1
|
Knol MGE, Wulfmeyer VC, Müller RU, Rinschen MM. Amino acid metabolism in kidney health and disease. Nat Rev Nephrol 2024; 20:771-788. [PMID: 39198707 DOI: 10.1038/s41581-024-00872-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/03/2024] [Indexed: 09/01/2024]
Abstract
Amino acids form peptides and proteins and are therefore considered the main building blocks of life. The kidney has an important but under-appreciated role in the synthesis, degradation, filtration, reabsorption and excretion of amino acids, acting to retain useful metabolites while excreting potentially harmful and waste products from amino acid metabolism. A complex network of kidney transporters and enzymes guides these processes and moderates the competing concentrations of various metabolites and amino acid products. Kidney amino acid metabolism contributes to gluconeogenesis, nitrogen clearance, acid-base metabolism and provision of fuel for tricarboxylic acid cycle and urea cycle intermediates, and is thus a central hub for homeostasis. Conversely, kidney disease affects the levels and metabolism of a variety of amino acids. Here, we review the metabolic role of the kidney in amino acid metabolism and describe how different diseases of the kidney lead to aberrations in amino acid metabolism. Improved understanding of the metabolic and communication routes that are affected by disease could provide new mechanistic insights into the pathogenesis of kidney diseases and potentially enable targeted dietary or pharmacological interventions.
Collapse
Affiliation(s)
- Martine G E Knol
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Roman-Ulrich Müller
- Department II of Internal Medicine and Center for Molecular Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Markus M Rinschen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- III Department of Medicine, University Medical Center Hamburg Eppendorf, Hamburg, Germany.
- Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark.
- Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
2
|
Chen H, Deng Y, Zhou H, Wu W, Bao J, Cao D, Li Y, Feng Y. Blood L-cystine levels positively related to increased risk of hypertension. J Clin Hypertens (Greenwich) 2024; 26:1411-1423. [PMID: 39403054 DOI: 10.1111/jch.14902] [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: 03/28/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 12/20/2024]
Abstract
Hypertension is one component of metabolic syndrome (MetS). Here, the study evaluated hypertension-associated metabolites in relation to other MetS components. Fasting plasma samples were collected from 22 hypertensive and 63 normotensive subjects for non-targeted metabolomics. Compared with normotensive subjects, hypertensive patients were more diabetic (6.3% vs. 36.4%) and had dyslipidemia (27.0% vs. 63.6%) (both p < .05). By non-targeted metabolomics, 758 metabolites in 22 classes were identified and 56 were differentially regulated between hypertensive and normotensive groups. Amongst these 56 metabolites, receiver operating characteristic analysis showed that 14 had an area under the curve above 0.6. Multivariate-adjusted logistic regression analysis demonstrated that per one-fold increase of L-glutmatic acid, L-cystine, (9S,10E,12Z,15Z)-9-Hydroxy-10,12,15-octadecatrienoic acid, deoxyribose 5-phosphate, and falcarinolone, the odds ratios were 3.64, 4.61, 0.26, 0.26, and 0.37 for having the risk of hypertension, respectively. Of five metabolites, by Spearman's correlation analysis, only L-glutmatic acid and L-cystine levels were positively associated with systolic and diastolic blood pressure (all p < .05). Spearman's correlation analysis further revealed that L-glutmatic acid levels were positively correlated with to body mass index (BMI), fasting blood glucose, and serum triglyceride but negatively associated with HDL-c (all p < .05) whereas L-cystine levels were not related to any of these components (p ≥ .13). Multivariate-adjusted linear regression analysis confirmed the positive correlation between L-cystine levels and systolic or diastolic blood pressure (β = 2.66 for SBP; β = 2.50 for DBP; both p < .05). In conclusion, L-cystine could be a potent metabolite for increased risk of hypertension.
Collapse
Affiliation(s)
- Haijun Chen
- Department of Computed Tomography, Heilongjiang Provincial Hospital, Harbin, Heilongjiang Province, China
| | - Yalan Deng
- Beijing Hepatitis Institute, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Department of Science and Technology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Hailing Zhou
- Department of Emergency, Heilongjiang Provincial Hospital, Harbin, Heilongjiang Province, China
| | - Wenzhong Wu
- Heilongiiang Red Cross Sengong General Hospital, Harbin, Heilongjiang Province, China
| | - Jinhua Bao
- Department of Clinical Nutrition, Heilongjiang Provincial Hospital, Harbin, Heilongjiang Province, China
| | - Deyou Cao
- People's Government of Hulan District in Harbin City, Harbin, Heilongjiang Province, China
| | - Yuze Li
- Department of Clinical Nutrition, Heilongjiang Provincial Hospital, Harbin, Heilongjiang Province, China
| | - Yingmei Feng
- Department of Science and Technology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Laboratory of Clinical Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Lin Z, Li J, Liu F, Cao J, Chen S, Chen J, Huang K, Wang Y, Li H, Wang Y, Huang J, Gu D, Lu X. Metabolomics signature of blood pressure salt sensitivity and its link to cardiovascular disease: A dietary salt-intervention trial. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1666-1675. [PMID: 38739172 DOI: 10.1007/s11427-023-2507-9] [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: 09/23/2023] [Accepted: 12/11/2023] [Indexed: 05/14/2024]
Abstract
Individuals with a high degree of salt sensitivity (SS) have a greater risk of cardiovascular disease (CVD), but whether SS fosters CVD by influencing metabolomics homeostasis remains unclear. This study aimed to reveal the role of the SS-related metabolomics signature in the development of CVDs, based on the MetaSalt study, which was a dietary salt-intervention trial conducted at four centers in China in 2019. A total of 528 participants were recruited and underwent 3 days of baseline observations, a 10-day low-salt intervention, and a 10-day high-salt intervention. Plasma untargeted metabolomics, lipidomics, and BP measurements were scheduled at each stage. Participants were grouped into extreme SS, moderate SS, and salt-resistant (SR) individuals according to their BP responses to salt. Linear mixed models were used to identify SS-related metabolites and determine the relationship between the SS-related metabolomics signature and arterial stiffness. Mendelian randomization (MR) analyses were applied to establish the causal pathways among the SS-related metabolites, BP, and CVDs. Among the 713 metabolites, 467 were significantly changed after the high-salt intervention. Among them, the changes in 30 metabolites from the low-salt to the high-salt intervention differed among the SS groups. Of the remaining nonsalt-related metabolites, the baseline levels of 11 metabolites were related to SS. These 41 metabolites explained 23% of the variance in SS. Moreover, SS and its metabolomics signature were positively correlated with arterial stiffness. MR analyses demonstrated that the SS-related metabolites may affect CVD risk by altering BP, indicating that the increase in BP was the consequence of the changes in SS-related metabolites rather than the cause. Our study revealed that the metabolomics signature of SS individuals differs from that of SR individuals and that the changes in SS-related metabolites may increase arterial stiffness and foster CVDs. This study provides insight into understanding the biology and targets of SS and its role in CVDs.
Collapse
Affiliation(s)
- Zhennan Lin
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Jianxin Li
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Fangchao Liu
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Jie Cao
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Shufeng Chen
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Jichun Chen
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Keyong Huang
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Yaqin Wang
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Hongfan Li
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Yan Wang
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Jianfeng Huang
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China
| | - Dongfeng Gu
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China.
