardiovascular DiseaseUniversity of Alabama at BirminghamBirminghamAL
- Section of CardiologyBirmingham Veterans Affairs Medical CenterBirminghamAL
| |
Collapse
|
86
|
Kurniansyah N, Goodman MO, Kelly TN, Elfassy T, Wiggins KL, Bis JC, Guo X, Palmas W, Taylor KD, Lin HJ, Haessler J, Gao Y, Shimbo D, Smith JA, Yu B, Feofanova EV, Smit RAJ, Wang Z, Hwang SJ, Liu S, Wassertheil-Smoller S, Manson JE, Lloyd-Jones DM, Rich SS, Loos RJF, Redline S, Correa A, Kooperberg C, Fornage M, Kaplan RC, Psaty BM, Rotter JI, Arnett DK, Morrison AC, Franceschini N, Levy D, Sofer T. A multi-ethnic polygenic risk score is associated with hypertension prevalence and progression throughout adulthood. Nat Commun 2022; 13:3549. [PMID: 35729114 PMCID: PMC9213527 DOI: 10.1038/s41467-022-31080-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/31/2022] [Indexed: 12/12/2022] Open
Abstract
In a multi-stage analysis of 52,436 individuals aged 17-90 across diverse cohorts and biobanks, we train, test, and evaluate a polygenic risk score (PRS) for hypertension risk and progression. The PRS is trained using genome-wide association studies (GWAS) for systolic, diastolic blood pressure, and hypertension, respectively. For each trait, PRS is selected by optimizing the coefficient of variation (CV) across estimated effect sizes from multiple potential PRS using the same GWAS, after which the 3 trait-specific PRSs are combined via an unweighted sum called "PRSsum", forming the HTN-PRS. The HTN-PRS is associated with both prevalent and incident hypertension at 4-6 years of follow up. This association is further confirmed in age-stratified analysis. In an independent biobank of 40,201 individuals, the HTN-PRS is confirmed to be predictive of increased risk for coronary artery disease, ischemic stroke, type 2 diabetes, and chronic kidney disease.
Collapse
Affiliation(s)
- Nuzulul Kurniansyah
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Matthew O Goodman
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Tali Elfassy
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Walter Palmas
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Henry J Lin
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jeffrey Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Yan Gao
- The Jackson Heart Study, University of Mississippi Medical Center, Jackson, MS, USA
| | - Daichi Shimbo
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Bing Yu
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Elena V Feofanova
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Roelof A J Smit
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhe Wang
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shih-Jen Hwang
- Department of Biostatistics, Boston University, Boston, MA, USA
| | - Simin Liu
- Center for Global Cardiometabolic Health and Departments of Epidemiology, Medicine, and Surgery, Brown University, Providence, RI, USA
| | - Sylvia Wassertheil-Smoller
- Department of Epidemiology & Population Health, Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - JoAnn E Manson
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Adolfo Correa
- Departments of Medicine and Pediatrics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Robert C Kaplan
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Donna K Arnett
- College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Daniel Levy
- The Population Sciences Branch of the National Heart, Lung and Blood Institute, Bethesda, MD, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
87
|
Sluysmans S, Salmaso A, Rouaud F, Méan I, Brini M, Citi S. The PLEKHA7-PDZD11 complex regulates the localization of the calcium pump PMCA and calcium handling in cultured cells. J Biol Chem 2022; 298:102138. [PMID: 35714771 PMCID: PMC9307954 DOI: 10.1016/j.jbc.2022.102138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 06/05/2022] [Accepted: 06/06/2022] [Indexed: 01/11/2023] Open
Abstract
The plasma membrane calcium ATPase (PMCA) extrudes calcium from the cytosol to the extracellular space to terminate calcium-dependent signaling. Although the distribution of PMCA is crucial for its function, the molecular mechanisms that regulate the localization of PMCA isoforms are not well understood. PLEKHA7 is implicated by genetic studies in hypertension and the regulation of calcium handling. PLEKHA7 recruits the small adapter protein PDZD11 to adherens junctions, and together they control the trafficking and localization of plasma membrane associated proteins, including the Menkes copper ATPase. Since PDZD11 binds to the C-terminal domain of b-isoforms of PMCA, PDZD11 and its interactor PLEKHA7 could control the localization and activity of PMCA. Here, we test this hypothesis using cultured cell model systems. We show using immunofluorescence microscopy and a surface biotinylation assay that KO of either PLEKHA7 or PDZD11 in mouse kidney collecting duct epithelial cells results in increased accumulation of endogenous PMCA at lateral cell–cell contacts and PDZ-dependent ectopic apical localization of exogenous PMCA4x/b isoform. In HeLa cells, coexpression of PDZD11 reduces membrane accumulation of overexpressed PMCA4x/b, and analysis of cytosolic calcium transients shows that PDZD11 counteracts calcium extrusion activity of overexpressed PMCA4x/b, but not PMCA4x/a, which lacks the PDZ-binding motif. Moreover, KO of PDZD11 in either endothelial (bEnd.3) or epithelial (mouse kidney collecting duct) cells increases the rate of calcium extrusion. Collectively, these results suggest that the PLEKHA7–PDZD11 complex modulates calcium homeostasis by regulating the localization of PMCA.
