1
|
Niu Y, Li X, Guo J, Luo S, Shang X, Liu J, Liu S, He M, Shi D, Huang Y, Zhang H. Comprehensive genome-wide analysis of retinal vessel caliber reveals microvascular-blood pressure pathways: advancing predictive, preventive, and personalized medicine. EPMA J 2025; 16:401-417. [PMID: 40438498 PMCID: PMC12106259 DOI: 10.1007/s13167-025-00411-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 04/20/2025] [Indexed: 06/01/2025]
Abstract
Background Retinal vessel caliber is strongly associated with systemic blood pressure (BP); however, the causal relationship between retinal vascular caliber and BP remains unclear. Understanding this relationship is essential for advancing predictive, preventive, and personalized medicine (PPPM) approaches to effectively manage hypertension and its related complications. Working hypothesis Microvessel morphology is causally related to blood pressure. By integrating genome-wide association studies, Mendelian randomization analysis, transcriptomic data, and multivariate genomic approaches, this study aims to identify predictive biomarkers, uncover preventive strategies, and develop personalized intervention targets, thereby advancing the principles of 3P medicine for improved cardiovascular health management. Methods and results We conducted a comprehensive investigation into the genetic factors underlying retinal vessel calibers and their complex relationship with BP traits. Our genome-wide association study (GWAS) assess retinal vessel calibers-central retinal arteriolar equivalent (CRAE), central retinal venular equivalent (CRVE), and the arteriole-to-venule ratio (AVR)-in a subset of 36,223 individuals of European descent from the UK Biobank. The analysis identified 9, 5, and 4 SNPs located in TNS, Y_RNA, PBLD, C10orf32-ASMT:AS3MT, GNB3:CDCA3, NTN4, COL4 A2, CTD-2378E21.1, WNT7B, VTA1, FCF1, NPLOC4, FUT1 and CSK region, which are significantly associated with CRAE, CRVE, and AVR, respectively. Genetic correlation analysis revealed shared heritability between BP traits and both CRAE and AVR, but not CRVE. Mendelian randomization analysis confirmed bidirectional causal relationships between CRAE and BP traits, whereas CRVE was neither influenced by nor influenced BP traits. To explore the potential regulatory mechanisms, we leveraged transcriptomic data and identified the following causal pathways in vessel tissue: The expression of MRPL23-AS1 and ULK3 was correlated with the elevation of blood pressure SBP and narrowing of the CRAE. Finally, we constructed a multivariable genetic model including CRAE, AVR, SBP, and DBP, suggesting a common driving factor which underlies these traits. Conclusions Our study elucidates the complex relationship between BP and retinal vessel caliber, highlighting potential intervention targets for lowering BP and vascular narrowing-related diseases. These findings contribute to the development of tailored prevention and treatment strategies aligned with PPPM principles. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-025-00411-w.
Collapse
Affiliation(s)
- Yongyi Niu
- Department of Ophthalmology, Guangdong Provincial People’s Hospital of Southern Medical University, Guangzhou, 510080 China
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Xue Li
- Department of Ophthalmology, The Second People’s Hospital of Foshan, Foshan, 528000 China
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Jingze Guo
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Songyuan Luo
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080 Guangdong China
| | - Xianwen Shang
- Experimental Ophthalmology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Jing Liu
- Department of Ophthalmology, Guangdong Provincial People’s Hospital of Southern Medical University, Guangzhou, 510080 China
| | - Shunming Liu
- Department of Ophthalmology, Guangdong Provincial People’s Hospital of Southern Medical University, Guangzhou, 510080 China
| | - Mingguang He
- Department of Ophthalmology, Guangdong Provincial People’s Hospital of Southern Medical University, Guangzhou, 510080 China
- Experimental Ophthalmology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Danli Shi
- Experimental Ophthalmology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yu Huang
- Department of Ophthalmology, Guangdong Provincial People’s Hospital of Southern Medical University, Guangzhou, 510080 China
- Division of Population Health and Genomics, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY UK
| | - Hongyang Zhang
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| |
Collapse
|
2
|
Liu Y, Hong J, Wang G, Mei Z. An emerging role of SNAREs in ischemic stroke: From pre-to post-diseases. Biochem Pharmacol 2025; 236:116907. [PMID: 40158821 DOI: 10.1016/j.bcp.2025.116907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/04/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Ischemic stroke is a debilitating condition characterized by high morbidity, disability, recurrence, and mortality rates on a global scale, posing a significant threat to public health and economic stability. Extensive research has thoroughly explored the molecular mechanisms underlying ischemic stroke, elucidating a strong association between soluble N-ethylmaleimide-sensitive factor (NSF) attachment protein receptor proteins (SNAREs) and the pathogenesis of this condition. SNAREs, a class of highly conserved proteins involved in membrane fusion, play a crucial role in modulating neuronal information transmission and promoting myelin formation in the central nervous system (CNS). Preventing the SNARE complex formation, malfunctions in SNARE-dependent exocytosis, and altered regulation of SNARE-mediated vesicle fusion are linked to excitotoxicity, endoplasmic reticulum (ER) stress, and programmed cell death (PCD) in ischemic stroke. However, its underlying mechanisms remain unclear. This study conducts a comprehensive review of the existing literature on SNARE proteins, encompassing the structure, classification, and expression of the SNARE protein family, as well as the assembly - disassembly cycle of SNARE complexes and their physiological roles in the CNS. We thoroughly examine the mechanisms by which SNAREs contribute to the pathological progression and associated risk factors of ischemic stroke (hypertension, hyperglycemia, dyslipidemia, and atherosclerosis). Furthermore, our findings highlight the promise of SNAREs as a viable target for pharmacological interventions in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yaxin Liu
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Jingyan Hong
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Guozuo Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410005, China.
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| |
Collapse
|
3
|
Onyenobi E, Zhong M, Soremekun O, Kamiza A, Boua R, Chikwore T, ACCME Research Group*, Fatumo S, Choudhury A, Hazelhurst S, Adebamowo C, Ramsay M, Tayo B, Albrecht JS, O'Connor TD, Zhang Y, Mitchell BD, Adebamowo SN. Development and Validation of Polygenic Risk Scores for Blood Pressure Traits in Continental African Populations. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2025:e005048. [PMID: 40421529 DOI: 10.1161/circgen.124.005048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 04/21/2025] [Indexed: 05/28/2025]
Abstract
BACKGROUND Most polygenic risk scores (PRS) have been developed in European populations, frequently leading to limited transferability across diverse ancestry populations. This study aimed to develop and evaluate PRS for blood pressure (BP) traits in continental African (African) populations and investigate how African genetic diversity influences PRS performance. METHODS We generated PRS for systolic BP, diastolic BP, pulse pressure, and hypertension. We used a pan-African cohort as the target population and compared singleancestry and multiancestry PRS methods. We compared the performance of African ancestry-derived PRS against multiancestry PRS on the entire data set and within South, East, and West African subpopulations. RESULTS Multiancestry PRS demonstrated significantly higher predictive accuracy compared with singleancestry PRS models. PRS predictive accuracy varied across different African regions, with the highest performance observed in East Africa. In the combined population, the difference in mean BP values between the first multiancestry PRS quartile and the top quartile was 6.53 (95% CI, 5.3-7.74), 3.81 (95% CI, 3.9-4.52), and 3.59 (95% CI, 2.4-4.32) mm Hg for systolic BP, diastolic BP, and pulse pressure, respectively. Individuals in the highest PRS risk quartile had odds of hypertension that were 1.47 (95% CI, 1.7-1.69) times greater than those in the lowest risk quartile. CONCLUSIONS These findings highlight the importance of integrating diverse ancestries in PRS development and accounting for subpopulation genetic variation to improve the predictive accuracy of BP PRS in African populations.
Collapse
Affiliation(s)
- Ebuka Onyenobi
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore. (E.O., M.Z., C.A., J.S.A., Y.Z., B.D.M., S.N.A.)
| | - Michael Zhong
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore. (E.O., M.Z., C.A., J.S.A., Y.Z., B.D.M., S.N.A.)
| | - Opeyemi Soremekun
- The African Computational Genomics Research Group, MRC/UVRI and LSHTM, Entebbe, Uganda (O.S., A.K., S.F.)
| | - Abram Kamiza
- The African Computational Genomics Research Group, MRC/UVRI and LSHTM, Entebbe, Uganda (O.S., A.K., S.F.)
| | - Romuald Boua
- Institut de Recherche en Sciences de la Santé, Clinical Research Unit of Nanoro, Burkina Faso (R.B.)
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa. (R.B., A.C., S.H., M.R.)
| | - Tinashe Chikwore
- Harvard Medical School (T.C.)
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA (T.C.)
| | | | - Segun Fatumo
- The African Computational Genomics Research Group, MRC/UVRI and LSHTM, Entebbe, Uganda (O.S., A.K., S.F.)
- Precision Healthcare University Research Institute, Queen Mary University of London, United Kingdom (S.F.)
| | - Ananyo Choudhury
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa. (R.B., A.C., S.H., M.R.)
| | - Scott Hazelhurst
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa. (R.B., A.C., S.H., M.R.)
- and School of Electrical and Information Engineering, University of the Witwatersrand, Johannesburg, South Africa. (S.H.)
| | - Clement Adebamowo
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore. (E.O., M.Z., C.A., J.S.A., Y.Z., B.D.M., S.N.A.)
- Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore. (C.A., S.N.A.)
| | - Michele Ramsay
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa. (R.B., A.C., S.H., M.R.)
| | - Bamidele Tayo
- Department of Public Health Sciences, Loyola University Parkinson School of Health Sciences and Public Health, Maywood, IL (B.T.)
| | - Jennifer S Albrecht
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore. (E.O., M.Z., C.A., J.S.A., Y.Z., B.D.M., S.N.A.)
| | - Timothy D O'Connor
- Department of Medicine, University of Maryland School of Medicine, Baltimore. (T.D.O., B.D.M.)
| | - Yuji Zhang
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore. (E.O., M.Z., C.A., J.S.A., Y.Z., B.D.M., S.N.A.)
| | - Braxton D Mitchell
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore. (E.O., M.Z., C.A., J.S.A., Y.Z., B.D.M., S.N.A.)
- Department of Medicine, University of Maryland School of Medicine, Baltimore. (T.D.O., B.D.M.)
| | - Sally N Adebamowo
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore. (E.O., M.Z., C.A., J.S.A., Y.Z., B.D.M., S.N.A.)
- Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore. (C.A., S.N.A.)
| |
Collapse
|
4
|
Riglea T, Dessy T, Kalubi J, Goulet D, Ikwa Ndol Mbutiwi F, Williams SM, Engert JC, Chen HY, O'Loughlin J, Sylvestre MP. Body mass index modifies genetic susceptibility to high systolic blood pressure in adolescents and young adults: results from an 18-year longitudinal study. J Hum Hypertens 2025; 39:334-342. [PMID: 40089570 DOI: 10.1038/s41371-025-01003-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/07/2025] [Accepted: 02/27/2025] [Indexed: 03/17/2025]
Abstract
Genome-wide association studies (GWAS) in adults have identified single nucleotide polymorphisms (SNPs) associated with systolic blood pressure (SBP), but it is unclear whether the findings apply in youth. Further, the role of body mass index (BMI) in these associations is understudied. Our objective was to determine whether BMI modifies genetic susceptibility to high SBP in young people. The sample comprised 714 participants of European ancestry recruited in 1999-2000 from 10 Montreal-area high schools for a longitudinal study. SBP was measured at ages 12, 15, 17, 24, and 30. Blood and saliva samples were collected at ages 14, 20, and 25. Two evidence-based genetic risk scores (GRS) were constructed based on GWAS results in adults: GRS22 used 22 SNPs and GRS182 added 160 additional SNPs to GRS22. Sex-specific associations between each GRS and repeated measures of SBP were estimated using linear mixed models including BMI and a GRS*BMI product term. GRS182 explained a greater proportion of SBP variance than GRS22, and a greater proportion in females than males. The associations increased monotonically with BMI values between 22 kg/m2 and 35 kg/m2. Results indicate that BMI modifies the association between a GRS and SBP levels from adolescence to adulthood.
Collapse
Affiliation(s)
- Teodora Riglea
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CrCHUM), Montréal, QC, Canada
- Department of Social and Preventive Medicine, Université de Montréal, Montréal, QC, Canada
| | - Tatiana Dessy
- Department of Social and Preventive Medicine, Université de Montréal, Montréal, QC, Canada
- Montreal Heart Institute, Montréal, QC, Canada
- Centre de Pharmacogénomique Beaulieu-Saucier de l'Université de Montréal, Montréal, QC, Canada
| | - Jodi Kalubi
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CrCHUM), Montréal, QC, Canada
- Department of Social and Preventive Medicine, Université de Montréal, Montréal, QC, Canada
- Centre de recherche en santé publique (CReSP), Université de Montréal & CIUSSS du Centre-Sud-de-l'Île-de-Montréal, Montreal, QC, Canada
| | - Danick Goulet
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | - Fiston Ikwa Ndol Mbutiwi
- Department of Social and Preventive Medicine, Université de Montréal, Montréal, QC, Canada
- Faculty of Medicine, University of Kikwit, Kikwit, Democratic Republic of the Congo
| | - Scott M Williams
- Case Western Reserve University School of Medicine Department of Population and Quantitative Health Sciences, Cleveland, OH, USA
- Cleveland Institute for Computational Biology, Cleveland, OH, USA
| | - James C Engert
- McGill University Department of Medicine, Montréal, QC, Canada
- McGill University Department of Human Genetics, Montréal, QC, Canada
| | - Hao Yu Chen
- McGill University Department of Medicine, Montréal, QC, Canada
| | - Jennifer O'Loughlin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CrCHUM), Montréal, QC, Canada
- Department of Social and Preventive Medicine, Université de Montréal, Montréal, QC, Canada
| | - Marie-Pierre Sylvestre
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CrCHUM), Montréal, QC, Canada.
- Department of Social and Preventive Medicine, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
5
|
Han Y, Huh R, Jung KJ, Kimm H, Jee SH. Dietary modulation for the hypertension risk group in Koreans: a cross-sectional study. Nutr Metab (Lond) 2025; 22:30. [PMID: 40211282 PMCID: PMC11987358 DOI: 10.1186/s12986-025-00921-4] [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: 11/11/2024] [Accepted: 03/21/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Hypertension (HTN) is a critical global health issue, contributing to high morbidity and mortality rates. Representative risk factors for HTN include aging, genetics, obesity, alcohol drinking, smoking, and diet. Dietary interventions like the Dietary Approaches to Stop HTN (DASH) diet plan effectively prevent and manage HTN. We intend to evaluate the influence of eating patterns on HTN, applying multiple risk factors. METHODS For cross-sectional design, study subjects were grouped into four groups: optimal (n = 7,712), normal (n = 1,220), high normal (n = 3,655), and HTN (n = 4,355) according to the 2022 HTN treatment guidelines of Korea. Factor analysis was performed to identify major dietary patterns based on nutritional data obtained from a brief dietary questionnaire, including 17 food items. Finally, we conducted a moderation analysis to evaluate the impact of dietary patterns on the HTN risk score, which is determined by genetic variables, body mass index, alcohol consumption, and smoking status. RESULTS We identified three principal dietary patterns (Korean, Western, and New diet) in the study population. Adherence to the New diet was linked to lower HTN risk in all models (p < 0.001), while the Western and Korean diets were associated with a higher risk of HTN in some models. In high HTN-risk individuals, adherence to the Western diet increased the HTN risk trend (p < 0.001), whereas the New diet showed a potential protective trend (p = 0.059). CONCLUSIONS The nutritional moderation effect was evident in the HTN high-risk group, where the Western diet increased risk, while the New diet showed a borderline protective effect. If the findings are validated by longitudinal investigation, our findings could serve as the basis for developing dietary guidelines for HTN.
Collapse
Affiliation(s)
- Youngmin Han
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Republic of Korea
- Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Republic of Korea
| | - Ryun Huh
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Republic of Korea
- Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Republic of Korea
| | - Keum Ji Jung
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Republic of Korea
- Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Republic of Korea
| | - Heejin Kimm
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Republic of Korea
- Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Republic of Korea
| | - Sun Ha Jee
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Republic of Korea.
- Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
6
|
Aubdool AA, Moyes AJ, Pérez-Ternero C, Baliga RS, Sanghera J, Syed MT, Jaigirdah K, Panesar AK, Tsui JC, Li Y, Vasquez HG, Shen YH, LeMaire SA, Raffort-Lareyre J, Mallat Z, Lu HS, Daugherty A, Hobbs AJ. Endothelium- and Fibroblast-Derived C-Type Natriuretic Peptide Prevents the Development and Progression of Aortic Aneurysms. Arterioscler Thromb Vasc Biol 2025. [PMID: 40177775 DOI: 10.1161/atvbaha.124.322350] [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: 12/18/2024] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Thoracic aortic aneurysm (AA) and abdominal AA are life-threatening diseases characterized by dilation, inflammation, and structural weakness; development of pharmacological therapies is desperately needed. CNP (C-type natriuretic peptide) plays a key role in vascular homeostasis, mediating vasodilator, anti-inflammatory, and antiatherogenic actions. Because such processes drive AA, we determined the role of endogenous CNP in offsetting pathogenesis. METHODS Tissue from patients with AA was analyzed to determine the consequences on CNP signaling. Ascending and suprarenal aortic diameters were assessed at baseline and following Ang II (angiotensin II; 1.44 mg/kg per day) infusion in wild-type, endothelium-restricted (ecCNP-/-), fibroblast-restricted (fbCNP-/-), global CNP (gbCNP-/-), or global NPR-C-/- mice infected with an adeno-associated virus expressing a proprotein convertase subtilisin/kexin type 9 gain-of-function mutation or backcrossed to an apoE-/- background. At 28 days, aortas were harvested for RT-qPCR and histological analyses. CNP (0.2 mg/kg per day) was infused to rescue any adverse phenotype. RESULTS Aneurysmal tissue from patients with thoracic AA and abdominal AA revealed that CNP and NPR-C (natriuretic peptide receptor-C) expression were overtly perturbed. ecCNP-/-, fbCNP-/-, and gbCNP-/- mice exhibited an aggravated phenotype compared with wild-type mice in both ascending and suprarenal aortas, exemplified by greater dilation, fibrosis, elastin degradation, and macrophage infiltration. CNP and NPR-C expression was also dysregulated in murine thoracic AA and abdominal AA, accompanied by increased accumulation of mRNA encoding markers of inflammation, extracellular matrix remodeling/calcification, fibrosis, and apoptosis. CNP also prevented activation of isolated macrophages and vascular smooth muscle cells. An essentially identical phenotype was observed in NPR-C-/- mice and while administration of CNP protected against disease severity in wild-type animals, this phenotypic rescue was not apparent in NPR-C-/- mice. CONCLUSIONS Endothelium- and fibroblast-derived CNP, via NPR-C activation, plays important roles in attenuating AA formation by preserving aortic structure and function. Therapeutic strategies aimed at mimicking CNP bioactivity hold potential to reduce the need for surgical intervention.
Collapse
Affiliation(s)
- Aisah A Aubdool
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Amie J Moyes
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Cristina Pérez-Ternero
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Reshma S Baliga
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Jasspinder Sanghera
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - M Taaha Syed
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Kareemah Jaigirdah
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Anmolpreet K Panesar
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Janice C Tsui
- Division of Surgery and Interventional Science, University College London and Royal Free London NHS Foundation Trust, United Kingdom (J.C.T.)
| | - Yanming Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (Y.L., H.G.V., Y.H.S., S.A.L.)
| | - Hernan G Vasquez
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (Y.L., H.G.V., Y.H.S., S.A.L.)
| | - Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (Y.L., H.G.V., Y.H.S., S.A.L.)
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (Y.L., H.G.V., Y.H.S., S.A.L.)
- Geisinger Research Institute and Heart & Vascular Institute (S.A.L.)
| | | | - Ziad Mallat
- Division of Cardiovascular Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, United Kingdom (Z.M.)
| | - Hong S Lu
- Department of Physiology, Saha Cardiovascular Research Center, Saha Aortic Center, University of Kentucky, Lexington (H.S.L., A.D.)
| | - Alan Daugherty
- Department of Physiology, Saha Cardiovascular Research Center, Saha Aortic Center, University of Kentucky, Lexington (H.S.L., A.D.)
| | - Adrian J Hobbs
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| |
Collapse
|
7
|
Zhang Y, Guo J, Zhang N. Mendelian randomization study investigating the relationship between nutrients and polycystic ovary syndrome. Int J Gynaecol Obstet 2025; 169:198-205. [PMID: 39569763 DOI: 10.1002/ijgo.16029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/22/2024] [Accepted: 10/29/2024] [Indexed: 11/22/2024]
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS) affects many women and is linked to nutrient intake, but the causal role of nutrients is unclear. Our study used Mendelian randomization (MR) to explore this relationship. METHODS We employed two-sample MR to investigate the causal effects of four macronutrients (carbohydrates, fats, sugars, proteins) and 12 micronutrients (vitamin A [retinol], folate, vitamin B6, vitamin B12, vitamin C [ascorbic acid], vitamin D, vitamin E [alpha-tocopherol], magnesium, calcium, iron, selenium, and zinc) on the risk of PCOS based on their circulating levels. We conducted analyses using the inverse variance-weighted method (IVW), MR-Egger regression, weighted median method, and simple model approach, with IVW as the primary method. To correct for multiple testing, the false discovery rate (FDR) method was applied. The reliability and potential bias of the results were assessed by sensitivity analysis and F-statistics. RESULTS From a genetic standpoint, higher circulating vitamin A (retinol) levels were found to be a protective factor for PCOS and closely associated with the risk of PCOS, consistent with recent research findings. The inverse variance-weighted (IVW) estimation indicated a protective effect of vitamin A (retinol) on PCOS (IVWOR = 0.97, 95% CI: 0.95-0.99, P = 0.002, FDR_P = 0.03). However, no causal relationship was found between other nutrients and PCOS. In addition, MR-PRESSO and MR-Egger regression showed that our results were not affected by horizontal pleiotropy (P > 0.05). Finally, leave-one-out sensitivity analysis confirmed the robustness of MR Results. CONCLUSION Our findings suggest that circulating vitamin A (retinol) levels influence the risk of PCOS, but further research is needed.
Collapse
Affiliation(s)
- Yujie Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Jiawen Guo
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Ning Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| |
Collapse
|
8
|
Khalafi M, Habibi Maleki A, Symonds ME, Rosenkranz SK, Ehsanifar M, Mohammadi Dinani S. The combined effects of omega-3 polyunsaturated fatty acid supplementation and exercise training on body composition and cardiometabolic health in adults: A systematic review and meta-analysis. Clin Nutr ESPEN 2025; 66:151-159. [PMID: 39848543 DOI: 10.1016/j.clnesp.2025.01.022] [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: 04/30/2024] [Revised: 11/19/2024] [Accepted: 01/03/2025] [Indexed: 01/25/2025]
Abstract
INTRODUCTION We performed a systematic review and meta-analysis to investigate the effects of combining omega-3 polyunsaturated fatty acids (n-3 PUFAs) supplementation with exercise training, as compared to exercise training alone, on body composition measures including body weight, body mass index (BMI), fat mass, body fat percentage, and lean body mass. Additionally, we determined the effects on cardiometabolic health outcomes including lipid profiles, blood pressure, glycemic markers, and inflammatory markers. METHOD Three primary electronic databases including PubMed, Web of Science, and Scopus were searched from inception to April 5th, 2023 to identify original articles comparing n-3 PUFA supplementation plus exercise training versus exercise training alone, that investigated at least one of the following outcomes: fat mass, body fat percentage, lean body mass, triglycerides (TG), total cholesterol (TC), low-density lipoprotein (LDL), high-density lipoprotein (HDL), systolic (SBP) and diastolic (DBP) blood pressures, fasting glucose and insulin, interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α). Standardized mean differences (SMD) or weighted mean differences (WMD), and 95 % confidence intervals (CIs) were calculated using random-effects models. RESULTS A total of 21 studies involving 673 participants with BMIs ranging from 24 to 37 kg.m2 and ages ranging from 30 to 70 years were included in the meta-analysis. Overall, the results indicated that as compared with exercise training alone, adding omega-3 supplementation to exercise training decreased fat mass [WMD: -1.05 kg (95 % CI: -1.88 to -0.22), p = 0.01], TG [WMD: -0.10 mmol/L (95 % CI: -0.19 to -0.02)], SBP [WMD: -4.09 mmHg (95 % CI: -7.79 to -2.16), p = 0.03], DBP [WMD: -4.26 mmHg (95 % CI: -6.46 to -2.07), p = 0.001], and TNF-α [SMD: -0.35 (95 % CI: -0.70 to -0.00), p = 0.04], and increased LDL [WMD: 0.14 mmol/L (95 % CI: 0.02 to 0.26), p = 0.01] and lower-body muscular strength [SMD: 0.42 (95 % CI: 0.01 to 0.84), p = 0.04]. However, omega-3 supplementation with exercise training had no additional effects compared with training alone, for other body composition or cardiometabolic outcomes. CONCLUSION This systematic review and meta-analyses suggestes that adding omega-3 supplementation to exercise training may augment some effects of exercise training on body composition and cardiometabolic health in adults, although such effects appear to be modest.
Collapse
Affiliation(s)
- Mousa Khalafi
- Department of Sport Sciences, Faculty of Humanities, University of Kashan, Kashan, Iran.
| | - Aref Habibi Maleki
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Michael E Symonds
- Academic Unit of Population and Lifespan Sciences, Centre for Perinatal Research, School of Medicine, University of Nottingham, Nottingham, United Kingdom.
| | - Sara K Rosenkranz
- Department of Kinesiology and Nutrition Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA.
| | - Mahsa Ehsanifar
- Department of Exercise Physiology and Corrective Exercises, Faculty of Sport Sciences, Urmia University, Urmia, Iran.
| | - Sanaz Mohammadi Dinani
- Department of Sport Sciences, Faculty of Humanities, University of Kashan, Kashan, Iran.
| |
Collapse
|
9
|
Alexander MR, Edwards TL, Harrison DG. GWAS for Defining the Pathogenesis of Hypertension: Have They Delivered? Hypertension 2025; 82:573-582. [PMID: 39936322 PMCID: PMC11922662 DOI: 10.1161/hypertensionaha.124.23451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Genome-wide association studies have identified >3500 associated single nucleotide polymorphisms and over 1000 independent loci associated with hypertension. These individually have small effect sizes, and few associated loci have been experimentally tested for causal roles in hypertension using animal models or in humans. Thus, methods to prioritize and maximize the relevance of identified single nucleotide polymorphisms and associated loci are critical to determine their importance in hypertension. We propose several approaches to aid in these efforts, including: (1) integration of genome-wide association study data with multiomic data sets, including proteomics, transcriptomics, and epigenomics, (2) utilizing linked clinical and genetic data sets to determine genetic contributions to hypertension subphenotypes with distinct drivers, and (3) performing whole exome/genome sequencing on cohorts of individuals with severe hypertension to enrich for rare variants with larger effect sizes. Rather than creating longer lists of hypertension-associated single nucleotide polymorphisms, these approaches are needed to identify key mediators of hypertension pathophysiology.
Collapse
Affiliation(s)
- Matthew R Alexander
- Department of Medicine, Division of Clinical Pharmacology (M.R.A., D.G.H.), Vanderbilt University Medical Center, Nashville, TN
- Division of Cardiovascular Medicine (M.R.A., D.G.H.), Vanderbilt University Medical Center, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN (M.R.A., D.G.H.)
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN (M.R.A., D.G.H.)
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine (T.L.E.), Vanderbilt University Medical Center, Nashville, TN
| | - David G Harrison
- Department of Medicine, Division of Clinical Pharmacology (M.R.A., D.G.H.), Vanderbilt University Medical Center, Nashville, TN
- Division of Cardiovascular Medicine (M.R.A., D.G.H.), Vanderbilt University Medical Center, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN (M.R.A., D.G.H.)
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Nashville, TN (M.R.A., D.G.H.)
| |
Collapse
|
10
|
Hiie L, Kolde A, Pervjakova N, Reigo A, Abner E, Võsa U, Esko T, Fischer K, Palta P, Kronberg J. Pathway level metabolomics analysis identifies carbon metabolism as a key factor of incident hypertension in the Estonian Biobank. Sci Rep 2025; 15:8470. [PMID: 40069276 PMCID: PMC11897224 DOI: 10.1038/s41598-025-92840-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
The purpose of this study was to find metabolic changes associated with incident hypertension in the volunteer-based Estonian Biobank. We used a subcohort of the Estonian Biobank where metabolite levels had been measured by mass-spectrometry (LC-MS, Metabolon platform). We divided annotated metabolites of 989 individuals into KEGG pathways, followed by principal component analysis of metabolites in each pathway, resulting in a dataset of 91 pathway components. Next, we defined incident hypertension cases and controls based on electronic health records, resulting in a dataset of 101 incident hypertension cases and 450 controls. We used Cox proportional hazards models and replicated the results in a separate cohort of the Estonian Biobank, assayed with LC-MS dataset of the Broad platform and including 582 individuals. Our results show that body mass index and a component of the carbon metabolism KEGG pathway are associated with incident hypertension in both discovery and replication cohorts. We demonstrate that a high-dimensional dataset can be meaningfully reduced into informative pathway components that can subsequently be analysed in an interpretable way, and replicated in a metabolomics dataset from a different platform.
Collapse
Affiliation(s)
- Liis Hiie
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Riia 23b, 51010, Tartu, Estonia
- Institute of Mathematics and Statistics, University of Tartu, Narva 18, 51009, Tartu, Estonia
| | - Anastassia Kolde
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Riia 23b, 51010, Tartu, Estonia
- Institute of Mathematics and Statistics, University of Tartu, Narva 18, 51009, Tartu, Estonia
| | - Natalia Pervjakova
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Riia 23b, 51010, Tartu, Estonia
| | - Anu Reigo
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Riia 23b, 51010, Tartu, Estonia
| | - Erik Abner
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Riia 23b, 51010, Tartu, Estonia
| | - Urmo Võsa
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Riia 23b, 51010, Tartu, Estonia
| | - Tõnu Esko
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Riia 23b, 51010, Tartu, Estonia
| | - Krista Fischer
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Riia 23b, 51010, Tartu, Estonia
- Institute of Mathematics and Statistics, University of Tartu, Narva 18, 51009, Tartu, Estonia
| | - Priit Palta
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Riia 23b, 51010, Tartu, Estonia
| | - Jaanika Kronberg
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Riia 23b, 51010, Tartu, Estonia.
| |
Collapse
|
11
|
Deng M, Guo J, Li B, Yang J, Zhang X, Liang D, Wang Y, Huang X. Intelligent risk stratification of hypertension based on ambulatory blood pressure monitoring and machine learning algorithms. Physiol Meas 2025; 46:035001. [PMID: 40009995 DOI: 10.1088/1361-6579/adbab0] [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: 10/31/2024] [Accepted: 02/26/2025] [Indexed: 02/28/2025]
Abstract
Objective. Risk stratification of hypertension plays a crucial role in the treatment decisions and medication guidance during clinical practices. Although fruitful achievements have been reported on risk stratification of hypertension, the potential use of ambulatory blood pressure monitoring data is not well investigated. Different from single measuring blood pressure data, long-term blood pressure monitoring data can provide more comprehensive dynamical blood pressure information. Therefore, this paper proposes an intelligent hypertension risk stratification method based on ambulatory blood pressure monitoring data and improved machine learning algorithms.Approach. A total of 262 patients with hypertension are enrolled at People's Hospital of Yangjiang, in which 93 subjects are with simple hypertension and 169 subjects have hypertension with complication. Time-domain features, frequency-domain features, nonlinear dynamics features and correlation features underlying time-varying ambulatory blood pressure monitoring data are extracted to obtain discriminative feature representations. Synthetic minority over-sampling algorithm is applied to solve the problem of data balancing. The particle swarm optimization combined with kernel extreme learning machine is employed for feature fusion and optimization.Main results. The proposed method can yield a diagnostic accuracy of 93.7%, 97.8%, and 98.4% under two-, five- and ten-fold cross-validation, which demonstrates hypertension risk stratification in an intuitive, quantizable manner using multi-dimensional feature representation and learning.Significance. The proposed method is expected to provide early warning for latent serious cardiovascular diseases before obvious symptoms are present.