- Medical School, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Xiangfeng Lu
- Key Laboratory of Cardiovascular Epidemiology, Department of Epidemiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100037, China.
| |
Collapse
|
4
|
Chrysopoulou M, Rinschen MM. Metabolic Rewiring and Communication: An Integrative View of Kidney Proximal Tubule Function. Annu Rev Physiol 2024; 86:405-427. [PMID: 38012048 DOI: 10.1146/annurev-physiol-042222-024724] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The kidney proximal tubule is a key organ for human metabolism. The kidney responds to stress with altered metabolite transformation and perturbed metabolic pathways, an ultimate cause for kidney disease. Here, we review the proximal tubule's metabolic function through an integrative view of transport, metabolism, and function, and embed it in the context of metabolome-wide data-driven research. Function (filtration, transport, secretion, and reabsorption), metabolite transformation, and metabolite signaling determine kidney metabolic rewiring in disease. Energy metabolism and substrates for key metabolic pathways are orchestrated by metabolite sensors. Given the importance of renal function for the inner milieu, we also review metabolic communication routes with other organs. Exciting research opportunities exist to understand metabolic perturbation of kidney and proximal tubule function, for example, in hypertension-associated kidney disease. We argue that, based on the integrative view outlined here, kidney diseases without genetic cause should be approached scientifically as metabolic diseases.
Collapse
Affiliation(s)
| | - Markus M Rinschen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark;
- III. Department of Medicine and Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark
| |
Collapse
|
5
|
Wu ZP, Wei W, Cheng Y, Chen JY, Liu Y, Liu S, Hu MD, Zhao H, Li XF, Chen X. Altered adolescents obesity metabolism is associated with hypertension: a UPLC-MS-based untargeted metabolomics study. Front Endocrinol (Lausanne) 2023; 14:1172290. [PMID: 37229452 PMCID: PMC10203610 DOI: 10.3389/fendo.2023.1172290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/19/2023] [Indexed: 05/27/2023] Open
Abstract
Objective This study aimed to explore the relationship between the plasma metabolites of adolescent obesity and hypertension and whether metabolite alterations had a mediating effort between adolescent obesity and hypertension. Methods We applied untargeted ultra-performance liquid chromatography-mass spectrometry (UPLC-MS) to detect the plasma metabolomic profiles of 105 adolescents. All participants were selected randomly based on a previous cross-sectional study. An orthogonal partial least squares- discriminant analysis (OPLS-DA), followed by univariate statistics and enrichment analysis, was used to identify differential metabolites. Using logistic regression for variable selection, an obesity-related metabolite score (OMS, OMS=∑k=1nβnmetabolite n) was constructed from the metabolites identified, and hypertension risk was estimated. Results In our study, based on P< 0.05, variable importance in projection (VIP) > 1.0, and impact value > 0.1, we identified a total of 12 differential metabolites. Significantly altered metabolic pathways were the sphingolipid metabolism, purine metabolism, pyrimidine metabolism, phospholipid metabolism, steroid hormone biosynthesis, tryptophan, tyrosine, and phenylalanine biosynthesis. The logistic regression selection resulted in a four-metabolite score (thymidine, sphingomyelin (SM) d40:1, 4-hydroxyestradiol, and L-lysinamide), which was positively associated with hypertension risk (odds ratio: 7.79; 95% confidence interval: 2.13, 28.47; for the quintile 4 compared with quartile 1 of OMS) after multivariable adjustment. Conclusions The OMS constructed from four differential metabolites was used to predict the risk of hypertension in adolescents. These findings could provide sensitive biomarkers for the early recognition of hypertension in adolescents with obesity.
Collapse
Affiliation(s)
- Zhi-Ping Wu
- Department of Epidemiology, School of Public Health, Dalian Medical University, Dalian, China
| | - Wei Wei
- Department of Neurosurgery, Central Hospital of Dalian University of Technology, Dalian, China
| | - Yuan Cheng
- Department of Epidemiology, School of Public Health, Dalian Medical University, Dalian, China
| | - Jing-Yi Chen
- Department of Epidemiology, School of Public Health, Dalian Medical University, Dalian, China
| | - Yang Liu
- Institute of Health Science, China Medical University, Shenyang, China
| | - Shan Liu
- Department of Epidemiology, School of Public Health, Dalian Medical University, Dalian, China
| | - Meng-Die Hu
- Department of Epidemiology, School of Public Health, Dalian Medical University, Dalian, China
| | - Heng Zhao
- Department of Epidemiology, School of Public Health, Dalian Medical University, Dalian, China
| | - Xiao-Feng Li
- Department of Epidemiology, School of Public Health, Dalian Medical University, Dalian, China
| | - Xin Chen
- Department of Epidemiology, School of Public Health, Dalian Medical University, Dalian, China
| |
Collapse
|
6
|
Zhang F, Xie Y, Yang X, Peng W, Qi H, Li B, Wen F, Li P, Sun Y, Zhang L. Association of Serum Metabolites and Salt Sensitivity of Blood Pressure in Chinese Population: The EpiSS Study. Nutrients 2023; 15:nu15030690. [PMID: 36771399 PMCID: PMC9921558 DOI: 10.3390/nu15030690] [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: 11/14/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND To identify novel metabolites associated with salt sensitivity of blood pressure (SSBP) in Chinese Han population. METHODS A case-control study was conducted with 25 salt sensitive (SS) and 26 salt resistant (SR) participants, which was selected from the Systems Epidemiology Study on Salt Sensitivity of Blood Pressure (EpiSS) study. The modified Sullivan's acute oral saline load and diuresis shrinkage test (MSAOSL-DST) was conducted to identify SS. Untargeted, ultra-high performance liquid chromatograph-high resolution mass spectrometer (UPLC-HRMS) was conducted and orthogonal partial least squares-discriminate analysis (OPLS-DA) and multivariable logistic regression model were used to screen the metabolites related to SS, mixed linear regressions models were used to examined the association of SSBP with metabolites during saline load period and diuresis shrinkage period. Receiver operating characteristic (ROC) curve analysis was performed. The area under the curve's (AUC) sensitivity and specificity were calculated to identified metabolites biomarkers for SS. RESULTS There were 39 differentially expressed metabolites (DE-metabolites) between SS and SR. Thirty-five and four of DE-metabolites were inversely or positively associated with SS, respectively. Four biochemical pathways demonstrated significant enrichment for identified metabolites. In single-metabolite analyses, L-Glutamine displayed the best diagnostic performance (AUC = 0.88, 95% CI: 0.78-0.97). In multi-metabolites analyses, L-Glutamine + Cholesterol ester 22:5n6 combination showed the best diagnostic performance (AUC = 0.96, 95% CI: 0.91-1.00). Adjusted for traditional risk factors, L-Glutamine and Cholesterol ester 22:5n6 explained an additional 38.3% of SS susceptibility. CONCLUSIONS This study provide potential evidence for clarifying the mechanism of SS and provide novel biological insights into salt sensitive hypertension.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ling Zhang
- Correspondence: ; Tel./Fax: +86-010-83911777
| |
Collapse
|
7
|
Maaliki D, Itani MM, Itani HA. Pathophysiology and genetics of salt-sensitive hypertension. Front Physiol 2022; 13:1001434. [PMID: 36176775 PMCID: PMC9513236 DOI: 10.3389/fphys.2022.1001434] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Most hypertensive cases are primary and heavily associated with modifiable risk factors like salt intake. Evidence suggests that even small reductions in salt consumption reduce blood pressure in all age groups. In that regard, the ACC/AHA described a distinct set of individuals who exhibit salt-sensitivity, regardless of their hypertensive status. Data has shown that salt-sensitivity is an independent risk factor for cardiovascular events and mortality. However, despite extensive research, the pathogenesis of salt-sensitive hypertension is still unclear and tremendously challenged by its multifactorial etiology, complicated genetic influences, and the unavailability of a diagnostic tool. So far, the important roles of the renin-angiotensin-aldosterone system, sympathetic nervous system, and immune system in the pathogenesis of salt-sensitive hypertension have been studied. In the first part of this review, we focus on how the systems mentioned above are aberrantly regulated in salt-sensitive hypertension. We follow this with an emphasis on genetic variants in those systems that are associated with and/or increase predisposition to salt-sensitivity in humans.