Collapse
Affiliation(s)
- Sophie Sluysmans
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Andrea Salmaso
- Department of Biology, University of Padua, Padua, Italy
| | - Florian Rouaud
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Isabelle Méan
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Marisa Brini
- Department of Biology, University of Padua, Padua, Italy.
| | - Sandra Citi
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
88
|
A Review of Vascular Traits and Assessment Techniques, and Their Heritability. Artery Res 2022. [DOI: 10.1007/s44200-022-00016-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
AbstractVarious tools are available to assess atherosclerosis, arterial stiffening, and endothelial function. They offer utility in the assessment of hypertensive phenotypes, in cardiovascular risk prediction, and as surrogate endpoints in clinical trials. We explore the relative influence of participant genetics, with reference to large-scale genomic studies, population-based cohorts, and candidate gene studies. We find heritability estimates highest for carotid intima-media thickness (CIMT 35–65%), followed by pulse wave velocity as a measure of arterial stiffness (26–43%), and flow mediated dilatation as a surrogate for endothelial function (14–39%); data were lacking for peripheral artery tonometry. We furthermore examine genes and polymorphisms relevant to each technique. We conclude that CIMT and pulse wave velocity dominate the existing evidence base, with fewer published genomic linkages for measures of endothelial function. We finally make recommendations regarding planning and reporting of data relating to vascular assessment techniques, particularly when genomic data are also available, to facilitate integration of these tools into cardiovascular disease research.
Collapse
|
89
|
Cong PK, Bai WY, Li JC, Yang MY, Khederzadeh S, Gai SR, Li N, Liu YH, Yu SH, Zhao WW, Liu JQ, Sun Y, Zhu XW, Zhao PP, Xia JW, Guan PL, Qian Y, Tao JG, Xu L, Tian G, Wang PY, Xie SY, Qiu MC, Liu KQ, Tang BS, Zheng HF. Genomic analyses of 10,376 individuals in the Westlake BioBank for Chinese (WBBC) pilot project. Nat Commun 2022; 13:2939. [PMID: 35618720 PMCID: PMC9135724 DOI: 10.1038/s41467-022-30526-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 05/05/2022] [Indexed: 01/04/2023] Open
Abstract
We initiate the Westlake BioBank for Chinese (WBBC) pilot project with 4,535 whole-genome sequencing (WGS) individuals and 5,841 high-density genotyping individuals, and identify 81.5 million SNPs and INDELs, of which 38.5% are absent in dbSNP Build 151. We provide a population-specific reference panel and an online imputation server ( https://wbbc.westlake.edu.cn/ ) which could yield substantial improvement of imputation performance in Chinese population, especially for low-frequency and rare variants. By analyzing the singleton density of the WGS data, we find selection signatures in SNX29, DNAH1 and WDR1 genes, and the derived alleles of the alcohol metabolism genes (ADH1A and ADH1B) emerge around 7,000 years ago and tend to be more common from 4,000 years ago in East Asia. Genetic evidence supports the corresponding geographical boundaries of the Qinling-Huaihe Line and Nanling Mountains, which separate the Han Chinese into subgroups, and we reveal that North Han was more homogeneous than South Han.