Collapse
Affiliation(s)
- Muqing Deng
- School of Automation, Guangdong University of Technology, Guangzhou, People's Republic of China
- Department of Cardiology and Center of Cardiovascular Disease, People's Hospital of Yangjiang, Yangjiang, People's Republic of China
| | - Junsheng Guo
- School of Automation, Guangdong University of Technology, Guangzhou, People's Republic of China
| | - Boyan Li
- School of Automation, Guangdong University of Technology, Guangzhou, People's Republic of China
| | - Jingfen Yang
- Department of Cardiology and Center of Cardiovascular Disease, People's Hospital of Yangjiang, Yangjiang, People's Republic of China
| | - Xiaobo Zhang
- School of Automation, Guangdong University of Technology, Guangzhou, People's Republic of China
| | - Dandan Liang
- Department of Cardiology and Center of Cardiovascular Disease, People's Hospital of Yangjiang, Yangjiang, People's Republic of China
| | - Yanjiao Wang
- School of Automation, Guangdong University of Technology, Guangzhou, People's Republic of China
| | - Xiaoyu Huang
- Department of Cardiology and Center of Cardiovascular Disease, People's Hospital of Yangjiang, Yangjiang, People's Republic of China
| |
Collapse
|
12
|
Ray A, Yang C, Stelloh C, Tutaj M, Liu P, Liu Y, Qiu Q, Auer PL, Lin CW, Widlansky ME, Geurts AM, Cowley AW, Liang M, Kwitek AE, Greene AS, Rao S. Chromatin State Maps of Blood Pressure-Relevant Renal Segments Reveal Potential Regulatory Role for SNPs. Hypertension 2025; 82:476-488. [PMID: 39723540 DOI: 10.1161/hypertensionaha.124.23873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Hypertension or elevated blood pressure (BP) is a worldwide clinical challenge and the leading primary risk factor for kidney dysfunctions, heart failure, and cerebrovascular disease. The kidney is a central regulator of BP by maintaining sodium-water balance. Multiple genome-wide association studies revealed that BP is a heritable quantitative trait, modulated by several genetic, epigenetic, and environmental factors. The SNPs identified in genome-wide association studies predominantly (>95%) reside within noncoding genomic regions, making it difficult to understand how they regulate BP. Given the central role of the kidney in regulating BP, we hypothesized that chromatin-accessible regions in renal tissue would be enriched for BP-associated single nucleotide polymorphisms. METHODS We manually dissected 2 important kidney segments that maintain the sodium-water balance: proximal tubules and medullary thick ascending limbs from the human and rat kidneys. To delineate their chromatin and transcriptomic profiles, we performed the assay for transposase-accessible chromatin and RNA sequencing, respectively. RESULTS The chromatin accessibility maps revealed the shared and unique cis-regulatory elements that modulate the chromatin accessibility in proximal tubule and medullary thick ascending limbs of humans and rats. We developed a visualization tool to compare the cross-species epigenomic maps to identify potential regulatory targets for hypertension pathogenesis. We also identified a significant enrichment of BP-associated single nucleotide polymorphisms (1064 for human proximal tubule and 1172 for human medullary thick ascending limbs) within accessible chromatin regions of both segments, including rs1173771 and rs1421811 at the NPR3 locus and rs1800470 at the TGFb1 locus. CONCLUSIONS Collectively, this study lays a foundation for interrogating how intergenic single nucleotide polymorphisms may regulate polygenic traits such as BP.
Collapse
Affiliation(s)
- Atrayee Ray
- Versiti Blood Research Institute, Milwaukee, WI (A.R., C.S., S.R.)
| | - Chun Yang
- Department of Physiology (C.Y., M.T., A.M.G., A.W.C., A.E.K.), Medical College of Wisconsin, Milwaukee
| | - Cary Stelloh
- Versiti Blood Research Institute, Milwaukee, WI (A.R., C.S., S.R.)
| | - Monika Tutaj
- Department of Physiology (C.Y., M.T., A.M.G., A.W.C., A.E.K.), Medical College of Wisconsin, Milwaukee
| | - Pengyuan Liu
- Department of Physiology, University of Arizona, Tucson (P.L., Y.L., Q.Q., M.L.)
| | - Yong Liu
- Department of Physiology, University of Arizona, Tucson (P.L., Y.L., Q.Q., M.L.)
| | - Qiongzi Qiu
- Department of Physiology, University of Arizona, Tucson (P.L., Y.L., Q.Q., M.L.)
| | - Paul L Auer
- The Institute for Health and Equity (P.L.A.), Medical College of Wisconsin, Milwaukee
| | - Chien-Wei Lin
- Division of Biostatistics, Data Science Institute (C.-W.L.), Medical College of Wisconsin, Milwaukee
| | | | - Aron M Geurts
- Department of Physiology (C.Y., M.T., A.M.G., A.W.C., A.E.K.), Medical College of Wisconsin, Milwaukee
| | - Allen W Cowley
- Department of Physiology (C.Y., M.T., A.M.G., A.W.C., A.E.K.), Medical College of Wisconsin, Milwaukee
| | - Mingyu Liang
- Department of Physiology, University of Arizona, Tucson (P.L., Y.L., Q.Q., M.L.)
| | - Anne E Kwitek
- Department of Physiology (C.Y., M.T., A.M.G., A.W.C., A.E.K.), Medical College of Wisconsin, Milwaukee
| | | | - Sridhar Rao
- Versiti Blood Research Institute, Milwaukee, WI (A.R., C.S., S.R.)
- Department of Pediatrics, Section of Hematology/Oncology/Transplantation (S.R.), Medical College of Wisconsin, Milwaukee
- Department of Cell Biology, Neurobiology, and Anatomy (S.R.), Medical College of Wisconsin, Milwaukee
| |
Collapse
|
13
|
Holzbach LC, Brandão-Lima PN, Duarte GBS, Rogero MM, Cominetti C. Nutrigenetics and Nutritional Strategies in Systemic Arterial Hypertension: Evidence From a Scoping Review. Nutr Rev 2025; 83:539-550. [PMID: 39164015 DOI: 10.1093/nutrit/nuae112] [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] [Indexed: 08/22/2024] Open
Abstract
Nutrition and genetics have individual roles in systemic arterial hypertension (SAH); however, they can interact, influencing the regulation of blood pressure (BP) levels. The aim of this study was to evaluate the available evidence regarding gene-nutrient interactions in modulating BP levels in adults with SAH. The review followed the recommendations of the Joanna Briggs Institute and the Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews. Twelve studies met the inclusion criteria for this review, reporting on 20 genes and 31 single nucleotide polymorphisms (SNPs), with 19 of them associated with BP variations. The most frequently evaluated SNPs were ACE rs4646994 and AT1R rs5186. Among the nutritional interventions, dietary sodium content was the focus of most studies (n = 11). Interactions with sodium consumption were observed for the following SNPs: KDM1A rs587168, EDNRB rs5351, LSS rs2254524, IRS1 rs1801278, KCNK9 rs6997709, ACE rs4646994, GNB3 rs5443, PPARG rs4684847, EDN1 rs5370, BCAT1 rs7961152, IL18 rs5744292, NOS3 rs2070744, and AT1R rs5186. In the presence of a diet rich in fruits and vegetables, moderate alcohol consumption, and reduced sodium intake, the SNP AT2R rs11091046 was associated with a decrease in BP levels. Furthermore, the SNP MTHFR rs1801133 exhibited an interaction with riboflavin supplementation in affecting BP levels. The evidence regarding the interaction between genetics and diet on BP levels remains limited. Among the existing findings, an interaction was observed between sodium, calcium, riboflavin, and specific polymorphisms; however, the underlying mechanisms for these interactions have yet to be identified. Note: This paper is part of the Nutrition Reviews Special Collection on Precision Nutrition.
Collapse
Affiliation(s)
- Luciana C Holzbach
- Nutritional Genomics Research Group, Graduate Program in Nutrition and Health, School of Nutrition, Federal University of Goiás, Goiânia 74605080, Brazil
- Nutrition Undergraduate Course, Federal University of Tocantins, Palmas 77001-090, Brazil
| | - Paula N Brandão-Lima
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo 01246-904, Brazil
| | - Graziela B S Duarte
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo 01246-904, Brazil
| | - Marcelo M Rogero
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo 01246-904, Brazil
| | - Cristiane Cominetti
- Nutritional Genomics Research Group, Graduate Program in Nutrition and Health, School of Nutrition, Federal University of Goiás, Goiânia 74605080, Brazil
| |
Collapse
|
14
|
Naderi H, Warren HR, Munroe PB. Harnessing the power of genomics in hypertension: tip of the iceberg? CAMBRIDGE PRISMS. PRECISION MEDICINE 2025; 3:e2. [PMID: 40071139 PMCID: PMC11894416 DOI: 10.1017/pcm.2025.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 12/18/2024] [Accepted: 01/09/2025] [Indexed: 03/14/2025]
Abstract
Despite the blaze of advancing knowledge on its complex genetic architecture, hypertension remains an elusive condition. Genetic studies of blood pressure have yielded bitter-sweet results thus far with the identification of more than 2,000 genetic loci, though the candidate causal genes and biological pathways remain largely unknown. The era of big data and sophisticated statistical tools has propelled insights into pathophysiology and causal inferences. However, new genetic risk tools for hypertension are the tip of the iceberg, and applications of genomic technology are likely to proliferate. We review the genomics of hypertension, exploring the significant milestones in our current understanding of this condition and the progress towards personalised treatment and management for hypertension.
Collapse
Affiliation(s)
- Hafiz Naderi
- William Harvey Research Institute, Queen Mary University of London, London, UK
- Barts Heart Centre, St Bartholomew’s Hospital, West Smithfield, London, UK
- National Institute of Health and Care Research Barts Biomedical Research Centre, Queen Mary University of London, London, UK
| | - Helen R. Warren
- William Harvey Research Institute, Queen Mary University of London, London, UK
- National Institute of Health and Care Research Barts Biomedical Research Centre, Queen Mary University of London, London, UK
| | - Patricia B. Munroe
- William Harvey Research Institute, Queen Mary University of London, London, UK
- National Institute of Health and Care Research Barts Biomedical Research Centre, Queen Mary University of London, London, UK
| |
Collapse
|
15
|
Chandler HL, Wheeler J, Escott‐Price V, Murphy K, Lancaster TM. Non-APOE variants predominately expressed in smooth muscle cells contribute to the influence of Alzheimer's disease genetic risk on white matter hyperintensities. Alzheimers Dement 2025; 21:e14455. [PMID: 39737667 PMCID: PMC11848156 DOI: 10.1002/alz.14455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 01/01/2025]
Abstract
INTRODUCTION White matter hyperintensity volumes (WMHVs) are disproportionally prevalent in individuals with Alzheimer's disease (AD), potentially reflecting neurovascular injury. We quantify the association between AD polygenic risk score (AD-PRS) and WMHV, exploring single-nucleotide polymorphisms (SNPs) that are proximal to genes overexpressed in cerebrovascular cell species. METHODS In a UK-Biobank sub-sample (mean age = 64, range = 45-81 years), we associate WMHV with (1) AD-PRS estimated via SNPs across the genome (minus apolipoprotein E [APOE] locus) and (2) AD-PRS estimated with SNPs proximal to specific genes that are overexpressed in cerebrovascular cell species. RESULTS We observed a positive association between non-APOE-AD-PRS and WMHVs. We further demonstrate an association between WMHVs and AD-PRS constructed with SNPs that are proximal to genes over-represented in smooth muscles cells (SMCs; β = 0.135, PFWE < 0.01) and internally replicated (PDISCOVERY+REPLICATION < 0.01). DISCUSSION Common AD genetic risk could explain physiological processes underlying vascular pathology in AD. SMC function may offer a treatment target to prevent WMHV-related AD pathophysiology prior to the onset of symptoms. HIGHLIGHTS Alzheimer's disease (AD) risk factors such as apolipoprotein E (APOE) ε4, link to increased white matter hyperintensity volume (WMHV). WMHVs indicate vascular risk and neurovascular injury in AD. The broader genetic link between AD risk and WMHV is not fully understood. We quantify AD polygenic risk score (PRS) associations with WMHV, excluding APOE. AD-PRS in smooth muscle cells (SMCs) shows a significant association with increased WMHV.
Collapse
Affiliation(s)
- Hannah Louise Chandler
- School of Physics and AstronomyCardiff University Brain Research Imaging Centre (CUBRIC)Cardiff UniversityCardiffUK
| | - Joshua Wheeler
- School of Clinical SciencesUniversity of BristolBristolUK
- Department of PsychologyUniversity of BathBathUK
| | - Valentina Escott‐Price
- Centre for Neuropsychiatric Genetics and GenomicsDepartment of Psychological Medicine and Clinical NeurosciencesCardiff UniversityCardiffUK
| | - Kevin Murphy
- School of Physics and AstronomyCardiff University Brain Research Imaging Centre (CUBRIC)Cardiff UniversityCardiffUK
| | | |
Collapse
|
16
|
Li H, Zeng J, Snyder MP, Zhang S. Modeling gene interactions in polygenic prediction via geometric deep learning. Genome Res 2025; 35:178-187. [PMID: 39562137 PMCID: PMC11789630 DOI: 10.1101/gr.279694.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
Polygenic risk score (PRS) is a widely used approach for predicting individuals' genetic risk of complex diseases, playing a pivotal role in advancing precision medicine. Traditional PRS methods, predominantly following a linear structure, often fall short in capturing the intricate relationships between genotype and phenotype. In this study, we present PRS-Net, an interpretable geometric deep learning-based framework that effectively models the nonlinearity of biological systems for enhanced disease prediction and biological discovery. PRS-Net begins by deconvoluting the genome-wide PRS at the single-gene resolution and then explicitly encapsulates gene-gene interactions leveraging a graph neural network (GNN) for genetic risk prediction, enabling a systematic characterization of molecular interplay underpinning diseases. An attentive readout module is introduced to facilitate model interpretation. Extensive tests across multiple complex traits and diseases demonstrate the superior prediction performance of PRS-Net compared with a wide range of conventional PRS methods. The interpretability of PRS-Net further enhances the identification of disease-relevant genes and gene programs. PRS-Net provides a potent tool for concurrent genetic risk prediction and biological discovery for complex diseases.
Collapse
Affiliation(s)
- Han Li
- School of Mathematical Sciences and LPMC, Nankai University, Tianjin, 300071, China
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, 100084, China
| | - Jianyang Zeng
- School of Engineering, Research Center for Industries of the Future, Westlake University, Hangzhou, 310030, Zhejiang, China;
| | - Michael P Snyder
- Department of Genetics, Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, California 94304, USA;
| | - Sai Zhang
- Department of Epidemiology, University of Florida, Gainesville, Florida 32603, USA;
- Departments of Biostatistics & Biomedical Engineering, UF Genetics Institute, University of Florida, Gainesville, Florida 32603, USA
| |
Collapse
|
17
|
Huang JX, Xu SZ, Tian T, Wang J, Jiang LQ, He T, Meng SY, Ni J, Pan HF. Genetic Links Between Metabolic Syndrome and Osteoarthritis: Insights From Cross-Trait Analysis. J Clin Endocrinol Metab 2025; 110:e461-e469. [PMID: 38482593 DOI: 10.1210/clinem/dgae169] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Indexed: 01/22/2025]
Abstract
CONTEXT Previous observational studies have indicated a bidirectional association between metabolic syndrome (MetS) and osteoarthritis (OA). However, it remains unclear whether these bidirectional associations reflect causal relationships or shared genetic factors, and the underlying biological mechanisms of this association are not fully understood. OBJECTIVE We aimed to explore the genetic connection between MetS and OA using genome-wide association study (GWAS) summary data. METHODS Leveraging summary statistics from GWAS conducted by the UK Biobank and the Glucose and Insulin-related Traits Consortium (MAGIC), we performed global genetic correlation analyses, genome-wide cross-trait meta-analyses, and a bidirectional two-sample Mendelian randomization analyses using summary statistics from GWAS to comprehensively assess the relationship of MetS and OA. RESULTS We first detected an extensive genetic correlation between MetS and OA (rg = 0.393, P = 1.52 × 10-18), which was consistent in 4 MetS components, including waist circumference, triglycerides, hypertension, and high-density lipoprotein cholesterol and OA with rg ranging from -0.229 to 0.490. We then discovered 32 variants jointly associated with MetS and OA through Multi-Trait Analysis of GWAS (MTAG). Co-localization analysis found 12 genes shared between MetS and OA, with functional implications in several biological pathways. Finally, Mendelian randomization analysis suggested genetic liability to MetS significantly increased the risk of OA, but no reverse causality was found. CONCLUSION Our results illustrate a common genetic architecture, pleiotropic loci, as well as causality between MetS and OA, potentially enhancing our knowledge of high comorbidity and genetic processes that overlap between the 2 disorders.