Collapse
Affiliation(s)
- Dina Maaliki
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Maha M. Itani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hana A. Itani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
8
|
Rinschen MM, Palygin O, El-Meanawy A, Domingo-Almenara X, Palermo A, Dissanayake LV, Golosova D, Schafroth MA, Guijas C, Demir F, Jaegers J, Gliozzi ML, Xue J, Hoehne M, Benzing T, Kok BP, Saez E, Bleich M, Himmerkus N, Weisz OA, Cravatt BF, Krüger M, Benton HP, Siuzdak G, Staruschenko A. Accelerated lysine metabolism conveys kidney protection in salt-sensitive hypertension. Nat Commun 2022; 13:4099. [PMID: 35835746 PMCID: PMC9283537 DOI: 10.1038/s41467-022-31670-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 06/27/2022] [Indexed: 01/07/2023] Open
Abstract
Hypertension and kidney disease have been repeatedly associated with genomic variants and alterations of lysine metabolism. Here, we combined stable isotope labeling with untargeted metabolomics to investigate lysine's metabolic fate in vivo. Dietary 13C6 labeled lysine was tracked to lysine metabolites across various organs. Globally, lysine reacts rapidly with molecules of the central carbon metabolism, but incorporates slowly into proteins and acylcarnitines. Lysine metabolism is accelerated in a rat model of hypertension and kidney damage, chiefly through N-alpha-mediated degradation. Lysine administration diminished development of hypertension and kidney injury. Protective mechanisms include diuresis, further acceleration of lysine conjugate formation, and inhibition of tubular albumin uptake. Lysine also conjugates with malonyl-CoA to form a novel metabolite Nε-malonyl-lysine to deplete malonyl-CoA from fatty acid synthesis. Through conjugate formation and excretion as fructoselysine, saccharopine, and Nε-acetyllysine, lysine lead to depletion of central carbon metabolites from the organism and kidney. Consistently, lysine administration to patients at risk for hypertension and kidney disease inhibited tubular albumin uptake, increased lysine conjugate formation, and reduced tricarboxylic acid (TCA) cycle metabolites, compared to kidney-healthy volunteers. In conclusion, lysine isotope tracing mapped an accelerated metabolism in hypertension, and lysine administration could protect kidneys in hypertensive kidney disease.
Collapse
Affiliation(s)
- Markus M Rinschen
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- III. Medical Clinic, University Hospital Hamburg Eppendorf, Hamburg, Germany.
- AIAS, Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Aarhus, Denmark.
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Ashraf El-Meanawy
- Division of Nephrology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Xavier Domingo-Almenara
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
- Omics Sciences Unit, EURECAT, Technology Centre of Catalonia, Reus, Catalonia, Spain
| | - Amelia Palermo
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Lashodya V Dissanayake
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, 33602, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Daria Golosova
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | | | - Carlos Guijas
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
| | - Fatih Demir
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Megan L Gliozzi
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Jingchuan Xue
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
| | - Martin Hoehne
- Center for Molecular Medicine Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
- Department II of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Thomas Benzing
- Center for Molecular Medicine Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
- Department II of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Bernard P Kok
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, 92037, USA
| | - Enrique Saez
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, 92037, USA
| | - Markus Bleich
- Institute of Physiology, University Kiel, Kiel, Germany
| | | | - Ora A Weisz
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | | | - Marcus Krüger
- Center for Molecular Medicine Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
| | - H Paul Benton
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
| | - Gary Siuzdak
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA.
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, 33602, USA.
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
- James A. Haley Veterans' Hospital, Tampa, FL, 33612, USA.
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL, 33602, USA.
| |
Collapse
|
9
|
Shi M, He J, Li C, Lu X, He WJ, Cao J, Chen J, Chen JC, Bazzano LA, Li JX, He H, Gu D, Kelly TN. Metabolomics study of blood pressure salt-sensitivity and hypertension. Nutr Metab Cardiovasc Dis 2022; 32:1681-1692. [PMID: 35599090 PMCID: PMC9596959 DOI: 10.1016/j.numecd.2022.04.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND AIMS Identify novel metabolite associations with blood pressure (BP) salt-sensitivity and hypertension. METHODS AND RESULTS The Genetic Epidemiology Network of Salt Sensitivity (GenSalt) Replication study includes 698 Chinese participants who underwent a 3-day baseline examination followed by a 7-day low-sodium feeding and 7-day high-sodium feeding. Latent mixture models identified three trajectories of blood pressure (BP) responses to the sodium interventions. We selected 50 most highly salt-sensitive and 50 most salt-resistant participants for untargeted metabolomics profiling. Multivariable adjusted mixed logistic regression models tested the associations of baseline metabolites with BP salt-sensitivity. Multivariable adjusted mixed linear regression models tested the associations of BP salt-sensitivity with metabolite changes during the sodium interventions. Identified metabolites were tested for associations with hypertension among 1249 Bogalusa Heart Study (BHS) participants using multiple logistic regression. Fifteen salt-sensitivity metabolites were associated with hypertension in the BHS. Baseline values of serine, 2-methylbutyrylcarnitine and isoleucine directly associated with high salt-sensitivity. Among them, serine indirectly associated with hypertension while 2-methylbutyrylcarnitine and isoleucine directly associated with hypertension. Baseline salt-sensitivity status predicted changes in 14 metabolites when switching to low-sodium or high-sodium interventions. Among them, glutamate, 1-carboxyethylvaline, 2-methylbutyrylcarnitine, 3-methoxytyramine sulfate, glucose, alpha-ketoglutarate, hexanoylcarnitine, gamma-glutamylisoleucine, gamma-glutamylleucine, and gamma-glutamylphenylalanine directly associated with hypertension. Conversely, serine, histidine, threonate and 5-methyluridine indirectly associated with hypertension. Together, these metabolites explained an additional 7% of hypertension susceptibility when added to a model including traditional risk factors. CONCLUSIONS Our findings contribute to the molecular characterization of BP response to sodium and provide novel biological insights into salt-sensitive hypertension.