Collapse
Affiliation(s)
- Pei-Kuan Cong
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Wei-Yang Bai
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Jin-Chen Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center for Medical Genetics & Hunan Key Laboratory, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Meng-Yuan Yang
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Saber Khederzadeh
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Si-Rui Gai
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Nan Li
- The High-Performance Computing Center, Westlake University, Hangzhou, Zhejiang, China
| | - Yu-Heng Liu
- The High-Performance Computing Center, Westlake University, Hangzhou, Zhejiang, China
| | - Shi-Hui Yu
- Clinical Genome Center, KingMed Diagnostics, Co., Ltd., Guangzhou, Guangdong, China
| | - Wei-Wei Zhao
- Clinical Genome Center, KingMed Diagnostics, Co., Ltd., Guangzhou, Guangdong, China
| | - Jun-Quan Liu
- Clinical Genome Center, KingMed Diagnostics, Co., Ltd., Guangzhou, Guangdong, China
| | - Yi Sun
- Clinical Genome Center, KingMed Diagnostics, Co., Ltd., Guangzhou, Guangdong, China
| | - Xiao-Wei Zhu
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Pian-Pian Zhao
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Jiang-Wei Xia
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Peng-Lin Guan
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Yu Qian
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Jian-Guo Tao
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Lin Xu
- WBBC Shandong Center, Binzhou Medical University, Yantai, Shandong, China
| | - Geng Tian
- WBBC Shandong Center, Binzhou Medical University, Yantai, Shandong, China
| | - Ping-Yu Wang
- WBBC Shandong Center, Binzhou Medical University, Yantai, Shandong, China
| | - Shu-Yang Xie
- WBBC Shandong Center, Binzhou Medical University, Yantai, Shandong, China
| | - Mo-Chang Qiu
- WBBC Jiangxi Center, Jiangxi Medical College, Shangrao, Jiangxi, China
| | - Ke-Qi Liu
- WBBC Jiangxi Center, Jiangxi Medical College, Shangrao, Jiangxi, China
| | - Bei-Sha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Hou-Feng Zheng
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| |
Collapse
|
90
|
Jiang S, Zeng J, Zhang X, Zhou S, Wang L, Xu S, Lu Q. Association of urinary rubidium concentrations with hypertension risk and blood pressure levels: A cross-sectional study in China. J Trace Elem Med Biol 2022; 71:126936. [PMID: 35092936 DOI: 10.1016/j.jtemb.2022.126936] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/20/2021] [Accepted: 01/19/2022] [Indexed: 12/12/2022]
Abstract
BACHGROUND Rubidium resembles potassium. We hypothesized that rubidium might play a role in blood pressure control. METHODS We measured urinary rubidium concentrations and blood pressure levels using validated techniques and methods in 2002 eligible participants. Multivariable logistic and linear regression models were applied to explore the associations. The restricted cubic spline model was utilized to investigate the dose-response relationship. Furthermore, we explored the associations of rubidium with risk factors (glomerular filtration rate, uric acid, and homocysteine) for hypertension and relevant biochemical indexes. RESULTS After adjustment for potential confounders and urinary potassium and sodium levels, doubling of urinary rubidium concentrations was significantly associated with decreased hypertension risk [odds ratio (OR), 0.76; 95% confidence interval (CI), 0.61, 0.93] and reduced systolic blood pressure (SBP) levels of 2.92 (95% CI: 1.56, 4.26) mm Hg. Each 1.00 mg/L increase in rubidium concentrations was associated with a 1.25 mm Hg decreased SBP levels, which was at least 200 times more effective than potassium. Furthermore, urinary rubidium concentrations were negatively associated with the risk factors for hypertension. CONCLUSIONS Rubidium might have more prominent effects on lowering blood pressure levels than potassium. Prospective studies and experimental research focusing on our findings are needed.