Collapse
Affiliation(s)
- Ji-Xiang Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Shu-Zhen Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Tian Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Jing Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Ling-Qiong Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Tian He
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Shi-Yin Meng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Jing Ni
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| |
Collapse
|
18
|
Lehners M, Schmidt H, Zaldivia MTK, Stehle D, Krämer M, Peter A, Adler J, Lukowski R, Feil S, Feil R. Single-cell analysis identifies the CNP/GC-B/cGMP axis as marker and regulator of modulated VSMCs in atherosclerosis. Nat Commun 2025; 16:429. [PMID: 39814746 PMCID: PMC11735800 DOI: 10.1038/s41467-024-55687-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 12/20/2024] [Indexed: 01/18/2025] Open
Abstract
A balanced activity of cGMP signaling contributes to the maintenance of cardiovascular homeostasis. Vascular smooth muscle cells (VSMCs) can generate cGMP via three ligand-activated guanylyl cyclases, the NO-sensitive guanylyl cyclase, the atrial natriuretic peptide (ANP)-activated GC-A, and the C-type natriuretic peptide (CNP)-stimulated GC-B. Here, we study natriuretic peptide signaling in murine VSMCs and atherosclerotic lesions. Correlative profiling of pathway activity and VSMC phenotype at the single-cell level shows that phenotypic modulation of contractile VSMCs to chondrocyte-like plaque cells during atherogenesis is associated with a switch from ANP/GC‑A to CNP/GC‑B signaling. Silencing of the CNP/GC-B axis in VSMCs results in an increase of chondrocyte-like plaque cells. These findings indicate that the CNP/GC-B/cGMP pathway is a marker and atheroprotective regulator of modulated VSMCs, limiting their transition to chondrocyte-like cells. Overall, this study highlights the plasticity of cGMP signaling in VSMCs and suggests analogies between CNP-dependent remodeling of bone and blood vessels.
Collapse
MESH Headings
- Animals
- Cyclic GMP/metabolism
- Natriuretic Peptide, C-Type/metabolism
- Natriuretic Peptide, C-Type/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/cytology
- Signal Transduction
- Mice
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Receptors, Atrial Natriuretic Factor/metabolism
- Receptors, Atrial Natriuretic Factor/genetics
- Single-Cell Analysis
- Male
- Mice, Inbred C57BL
- Biomarkers/metabolism
Collapse
Affiliation(s)
- Moritz Lehners
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Hannes Schmidt
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Maria T K Zaldivia
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Daniel Stehle
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Michael Krämer
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Andreas Peter
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany
| | - Julia Adler
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Susanne Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Robert Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
19
|
Gurholt TP, Elvsåshagen T, Bahrami S, Rahman Z, Shadrin A, Askeland-Gjerde DE, van der Meer D, Frei O, Kaufmann T, Sønderby IE, Halvorsen S, Westlye LT, Andreassen OA. Large-scale brainstem neuroimaging and genetic analyses provide new insights into the neuronal mechanisms of hypertension. HGG ADVANCES 2025; 6:100392. [PMID: 39663699 PMCID: PMC11731578 DOI: 10.1016/j.xhgg.2024.100392] [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: 10/25/2023] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024] Open
Abstract
While brainstem regions are central regulators of blood pressure, the neuronal mechanisms underlying their role in hypertension remain poorly understood. Here, we investigated the structural and genetic relationships between global and regional brainstem volumes and blood pressure. We used magnetic resonance imaging data from n = 32,666 UK Biobank participants, and assessed the association of volumes of the whole brainstem and its main regions with blood pressure. We applied powerful statistical genetic tools, including bivariate causal mixture modeling (MiXeR) and conjunctional false discovery rate (conjFDR), to non-overlapping genome-wide association studies of brainstem volumes (n = 27,034) and blood pressure (n = 321,843) in the UK Biobank cohort. We observed negative associations between the whole brainstem and medulla oblongata volumes and systolic blood and pulse pressure, and positive relationships between midbrain and pons volumes and blood pressure traits when adjusting for the whole brainstem volume (all partial correlation coefficients ∣r∣ effects between 0.03 and 0.05, p ≤ 0.0042). We observed the largest genetic overlap for the whole brainstem, sharing 77% of its trait-influencing variants with blood pressure. We identified 65 shared loci between brainstem volumes and blood pressure traits and mapped these to 71 genes, implicating molecular pathways linked to sympathetic nervous system development, metal ion transport, and vascular homeostasis. The present findings support a link between brainstem structures and blood pressure and provide insights into their shared genetic underpinnings. The overlapping genetic architectures and mapped genes offer mechanistic information about the roles of brainstem regions in hypertension.
Collapse
Affiliation(s)
- Tiril P Gurholt
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Section for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, 0424 Oslo, Norway.
| | - Torbjørn Elvsåshagen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Section for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, 0424 Oslo, Norway; Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; Department of Neurology, Oslo University Hospital, Oslo, Norway; Department of Behavioural Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway
| | - Shahram Bahrami
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Center for Precision Psychiatry, Division of Mental Health and Addiction, Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway
| | - Zillur Rahman
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Center for Precision Psychiatry, Division of Mental Health and Addiction, Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway
| | - Alexey Shadrin
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Center for Precision Psychiatry, Division of Mental Health and Addiction, Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway
| | - Daniel E Askeland-Gjerde
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Section for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, 0424 Oslo, Norway
| | - Dennis van der Meer
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Center for Precision Psychiatry, Division of Mental Health and Addiction, Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway; School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht 6200 MD, the Netherlands
| | - Oleksandr Frei
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Center for Precision Psychiatry, Division of Mental Health and Addiction, Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway
| | - Tobias Kaufmann
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany; German Center for Mental Health (DZPG), Partner Site Tübingen, Tübingen, Germany
| | - Ida E Sønderby
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, 0424 Oslo, Norway
| | - Sigrun Halvorsen
- Department of Cardiology, Oslo University Hospital Ullevål and University of Oslo, 0424 Oslo, Norway
| | - Lars T Westlye
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Department of Psychology, University of Oslo, 0373 Oslo, Norway
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway; Section for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, 0424 Oslo, Norway; Center for Precision Psychiatry, Division of Mental Health and Addiction, Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway
| |
Collapse
|
20
|
Kim S, Ko H, Myung W, Yoon J, Kim K, Jung SH, Shim I, Cha S, Kim B, Kang JM, Park WY, Natarajan P, Do R, Won HH. Association between genetically predicted leisure and social activities and cardiovascular disease and other health outcomes. NATURE CARDIOVASCULAR RESEARCH 2025; 4:15-25. [PMID: 39747453 DOI: 10.1038/s44161-024-00581-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 11/01/2024] [Indexed: 01/04/2025]
Abstract
Participation in leisure and social activities (LSA) is associated with better health outcomes and lower mortality1-3. Previous observational studies demonstrated a relationship between engagement in LSA and both mental and physical health4,5. Although several studies6 examined the association between LSA and health outcomes, including cardiovascular disease, their possible causal relationship has not been studied. In this study, we investigated the causal relationship between LSA and various health outcomes, including cardiovascular disease, using data from genome-wide association study summary statistics (sample size = 63,926-1,557,411) and a Mendelian randomization approach. Genetically predicted LSA were associated with a reduced risk of several health outcomes, including coronary artery disease and coronary atherosclerosis. Mediation analysis indicated that these effects were partly mediated by modifiable risk factors, such as body mass index, smoking and lipid levels. These findings highlight the importance of LSA in disease prevention and health promotion.
Collapse
Affiliation(s)
- Soyeon Kim
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hyunwoong Ko
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Woojae Myung
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea.
- Department of Psychiatry, Seoul National University, College of Medicine, Seoul, South Korea.
| | - Joohyun Yoon
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Kiwon Kim
- Department of Psychiatry, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, South Korea
| | - Sang-Hyuk Jung
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Injeong Shim
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Soojin Cha
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Beomsu Kim
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Jae Myeong Kang
- Department of Psychiatry, Gil Medical Center, Gachon University, Incheon, South Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Pradeep Natarajan
- Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Ron Do
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hong-Hee Won
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea.
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| |
Collapse
|
21
|
He Z, IJpma AS, Vreeken D, Heijsman D, Rosier K, Verhagen HJM, de Bruin JL, Brüggenwirth HT, Roos-Hesselink JW, Bekkers JA, Huylebroeck DFE, van Beusekom HMM, Creemers JWM, Majoor-Krakauer D. The proprotein convertase FURIN is a novel aneurysm predisposition gene impairing TGF-β signalling. Cardiovasc Res 2024; 120:2278-2292. [PMID: 38636100 PMCID: PMC11687399 DOI: 10.1093/cvr/cvae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 01/18/2024] [Accepted: 02/25/2024] [Indexed: 04/20/2024] Open
Abstract
AIMS Aortic aneurysms (AA) frequently involve dysregulation of transforming growth factor β (TGF-β)-signalling in the aorta. Here, FURIN was tested as aneurysm predisposition gene given its role as proprotein convertase in pro-TGF-β maturation. METHODS AND RESULTS Rare FURIN variants were detected by whole-exome sequencing of 781 unrelated aortic aneurysm patients and affected relatives. Thirteen rare heterozygous FURIN variants occurred in 3.7% (29) unrelated index AA patients, of which 72% had multiple aneurysms or a dissection. FURIN maturation and activity of these variants were decreased in vitro. Patient-derived fibroblasts showed decreased pro-TGF-β processing, phosphorylation of downstream effector SMAD2 and kinases ERK1/2, and steady-state mRNA levels of the TGF-β-responsive ACTA2 gene. In aortic tissue, collagen and fibrillin fibres were affected. One variant (R745Q), observed in 10 unrelated cases, affected TGF-β signalling variably, indicating effect modification by individual genetic backgrounds. CONCLUSION FURIN is a novel, frequent genetic predisposition for abdominal-, thoracic-, and multiple aortic or middle sized artery aneurysms in older patients, by affecting intracellular TGF-β signalling, depending on individual genetic backgrounds.
Collapse
Affiliation(s)
- Zongsheng He
- Laboratory of Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Gasthuisberg O/N 06, Herestraat 49, Box 607, Leuven B-3000, Belgium
| | - Arne S IJpma
- Department of Pathology, Erasmus MC University Medical Center, Dr. Molewater 40, PO BOX 2040, Rotterdam 3000 CA, The Netherlands
| | - Dianne Vreeken
- Department of Cardiology, Erasmus MC University Medical Center, Dr. Molewaterplein 40, PO BOX 2040, Rotterdam 3015 GD, The Netherlands
| | - Daphne Heijsman
- Department of Clinical Genetics, Erasmus MC University Medical Center, Dr Molewaterplein 40 PO BOX 2040, 3000CA Rotterdam, The Netherlands
| | - Karen Rosier
- Laboratory of Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Gasthuisberg O/N 06, Herestraat 49, Box 607, Leuven B-3000, Belgium
| | - Hence J M Verhagen
- Department of Surgery, Erasmus MC University Medical Center, Dr. Molewaterplein 40, PO BOX 2040, 3000 CA Rotterdam, The Netherlands
| | - Jorg L de Bruin
- Department of Surgery, Erasmus MC University Medical Center, Dr. Molewaterplein 40, PO BOX 2040, 3000 CA Rotterdam, The Netherlands
| | - Hennie T Brüggenwirth
- Department of Clinical Genetics, Erasmus MC University Medical Center, Dr Molewaterplein 40 PO BOX 2040, 3000CA Rotterdam, The Netherlands
| | - Jolien W Roos-Hesselink
- Department of Cardiology, Erasmus MC University Medical Center, Dr. Molewaterplein 40, PO BOX 2040, Rotterdam 3015 GD, The Netherlands
| | - Jos A Bekkers
- Department of Cardiothoracic Surgery, Erasmus MC University Medical Center, Dr. Molewaterplein 40, PO BOX 2040, 3000 CA Rotterdam, The Netherlands
| | - Danny F E Huylebroeck
- Department of Cell Biology, Erasmus MC University Medical Center, Dr. Molewaterplein 40, PO BOX 2040, 3000 CA Rotterdam, The Netherlands
| | - Heleen M M van Beusekom
- Department of Cardiology, Erasmus MC University Medical Center, Dr. Molewaterplein 40, PO BOX 2040, Rotterdam 3015 GD, The Netherlands
| | - John W M Creemers
- Laboratory of Biochemical Neuroendocrinology, Department of Human Genetics, KU Leuven, Gasthuisberg O/N 06, Herestraat 49, Box 607, Leuven B-3000, Belgium
| | - Danielle Majoor-Krakauer
- Department of Clinical Genetics, Erasmus MC University Medical Center, Dr Molewaterplein 40 PO BOX 2040, 3000CA Rotterdam, The Netherlands
| |
Collapse
|
22
|
Xu Y, Wang X, Wang G, Wei W, Li N. Relationship between hypothyroidism and chronic kidney disease: Results from the National Health and Nutrition Examination Survey 2007 to 2012 and Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40925. [PMID: 39705485 PMCID: PMC11666227 DOI: 10.1097/md.0000000000040925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 12/22/2024] Open
Abstract
Chronic kidney disease (CKD) and hypothyroidism are prevalent chronic conditions with a generally believed correlation between them. However, large-scale population studies and investigations into causation are lacking. This study analyzed CKD and thyroid function data from the National Health and Nutrition Examination Survey database spanning 2007 to 2012 using multiple regression analyses to examine the correlation between CKD and hypothyroidism. Bidirectional Mendelian randomization analysis was employed to investigate the causal association between the two conditions. As CKD stages deteriorated, there was a significant decrease in total triiodothyronine (TT3) and free triiodothyronine (P < .05). However, no significant decrease was observed in total thyroxine and free thyroxine. Notably, there was a significant increase in thyroid-stimulating hormone levels (P < .05). However, no significant changes were observed in thyroglobulin, thyroglobulin antibody, and thyroid peroxidase antibody levels. A causal relationship between CKD and reduced thyroid function was observed (odds ratio [OR] = 1.0041, 95% confidence interval [CI]: 1.0007-1.0075, P = .0186). Conversely, reverse causality was not statistically significant (OR = 2.540, 95%CI: 0.8680-4.8603, P = .1014). As CKD progressed, the risk of hypothyroidism increased. A causal correlation was observed between CKD and reduced thyroid function. Chronic kidney disease (CKD) and hypothyroidism are prevalent chronic conditions with a generally believed correlation between them. There is no large-scale population studies and the causation relationship between CKD and hypothyroidism are lacking. The finding of the causal relationship between CKD and hypothyroidism may be beneficial to the prevention of the disease and the prognosis of the patients.
Collapse
Affiliation(s)
- Yin Xu
- Department of Gerontology, The 960th Hospital of PLA, Jinan, China
| | - Xinmei Wang
- Beijing Academy of Military Medical Sciences, China
| | - Guofeng Wang
- Department of Gerontology, The 960th Hospital of PLA, Jinan, China
| | - Wei Wei
- Department of Gerontology, The 960th Hospital of PLA, Jinan, China
| | - Ning Li
- Department of Gerontology, The 960th Hospital of PLA, Jinan, China
| |
Collapse
|
23
|
Ahmed W, Dixit P, Halli S. Additive interaction of family medical history of cardiovascular diseases with hypertension and diabetes on the diagnosis of cardiovascular diseases among older adults in India. Front Cardiovasc Med 2024; 11:1386378. [PMID: 39713213 PMCID: PMC11659756 DOI: 10.3389/fcvm.2024.1386378] [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: 02/19/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Introduction The present study aimed to examine the additive interaction of family medical history of cardiovascular disease (CVD) and self-reported hypertension and diabetes on the diagnosis of CVD among older adults aged 45 years and above in India. A family medical history of CVD in individuals with hypertension and diabetes could identify a subpopulation with a higher risk of CVD. Methods The study used the data from the Longitudinal Ageing Study in India (LASI) Wave 1 (2017-2018). The total sample size for the study was 58,734 older adults aged 45 years and above. An additive model was applied to determine the additive interaction effect of the family medical history of CVD with hypertension and diabetes on the diagnosis of CVD by calculating three different measures of additive interaction: the relative excess risk due to interaction (RERI), attribution proportion due to interaction (AP), and synergy index (S). Results The prevalence of CVD was higher among hypertensive individuals with a family medical history of CVD (18.6%) than individuals without the coexistence of family medical history of CVD and hypertension (4.7%), and hypertensive individuals without family medical history of CVD (11.3%). On the other hand, the prevalence of CVD was higher among individuals with diabetes and family history of CVD (20.5%) than individuals without the coexistence of family history of CVD and diabetes (5.0%). Individuals with parental and sibling medical history had two times higher odds of having chronic heart diseases and strokes, respectively than those without parental and sibling history. In the adjusted model, RERI, AP, and S for CVD were 2.30 (95% CI: 0.87-3.74), 35% (0.35; 95% CI: 0.20-0.51), and 1.71 (95% CI: 1.27-2.28) respectively, demonstrating significant positive interaction between family medical history and hypertension on the diagnosis of cardiovascular diseases. Conclusions The present study revealed that in the additive model, the interaction effects of family medical history and hypertension were significantly positive on cardiovascular diseases even after adjustment with potential confounding factors. Therefore, it is crucial to consider the presence of family medical history of CVD among individuals with hypertension and diabetes measured in research and clinical practice.