Collapse
Affiliation(s)
- Mengyao Shi
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States; Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Changwei Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Xiangfeng Lu
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing, China
| | - William J He
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, United States; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Jie Cao
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Chen
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States; Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Ji-Chun Chen
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing, China
| | - Lydia A Bazzano
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Jian-Xin Li
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing, China
| | - Hua He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Dongfeng Gu
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing, China
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States.
| |
Collapse
|
10
|
Serum Metabolites Associated with Blood Pressure in Chronic Kidney Disease Patients. Metabolites 2022; 12:metabo12040281. [PMID: 35448468 PMCID: PMC9027690 DOI: 10.3390/metabo12040281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 02/05/2023] Open
Abstract
Blood pressure is one of the most basic health screenings and it has a complex relationship with chronic kidney disease (CKD). Controlling blood pressure for CKD patients is crucial for curbing kidney function decline and reducing the risk of cardiovascular disease. Two independent CKD cohorts, including matched controls (discovery n = 824; validation n = 552), were recruited. High-throughput metabolomics was conducted with the patients’ serum samples using mass spectrometry. After controlling for CKD severity and other clinical hypertension risk factors, we identified ten metabolites that have significant associations with blood pressure. The quantitative importance of these metabolites was verified in a fully connected neural network model. Of the ten metabolites, seven have not previously been associated with blood pressure. The metabolites that had the strongest positive association with blood pressure were aspartylglycosamine (p = 4.58 × 10−5), fructose-1,6-diphosphate (p = 1.19 × 10−4) and N-Acetylserine (p = 3.27 × 10−4). Three metabolites that were negatively associated with blood pressure (phosphocreatine, p = 6.39 × 10−3; dodecanedioic acid, p = 0.01; phosphate, p = 0.04) have been reported previously to have beneficial effects on hypertension. These results suggest that intake of metabolites as supplements may help to control blood pressure in CKD patients.
Collapse
|
11
|
Correia MJ, Pimpão AB, Fernandes DGF, Morello J, Sequeira CO, Calado J, Antunes AMM, Almeida MS, Branco P, Monteiro EC, Vicente JB, Serpa J, Pereira SA. Cysteine as a Multifaceted Player in Kidney, the Cysteine-Related Thiolome and Its Implications for Precision Medicine. Molecules 2022; 27:1416. [PMID: 35209204 PMCID: PMC8874463 DOI: 10.3390/molecules27041416] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 11/16/2022] Open
Abstract
In this review encouraged by original data, we first provided in vivo evidence that the kidney, comparative to the liver or brain, is an organ particularly rich in cysteine. In the kidney, the total availability of cysteine was higher in cortex tissue than in the medulla and distributed in free reduced, free oxidized and protein-bound fractions (in descending order). Next, we provided a comprehensive integrated review on the evidence that supports the reliance on cysteine of the kidney beyond cysteine antioxidant properties, highlighting the relevance of cysteine and its renal metabolism in the control of cysteine excess in the body as a pivotal source of metabolites to kidney biomass and bioenergetics and a promoter of adaptive responses to stressors. This view might translate into novel perspectives on the mechanisms of kidney function and blood pressure regulation and on clinical implications of the cysteine-related thiolome as a tool in precision medicine.
Collapse
Affiliation(s)
- Maria João Correia
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - António B. Pimpão
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - Dalila G. F. Fernandes
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA), 2780-157 Oeiras, Portugal; (D.G.F.F.); (J.B.V.)
| | - Judit Morello
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - Catarina O. Sequeira
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - Joaquim Calado
- Centre for Toxicogenomics and Human Health (ToxOmics), Genetics, Oncology and Human Toxicology, Nova Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal;
- Nephrology Department, Centro Hospitalar Universitário de Lisboa Central, 1069-166 Lisboa, Portugal
| | - Alexandra M. M. Antunes
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, 1049-001 Lisboa, Portugal;
| | - Manuel S. Almeida
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
- Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, 2790-134 Carnaxide, Portugal
| | - Patrícia Branco
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
- Hospital de Santa Cruz, Centro Hospitalar de Lisboa Ocidental, 2790-134 Carnaxide, Portugal
| | - Emília C. Monteiro
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| | - João B. Vicente
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA), 2780-157 Oeiras, Portugal; (D.G.F.F.); (J.B.V.)
| | - Jacinta Serpa
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), 1099-023 Lisboa, Portugal
| | - Sofia A. Pereira
- CEDOC, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal; (M.J.C.); (A.B.P.); (J.M.); (C.O.S.); (M.S.A.); (P.B.); (E.C.M.); (J.S.)
| |
Collapse
|
12
|
Zhang X, Tomar N, Kandel SM, Audi SH, Cowley AW, Dash RK. Substrate- and Calcium-Dependent Differential Regulation of Mitochondrial Oxidative Phosphorylation and Energy Production in the Heart and Kidney. Cells 2021; 11:131. [PMID: 35011693 PMCID: PMC8750792 DOI: 10.3390/cells11010131] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/26/2021] [Accepted: 12/28/2021] [Indexed: 11/16/2022] Open
Abstract
Mitochondrial dehydrogenases are differentially stimulated by Ca2+. Ca2+ has also diverse regulatory effects on mitochondrial transporters and other enzymes. However, the consequences of these regulatory effects on mitochondrial oxidative phosphorylation (OxPhos) and ATP production, and the dependencies of these consequences on respiratory substrates, have not been investigated between the kidney and heart despite the fact that kidney energy requirements are second only to those of the heart. Our objective was, therefore, to elucidate these relationships in isolated mitochondria from the kidney outer medulla (OM) and heart. ADP-induced mitochondrial respiration was measured at different CaCl2 concentrations in the presence of various respiratory substrates, including pyruvate + malate (PM), glutamate + malate (GM), alpha-ketoglutarate + malate (AM), palmitoyl-carnitine + malate (PCM), and succinate + rotenone (SUC + ROT). The results showed that, in both heart and OM mitochondria, and for most complex I substrates, Ca2+ effects are biphasic: small increases in Ca2+ concentration stimulated, while large increases inhibited mitochondrial respiration. Furthermore, significant differences in substrate- and Ca2+-dependent O2 utilization towards ATP production between heart and OM mitochondria were observed. With PM and PCM substrates, Ca2+ showed more prominent stimulatory effects in OM than in heart mitochondria, while with GM and AM substrates, Ca2+ had similar biphasic regulatory effects in both OM and heart mitochondria. In contrast, with complex II substrate SUC + ROT, only inhibitory effects on mitochondrial respiration was observed in both the heart and the OM. We conclude that the regulatory effects of Ca2+ on mitochondrial OxPhos and ATP synthesis are biphasic, substrate-dependent, and tissue-specific.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
| | - Namrata Tomar
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
| | - Sunil M. Kandel
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
| | - Said H. Audi
- Department of Biomedical Engineering, Marquette University, Milwaukee, WI 53223, USA;
| | - Allen W. Cowley
- Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ranjan K. Dash
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (X.Z.); (N.T.); (S.M.K.)
- Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
13
|
Zheng X, Zhou L, Jin Y, Zhao X, Ahmad H, OuYang Y, Chen S, Du J, Chen X, Chen L, Gao D, Yang Z, Tian Z. β-Aminoisobutyric acid supplementation attenuated salt-sensitive hypertension in Dahl salt-sensitive rats through prevention of insufficient fumarase. Amino Acids 2021; 54:169-180. [PMID: 34837556 DOI: 10.1007/s00726-021-03092-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/13/2021] [Indexed: 10/19/2022]
Abstract
The human Dietary Approaches to Stop Hypertension-Sodium Trial has shown that β-aminoisobutyric acid (BAIBA) may prevent the development of salt-sensitive hypertension (SSHT). However, the specific antihypertensive mechanism remains unclear in the renal tissues of salt-sensitive (SS) rats. In this study, BAIBA (100 mg/kg/day) significantly attenuated SSHT via increased nitric oxide (NO) content in the renal medulla, and it induced a significant increase in NO synthesis substrates (L-arginine and malic acid) in the renal medulla. BAIBA enhanced the activity levels of total NO synthase (NOS), inducible NOS, and constitutive NOS. BAIBA resulted in increased fumarase activity and decreased fumaric acid content in the renal medulla. The high-salt diet (HSD) decreased fumarase expression in the renal cortex, and BAIBA increased fumarase expression in the renal medulla and renal cortex. Furthermore, in the renal medulla, BAIBA increased the levels of ATP, ADP, AMP, and ADP/ATP ratio, thus further activating AMPK phosphorylation. BAIBA prevented the decrease in renal medullary antioxidative defenses induced by the HSD. In conclusion, BAIBA's antihypertensive effect was underlined by the phosphorylation of AMPK, the prevention of fumarase's activity reduction caused by the HSD, and the enhancement of NO content, which in concert attenuated SSHT in SS rats.
Collapse
Affiliation(s)
- Xuewei Zheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Luxin Zhou
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yuexin Jin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xinrui Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Hussain Ahmad
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yanan OuYang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Sa Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jie Du
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xiangbo Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Lan Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Di Gao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhongmin Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
14
|
Ruan Z, Li J, Liu F, Cao J, Chen S, Chen J, Huang K, Wang Y, Li H, Wang Y, Xue Z, Wang L, Huang J, Gu D, Lu X. Study design, general characteristics of participants, and preliminary findings from the metabolome, microbiome, and dietary salt intervention study (MetaSalt). Chronic Dis Transl Med 2021; 7:227-234. [PMID: 34786542 PMCID: PMC8579015 DOI: 10.1016/j.cdtm.2021.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Indexed: 12/05/2022] Open
Abstract
Background High sodium intake is an important risk factor for hypertension and cardiovascular disease. However, the association between gut microbiota composition and metabolomic profiles with dietary sodium intake and blood pressure (BP) is not well-understood. The metabolome, microbiome, and dietary salt intervention (MetaSalt) study aimed to investigate microbial and metabolomic profiles related to dietary sodium intake and BP regulation. Methods This family-based intervention study was conducted in four communities across three provinces in rural northern China in 2019. Probands with untreated prehypertension or stage-1 hypertension were identified through community-based BP screening, and family members including siblings, offspring, spouses, and parents were subsequently included. All participants participated in a 3-day baseline examination with usual diet consumption, followed by a 10-day low-salt diet (3 g/d of salt or 51.3 mmol/d of sodium) and a 10-day high-salt diet (18 g/d of salt or 307.8 mmol/d of sodium). Differences in mean BP levels were compared according to the intervention phases using a paired Student's t-test. Results A total of 528 participants were included in this study, with a mean age of 48.1 years, 36.7% of whom were male, 76.8% had a middle school (69.7%) or higher (7.1%) diploma, 23.4% had a history of smoking, and 24.4% were current drinkers. The mean arterial pressure at baseline was 97.2 ± 10.5 mm Hg for all participants, and significantly decreased during the low-salt intervention (93.8 ± 9.3, P < 0.0001) and subsequently increased during the high-salt intervention (96.4 ± 10.0, P < 0.0001). Conclusions Our dietary salt intervention study has successfully recruited participants and will facilitate to evaluate the effects of gut microbiota and metabolites on BP regulation in response to sodium burden, which will provide important evidence for investigating the underlying mechanisms in the development of hypertension and subsequent cardiovascular diseases. Trial registration The study was registered in the Chinese Clinical Trial Registry database (ChiCTR1900025171).
Collapse
Affiliation(s)
- Zengliang Ruan
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jianxin Li
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Fangchao Liu
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jie Cao
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Shufeng Chen
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jichun Chen
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Keyong Huang
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Yaqin Wang
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Hongfan Li
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Yan Wang
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Zhongyu Xue
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Laiyuan Wang
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jianfeng Huang
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Dongfeng Gu
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China.,School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Xiangfeng Lu
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| |
Collapse
|
15
|
Razavi MA, Bazzano LA, Nierenberg J, Huang Z, Fernandez C, Razavi AC, Whelton SP, He J, Kelly TN. Advances in Genomics Research of Blood Pressure Responses to Dietary Sodium and Potassium Intakes. Hypertension 2021; 78:4-15. [PMID: 33993724 DOI: 10.1161/hypertensionaha.121.16509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
More than half of US adults have hypertension by 40 years of age and a subsequent increase in atherosclerotic cardiovascular disease risk. Dietary sodium and potassium are intricately linked to the pathophysiology of hypertension. However, blood pressure responses to dietary sodium and potassium, phenomena known as salt and potassium sensitivity of blood pressure, respectively, are heterogenous and normally distributed in the general population. Like blood pressure, salt and potassium sensitivity are complex phenotypes, and previous research has shown that up to 75% of individuals experience a blood pressure change in response to such dietary minerals. Previous research has also implicated both high salt sensitivity and low salt sensitivity (or salt resistance) of blood pressure to an increased risk of hypertension and potentially atherosclerotic cardiovascular disease risk. Given the clinical challenges required to accurately measure the sodium and potassium response phenotypes, genomic characterization of these traits has become of interest for hypertension prevention initiatives on both the individual and population levels. Here, we review advances in human genomics research of blood pressure responses to dietary sodium and potassium by focusing on 3 main areas, including the phenotypic characterization of salt sensitivity and resistance, clinical challenges in diagnosing such phenotypes, and the genomic mechanisms that may help to explain salt and potassium sensitivity and resistance. Through this process, we hope to further underline the value of leveraging genomics and broader multiomics for characterizing the blood pressure response to sodium and potassium to improve precision in lifestyle approaches for primordial and primary atherosclerotic cardiovascular disease prevention.
Collapse
Affiliation(s)
| | - Lydia A Bazzano
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (L.A.B., Z.H., C.F., A.C.R., J.H., T.N.K.)
| | - Jovia Nierenberg
- Department of Epidemiology and Biostatistics, University of California, San Francisco School of Medicine (J.N.)
| | - Zhijie Huang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (L.A.B., Z.H., C.F., A.C.R., J.H., T.N.K.)
| | - Camilo Fernandez
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA (C.F., A.C.R., J.H.).,Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (L.A.B., Z.H., C.F., A.C.R., J.H., T.N.K.)
| | - Alexander C Razavi
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA (C.F., A.C.R., J.H.).,Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (L.A.B., Z.H., C.F., A.C.R., J.H., T.N.K.)
| | - Seamus P Whelton
- The Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD (S.P.W.)
| | - Jiang He
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA (C.F., A.C.R., J.H.).,Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (L.A.B., Z.H., C.F., A.C.R., J.H., T.N.K.)