Collapse
Affiliation(s)
- Shunli Jiang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Junchao Zeng
- Healthcare Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, #1277 Jiefang Road, Wuhan, Hubei, 430022, China
| | - Xu Zhang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuang Zhou
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lin Wang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Sanping Xu
- Healthcare Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, #1277 Jiefang Road, Wuhan, Hubei, 430022, China.
| | - Qing Lu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
91
|
Jung J, Kim H. Shared genetic etiology and antagonistic relationship of plasma renin activity and systolic blood pressure in a Korean cohorts. Genomics 2022; 114:110334. [PMID: 35278618 DOI: 10.1016/j.ygeno.2022.110334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/11/2022] [Accepted: 03/06/2022] [Indexed: 01/14/2023]
Abstract
Despite extensive studies on blood pressure, its genetic risk factors remain uncertain. Even one of the most researched blood pressure-related traits - renin - is not fully understood genetically. Here, we determine the genetic relationship and associated predisposition between blood pressure and baseline renin. In 8840 Korean individuals, we observed a strong negative genome-wide genetic correlation (rg = -0.484) between systolic blood pressure (SBP) and plasma renin activity (PRA), suggesting that antagonistic genetic signals explain the variance in the two traits. We found 51 significant pleiotropic SNPs affecting the two traits, which could contribute to the Renin-Angiotensin-Aldosterone System (RAAS). Our findings provide insight into studies on RAAS by identifying the genome-wide relationship and susceptibility loci of SBP and PRA.
Collapse
Affiliation(s)
- Jaehoon Jung
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 151-742, Republic of Korea; eGnome, 26 Beobwon-ro, Songpa-gu, Seoul 05836, Republic of Korea.
| | - Heebal Kim
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 151-742, Republic of Korea; eGnome, 26 Beobwon-ro, Songpa-gu, Seoul 05836, Republic of Korea; Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 151-742, Republic of Korea.
| |
Collapse
|
92
|
Adebamowo CA, Adeyemo A, Ashaye A, Akpa OM, Chikowore T, Choudhury A, Fakim YJ, Fatumo S, Hanchard N, Hauser M, Mitchell B, Mulder N, Ofori-Acquah SF, Owolabi M, Ramsay M, Tayo B, VasanthKumar AB, Zhang Y, Adebamowo SN. Polygenic risk scores for CARDINAL study. Nat Genet 2022; 54:527-530. [PMID: 35513726 PMCID: PMC9907721 DOI: 10.1038/s41588-022-01074-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The Cardiometabolic Disorders in African-Ancestry Populations (CARDINAL) study site is a well-powered, first-of-its-kind resource for developing, refining and validating methods for research into polygenic risk scores that accounts for local ancestry, to improve risk prediction in diverse populations.
Collapse
Affiliation(s)
- Clement A Adebamowo
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
- University of Maryland Marlene and Stewart Greenbaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Adeyinka Ashaye
- Department of Ophthalmology, University of Ibadan, Ibadan, Nigeria
| | - Onoja M Akpa
- Department of Epidemiology and Medical Statistics, University of Ibadan, Ibadan, Nigeria
| | - Tinashe Chikowore
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ananyo Choudhury
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Yasmina J Fakim
- Department of Biotechnology, University of Mauritius, Reduit, MU, Mauritius
| | - Segun Fatumo
- The African Computational Genomics (TACG) Research Group, MRC/UVRI and LSHTM (Uganda Research Unit), Entebbe, Uganda
- London School of Hygiene and Tropical Medicine, London, UK
| | - Neil Hanchard
- National Human Genome Research Institute, Rockville, MD, USA
| | - Michael Hauser
- Department of Medicine, Duke University, Durham, NC, USA
| | - Braxton Mitchell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Nicola Mulder
- Computational Biology Division, IDM, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Solomon F Ofori-Acquah
- West African Genetic Medicine Centre (WAGMC), University of Ghana, Accra, Ghana
- Center for Translational and International Hematology, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mayowa Owolabi
- Department of Medicine and Center for Genomic and Precision Medicine, University of Ibadan, Ibadan, Nigeria
| | - Michèle Ramsay
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Bamidele Tayo
- Department of Public Health Sciences, Loyola University Chicago, Maywood, IL, USA
| | | | - Yuji Zhang
- University of Maryland Marlene and Stewart Greenbaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sally N Adebamowo
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, USA.