Collapse
Affiliation(s)
- Waquar Ahmed
- School of Health Systems Studies, Tata Institute of Social Sciences, Mumbai, India
| | - Priyanka Dixit
- School of Health Systems Studies, Tata Institute of Social Sciences, Mumbai, India
| | - Shiva Halli
- Department of Community Health Sciences, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
24
|
Baldwin J, Burnier M, Ponte B, Ackermann D, Pruijm M, Vogt B, Bochud M. Association of mid-regional pro-adrenomedullin with office and 24-h ambulatory blood pressure in a Swiss general population sample. J Hypertens 2024; 42:2187-2195. [PMID: 39469923 PMCID: PMC11556881 DOI: 10.1097/hjh.0000000000003866] [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: 04/08/2024] [Revised: 07/20/2024] [Accepted: 08/23/2024] [Indexed: 10/30/2024]
Abstract
OBJECTIVE Adrenomedullin (ADM) is a potent vasodilator. The association between plasma ADM levels and blood pressure (BP) remains unclear. We assessed the association between mid-regional-pro-ADM (MR-proADM) and BP in a multicenter population- and family-based cohort. METHODS We used data from the Swiss Kidney Project on Genes in Hypertension (SKIPOGH). We included participants present at both baseline and 3-year follow-up (N = 843). We examined the association of baseline MR-proADM with baseline office and 24 h ambulatory BP as well as the 3-year change in office BP. In secondary analyses, we studied the association between baseline MR-proADM and 3-year changes in pulse wave velocity (PWV), renal resistive index (RRI) and augmentation index (AI). Mixed-effects linear regression models were used. RESULTS In cross-sectional analyses, MR-proADM was negatively associated with office, 24-h and daytime diastolic BP (DBP). MR-proADM was positively associated with nighttime systolic BP (SBP). In longitudinal analyses, baseline MR-proADM was associated with an increase in office SBP and pulse pressure (PP) over 3 years [β (95% CI): 8.2 (0.4, 15.9) and β (95% CI): 6.4 (0.3, 12.4), respectively] but not with changes in PWV, RRI and AI. CONCLUSIONS The cross-sectional negative association of MR-proADM with DBP is in line with known vasodilatory properties of ADM. The positive association between MR-proADM and nighttime SBP at baseline may reflect endothelial dysfunction believed to be part of the pathogenesis of nocturnal hypertension. The association of higher baseline MR-proADM levels with increased SBP and PP at 3-year follow-up suggests that ADM levels could be a marker of cardiovascular risk.
Collapse
Affiliation(s)
- Julia Baldwin
- Department of Epidemiology and Health Systems, Unisanté
| | | | - Belen Ponte
- Department of Nephrology, University Hospital of Geneva (HUG)
| | - Daniel Ackermann
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital and University of Bern
| | - Menno Pruijm
- Service of Nephrology and Hypertension, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Switzerland
| | - Bruno Vogt
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital and University of Bern
| | | |
Collapse
|
25
|
Rudman-Melnick V, Vanhoutte D, Stowers K, Sargent M, Adam M, Ma Q, Perl AKT, Miethke AG, Burg A, Shi T, Hildeman DA, Woodle ESS, Kofron JM, Devarajan P. Gucy1α1 specifically marks kidney, heart, lung and liver fibroblasts. Sci Rep 2024; 14:29307. [PMID: 39592775 PMCID: PMC11599588 DOI: 10.1038/s41598-024-80930-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/22/2024] [Indexed: 11/28/2024] Open
Abstract
Fibrosis is a common outcome of numerous pathologies, including chronic kidney disease (CKD), a progressive renal function deterioration. Current approaches to target activated fibroblasts, key effector contributors to fibrotic tissue remodeling, lack specificity. Here, we report Gucy1α1 as a specific kidney fibroblast marker. Gucy1α1 levels significantly increased over the course of two clinically relevant murine CKD models and directly correlated with established fibrosis markers. Immunofluorescent (IF) imaging showed that Gucy1α1 comprehensively labelled cortical and medullary quiescent and activated fibroblasts in the control kidney and throughout injury progression, respectively. Unlike traditionally used markers platelet derived growth factor receptor beta (Pdgfrβ) and vimentin (Vim), Gucy1α1 did not overlap with off-target populations such as podocytes. Notably, Gucy1α1 labelled kidney fibroblasts in both male and female mice. Furthermore, we observed elevated GUCY1α1 expression in the human fibrotic kidney and lung. Studies in the murine models of cardiac and liver fibrosis revealed Gucy1α1 elevation in activated Pdgfrβ-, Vim- and alpha smooth muscle actin (αSma)-expressing fibroblasts paralleling injury progression and resolution. Overall, we demonstrate Gucy1α1 as an exclusive fibroblast marker in both sexes. Due to its multiorgan translational potential, GUCY1α1 might provide a novel promising strategy to specifically target and mechanistically examine fibroblasts.
Collapse
Affiliation(s)
- Valeria Rudman-Melnick
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 268-280 Albert Sabin Way, location T, floor 6, suite 272, Cincinnati, OH, 45229, USA
| | - Davy Vanhoutte
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kaitlynn Stowers
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 268-280 Albert Sabin Way, location T, floor 6, suite 272, Cincinnati, OH, 45229, USA
| | - Michelle Sargent
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Mike Adam
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Qing Ma
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 268-280 Albert Sabin Way, location T, floor 6, suite 272, Cincinnati, OH, 45229, USA
| | - Anne Karina T Perl
- Division of Neonatology and Pulmonary biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Alexander G Miethke
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ashley Burg
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tiffany Shi
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - David A Hildeman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - E Steve S Woodle
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - J Matthew Kofron
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Prasad Devarajan
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 268-280 Albert Sabin Way, location T, floor 6, suite 272, Cincinnati, OH, 45229, USA.
| |
Collapse
|
26
|
Zhang Y, Gao D, Liang J, Ji M, Zhang W, Pan Y, Zheng F, Xie W. Association between folate deficiency and hypertension: evidence from an observational and Mendelian randomization study. Eur J Prev Cardiol 2024:zwae386. [PMID: 39590514 DOI: 10.1093/eurjpc/zwae386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 09/07/2024] [Accepted: 11/25/2024] [Indexed: 11/28/2024]
Abstract
AIMS Although folate intake might affect hypertension risk, evidence about the risk of hypertension according to an individual's folate deficiency status is scarce. Therefore, we aimed to investigate the relationship between folate deficiency and hypertension, and their causal associations. METHODS AND RESULTS A cross-sectional and prospective cohort study were performed in the UK Biobank (UKB) to investigate the associations between folate deficiency and hypertension prevalence and incidence, using logistic regression and Cox proportional hazard regression, respectively. Subsequently, we conducted one-sample Mendelian randomization (MR) with individual-level data from the UKB to further validate their causal associations. Finally, two-sample MR analyses were applied using summary-level data to further assess the causal relationships of serum folate with blood pressure (BP) and hypertension. In a total of up to 219,089 participants free of hypertension at enrollment, 17,670 participants developed hypertension after a median 12.8 years of follow-up. Compared with participants without folate deficiency, those with folate deficiency had a higher risk of hypertension (HR=1.42, 95% CI=1.24-1.63). One-sample MR analysis in the UKB provided supportive evidence for a causal effect of folate deficiency on hypertension risk (OR for the highest quantile=1.07, 95% CI=1.04-1.10, Ptrend<0.001). Furthermore, two-sample MR also supported a protective effect of higher levels of serum folate on BP (For systolic BP: β =-2.313, 95% CI=-3.532, -1.094; for diastolic BP: β = -1.648, 95% CI= -3.085, -0.211) and hypertension (β =-0.049, 95% CI=-0.069, -0.029). CONCLUSIONS Observational and genetically determined folate deficiency were associated with hypertension, suggesting that folate deficiency might be a causal risk factor for hypertension.
Collapse
Affiliation(s)
- Yanyu Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
- Clinical Research Institute, Institute of Advanced Clinical Medicine, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Darui Gao
- Department of Endocrinology, Peking University First Hospital, Beijing, China
- Clinical Research Institute, Institute of Advanced Clinical Medicine, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Jie Liang
- School of Nursing, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Mengmeng Ji
- Department of Endocrinology, Peking University First Hospital, Beijing, China
- Clinical Research Institute, Institute of Advanced Clinical Medicine, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Wenya Zhang
- School of Nursing, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yang Pan
- School of Nursing, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Fanfan Zheng
- School of Nursing, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wuxiang Xie
- Department of Endocrinology, Peking University First Hospital, Beijing, China
- Clinical Research Institute, Institute of Advanced Clinical Medicine, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| |
Collapse
|
27
|
Wei Z, Hu Y, Zuo F, Wen X, Wu D, Sun X, Liu C. The association between metabolic syndrome and lung cancer risk: a Mendelian randomization study. Sci Rep 2024; 14:28494. [PMID: 39558018 PMCID: PMC11574301 DOI: 10.1038/s41598-024-79260-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
Metabolic syndrome (MetS) is closely linked to cancer development, with emerging evidence suggesting its association with pulmonary carcinoma. However, causal relationships remain unclear due to observational study limitations. Employing Mendelian randomization, we investigated the causal link between MetS and lung cancer (LC) susceptibility. The data utilized in this study were obtained from the publicly available genetic variation summary database. The causal relationship was assessed using the inverse variance weighting method (IVW), weighted median method, and MR-Egger regression. A sensitivity analysis was carried out to confirm the robustness of the findings. Furthermore, risk factor analyses were conducted to explore potential mediators. Utilizing various analyses, MetS demonstrated a significant positive association with LC (OR, 1.22; 95% CI, 1.09-1.37, p = 7.57 × 10- 4), lung squamous cell carcinoma (LUSC) (OR, 1.47; 95%, 1.23-1.75, p = 2.22 × 10- 5), and small cell lung cancer (SCLC) (OR, 1.76; 95% CI, 1.37-2.26, p = 8.20 × 10- 6) but not lung adenocarcinoma (LUAD) (OR, 1.08; 95% CI, 0.94-1.24, p = 0.28). Risk factor analyses indicated that smoking, alcohol, body mass index, education, and type 2 diabetes might mediate the association. This study genetically validates and reinforces the evidence of MetS increasing the incidence of LC, including both LUSC) and SCLC, especially among individuals with abdominal obesity. It provides valuable insights for the development of lung cancer prevention strategies and directions for future research.
Collapse
Affiliation(s)
- Zhicheng Wei
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, 635000, People's Republic of China
| | - Yunyun Hu
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, 635000, People's Republic of China
| | - Fang Zuo
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Xiushu Wen
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, 635000, People's Republic of China
| | - Desheng Wu
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, 635000, People's Republic of China
| | - Xiaodong Sun
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, 635000, People's Republic of China
| | - Conghai Liu
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, 635000, People's Republic of China.
| |
Collapse
|
28
|
Rroji M, Spasovski G. Omics Studies in CKD: Diagnostic Opportunities and Therapeutic Potential. Proteomics 2024:e202400151. [PMID: 39523931 DOI: 10.1002/pmic.202400151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Omics technologies have significantly advanced the prediction and therapeutic approaches for chronic kidney disease (CKD) by providing comprehensive molecular insights. This is a review of the current state and future prospects of integrating biomarkers into the clinical practice for CKD, aiming to improve patient outcomes by targeted therapeutic interventions. In fact, the integration of genomic, transcriptomic, proteomic, and metabolomic data has enhanced our understanding of CKD pathogenesis and identified novel biomarkers for an early diagnosis and targeted treatment. Advanced computational methods and artificial intelligence (AI) have further refined multi-omics data analysis, leading to more accurate prediction models for disease progression and therapeutic responses. These developments highlight the potential to improve CKD patient care with a precise and individualized treatment plan .
Collapse
Affiliation(s)
- Merita Rroji
- Faculty of Medicine, Department of Nephrology, University of Medicine Tirana, Tirana, Albania
| | - Goce Spasovski
- Medical Faculty, Department of Nephrology, University of Skopje, Skopje, North Macedonia
| |
Collapse
|
29
|
Wei P, Xie H, Sun J, Zhuang Q, Xie J, Yin Y, Liu F, Li W, Chen C, Wang F, Han X, Xu L, Zhao X, Chen Y, Yang S, Shen C. Associations of genetic variation and mRNA expression of PDGF/PDGFRB pathway genes with coronary artery disease in the Chinese population. J Cell Mol Med 2024; 28:e70193. [PMID: 39569832 PMCID: PMC11579943 DOI: 10.1111/jcmm.70193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/04/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024] Open
Abstract
Platelet-derived growth factors (PDGFs) and receptors (PDGFR) play a key role in the process of coronary atherosclerosis. We aimed to investigate the association of genetic variations and mRNA expressions of PDGF/PDGFRB pathway genes with coronary artery disease (CAD). In this case-control study (3139 CAD vs. 3270 controls), 13 single nucleotide polymorphisms (SNPs) at five pathway genes were genotyped and combined to construct a weighted genetic risk score (wGRS). Three hundred and six pairs of cases and controls were selected for mRNA quantification. Restricted cubic spline (RCS) analyses were conducted for the dose-response relationship between wGRS, mRNAs and CAD. Area under the curve (AUC) was estimated to evaluate the discrimination of wGRS, mRNAs, and traditional risk factors (TRF) for CAD. The wGRS exhibited a positive linear relationship with CAD (p for linearity <0.001), and the medium and high wGRS had 37% and 50% increased risk of CAD compared to the low wGRS group (p = 1.5 × 10-4; p = 5.7 × 10-5). mRNA expression levels of five genes in peripheral blood leukocytes were all lower among patients at admission than controls (p < 0.001). The PDGF/PDGFRB mRNA expressions had significant non-linear correlations with AMI, with "U"-shaped trend for PDGFA, PDGFB and "L"-shaped trend for PDGFC, PDGFD and PDGFRB. Adding wGRS and mRNAs to the TRF model significantly improved the discrimination for CAD with an AUC of 0.921 (95% CI, 0.898-0.943). Genetic variations in the PDGF/PDGFRB pathway contribute to CAD susceptibility with a significantly joint effect. The down-regulated PDGF/PDGFRB mRNAs in peripheral leukocytes have the potential as blood-based biomarkers for CAD with high discriminative value.