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (L.A.B., Z.H., C.F., A.C.R., J.H., T.N.K.)
| |
Collapse
|
16
|
A Systematic Review of the Efficacy of DASH Diet in Lowering Blood Pressure Among Hypertensive Adults. TOP CLIN NUTR 2021. [DOI: 10.1097/tin.0000000000000238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
17
|
Abstract
Hypertension is a leading risk factor for disease burden worldwide. The kidneys, which have a high specific metabolic rate, play an essential role in the long-term regulation of arterial blood pressure. In this review, we discuss the emerging role of renal metabolism in the development of hypertension. Renal energy and substrate metabolism is characterized by several important and, in some cases, unique features. Recent advances suggest that alterations of renal metabolism may result from genetic abnormalities or serve initially as a physiological response to environmental stressors to support tubular transport, which may ultimately affect regulatory pathways and lead to unfavorable cellular and pathophysiological consequences that contribute to the development of hypertension.
Collapse
Affiliation(s)
- Zhongmin Tian
- grid.43169.390000 0001 0599 1243The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, Shaanxi China
| | - Mingyu Liang
- grid.30760.320000 0001 2111 8460Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin, Milwaukee, WI USA
| |
Collapse
|
18
|
Xu F, Zhu J, Chen Y, He K, Guo J, Bai S, Zhao R, Du J, Shen B. Physical interaction of tropomyosin 3 and STIM1 regulates vascular smooth muscle contractility and contributes to hypertension. Biomed Pharmacother 2021; 134:111126. [PMID: 33341060 DOI: 10.1016/j.biopha.2020.111126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/26/2020] [Accepted: 12/08/2020] [Indexed: 12/01/2022] Open
Abstract
SCOPE Tropomyosin (TPM), an actin-binding protein widely expressed across different cell types, is primarily involved in cellular contractile processes. We investigated whether TPM3 physically and functionally interacts with stromal interaction molecule 1 (STIM1) to contribute to vascular smooth muscle cell (VSMC) contraction, store-operated calcium entry (SOCE), and high-salt intake-induced hypertension in rats. METHODS AND RESULTS Analysis of a rat RNA-seq data set of 80 samples showed that the STIM1 and Tpm3 transcriptome expression pattern is highly correlated, and co-immunoprecipitation results indicated that TPM3 and STIM1 proteins physically interacted in rat VSMCs. Immunohistochemical data displayed obvious co-localization of TPM3 and STIM1 in rat VSMCs. Knockdown of TPM3 or STIM1 in VSMCs with specific small interfering RNA significantly suppressed contractions in tension measurement assays and decreased SOCE in calcium assays. Rats fed a high-salt diet for 4 weeks had significantly higher systolic blood pressure than controls, with significantly increased contractility and markedly increased TPM3 and STIM1 expression levels in the mesenteric resistance artery (shown by tension measurements and immunoblotting, respectively). Additionally, high salt environment in vitro induced significant enhancement of TPM3 and STIM1 expression levels in VSMCs. CONCLUSIONS We showed for the first time that TPM3 and STIM1 physically and functionally interact to contribute to VSMC contraction, SOCE, and high-salt intake-induced hypertension. Our findings provide mechanistic insights and offer a potential therapeutic target for high-salt intake-induced hypertension.
Collapse
MESH Headings
- Animals
- Blood Pressure
- Cells, Cultured
- Databases, Genetic
- Disease Models, Animal
- Hypertension/chemically induced
- Hypertension/genetics
- Hypertension/metabolism
- Hypertension/physiopathology
- Male
- Mesenteric Arteries/metabolism
- Mesenteric Arteries/physiopathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Protein Binding
- Rats, Sprague-Dawley
- Signal Transduction
- Sodium Chloride, Dietary
- Stromal Interaction Molecule 1/genetics
- Stromal Interaction Molecule 1/metabolism
- Transcriptome
- Tropomyosin/genetics
- Tropomyosin/metabolism
- Vasoconstriction
- Rats
Collapse
Affiliation(s)
- Fangfang Xu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Jinhang Zhu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Ye Chen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Ke He
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, China
| | - Jizheng Guo
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Suwen Bai
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Ren Zhao
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, China
| | - Juan Du
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Bing Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
19
|
Zheng X, Zhao X, Jin Y, Zhou L, Yang P, Ahmad H, Tian Z. High salt diet contributes to hypertension by weakening the medullary tricarboxylic acid cycle and antioxidant system in Dahl salt-sensitive rats. Biochimie 2020; 181:154-161. [PMID: 33347925 DOI: 10.1016/j.biochi.2020.12.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/04/2020] [Accepted: 12/14/2020] [Indexed: 01/11/2023]
Abstract
High salt diet (HSD, 8% NaCl) contributes to salt-sensitive hypertension, this study aimed to determine the effect of HSD on salt-sensitive hypertension by combining proteomic with metabolomics methods. Salt-sensitive rats were fed on HSD and normal salt diet (NSD, 0.4% NaCl) for two weeks before further analysis. Proteomic analysis showed the differential expression proteins (DEPs) were primarily mapped in the tricarboxylic acid (TCA)-cycle, glycolysis/gluconeogenesis, and other pathways associated with multiple amino acids. HSD decreased the medullary activities and protein expression level of two key enzymes of TCA-cycle, MDH and NADP+-IDH. Metabolomics showed three serous TCA-cycle-associated compounds, including decreased malic acid, decreased citric acid, and increased fumaric acid were differentially detected, which resulted in a decrease in NO content and an increase in H2O2 content in serum. The content of GSH, GSH/GSSG ratio, and synthesis substrates of GSH-cysteine and glycine, were significantly decreased by HSD, thus attenuated the antioxidant system in the renal medulla. HSD enhanced the medullary pentose phosphate pathway, which finally increased the concentration of NADPH and NADP+, NADPH/NADP+, and the activity of NADPH oxidase in the renal medulla. Additionally, HSD enhanced the glycolysis pathway in the renal medulla. In summary, HSD significantly weakened the TCA cycle, and attenuated the antioxidant system in the renal medulla, which finally contributed to salt-sensitive hypertension.
Collapse
Affiliation(s)
- Xuewei Zheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xinrui Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yuexin Jin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Luxin Zhou
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Pengfei Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Hussain Ahmad
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhongmin Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
20
|
He FJ, Tan M, Ma Y, MacGregor GA. Salt Reduction to Prevent Hypertension and Cardiovascular Disease: JACC State-of-the-Art Review. J Am Coll Cardiol 2020; 75:632-647. [PMID: 32057379 DOI: 10.1016/j.jacc.2019.11.055] [Citation(s) in RCA: 286] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/04/2019] [Accepted: 11/19/2019] [Indexed: 12/21/2022]
Abstract
There is strong evidence for a causal relationship between salt intake and blood pressure. Randomized trials demonstrate that salt reduction lowers blood pressure in both individuals who are hypertensive and those who are normotensive, additively to antihypertensive treatments. Methodologically robust studies with accurate salt intake assessment have shown that a lower salt intake is associated with a reduced risk of cardiovascular disease, all-cause mortality, and other conditions, such as kidney disease, stomach cancer, and osteoporosis. Multiple complex and interconnected physiological mechanisms are implicated, including fluid homeostasis, hormonal and inflammatory mechanisms, as well as more novel pathways such as the immune response and the gut microbiome. High salt intake is a top dietary risk factor. Salt reduction programs are cost-effective and should be implemented or accelerated in all countries. This review provides an update on the evidence relating salt to health, with a particular focus on blood pressure and cardiovascular disease, as well as the potential mechanisms.