- University of Maryland Marlene and Stewart Greenbaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
93
|
Luo S, Zheng N, Lang B. ULK4 in Neurodevelopmental and Neuropsychiatric Disorders. Front Cell Dev Biol 2022; 10:873706. [PMID: 35493088 PMCID: PMC9039724 DOI: 10.3389/fcell.2022.873706] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/29/2022] [Indexed: 11/21/2022] Open
Abstract
The gene Unc51-like kinase 4 (ULK4) belongs to the Unc-51-like serine/threonine kinase family and is assumed to encode a pseudokinase with unclear function. Recently, emerging evidence has suggested that ULK4 may be etiologically involved in a spectrum of neuropsychiatric disorders including schizophrenia, but the underlying mechanism remains unaddressed. Here, we summarize the key findings of the structure and function of the ULK4 protein to provide comprehensive insights to better understand ULK4-related neurodevelopmental and neuropsychiatric disorders and to aid in the development of a ULK4-based therapeutic strategy.
Collapse
Affiliation(s)
- Shilin Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Center of Translational Medicine and Innovative Drug, Changsha, China
| | - Nanxi Zheng
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Nanxi Zheng, ; Bing Lang,
| | - Bing Lang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Nanxi Zheng, ; Bing Lang,
| |
Collapse
|
94
|
Ward JL, Guillot E, Domenig O, Ware WA, Yuan L, Mochel JP. Circulating renin‐angiotensin‐aldosterone system activity in cats with systemic hypertension or cardiomyopathy. J Vet Intern Med 2022; 36:897-909. [PMID: 35285549 PMCID: PMC9151484 DOI: 10.1111/jvim.16401] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 02/19/2022] [Accepted: 02/23/2022] [Indexed: 12/22/2022] Open
Abstract
Background Activity of the circulating renin‐angiotensin‐aldosterone system (RAAS) has not been comprehensively characterized in cats with systemic hypertension (SH) or cardiomyopathy (CM), and the effects of furosemide or amlodipine treatment on the RAAS have not been fully evaluated in cats. Hypothesis/Objectives To document RAAS activity in cats with SH or CM compared to healthy cats and determine how RAAS profiles change with furosemide or amlodipine treatment. Animals Sixty‐six client‐owned cats: 15 with SH (7 amlodipine‐treated, 8 untreated), 17 with advanced CM (7 furosemide‐treated, 10 not furosemide‐treated), and 34 healthy cats. Methods Equilibrium concentrations of RAAS peptides and aldosterone were quantified in serum samples by liquid chromatography‐mass spectrometry. Variables were compared between groups using Kruskal‐Wallis analysis with post hoc Holms‐corrected Dunn's testing. Results Compared with healthy cats, cats with CM had higher concentrations of angiotensin I, aldosterone, and plasma renin activity (all P < .01), and these differences remained significant (P < .03) after considering subgroups of untreated or furosemide‐treated cats. Compared with healthy cats, untreated cats with SH showed no differences in RAAS biomarkers, whereas amlodipine‐treated cats had higher concentrations of angiotensins I, II, III, IV, and 1‐7, aldosterone, and plasma renin activity (all P < .03). Multivariable analysis determined that furosemide and amlodipine treatments were independent predictors of increased RAAS biomarker concentrations. Conclusions and Clinical Importance Cats with CM had increased RAAS activity, whereas cats with untreated SH did not. Furosemide and amlodipine both led to nonspecific activation of both classical and alternative RAAS pathways in cats.
Collapse
Affiliation(s)
- Jessica L. Ward
- Department of Veterinary Clinical Sciences College of Veterinary Medicine, Iowa State University Ames Iowa USA
| | - Emilie Guillot
- Ceva Santé Animale Companion Animal Franchise Libourne France
| | | | - Wendy A. Ware
- Department of Veterinary Clinical Sciences College of Veterinary Medicine, Iowa State University Ames Iowa USA
| | - Lingnan Yuan
- Department of Veterinary Biomedical Sciences, SMART Pharmacology College of Veterinary Medicine, Iowa State University Ames Iowa USA
| | - Jonathan P. Mochel
- Department of Veterinary Biomedical Sciences, SMART Pharmacology College of Veterinary Medicine, Iowa State University Ames Iowa USA
| |
Collapse
|
95
|
Cataloging the potential SNPs (single nucleotide polymorphisms) associated with quantitative traits, viz. BMI (body mass index), IQ (intelligence quotient) and BP (blood pressure): an updated review. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00266-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Single nucleotide polymorphism (SNP) variants are abundant, persistent and widely distributed across the genome and are frequently linked to the development of genetic diseases. Identifying SNPs that underpin complex diseases can aid scientists in the discovery of disease-related genes by allowing for early detection, effective medication and eventually disease prevention.