Collapse
Affiliation(s)
- Pengfei Wei
- Department of CardiologyAffiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing CityYixingChina
| | - Hankun Xie
- Department of Epidemiology, Center for Global Health, School of Public HealthNanjing Medical UniversityNanjingChina
| | - Junxiang Sun
- Department of CardiologyAffiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing CityYixingChina
| | - Qian Zhuang
- Department of CardiologyAffiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing CityYixingChina
| | - Jichao Xie
- Department of CardiologyAffiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing CityYixingChina
| | - Yunjie Yin
- Department of CardiologyAffiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing CityYixingChina
| | - Fangyuan Liu
- Department of Epidemiology, Center for Global Health, School of Public HealthNanjing Medical UniversityNanjingChina
| | - Wen Li
- Department of Epidemiology, Center for Global Health, School of Public HealthNanjing Medical UniversityNanjingChina
| | - Changying Chen
- Department of Epidemiology, Center for Global Health, School of Public HealthNanjing Medical UniversityNanjingChina
| | - Feifan Wang
- Department of Epidemiology, Center for Global Health, School of Public HealthNanjing Medical UniversityNanjingChina
| | - Xu Han
- Department of Epidemiology, Center for Global Health, School of Public HealthNanjing Medical UniversityNanjingChina
| | - Liang Xu
- Department of CardiologyAffiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing CityYixingChina
| | - Xianghai Zhao
- Department of CardiologyAffiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing CityYixingChina
| | - Yanchun Chen
- Department of CardiologyAffiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing CityYixingChina
| | - Song Yang
- Department of CardiologyAffiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing CityYixingChina
| | - Chong Shen
- Department of Epidemiology, Center for Global Health, School of Public HealthNanjing Medical UniversityNanjingChina
| |
Collapse
|
30
|
Curtis D. Assessment of ability of AlphaMissense to identify variants affecting susceptibility to common disease. Eur J Hum Genet 2024; 32:1419-1427. [PMID: 39097650 DOI: 10.1038/s41431-024-01675-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/28/2024] [Accepted: 07/23/2024] [Indexed: 08/05/2024] Open
Abstract
An important issue in the analysis of rare variant association studies is the ability to annotate nonsynonymous variants in terms of their likely importance as affecting protein function. To address this, AlphaMissense was recently released and was shown to have good performance using benchmarks based on variants causing severe disease and on functional assays. Here, we assess the performance of AlphaMissense across 18 genes which had previously demonstrated association between rare coding variants and hyperlipidaemia, hypertension or type 2 diabetes. The strength of evidence in favour of association, expressed as the signed log p value (SLP), was compared between AlphaMissense and 43 other annotation methods. The results demonstrated marked variability between genes regarding the extent to which nonsynonymous variants contributed to evidence for association and also between the performance of different methods of annotating the nonsynonymous variants. Although AlphaMissense produced the highest SLP on average across genes, it produced the maximum SLP for only 4 genes. For some genes, other methods produced a considerably higher SLP and there were examples of genes where AlphaMissense produced no evidence for association while another method performed well. The marked inconsistency across genes means that it is difficult to decide on an optimal method of analysis of sequence data. The fact that different methods perform well for different genes suggests that if one wished to use sequence data for individual risk prediction then gene-specific annotation methods should be used.
Collapse
Affiliation(s)
- David Curtis
- UCL Genetics Institute, University College London, London, UK.
| |
Collapse
|
31
|
Lu L, Gu X, Yang D, Wang B, Long G. Circulating fatty acids, genetic susceptibility and hypertension: a prospective cohort study. Front Nutr 2024; 11:1454364. [PMID: 39545052 PMCID: PMC11562856 DOI: 10.3389/fnut.2024.1454364] [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: 06/25/2024] [Accepted: 10/07/2024] [Indexed: 11/17/2024] Open
Abstract
Background Combining genetic risk factors and plasma fatty acids (FAs) can be used as an effective method of precision medicine to prevent hypertension risk. Methods A total of 195,250 participants in the UK Biobank cohort were included in this study from 2006-2010. Polygenic risk scores (PRSs) were calculated for hypertension using single-nucleotide polymorphisms (SNPs). Concentrations of plasma FAs, including polyunsaturated fatty acids (PUFAs), monounsaturated fatty acids (MUFAs) and saturated fatty acids (SFAs), were tested by nuclear magnetic resonance. The Cox model was used to test for the main effects of PRS, different plasma FAs and their joint effects on hypertension. Relative excess risk due to interaction (RERI) and the attributable proportion due to interaction (AP) were used to test the additive interaction. Results Plasma PUFAs, n-3 PUFAs, MUFAs and SFAs were related to the risk of hypertension (PUFAs: HR, 0.878; 95% CI, 0.868-0.888; MUFAs: HR, 1.13; 95% CI, 1.123-1.150; SFAs: HR, 1.086; 95% CI, 1.074-1.098; n-3 PUFAs: HR, 0.984; 95% CI, 0.973-0.995). Moreover, an additive interaction was found between PRS and plasma FAs, which could contribute to an approximately 10-18% risk of hypertension, and the associations between high plasma MUFAs and a high PRS of hypertension were the strongest positive [RERI: 0.178 (95% CI: 0.062, 0.294), AP: 0.079 (95% CI: 0.027, 0.130)]. Conclusion Increased plasma MUFAs or SFAs and decreased plasma PUFAs or n-3 PUFAs were associated with hypertension risk, especially among people at high genetic risk.
Collapse
Affiliation(s)
- Lingling Lu
- Department of Infectious Disease, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoli Gu
- Department of Party and Government Office, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Daheng Yang
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Bingjian Wang
- Department of Cardiology, Huai’an First People’s Hospital Affiliated with Nanjing Medical University, Huai’an, China
| | - Guangfeng Long
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
32
|
Tang YY, Liu JJ, Gu HJ, Wang XS, Tan CM. Leisure screen time and diabetic retinopathy risk: A Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40099. [PMID: 39470559 PMCID: PMC11521079 DOI: 10.1097/md.0000000000040099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 06/29/2024] [Accepted: 09/26/2024] [Indexed: 10/30/2024] Open
Abstract
The aim of this study was to investigate whether leisure screen time (LST) increases the risk of diabetic retinopathy (DR) using the Mendelian randomization (MR). This study employed a two-sample MR analysis, utilizing 63 single-nucleotide polymorphisms as instrumental variables (IVs) to assess the causal relationship between LST and the risk of Dr. To ensure the robustness of the results, a multi-effect test was conducted to evaluate the validity of the IVs. Additionally, heterogeneity tests were performed to explore differences among sub-samples. Sensitivity analyses were also conducted to further validate our findings. The impact of LST on the risk of DR was observed in both inverse variance weighted (odds ratio [OR]: 1.22, 95% confidence interval [CI]: 1.04-1.43, P = 1.38 × 10-2) and weighted median (OR: 1.30, 95% CI: 1.05-1.61, P = 1.46 × 10-2) analyses. However, the MR-Egger method (OR: 0.66, 95% CI: 0.32-1.36, P = .273) did not find an increased risk of DR with increased LST. The pleiotropy test yielded a P-value of P = .09. Heterogeneity tests showed that the Q value for the inverse variance weighted method was 71.39 with a P-value of 0.17, indicating no significant heterogeneity. These results suggest that the IVs might be appropriate, and the analysis results could be robust. A large-scale MR analysis suggests a causal relationship between LST and the risk of Dr.
Collapse
Affiliation(s)
- Yuan-Yuan Tang
- Department of Nephroendocrinology, Guang’an People’s Hospital, Guan’an, Sichuan, China
| | - Jun-Jie Liu
- Department of Nephroendocrinology, Guang’an People’s Hospital, Guan’an, Sichuan, China
| | - Hong-Jing Gu
- Department of Nephroendocrinology, Guang’an People’s Hospital, Guan’an, Sichuan, China
| | - Xiao-Shu Wang
- Department of Nephroendocrinology, Guang’an People’s Hospital, Guan’an, Sichuan, China
| | - Chun-Mei Tan
- Department of Nephroendocrinology, Guang’an People’s Hospital, Guan’an, Sichuan, China
| |
Collapse
|
33
|
Funato Y, Mimura M, Nunomura K, Lin B, Fujii S, Haruta J, Miki H. Development of a high-throughput screening system targeting the protein-protein interactions between PRL and CNNM. Sci Rep 2024; 14:25432. [PMID: 39455715 PMCID: PMC11511866 DOI: 10.1038/s41598-024-76269-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Phosphatase of regenerating liver (PRL) is an oncogenic protein that promotes tumor progression by directly binding to cyclin M (CNNM) membrane proteins and inhibiting their Mg2+ efflux activity. In this study, we have developed a high-throughput screening system to detect the interactions between PRL and CNNM proteins based on homogenous time-resolved fluorescence resonance energy transfer (HTR-FRET, HTRF). We optimized the tag sequences attached to the recombinant proteins of the CNNM4 CBS domains and PRL3 lacking the carboxyl terminal CAAX motif, and successfully detected the interaction by observing the FRET signal in the mixture of the tagged proteins and fluorophore-conjugated antibodies. Moreover, we performed compound library screening using this system and discovered several compounds that could efficiently inhibit the PRL-CNNM interaction. Characterization of one candidate compound revealed that it was relatively stable compared with thienopyridone, a known inhibitor of the PRL-CNNM interaction. The candidate compound can also inhibit PRL function in cells: suppression of CNNM-dependent Mg2+ efflux, and has sufficient in vitro drug metabolism and pharmacokinetic properties. Overall, these results demonstrate the effectiveness of this screening system for identifying novel inhibitors of the PRL-CNNM interaction, which could contribute to the development of novel anti-cancer drugs.
Collapse
Affiliation(s)
- Yosuke Funato
- Laboratory of Biorecognition Chemistry, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan.
| | - Mai Mimura
- Department of Cellular Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871, Osaka, Japan
| | - Kazuto Nunomura
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, 565-0871, Osaka, Japan
| | - Bangzhong Lin
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, 565-0871, Osaka, Japan
| | - Shintarou Fujii
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, 565-0871, Osaka, Japan
| | - Junichi Haruta
- Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, 565-0871, Osaka, Japan
| | - Hiroaki Miki
- Laboratory of Biorecognition Chemistry, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan.
| |
Collapse
|
34
|
Gonçalves L, Zanlorenci S, Pelegrini A, de Lima TR, Silva DAS. Individual and Joint Association between Cardiovascular Disease Risk Factors and Inadequate Lifestyle Behaviors in a Sample from Brazil. Arq Bras Cardiol 2024; 121:e20240149. [PMID: 39442139 PMCID: PMC11634217 DOI: 10.36660/abc.20240149] [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: 03/04/2024] [Revised: 06/05/2024] [Accepted: 07/31/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Individual and Joint Association between Cardiovascular Disease Risk Factors and Inadequate Lifestyle Behaviors in a Sample from Brazil. CVD: cardiovascular diseases. BACKGROUND Cardiovascular diseases (CVD) are often influenced by modifiable factors, notably individuals' lifestyle choices, which play a crucial role in modulating cardiovascular risk. OBJECTIVE To investigate the individual and simultaneous association between inadequate lifestyle behaviors and risk factors for CVD in adults and older adults. METHOD A cross-sectional study with 1079 users of the Health Academy Program in Brazil. Information related to inadequate diet, excessive alcohol consumption, smoking, and physical inactivity were individually and collectively investigated (0, 1, or ≥ 2 factors) in association with CVD risk factors (hypertension, hypercholesterolemia, diabetes mellitus, and obesity), considering the following two outcomes: presence of CVD risk factors and number of CVD risk factors present in the same individual (0, 1, 2, or ≥ 3 risk factors). Logistic and multinomial logistic regression analyses were used. The statistical significance adopted was 5%. RESULTS A higher number of inadequate lifestyle behavior was associated with greater odds of simultaneous presence of 1, 2, or ≥ 3 CVD risk factors. The simultaneous adoption of 1 and ≥ 2 inadequate lifestyle behaviors was associated with greater odds of hypercholesterolemia. Simultaneous adherence to ≥ 2 inadequate lifestyle behaviors was associated with lower odds of hypertension. CONCLUSION A greater number of inadequate lifestyle behaviors was associated with higher odds of simultaneous presence of multiple CVD risk factors.
Collapse
Affiliation(s)
- Letícia Gonçalves
- Universidade Federal de Santa CatarinaFlorianópolisSCBrasilUniversidade Federal de Santa Catarina, Florianópolis, SC – Brasil
| | - Suellem Zanlorenci
- Universidade Federal de Santa CatarinaFlorianópolisSCBrasilUniversidade Federal de Santa Catarina, Florianópolis, SC – Brasil
| | - Andreia Pelegrini
- Universidade do Estado de Santa CatarinaFlorianópolisSCBrasilUniversidade do Estado de Santa Catarina, Florianópolis, SC – Brasil
| | - Tiago Rodrigues de Lima
- Universidade do Estado de Santa CatarinaFlorianópolisSCBrasilUniversidade do Estado de Santa Catarina, Florianópolis, SC – Brasil
| | - Diego Augusto Santos Silva
- Universidade Federal de Santa CatarinaFlorianópolisSCBrasilUniversidade Federal de Santa Catarina, Florianópolis, SC – Brasil
| |
Collapse
|
35
|
Lind L, Mazidi M, Clarke R, Bennett DA, Zheng R. Measured and genetically predicted protein levels and cardiovascular diseases in UK Biobank and China Kadoorie Biobank. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1189-1198. [PMID: 39322770 PMCID: PMC11473359 DOI: 10.1038/s44161-024-00545-6] [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: 01/08/2024] [Accepted: 08/28/2024] [Indexed: 09/27/2024]
Abstract
Several large-scale studies have measured plasma levels of the proteome in individuals with cardiovascular diseases (CVDs)1-7. However, since the majority of such proteins are interrelated2, it is difficult for observational studies to distinguish which proteins are likely to be of etiological relevance. Here we evaluate whether plasma levels of 2,919 proteins measured in 52,164 UK Biobank participants are associated with incident myocardial infarction, ischemic stroke or heart failure. Of those proteins, 126 were associated with all three CVD outcomes and 118 were associated with at least one CVD in the China Kadoorie Biobank. Mendelian randomization and colocalization analyses indicated that genetically determined levels of 47 and 18 proteins, respectively, were associated with CVDs, including FGF5, PROCR and FURIN. While the majority of protein-CVD observational associations were noncausal, these three proteins showed evidence to support potential causality and are therefore promising targets for drug treatment for CVD outcomes.
Collapse
Affiliation(s)
- Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Mohsen Mazidi
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Robert Clarke
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Derrick A Bennett
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Rui Zheng
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
36
|
Zhang W, Zhu J, Wu X, Feng T, Liao W, Li X, Chen J, Zhang L, Xiao C, Cui H, Yang C, Yan P, Wang Y, Tang M, Chen L, Liu Y, Zou Y, Wu X, Zhang L, Yang C, Yao Y, Li J, Liu Z, Jiang X, Zhang B. Phenotypic and genetic effect of carotid intima-media thickness on the risk of stroke. Hum Genet 2024; 143:1131-1143. [PMID: 38578439 DOI: 10.1007/s00439-024-02666-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 03/05/2024] [Indexed: 04/06/2024]
Abstract
While carotid intima-media thickness (cIMT) as a noninvasive surrogate measure of atherosclerosis is widely considered a risk factor for stroke, the intrinsic link underlying cIMT and stroke has not been fully understood. We aimed to evaluate the clinical value of cIMT in stroke through the investigation of phenotypic and genetic relationships between cIMT and stroke. We evaluated phenotypic associations using observational data from UK Biobank (N = 21,526). We then investigated genetic relationships leveraging genomic data conducted in predominantly European ancestry for cIMT (N = 45,185) and any stroke (AS, Ncase/Ncontrol=40,585/406,111). Observational analyses suggested an increased hazard of stroke per one standard deviation increase in cIMT (cIMTmax-AS: hazard ratio (HR) = 1.39, 95%CI = 1.09-1.79; cIMTmean-AS: HR = 1.39, 95%CI = 1.09-1.78; cIMTmin-AS: HR = 1.32, 95%CI = 1.04-1.68). A positive global genetic correlation was observed (cIMTmax-AS: [Formula: see text]=0.23, P=9.44 × 10-5; cIMTmean-AS: [Formula: see text]=0.21, P=3.00 × 10-4; cIMTmin-AS: [Formula: see text]=0.16, P=6.30 × 10-3). This was further substantiated by five shared independent loci and 15 shared expression-trait associations. Mendelian randomization analyses suggested no causal effect of cIMT on stroke (cIMTmax-AS: odds ratio (OR)=1.12, 95%CI=0.97-1.28; cIMTmean-AS: OR=1.09, 95%CI=0.93-1.26; cIMTmin-AS: OR=1.03, 95%CI = 0.90-1.17). A putative association was observed for genetically predicted stroke on cIMT (AS-cIMTmax: beta=0.07, 95%CI = 0.01-0.13; AS-cIMTmean: beta=0.08, 95%CI = 0.01-0.15; AS-cIMTmin: beta = 0.08, 95%CI = 0.01-0.16) in the reverse direction MR, which attenuated to non-significant in sensitivity analysis. Our work does not find evidence supporting causal associations between cIMT and stroke. The pronounced cIMT-stroke association is intrinsic, and mostly attributed to shared genetic components. The clinical value of cIMT as a surrogate marker for stroke risk in the general population is likely limited.
Collapse
Affiliation(s)
- Wenqiang Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Jingwei Zhu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Xuan Wu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Tianle Feng
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Wei Liao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Xuan Li
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Jianci Chen
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Li Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Chenghan Xiao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
- Department of Maternal, Child and Adolescent Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Huijie Cui
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Chao Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Peijing Yan
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Yutong Wang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Mingshuang Tang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Lin Chen
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Yunjie Liu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Yanqiu Zou
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Xueyao Wu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Ling Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
- Department of Iatrical Polymer Material and Artificial Apparatus, School of Polymer Science and Engineering, Sichuan University, Chengdu, China
| | - Chunxia Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Yuqin Yao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
- Department of Occupational and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jiayuan Li
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
| | - Zhenmi Liu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China
- Department of Maternal, Child and Adolescent Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xia Jiang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, West China- PUMC C. C. Chen Institute of Health, Sichuan University, No. 16, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, China.