Collapse
Affiliation(s)
- Feng J He
- Wolfson Institute of Preventive Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.
| | - Monique Tan
- Wolfson Institute of Preventive Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Yuan Ma
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Graham A MacGregor
- Wolfson Institute of Preventive Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
21
|
Gomes A, Godinho-Pereira J, Oudot C, Sequeira CO, Macià A, Carvalho F, Motilva MJ, Pereira SA, Matzapetakis M, Brenner C, Santos CN. Berry fruits modulate kidney dysfunction and urine metabolome in Dahl salt-sensitive rats. Free Radic Biol Med 2020; 154:119-131. [PMID: 32437928 DOI: 10.1016/j.freeradbiomed.2020.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/21/2020] [Accepted: 05/02/2020] [Indexed: 01/12/2023]
Abstract
Berries are rich sources of (poly)phenols which have been associated with the prevention of cardiovascular diseases in animal models and in human clinical trials. Recently, a berry enriched diet was reported to decrease blood pressure and attenuate kidney disease progression on Dahl salt-sensitive rats. However, the relationship between kidney function, metabolism and (poly)phenols was not evaluated. We hypothesize that berries promote metabolic alterations concomitantly with an attenuation of the progression of renal disease. For that, kidney and urinary metabolomic changes induced by the berry enriched diet in hypertensive rats (Dahl salt-sensitive) were analyzed using liquid chromatography (UPLC-MS/MS) and 1H NMR techniques. Moreover, physiological and metabolic parameters, and kidney histopathological data were also collected. The severity of the kidney lesions promoted in Dahl rats by a high salt diet was significantly reduced by berries, namely a decrease in sclerotic glomeruli. In addition, was observed a high urinary excretion of metabolites that are indicators of alterations in glycolysis/gluconeogenesis, citrate cycle, and pyruvate metabolism in the salt induced-hypertensive rats, a metabolic profile counteracted by berries consumption. We also provide novel insights that relates (poly)phenols consumption with alterations in cysteine redox pools. Cysteine contribute to the redox signaling that is normally disrupted during kidney disease onset and progression. Our findings provide a vision about the metabolic responses of hypertensive rats to a (poly)phenol enriched diet, which may contribute to the understanding of the beneficial effects of (poly)phenols in salt-induced hypertension.
Collapse
Affiliation(s)
- A Gomes
- Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade NOVA de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | - J Godinho-Pereira
- Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade NOVA de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | - C Oudot
- INSERM UMR-S 1180, University of Paris-Sud, University of Paris-Saclay, 5 rue Jean-Baptiste Clément, 92296, Châtenay Malabry, France
| | - C O Sequeira
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisboa, Portugal
| | - A Macià
- Food Technology Department, Agrotecnio Center, Escuela Técnica Superior de Ingeniería Agraria, University of Lleida, Lleida, Spain
| | - F Carvalho
- Laboratório de Morfologia Renal, Hospital Curry Cabral, EPE, Rua da Beneficência n. 8, 1069-166, Lisboa, Portugal
| | - M J Motilva
- Instituto de Ciencias de la Vid y del Vino-ICVV, CSIC-Universidad de La Rioja-Gobierno de La Rioja, Finca "La Grajera", Carretera de Burgos km 6, 26007, Logroño, La Rioja, Spain
| | - S A Pereira
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisboa, Portugal
| | - M Matzapetakis
- Instituto de Tecnologia Química e Biológica, Universidade NOVA de Lisboa, Av. da República, 2780-157, Oeiras, Portugal
| | - C Brenner
- Université Paris-Saclay, CNRS, Institut Gustave Roussy, Aspects métaboliques et systémiques de l'oncogénèse pour de nouvelles approches thérapeutiques, 94805, Villejuif, France
| | - C N Santos
- Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901, Oeiras, Portugal; Instituto de Tecnologia Química e Biológica, Universidade NOVA de Lisboa, Av. da República, 2780-157, Oeiras, Portugal; CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisboa, Portugal.
| |
Collapse
|
22
|
Metabolomics Elucidates Dose-Dependent Molecular Beneficial Effects of Hesperidin Supplementation in Rats Fed an Obesogenic Diet. Antioxidants (Basel) 2020; 9:antiox9010079. [PMID: 31963315 PMCID: PMC7023145 DOI: 10.3390/antiox9010079] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/07/2020] [Accepted: 01/14/2020] [Indexed: 02/07/2023] Open
Abstract
Metabolic syndrome (MetS) is a global epidemic concern. Polyphenols are proposed as good candidates for its prevention, although their mechanisms are not fully understood. The gut microbiota seems to play a key role in polyphenol beneficial effects. Here, we assessed the effects of the citrus polyphenol hesperidin combining an untargeted metabolomics approach, which has an inherent potential to elucidate the host-microbiome interplay, with extensive anthropometric and biochemical characterizations and integrating metabolomics results with our previous 16S rRNA bacterial sequencing data. The rats were fed either a standard or an obesogenic cafeteria diet (CAF) for 17 weeks. After nine weeks, rats were supplemented with vehicle; low- (H1), or high- (H2) hesperidin doses. CAF animals developed MetS features. Hesperidin supplementation in CAF rats decreased the total cholesterol, LDL-C, and free fatty acids. The highest hesperidin dose also ameliorated blood pressure, insulin sensitivity, and decreased markers of arterial stiffness and inflammation. Metabolomics revealed an improvement of the lipidomic profile, decreases in circulating amino acids, and lower excretions of inflammation- and oxidative stress-related metabolites. Bacteroidaceae increases in the CAF-H2 group paralleled higher excretions of microbial-derived metabolites. Overall, our results provide detailed insights into the molecular effects of hesperidin on MetS and suggest that it is a promising prebiotic for the treatment of MetS and related conditions.
Collapse
|
23
|
Zheng X, Li X, Chen M, Yang P, Zhao X, Zeng L, OuYang Y, Yang Z, Tian Z. The protective role of hawthorn fruit extract against high salt-induced hypertension in Dahl salt-sensitive rats: impact on oxidative stress and metabolic patterns. Food Funct 2019; 10:849-858. [PMID: 30681096 DOI: 10.1039/c8fo01818a] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In the present study, the renal-protective effect of hawthorn fruit extract (HW) on high-salt hypertension and its effect on metabolic patterns are determined. High salt causes hypertension in Dahl salt sensitive (SS) rats, while HW can effectively attenuate high-salt induced hypertension, and, various antihypertensive ingredients of HW have also been successfully identified using GC/MS. Of note, the biochemical assay indicates that HW significantly increases the concentration of nitric oxide (NO) and decreases the concentration of H2O2 and malonaldehyde. Especially, HW increases the activities of NO synthase and catalase in the renal medulla. Simultaneously, the renal cortex and medulla, harvested from SS rats, are used to perform the metabolomics analysis, and then, 11 and 8 differential metabolites are identified in the renal medulla and cortex with the HW gavage, respectively. All differential metabolites are then used to perform the pathway enrichment analysis. The results show that many metabolic pathways are enriched in both the renal medulla and cortex, especially those in the medulla including 23 enriched pathways. Therefore, it provides evidence that HW confers an antioxidant effect on high-salt induced hypertension and dramatically alters the metabolic patterns of SS rats, and the antihypertensive ingredients of HW also further indicate that it may be used as a nutritional supplemental therapeutic drug to protect against high-salt induced hypertension in the renal medulla.