Main body
Various SNP or polymorphism-based studies were used to categorize different SNPs potentially related to three quantitative traits: body mass index (BMI), intelligence quotient (IQ) and blood pressure, and then uncovered common SNPs for these three traits. We employed SNPedia, RefSNP Report, GWAS Catalog, Gene Cards (Data Bases), PubMed and Google Scholar search engines to find relevant material on SNPs associated with three quantitative traits. As a result, we detected three common SNPs for all three quantitative traits in global populations: SNP rs6265 of the BDNF gene on chromosome 11p14.1, SNP rs131070325 of the SL39A8 gene on chromosome 4p24 and SNP rs4680 of the COMT gene on chromosome 22q11.21.
Conclusion
In our review, we focused on the prevalent SNPs and gene expression activities that influence these three quantitative traits. These SNPs have been used to detect and map complex, common illnesses in communities for homogeneity testing and pharmacogenetic studies. High blood pressure, diabetes and heart disease, as well as BMI, schizophrenia and IQ, can all be predicted using common SNPs. Finally, the results of our work can be used to find common SNPs and genes that regulate these three quantitative features across the genome.
Collapse
|
96
|
Lysine-specific demethylase 1 as a corepressor of mineralocorticoid receptor. Hypertens Res 2022; 45:641-649. [PMID: 35177789 DOI: 10.1038/s41440-022-00859-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/26/2021] [Accepted: 12/10/2021] [Indexed: 12/15/2022]
Abstract
Mineralocorticoid receptor (MR) and its ligand aldosterone play a central role in controlling blood pressure by promoting sodium reabsorption in the kidney. Coregulators are recruited to regulate the activation of steroid hormone receptors. In our previous study, we identified several new candidates for MR coregulators through liquid chromatography-tandem mass spectrometry analysis using a biochemical approach. Lysine-specific demethylase 1 (LSD1) was identified as a candidate. The relationship between LSD1 and salt-sensitive hypertension has been reported; however, the role of MR in this condition is largely unknown. Here, we investigated the functions of LSD1 as a coregulator of MR. First, a coimmunoprecipitation assay using HEK293F cells showed specific interactions between MR and LSD1. A chromatin immunoprecipitation study demonstrated LSD1 recruitment to the gene promoter of epithelial Na+ channel (ENaC), a target gene of MR. Reduced LSD1 expression by treatment with shRNA potentiated the hormonal activation of ENaC and serum/glucocorticoid-regulated kinase 1, another target gene of MR, indicating that LSD1 is a corepressor of MR. In an animal study, mice with kidney-specific LSD1 knockout (LSD1flox/floxKSP-Cre mice) developed hypertension after a high-salt diet without elevation of aldosterone levels, which was counteracted by cotreatment with spironolactone, an MR antagonist. In conclusion, our in vitro and in vivo studies demonstrated that LSD1 is a newly identified corepressor of MR.