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
- Department of Clinical Neuroscience, Karolinskaa Institutet, Stockholm, Sweden.
| | - Ben Zhang
- Hainan General Hospital and Hainan Affiliated Hospital, Hainan Medical University, Haikou, China; West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
37
|
Dickinson YA, Moyes AJ, Hobbs AJ. C-type natriuretic peptide (CNP): The cardiovascular system and beyond. Pharmacol Ther 2024; 262:108708. [PMID: 39154787 DOI: 10.1016/j.pharmthera.2024.108708] [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: 03/28/2024] [Revised: 07/30/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
C-type natriuretic peptide (CNP) represents the 'local' member of the natriuretic peptide family, functioning in an autocrine or paracrine capacity to modulate a hugely diverse portfolio of physiological processes. Whilst the best-characterised of these regulatory roles are in the cardiovascular system, akin to its predominantly endocrine siblings atrial (ANP) and brain (BNP) natriuretic peptides, CNP governs many additional, unrelated mechanisms including bone growth, gamete maturation, auditory processing, and neuronal integrity. Furthermore, there is currently great interest in mimicking the biological activity of CNP for therapeutic gain in many of these disparate organ systems. Herein, we provide an overview of the physiology, pathophysiology and pharmacology of CNP in both cardiovascular and non-cardiovascular settings.
Collapse
Affiliation(s)
- Yasmin A Dickinson
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Amie J Moyes
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Adrian J Hobbs
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
38
|
Zheng X, Qian Y, Wang L. Causal relationship between gut microbiota and insulin-like growth factor 1: a bidirectional two-sample Mendelian randomization study. Front Cell Infect Microbiol 2024; 14:1406132. [PMID: 39386166 PMCID: PMC11463061 DOI: 10.3389/fcimb.2024.1406132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 08/29/2024] [Indexed: 10/12/2024] Open
Abstract
Background The causal relationship between gut microbiota and insulin-like growth factor 1 (IGF-1) remains unclear. The purpose of this study was to explore the causal relationship between gut microbiota and IGF-1 in men and women. Methods Single-nucleotide polymorphisms (SNPs) related to gut microbiota were derived from pooled statistics from large genome-wide association studies (GWASs) published by the MiBioGen consortium. Pooled data for IGF-1 were obtained from a large published GWAS. We conducted Mendelian randomization (MR) analysis, primarily using the inverse variance weighted (IVW) method. Additionally, we performed sensitivity analyses to enhance the robustness of our results, focusing on assessing heterogeneity and pleiotropy. Results In forward MR analysis, 11 bacterial taxa were found to have a causal effect on IGF-1 in men; 14 bacterial taxa were found to have a causal effect on IGF-1 in women (IVW, all P < 0.05). After false discovery rate (FDR) correction, all bacterial traits failed to pass the FDR correction. In reverse MR analysis, IGF-1 had a causal effect on nine bacterial taxa in men and two bacterial taxa in women respectively (IVW, all P < 0.05). After FDR correction, the causal effect of IGF-1 on order Actinomycetales (PFDR = 0.049) remains in men. The robustness of the IVW results was further confirmed after heterogeneity and pleiotropy analysis. Conclusion Our study demonstrates a bidirectional causal link between the gut microbiota and IGF-1, in both men and women.
Collapse
Affiliation(s)
- Xuejie Zheng
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yuping Qian
- Department of Neonatology, Anhui Provincial Children’s Hospital, Hefei, Anhui, China
| | - Lili Wang
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
39
|
Hassan MA, Zhou W, Ye M, He H, Gao Z. The effectiveness of physical activity interventions on blood pressure in children and adolescents: A systematic review and network meta-analysis. JOURNAL OF SPORT AND HEALTH SCIENCE 2024; 13:699-708. [PMID: 38244922 PMCID: PMC11282348 DOI: 10.1016/j.jshs.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/18/2023] [Accepted: 11/09/2023] [Indexed: 01/22/2024]
Abstract
BACKGROUND High blood pressure (BP) is a major contributor to mortality and cardiovascular diseases. Despite the known benefits of exercise for reducing BP, it is crucial to identify the most effective physical activity (PA) intervention. This systematic review and network meta-analysis (NMA) aimed to evaluate the available evidence on the effectiveness of various PA interventions for reducing BP and to determine their hierarchy based on their impact on BP. METHODS A search of PubMed, SPORTDiscus, PsycINFO, Web of Science, CINAHL, Cochrane, and Eric databases was conducted up to December 2022 for this systematic review and NMA. Randomized controlled trials and quasi-experimental studies targeting healthy children and adolescents aged 6-12 years old were included in this study. Only studies that compared controlled and intervention groups using PA or exercise as the major influence were included. We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Three independent investigators performed the literature screening, data extraction, and risk of bias assessment. We used Bayesian arm-based NMA to synthesize the data. The primary outcomes were systolic BP and diastolic BP. We calculated the mean differences (MDs) in systolic BP and diastolic BP before and after treatment. Mean treatment differences were estimated using NMA and random-effect models. RESULTS We synthesized 27 studies involving 15,220 children and adolescents. PA combined with nutrition and behavior change was the most effective intervention for reducing both systolic BP and diastolic BP (MD = -8.64, 95% credible interval (95%CI):-11.44 to -5.84; MD = -6.75, 95%CI: -10.44 to -3.11), followed by interventions with multiple components (MD = -1.39, 95%CI: -1.94 to -0.84; MD = -2.54, 95%CI: -4.89 to -0.29). CONCLUSION Our findings suggest that PA interventions incorporating nutrition and behavior change, followed by interventions with multiple components, are most effective for reducing both systolic BP and diastolic BP in children and adolescents.
Collapse
Affiliation(s)
- Mohamed A Hassan
- School of Kinesiology, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA; Department of Methods and Curriculum, Physical Education College for Men, Helwan University, Cairo 12552, Egypt
| | - Wanjiang Zhou
- School of Kinesiology, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Mingyi Ye
- China Institute of Sport and Health Science, Beijing Sport University, Beijing 100084, China
| | - Hui He
- China Institute of Sport and Health Science, Beijing Sport University, Beijing 100084, China
| | - Zan Gao
- Department of Kinesiology, Recreation, and Sport Studies, University of Tennessee, Knoxville, TN 37996, USA.
| |
Collapse
|
40
|
Fry H, Mazidi M, Kartsonaki C, Clarke R, Walters RG, Chen Z, Millwood IY. The Role of Furin and Its Therapeutic Potential in Cardiovascular Disease Risk. Int J Mol Sci 2024; 25:9237. [PMID: 39273186 PMCID: PMC11394739 DOI: 10.3390/ijms25179237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/15/2024] Open
Abstract
Furin is an important proteolytic enzyme, converting several proteins from inactive precursors to their active forms. Recently, proteo-genomic analyses in European and East Asian populations suggested a causal association of furin with ischaemic heart disease, and there is growing interest in its role in cardiovascular disease (CVD) aetiology. In this narrative review, we present a critical appraisal of evidence from population studies to assess furin's role in CVD risk and potential as a drug target for CVD. Whilst most observational studies report positive associations between furin expression and CVD risk, some studies report opposing effects, which may reflect the complex biological roles of furin and its substrates. Genetic variation in FURIN is also associated with CVD and its risk factors. We found no evidence of current clinical development of furin as a drug target for CVD, although several phase 1 and 2 clinical trials of furin inhibitors as a type of cancer immunotherapy have been completed. The growing field of proteo-genomics in large-scale population studies may inform the future development of furin and other potential drug targets to improve the treatment and prevention of CVD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Iona Y. Millwood
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK; (H.F.); (M.M.); (C.K.); (R.C.); (R.G.W.); (Z.C.)
| |
Collapse
|
41
|
Nishito Y, Fujishiro H, Nagamatsu S, Kambe T. Reduced Mn uptake of pleiotropic ZIP8 SNP is caused by its loss of Mn-responsive accumulation on the cell-surface. Biosci Biotechnol Biochem 2024; 88:1019-1026. [PMID: 38821503 DOI: 10.1093/bbb/zbae076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
Zrt/Irt-like protein 8 (ZIP8), which is a Zn transporter, plays a pivotal role as a Mn transporter. Recent studies have shown that a ZIP8 SNP (rs13107325 C→T, A391T) is associated with multiple diseases, likely by causing systemic Mn deficiency. However, the underlying molecular mechanisms remain unclear. We attempted to address this issue in cell-based experiments using Madin-Darby canine kidney cells stably expressing ZIP8 WT or the A391T SNP mutant under the control of the Tet-regulatable promoter. We showed that the A391T mutant lost the property of Mn-responsive accumulation on the cell surface, which was observed in WT ZIP8. We also showed that the loss of Mn-responsive accumulation of A391T mutant was associated with its reduced Mn uptake, compared with WT ZIP8, in the Mn uptake assay using the radioisotope 54Mn. Our results potentially explain how the ZIP8 A391T substitution is associated with disease pathogenesis caused by Mn deficiency.
Collapse
Affiliation(s)
- Yukina Nishito
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Hitomi Fujishiro
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan
| | - Shino Nagamatsu
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Taiho Kambe
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
42
|
Rudman-Melnick V, Vanhoutte D, Stowers K, Sargent M, Adam M, Ma Q, Perl AKT, Miethke AG, Burg A, Shi T, Hildeman DA, Woodle ESS, Kofron JM, Devarajan P. Gucy1α1 specifically marks kidney, heart, lung and liver fibroblasts. RESEARCH SQUARE 2024:rs.3.rs-4746078. [PMID: 39184103 PMCID: PMC11343171 DOI: 10.21203/rs.3.rs-4746078/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Fibrosis is a common outcome of numerous pathologies, including chronic kidney disease (CKD), a progressive renal function deterioration. Current approaches to target activated fibroblasts, key effector contributors to fibrotic tissue remodeling, lack specificity. Here, we report Gucy1α1 as a specific kidney fibroblast marker. Gucy1α1 levels significantly increased over the course of two clinically relevant murine CKD models and directly correlated with established fibrosis markers. Immunofluorescent (IF) imaging showed that Gucy1α1 comprehensively labelled cortical and medullary quiescent and activated fibroblasts in the control kidney and throughout injury progression, respectively. Unlike traditionally used markers platelet derived growth factor receptor beta (Pdgfrβ) and vimentin (Vim), Gucy1α1 did not overlap with off-target populations such as podocytes. Notably, Gucy1α1 labelled kidney fibroblasts in both male and female mice. Furthermore, we observed elevated GUCY1α1 expression in the human fibrotic kidney and lung. Studies in the murine models of cardiac and liver fibrosis revealed Gucy1α1 elevation in activated Pdgfrβ-, Vim- and alpha smooth muscle actin (αSma)-expressing fibroblasts paralleling injury progression and resolution. Overall, we demonstrate Gucy1α1 as an exclusive fibroblast marker in both sexes. Due to its multiorgan translational potential, GUCY1α1 might provide a novel promising strategy to specifically target and mechanistically examine fibroblasts.
Collapse
|
43
|
Kelly J, Xu X, Eales JM, Keavney B, Berzuini C, Tomaszewski M, Guo H. Interactive molecular causal networks of hypertension using a fast machine learning algorithm MRdualPC. BMC Med Res Methodol 2024; 24:168. [PMID: 39095705 PMCID: PMC11295895 DOI: 10.1186/s12874-024-02229-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 04/23/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Understanding the complex interactions between genes and their causal effects on diseases is crucial for developing targeted treatments and gaining insight into biological mechanisms. However, the analysis of molecular networks, especially in the context of high-dimensional data, presents significant challenges. METHODS This study introduces MRdualPC, a computationally tractable algorithm based on the MRPC approach, to infer large-scale causal molecular networks. We apply MRdualPC to investigate the upstream causal transcriptomics influencing hypertension using a comprehensive dataset of kidney genome and transcriptome data. RESULTS Our algorithm proves to be 100 times faster than MRPC on average in identifying transcriptomics drivers of hypertension. Through clustering, we identify 63 modules with causal driver genes, including 17 modules with extensive causal networks. Notably, we find that genes within one of the causal networks are associated with the electron transport chain and oxidative phosphorylation, previously linked to hypertension. Moreover, the identified causal ancestor genes show an over-representation of blood pressure-related genes. CONCLUSIONS MRdualPC has the potential for broader applications beyond gene expression data, including multi-omics integration. While there are limitations, such as the need for clustering in large gene expression datasets, our study represents a significant advancement in building causal molecular networks, offering researchers a valuable tool for analyzing big data and investigating complex diseases.
Collapse
Affiliation(s)
- Jack Kelly
- Centre for Biostatistics, School of Health 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
| | - James M Eales
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Bernard Keavney
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
- Division of Cardiology and Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Carlo Berzuini
- Centre for Biostatistics, School of Health Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - 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
| | - Hui Guo
- Centre for Biostatistics, School of Health Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
44
|
Yamamoto M, Takata T, Hanada H, Taniguchi S, Hamada S, Mae Y, Iyama T, Kanda T, Isomoto H. Zinc deficiency induces hypertension by paradoxically amplifying salt sensitivity under high salt intake in mice. Clin Exp Nephrol 2024; 28:728-739. [PMID: 38581621 DOI: 10.1007/s10157-024-02478-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 02/15/2024] [Indexed: 04/08/2024]
Abstract
BACKGROUND Hypertension is one of the major etiologies that cause chronic kidney disease (CKD) and can exacerbate kidney dysfunction. Zinc is an essential trace element playing a role in blood pressure regulation, and zinc deficiency, a common comorbidity in patients with CKD, can cause hypertension. However, the precise mechanism underlying zinc deficiency-induced hypertension is unknown. Sodium (Na+) retention due to inappropriate Na+ reabsorption in the renal tubule is the principal pathophysiology of hypertension. Therefore, this study aimed to investigate the association between zinc deficiency and salt sensitivity. METHODS Adult mice were fed a zinc-adequate (ZnA) or zinc-deficient (ZnD) diet combined with/without high salt in drinking water (HS) for 4 weeks (n = 6 each). Changes in blood pressure, urinary sodium excretion, and the expressions of the proximal tubular Na+ transporter, Na+/H+ exchanger 3 (NHE3), which mostly contributes to filtered Na+ reabsorption and the downstream Na+-Cl- transporter (NCC) were analyzed. RESULTS Urinary Na+ excretion significantly increased in ZnD mice, indicating that zinc deficiency causes natriuresis. NHE3 expressions were significantly suppressed, whereas NCC was upregulated in ZnD mice. Interestingly, the combination of high salt and ZnD diet (HS-ZnD) reversed the urinary Na+ loss. The NCC remained activated and NHE3 expressions paradoxically increased in HS-ZnD mice compared with those fed the combination of high salt and ZnA diet. In addition, blood pressure significantly increased only in HS-ZnD mice. CONCLUSION The combination of zinc deficiency and high salt causes hypertension. Zinc is associated with salt-sensitivity, potentially through NHE3 and NCC regulation.