Collapse
Affiliation(s)
- Xuewei Zheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Poggiogalle E, Fontana M, Giusti AM, Pinto A, Iannucci G, Lenzi A, Donini LM. Amino Acids and Hypertension in Adults. Nutrients 2019; 11:nu11071459. [PMID: 31252583 PMCID: PMC6683075 DOI: 10.3390/nu11071459] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/12/2019] [Accepted: 06/19/2019] [Indexed: 01/19/2023] Open
Abstract
Accumulating evidence suggests a potential role of dietary protein among nutritional factors interfering with the regulation of blood pressure. Dietary protein source (plant versus animal protein), and especially, protein composition in terms of amino acids has been postulated to interfere with mechanisms underlying the development of hypertension. Recently, mounting interest has been directed at amino acids in hypertension focusing on habitual dietary intake and their circulating levels regardless of single amino acid dietary supplementation. The aim of the present review was to summarize epidemiological evidence concerning the connection between amino acids and hypertension. Due to the large variability in methodologies used for assessing amino acid levels and heterogeneity in the results obtained, it was not possible to draw robust conclusions. Indeed, some classes of amino acids or individual amino acids showed non-causative association with blood pressure as well as the incidence of hypertension, but the evidence was far from being conclusive. Further research should be prompted for a thorough understanding of amino acid effects and synergistic actions of different amino acid classes on blood pressure regulation.
Collapse
Affiliation(s)
- Eleonora Poggiogalle
- Department of Experimental Medicine-Medical Pathophysiology, Food Science and Endocrinology Section; Sapienza University of Rome, 00185 Rome, Italy.
| | - Mario Fontana
- Department of Biochemical Sciences "A. Rossi-Fanelli"; Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Anna Maria Giusti
- Department of Experimental Medicine-Medical Pathophysiology, Food Science and Endocrinology Section; Sapienza University of Rome, 00185 Rome, Italy
| | - Alessandro Pinto
- Department of Experimental Medicine-Medical Pathophysiology, Food Science and Endocrinology Section; Sapienza University of Rome, 00185 Rome, Italy
| | - Gino Iannucci
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Viale del Policlinico 155, 00165 Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine-Medical Pathophysiology, Food Science and Endocrinology Section; Sapienza University of Rome, 00185 Rome, Italy
| | - Lorenzo Maria Donini
- Department of Experimental Medicine-Medical Pathophysiology, Food Science and Endocrinology Section; Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
25
|
Xue H, Geurts AM, Usa K, Wang F, Lin Y, Phillips J, Henderson L, Baker MA, Tian Z, Liang M. Fumarase Overexpression Abolishes Hypertension Attributable to endothelial NO synthase Haploinsufficiency in Dahl Salt-Sensitive Rats. Hypertension 2019; 74:313-322. [PMID: 31230549 DOI: 10.1161/hypertensionaha.119.12723] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Human blood pressure salt sensitivity is associated with changes in urinary metabolites related to fumarase (Fh) and nitric oxide (NO) metabolism, and fumarase promotes NO production through an arginine regeneration pathway. We examined the role of the fumarase-NO pathway in the development of hypertension using genetically engineered rat models. Dahl salt-sensitive (SS) rats with heterozygous mutation of eNOS (endothelial NO synthase or Nos3; SS-Nos3+/-) were bred with SS rats with a hemizygous Fh transgene. SS-Nos3+/- rats without the Fh transgene (SS-Nos3+/-/Fh0/0) developed substantial hypertension with a mean arterial pressure of 134.2±3.7 mm Hg on a 0.4% NaCl diet and 178.0±3.5 mm Hg after 14 days on a 4% NaCl diet. Mean arterial pressure decreased remarkably to 123.1±1.4 mm Hg on 0.4% NaCl, and 143.3±1.5 mm Hg on 4% NaCl in SS-Nos3+/- rats with a Fh transgene (SS-Nos3+/-/Fh0/1), and proteinuria, renal fibrosis, and tubular casts were attenuated in SS-Nos3+/-/Fh0/1 rats compared with SS-Nos3+/-/Fh0/0 rats. eNOS protein abundance decreased in rats with the Nos3 heterozygous mutation, which was not influenced by Fh overexpression in rats on the 0.4% NaCl diet. However, the decrease in NO metabolite in the renal outer medulla of SS-Nos3+/-/Fh0/0 rats on the 0.4% NaCl diet was reversed in SS-Nos3+/-/Fh0/1 rats, and levels of L-arginine, but not the other 12 amino acids analyzed, were significantly higher in SS-Nos3+/-/Fh0/1 rats than in SS-Nos3+/+/Fh0/0 rats. In conclusion, fumarase has potent effects in restoring NO production and blunting the development of hypertension attributable to eNOS haploinsufficiency.
Collapse
Affiliation(s)
- Hong Xue
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China (H.X.).,Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.)
| | - Aron M Geurts
- Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.).,Genomic Sciences and Precision Medicine Center Medical College of Wisconsin, Milwaukee (A.M.G.)
| | - Kristie Usa
- Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.)
| | - Feng Wang
- Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.).,Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China (F.W., Y.L.)
| | - Yingying Lin
- Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.).,Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China (F.W., Y.L.)
| | - Jenifer Phillips
- Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.)
| | - Lisa Henderson
- Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.)
| | - Maria Angeles Baker
- Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.)
| | - Zhongmin Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, China (Z.T.)
| | - Mingyu Liang
- Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.)
| |
Collapse
|
26
|
Williams AM, Liu Y, Regner KR, Jotterand F, Liu P, Liang M. Artificial intelligence, physiological genomics, and precision medicine. Physiol Genomics 2018; 50:237-243. [PMID: 29373082 DOI: 10.1152/physiolgenomics.00119.2017] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Big data are a major driver in the development of precision medicine. Efficient analysis methods are needed to transform big data into clinically-actionable knowledge. To accomplish this, many researchers are turning toward machine learning (ML), an approach of artificial intelligence (AI) that utilizes modern algorithms to give computers the ability to learn. Much of the effort to advance ML for precision medicine has been focused on the development and implementation of algorithms and the generation of ever larger quantities of genomic sequence data and electronic health records. However, relevance and accuracy of the data are as important as quantity of data in the advancement of ML for precision medicine. For common diseases, physiological genomic readouts in disease-applicable tissues may be an effective surrogate to measure the effect of genetic and environmental factors and their interactions that underlie disease development and progression. Disease-applicable tissue may be difficult to obtain, but there are important exceptions such as kidney needle biopsy specimens. As AI continues to advance, new analytical approaches, including those that go beyond data correlation, need to be developed and ethical issues of AI need to be addressed. Physiological genomic readouts in disease-relevant tissues, combined with advanced AI, can be a powerful approach for precision medicine for common diseases.
Collapse
Affiliation(s)
- Anna Marie Williams
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Yong Liu
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Kevin R Regner
- Division of Nephrology, Department of Medicine, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Fabrice Jotterand
- Center for Bioethics and Medical Humanities, Institute for Health & Equity, Medical College of Wisconsin , Milwaukee, Wisconsin.,Institute for Biomedical Ethics , University of Basel, Basel, Switzerland
| | - Pengyuan Liu
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin.,Institute for Biomedical Ethics , University of Basel, Basel, Switzerland.,Sir Run Run Shaw Hospital and Institute of Translational Medicine, Zhejiang University , Zhejiang , China
| | - Mingyu Liang
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| |
Collapse
|