Collapse
|
97
|
Morciano G, Rimessi A, Patergnani S, Vitto VAM, Danese A, Kahsay A, Palumbo L, Bonora M, Wieckowski MR, Giorgi C, Pinton P. Calcium dysregulation in heart diseases: Targeting calcium channels to achieve a correct calcium homeostasis. Pharmacol Res 2022; 177:106119. [PMID: 35131483 DOI: 10.1016/j.phrs.2022.106119] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 12/16/2022]
Abstract
Intracellular calcium signaling is a universal language source shared by the most part of biological entities inside cells that, all together, give rise to physiological and functional anatomical units, the organ. Although preferentially recognized as signaling between cell life and death processes, in the heart it assumes additional relevance considered the importance of calcium cycling coupled to ATP consumption in excitation-contraction coupling. The concerted action of a plethora of exchangers, channels and pumps inward and outward calcium fluxes where needed, to convert energy and electric impulses in muscle contraction. All this without realizing it, thousands of times, every day. An improper function of those proteins (i.e., variation in expression, mutations onset, dysregulated channeling, differential protein-protein interactions) being part of this signaling network triggers a short circuit with severe acute and chronic pathological consequences reported as arrhythmias, cardiac remodeling, heart failure, reperfusion injury and cardiomyopathies. By acting with chemical, peptide-based and pharmacological modulators of these players, a correction of calcium homeostasis can be achieved accompanied by an amelioration of clinical symptoms. This review will focus on all those defects in calcium homeostasis which occur in the most common cardiac diseases, including myocardial infarction, arrhythmia, hypertrophy, heart failure and cardiomyopathies. This part will be introduced by the state of the art on the proteins involved in calcium homeostasis in cardiomyocytes and followed by the therapeutic treatments that to date, are able to target them and to revert the pathological phenotype.
Collapse
Affiliation(s)
- Giampaolo Morciano
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, RA, Italy.
| | - Alessandro Rimessi
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Simone Patergnani
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Veronica A M Vitto
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Alberto Danese
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Asrat Kahsay
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Laura Palumbo
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Massimo Bonora
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Mariusz R Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism. Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Pinton
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, RA, Italy.
| |
Collapse
|
98
|
Leopold JA. Personalizing treatments for patients based on cardiovascular phenotyping. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2022; 7:4-16. [PMID: 36778892 PMCID: PMC9913616 DOI: 10.1080/23808993.2022.2028548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Introduction Cardiovascular disease persists as the leading cause of death worldwide despite continued advances in diagnostics and therapeutics. Our current approach to patients with cardiovascular disease is rooted in reductionism, which presupposes that all patients share a similar phenotype and will respond the same to therapy; however, this is unlikely as cardiovascular diseases exhibit complex heterogeneous phenotypes. Areas covered With the advent of high-throughput platforms for omics testing, phenotyping cardiovascular diseases has advanced to incorporate large-scale molecular data with classical history, physical examination, and laboratory results. Findings from genomics, proteomics, and metabolomics profiling have been used to define more precise cardiovascular phenotypes and predict adverse outcomes in population-based and disease-specific patient cohorts. These molecular data have also been utilized to inform drug efficacy based on a patient's unique phenotype. Expert opinion Multiscale phenotyping of cardiovascular disease has revealed diversity among patients that can be used to personalize pharmacotherapies and predict outcomes. Nonetheless, precision phenotyping for cardiovascular disease remains a nascent field that has not yet translated into widespread clinical practice despite its many potential advantages for patient care. Future endeavors that demonstrate improved pharmacotherapeutic responses and associated reduction in adverse events will facilitate mainstream adoption of precision cardiovascular phenotyping.
Collapse
Affiliation(s)
- Jane A. Leopold
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, 77 Ave Louis Pasteur, NRB0630K, Boston, Massachusetts, USA
| |
Collapse
|
99
|
Jeung HC, Puentes R, Aleshin A, Indarte M, Correa RG, Bankston LA, Layng FIAL, Ahmed Z, Wistuba I, Yao Y, Duenas DG, Zhang S, Meuillet EJ, Marassi F, Liddington RC, Kirkpatrick L, Powis G. PLEKHA7 signaling is necessary for the growth of mutant KRAS driven colorectal cancer. Exp Cell Res 2021; 409:112930. [PMID: 34800542 DOI: 10.1016/j.yexcr.2021.112930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/26/2021] [Accepted: 11/14/2021] [Indexed: 10/19/2022]
Abstract
Plekha7 (Pleckstrin homology [PH] domain containing, family A member 7) regulates the assembly of proteins of the cytoplasmic apical zonula adherens junction (AJ), thus ensuring cell-cell adhesion and tight-junction barrier integrity. Little is known of Plekha7 function in cancer. In colorectal cancer (CRC) Plekha7 expression is elevated compared to adjacent normal tissue levels, increasing with clinical stage. Plekha7 was present at plasma membrane AJ with wild-type KRas (wt-KRas) but was dispersed in cells expressing mutant KRas (mut-KRas). Fluorescence lifetime imaging microscopy (FLIM) indicated a direct Plekha7 interaction with wt-KRas but scantily with mut-KRas. Inhibiting Plekha7 specifically decreased mut-KRas cell signaling, proliferation, attachment, migration, and retarded mut-KRAS CRC tumor growth. Binding of diC8-phosphoinositides (PI) to the PH domain of Plekha7 was relatively low affinity. This may be because a D175 amino acid residue plays a "sentry" role preventing PI(3,4)P2 and PI(3,4,5)P3 binding. Molecular or pharmacological inhibition of the Plekha7 PH domain prevented the growth of mut-KRas but not wt-KRas cells. Taken together the studies suggest that Plekha7, in addition to maintaining AJ structure plays a role in mut-KRas signaling and phenotype through interaction of its PH domain with membrane mut-KRas, but not wt-KRas, to increase the efficiency of mut-KRas downstream signaling.