Collapse
Affiliation(s)
- Marie Yamamoto
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan
| | - Tomoaki Takata
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan.
| | - Hinako Hanada
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan
| | - Sosuke Taniguchi
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan
| | - Shintaro Hamada
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan
| | - Yukari Mae
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan
| | - Takuji Iyama
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan
| | - Tsutomu Kanda
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan
| | - Hajime Isomoto
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan
| |
Collapse
|
45
|
Takase M, Hirata T, Nakaya N, Nakamura T, Kogure M, Hatanaka R, Nakaya K, Chiba I, Kanno I, Nochioka K, Tsuchiya N, Narita A, Metoki H, Satoh M, Obara T, Ishikuro M, Ohseto H, Uruno A, Kobayashi T, Kodama EN, Hamanaka Y, Orui M, Ogishima S, Nagaie S, Fuse N, Sugawara J, Kuriyama S, Tamiya G, Hozawa A, Yamamoto M. Associations of combined genetic and lifestyle risks with hypertension and home hypertension. Hypertens Res 2024; 47:2064-2074. [PMID: 38914703 PMCID: PMC11298407 DOI: 10.1038/s41440-024-01705-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 06/26/2024]
Abstract
No study, to our knowledge, has constructed a polygenic risk score based on clinical blood pressure and investigated the association of genetic and lifestyle risks with home hypertension. We examined the associations of combined genetic and lifestyle risks with hypertension and home hypertension. In a cross-sectional study of 7027 Japanese individuals aged ≥20 years, we developed a lifestyle score based on body mass index, alcohol consumption, physical activity, and sodium-to-potassium ratio, categorized into ideal, intermediate, and poor lifestyles. A polygenic risk score was constructed with the target data (n = 1405) using publicly available genome-wide association study summary statistics from BioBank Japan. Using the test data (n = 5622), we evaluated polygenic risk score performance and examined the associations of combined genetic and lifestyle risks with hypertension and home hypertension. Hypertension and home hypertension were defined as blood pressure measured at a community-support center ≥140/90 mmHg or at home ≥135/85 mmHg, respectively, or self-reported treatment for hypertension. In the test data, 2294 and 2322 participants had hypertension and home hypertension, respectively. Both polygenic risk and lifestyle scores were independently associated with hypertension and home hypertension. Compared with those of participants with low genetic risk and an ideal lifestyle, the odds ratios for hypertension and home hypertension in the low genetic risk and poor lifestyle group were 1.94 (95% confidence interval, 1.34-2.80) and 2.15 (1.60-2.90), respectively. In summary, lifestyle is important to prevent hypertension; nevertheless, participants with high genetic risk should carefully monitor their blood pressure despite a healthy lifestyle.
Collapse
Affiliation(s)
- Masato Takase
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Takumi Hirata
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Human Care Research Team, Tokyo metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Naoki Nakaya
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Tomohiro Nakamura
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Kyoto Women's University, Kyoto, Japan
| | - Mana Kogure
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Rieko Hatanaka
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Kumi Nakaya
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Ippei Chiba
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Ikumi Kanno
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Kotaro Nochioka
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku University Hospital, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Naho Tsuchiya
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Akira Narita
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Hirohito Metoki
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical and Pharmaceutical University, Miyagino-ku, Sendai, Japan
| | - Michihiro Satoh
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical and Pharmaceutical University, Miyagino-ku, Sendai, Japan
| | - Taku Obara
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Mami Ishikuro
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Hisashi Ohseto
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Akira Uruno
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Tomoko Kobayashi
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku University Hospital, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Eiichi N Kodama
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- International Research Institute of Disaster Science, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Yohei Hamanaka
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Masatsugu Orui
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Soichi Ogishima
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Satoshi Nagaie
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Nobuo Fuse
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Junichi Sugawara
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku University Hospital, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Suzuki Memorial Hospital, Satonomori, Iwanumashi, Miyagi, Japan
| | - Shinichi Kuriyama
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- International Research Institute of Disaster Science, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Gen Tamiya
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Atsushi Hozawa
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan.
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan.
| | - Masayuki Yamamoto
- Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| |
Collapse
|
46
|
Li X, Guo Y, Liang H, Wang J, Qi L. Genome-wide association analysis of hypertension and epigenetic aging reveals shared genetic architecture and identifies novel risk loci. Sci Rep 2024; 14:17792. [PMID: 39090212 PMCID: PMC11294447 DOI: 10.1038/s41598-024-68751-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024] Open
Abstract
Hypertension is a disease associated with epigenetic aging. However, the pathogenic mechanism underlying this relationship remains unclear. We aimed to characterize the shared genetic architecture of hypertension and epigenetic aging, and identify novel risk loci. Leveraging genome-wide association studies (GWAS) summary statistics of hypertension (129,909 cases and 354,689 controls) and four epigenetic clocks (N = 34,710), we investigated genetic architectures and genetic overlap using bivariate casual mixture model and conditional/conjunctional false discovery rate methods. Functional gene-sets pathway analyses were performed by functional mapping and gene annotation (FUMA) protocol. Hypertension was polygenic with 2.8 K trait-influencing genetic variants. We observed cross-trait genetic enrichment and genetic overlap between hypertension and all four measures of epigenetic aging. Further, we identified 32 distinct genomic loci jointly associated with hypertension and epigenetic aging. Notably, rs1849209 was shared between hypertension and three epigenetic clocks (HannumAge, IEAA, and PhenoAge). The shared loci exhibited a combination of concordant and discordant allelic effects. Functional gene-set analyses revealed significant enrichment in biological pathways related to sensory perception of smell and nervous system processes. We observed genetic overlaps with mixed effect directions between hypertension and all four epigenetic aging measures, and identified 32 shared distinct loci with mixed effect directions, 25 of which were novel for hypertension. Shared genes enriched in biological pathways related to olfaction.
Collapse
Affiliation(s)
- Xin Li
- The Sino-Russian Medical Research Center of Jinan University, The Institute of Chronic Disease of Jinan University, The First Affiliated Hospital of Jinan University, Guangzhou, 511436, China
| | - Yu Guo
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, 150086, China
| | - Haihai Liang
- The Sino-Russian Medical Research Center of Jinan University, The Institute of Chronic Disease of Jinan University, The First Affiliated Hospital of Jinan University, Guangzhou, 511436, China.
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China.
| | - Jinghao Wang
- Department of Pharmacy, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China.
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, 510630, Guangdong, China.
| | - Lishuang Qi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150086, China.
| |
Collapse
|
47
|
Patel M, Harris N, Kasztan M, Hyndman K. Comprehensive analysis of the endothelin system in the kidneys of mice, rats, and humans. Biosci Rep 2024; 44:BSR20240768. [PMID: 38904098 PMCID: PMC11249498 DOI: 10.1042/bsr20240768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 06/20/2024] [Accepted: 06/20/2024] [Indexed: 06/22/2024] Open
Abstract
The intrarenal endothelin (ET) system is an established moderator of kidney physiology and mechanistic contributor to the pathophysiology and progression of chronic kidney disease in humans and rodents. The aim of the present study was to characterize ET system by combining single cell RNA sequencing (scRNA-seq) data with immunolocalization in human and rodent kidneys of both sexes. Using publicly available scRNA-seq data, we assessed sex and kidney disease status (human), age and sex (rats), and diurnal expression (mice) on the kidney ET system expression. In normal human biopsies of both sexes and in rodent kidney samples, the endothelin-converting enzyme-1 (ECE1) and ET-1 were prominent in the glomeruli and endothelium. These data agreed with the scRNA-seq data from these three species, with ECE1/Ece1 mRNA enriched in the endothelium. However, the EDN1/Edn1 gene (encodes ET-1) was rarely detected, even though it was immunolocalized within the kidneys, and plasma and urinary ET-1 excretion are easily measured. Within each species, there were some sex-specific differences. For example, in kidney biopsies from living donors, men had a greater glomerular endothelial cell endothelin receptor B (Ednrb) compared with women. In mice, females had greater kidney endothelial cell Ednrb than male mice. As commercially available antibodies did not work in all species, and RNA expression did not always correlate with protein levels, multiple approaches should be considered to maintain required rigor and reproducibility of the pre- and clinical studies evaluating the intrarenal ET system.
Collapse
Affiliation(s)
- Margi Patel
- Department of Medicine, Division of Nephrology, Section of Cardio-Renal Physiology and Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, U.K
| | - Nicholas Harris
- Department of Medicine, Division of Nephrology, Section of Cardio-Renal Physiology and Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, U.K
| | - Malgorzata Kasztan
- Department of Pediatrics, Division of Hematology-Oncology, Section of Cardio-Renal Physiology and Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, U.K
| | - Kelly A. Hyndman
- Department of Medicine, Division of Nephrology, Section of Cardio-Renal Physiology and Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, U.K
| |
Collapse
|
48
|
Zhang J, Song X, Li Z, Xu H, Shu H, Li J, Zhang Y. Association of apolipoprotein levels with all-cause and cardiovascular mortality. Eur J Prev Cardiol 2024; 31:1183-1194. [PMID: 38417834 DOI: 10.1093/eurjpc/zwae080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/30/2024] [Accepted: 02/21/2024] [Indexed: 03/01/2024]
Abstract
AIMS Research has shown that apolipoproteins (Apos) are potential indicators of heart health and death. We investigated the associations of Apo levels with all-cause and cardiovascular mortality. METHODS AND RESULTS We systematically searched the Cochrane Library, PubMed, and Web of Science for English language studies up to 28 November 2022. We used Stata 17.0 to summarize the estimated effects with 95% confidence intervals (CIs). We also conducted subgroup analyses according to study location, year of publication, individual age, follow-up years, and sample size. Moreover, we performed a sensitivity analysis to evaluate bias in our study. This study included 23 studies with 152 854 individuals in total. The level of ApoA was negatively related to cardiovascular mortality [odds ratio (OR) = 0.69, 95% CI = 0.52-0.93]. An increased ratio of ApoB/A1 was a risk factor for cardiovascular mortality (OR = 2.13, 95% CI = 1.48-3.07) and all-cause mortality (OR = 2.05, 95% CI = 1.52-2.77). The level of ApoB was positively related to cardiovascular mortality (OR = 1.12, 95% CI = 0.85-1.47), but the difference was not statistically significant. However, the associations between ApoB or ApoA1 and all-cause mortality were not obvious. Our subgroup analyses showed that the location, year of publication, individual age, and follow-up years of the studies affected the heterogeneity of our study to varying degrees. The sensitivity analysis showed that our results were almost robust, apart from excluding the article by Nomikos (OR = 0.77, 95% CI = 0.65-0.92) and Zeng (OR = 0.77, 95% CI = 0.65-0.91), when investigating the relationship between ApoA1 and all-cause mortality. CONCLUSION In this study, we found that Apo levels were linked to cardiovascular and all-cause mortality. Our study strengthens the evidence on the association between the level of Apos and cardiac health and may provide ideas for regulating the level of Apos to promote public health.
Collapse
Affiliation(s)
- Jiarong Zhang
- School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Xinru Song
- Department of General Surgery, The Affiliated Jiangning Hospital of Nanjing Medical University, 169 Hushan Road, Nanjing 211166, China
| | - Zhi Li
- School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Haibo Xu
- School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Haotian Shu
- School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Jun Li
- Department of General Surgery, The Affiliated Jiangning Hospital of Nanjing Medical University, 169 Hushan Road, Nanjing 211166, China
| | - Yan Zhang
- School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| |
Collapse
|
49
|
Mangum KD, Li Q, Bauer TM, Wolf SJ, Shadiow J, Moon JY, Barrett EC, Joshi AD, Ahmed Z, Wasikowski R, Boyer K, Obi AT, Davis FM, Chang L, Tsoi LC, Gudjonsson J, Gallagher KA. Epigenetic Alteration of Smooth Muscle Cells Regulates Endothelin-Dependent Blood Pressure and Hypertensive Arterial Remodeling. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.09.24310178. [PMID: 39040193 PMCID: PMC11261912 DOI: 10.1101/2024.07.09.24310178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Long-standing hypertension (HTN) affects multiple organ systems and leads to pathologic arterial remodeling, which is driven largely by smooth muscle cell (SMC) plasticity. Although genome wide association studies (GWAS) have identified numerous variants associated with changes in blood pressure in humans, only a small percentage of these variants actually cause HTN. In order to identify relevant genes important in SMC function in HTN, we screened three separate human GWAS and Mendelian randomization studies to identify SNPs located within non-coding gene regions, focusing on genes encoding epigenetic enzymes, as these have been recently identified to control SMC fate in cardiovascular disease. We identified SNPs rs62059712 and rs74480102 in the promoter of the human JMJD3 gene and show that the minor C allele increases JMJD3 transcription in SMCs via increased SP1 binding to the JMJD3 promoter. Using our novel SMC-specific Jmjd3-deficient murine model ( Jmjd3 flox/flox Myh11 CreERT ), we show that loss of Jmjd3 in SMCs results in HTN, mechanistically, due to decreased EDNRB expression and a compensatory increase in EDNRA expression. As a translational corollary, through single cell RNA-sequencing (scRNA-seq) of human arteries, we found strong correlation between JMJD3 and EDNRB expression in SMCs. Further, we identified that JMJD3 is required for SMC-specific gene expression, and loss of JMJD3 in SMCs in the setting of HTN results in increased arterial remodeling by promoting the SMC synthetic phenotype. Our findings link a HTN-associated human DNA variant with regulation of SMC plasticity, revealing therapeutic targets that may be used in the screening and/or personalized treatment of HTN.
Collapse
|
50
|
Murphy MB, Yang Z, Subati T, Farber-Eger E, Kim K, Blackwell DJ, Fleming MR, Stark JM, Van Amburg JC, Woodall KK, Van Beusecum JP, Agrawal V, Smart CD, Pitzer A, Atkinson JB, Fogo AB, Bastarache JA, Kirabo A, Wells QS, Madhur MS, Barnett JV, Murray KT. LNK/SH2B3 loss of function increases susceptibility to murine and human atrial fibrillation. Cardiovasc Res 2024; 120:899-913. [PMID: 38377486 PMCID: PMC11218690 DOI: 10.1093/cvr/cvae036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/31/2023] [Accepted: 10/07/2023] [Indexed: 02/22/2024] Open
Abstract
AIMS The lymphocyte adaptor protein (LNK) is a negative regulator of cytokine and growth factor signalling. The rs3184504 variant in SH2B3 reduces LNK function and is linked to cardiovascular, inflammatory, and haematologic disorders, including stroke. In mice, deletion of Lnk causes inflammation and oxidative stress. We hypothesized that Lnk-/- mice are susceptible to atrial fibrillation (AF) and that rs3184504 is associated with AF and AF-related stroke in humans. During inflammation, reactive lipid dicarbonyls are the major components of oxidative injury, and we further hypothesized that these mediators are critical drivers of the AF substrate in Lnk-/- mice. METHODS AND RESULTS Lnk-/- or wild-type (WT) mice were treated with vehicle or 2-hydroxybenzylamine (2-HOBA), a dicarbonyl scavenger, for 3 months. Compared with WT, Lnk-/- mice displayed increased AF duration that was prevented by 2-HOBA. In the Lnk-/- atria, action potentials were prolonged with reduced transient outward K+ current, increased late Na+ current, and reduced peak Na+ current, pro-arrhythmic effects that were inhibited by 2-HOBA. Mitochondrial dysfunction, especially for Complex I, was evident in Lnk-/- atria, while scavenging lipid dicarbonyls prevented this abnormality. Tumour necrosis factor-α (TNF-α) and interleukin-1 beta (IL-1β) were elevated in Lnk-/- plasma and atrial tissue, respectively, both of which caused electrical and bioenergetic remodelling in vitro. Inhibition of soluble TNF-α prevented electrical remodelling and AF susceptibility, while IL-1β inhibition improved mitochondrial respiration but had no effect on AF susceptibility. In a large database of genotyped patients, rs3184504 was associated with AF, as well as AF-related stroke. CONCLUSION These findings identify a novel role for LNK in the pathophysiology of AF in both experimental mice and humans. Moreover, reactive lipid dicarbonyls are critical to the inflammatory AF substrate in Lnk-/- mice and mediate the pro-arrhythmic effects of pro-inflammatory cytokines, primarily through electrical remodelling.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Action Potentials/drug effects
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Atrial Fibrillation/metabolism
- Atrial Fibrillation/physiopathology
- Atrial Fibrillation/genetics
- Benzylamines/pharmacology
- Disease Models, Animal
- Genetic Predisposition to Disease
- Heart Rate/drug effects
- Inflammation Mediators/metabolism
- Interleukin-1beta/metabolism
- Interleukin-1beta/genetics
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Mitochondria, Heart/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Oxidative Stress/drug effects
- Phenotype
- Signal Transduction
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/genetics
Collapse
Affiliation(s)
- Matthew B Murphy
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Zhenjiang Yang
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Tuerdi Subati
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Eric Farber-Eger
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
| | - Kyungsoo Kim
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Daniel J Blackwell
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Matthew R Fleming
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
| | - Joshua M Stark
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Joseph C Van Amburg
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Kaylen K Woodall
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Justin P Van Beusecum
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Vineet Agrawal
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
| | - Charles D Smart
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Ashley Pitzer
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - James B Atkinson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, 1161 21 Avenue South, Nashville, TN 37232, USA
| | - Agnes B Fogo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, 1161 21 Avenue South, Nashville, TN 37232, USA
| | - Julie A Bastarache
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Quinn S Wells
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, 2525 West End Avenue, Nashville, TN 37203, USA
| | - Meena S Madhur
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Joey V Barnett
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Katherine T Murray
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 559 PRB, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Nashville, TN 37232, USA
| |
Collapse
|