Collapse
Affiliation(s)
- Hei-Cheul Jeung
- MD Anderson Cancer Center, Houston, TX, USA; Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Gangnam-Gu, Seoul, South Korea
| | - Roisin Puentes
- Sanford Burnham Prebys Medical Discovery Institute Cancer Center, La Jolla, CA, USA
| | - Alexander Aleshin
- Sanford Burnham Prebys Medical Discovery Institute Cancer Center, La Jolla, CA, USA
| | | | - Ricardo G Correa
- Sanford Burnham Prebys Medical Discovery Institute Cancer Center, La Jolla, CA, USA
| | - Laurie A Bankston
- Sanford Burnham Prebys Medical Discovery Institute Cancer Center, La Jolla, CA, USA
| | - Fabiana I A L Layng
- Sanford Burnham Prebys Medical Discovery Institute Cancer Center, La Jolla, CA, USA
| | | | | | - Yong Yao
- Sanford Burnham Prebys Medical Discovery Institute Cancer Center, La Jolla, CA, USA
| | - Daniela G Duenas
- Sanford Burnham Prebys Medical Discovery Institute Cancer Center, La Jolla, CA, USA
| | | | | | - Francesca Marassi
- Sanford Burnham Prebys Medical Discovery Institute Cancer Center, La Jolla, CA, USA
| | - Robert C Liddington
- Sanford Burnham Prebys Medical Discovery Institute Cancer Center, La Jolla, CA, USA
| | | | - Garth Powis
- Sanford Burnham Prebys Medical Discovery Institute Cancer Center, La Jolla, CA, USA; PHusis Therapeutics, La Jolla, CA, USA.
| |
Collapse
|
100
|
Sheikhy A, Fallahzadeh A, Aghaei Meybodi HR, Hasanzad M, Tajdini M, Hosseini K. Personalized medicine in cardiovascular disease: review of literature. J Diabetes Metab Disord 2021; 20:1793-1805. [PMID: 34900826 DOI: 10.1007/s40200-021-00840-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/18/2021] [Indexed: 12/13/2022]
Abstract
Purpose Personalized medicine (PM) is the concept of managing patients based on their characteristics, including genotypes. In the field of cardiology, advantages of PM could be found in the diagnosis and treatment of several conditions such as arrhythmias and cardiomyopathies; moreover, it may be beneficial to prevent adverse drug reactions (ADR) and select the best medication. Genetic background can help us in selecting effective treatments, appropriate dose requirements, and preventive strategies in individuals with particular genotypes. Method In this review, we provide examples of personalized medicine based on human genetics for the most used pharmaceutics in cardiology, including warfarin, clopidogrel, and statins. We also review cardiovascular diseases, including coronary artery disease, arrhythmia, and cardiomyopathies. Conclusion Genetic factors are as important as environmental factors and they should be tested and evaluated more in the future by improving in genetic testing tools. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-021-00840-0.
Collapse
Affiliation(s)
- Ali Sheikhy
- Research Department, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Aida Fallahzadeh
- Research Department, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Aghaei Meybodi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mandana Hasanzad
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Masih Tajdini
- Cardiology Department, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Kaveh Hosseini
- Cardiology Department, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|