1
|
Zhao S, Chen Z, Liu H, Wang X, Zhang X, Shi H. Maternal nutrition and offspring lung health: sex-specific pathway modulation in fibrosis, metabolism, and immunity. Food Nutr Res 2025; 69:11035. [PMID: 39790857 PMCID: PMC11708518 DOI: 10.29219/fnr.v69.11035] [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: 09/08/2024] [Revised: 10/26/2024] [Accepted: 11/26/2024] [Indexed: 01/12/2025] Open
Abstract
Background Maternal nutrition profoundly influences offspring health, impacting both prenatal and early postnatal development. Previous studies have demonstrated that maternal dietary habits can affect key developmental pathways in the offsprings, including those related to lung function and disease susceptibility. However, the sex-specific impact of a maternal high-salt diet (HSD) on offspring lung injury remains poorly understood. Objective This study aimed to investigate the sex-specific effects of maternal HSD on lung injury in mouse offsprings, focusing on pathways related to fibrosis, metabolism, immunity, and apoptosis. Design Pregnant C57BL/6J mice were subjected to either normal or HSD conditions during gestation. Lung tissues from the male and female offsprings were analyzed using high-throughput RNA sequencing and bioinformatics tools to examine transcriptomic changes. Wet-lab validation, including Masson trichrome staining, immunofluorescence for α-SMA, and qRT-PCR for fibrotic markers (α-SMA, collagen I, Fn1, and TGF-β), was conducted to confirm fibrosis and other injury markers. Lung structure and weight were also evaluated to assess physical alterations due to maternal diet. Results Maternal HSD significantly altered lung transcriptomes in a sex-specific manner. Male offsprings showed increased susceptibility to fibrosis, as confirmed by histological and molecular analyses, including elevated expression of α-SMA, collagen I, Fn1, and TGF-β. In contrast, female offsprings exhibited distinct changes in metabolic and immune pathways. These findings highlight the differential regulation of pulmonary injury mechanisms between male and female offsprings exposed to HSD. Conclusions Maternal HSD induces sex-specific lung injury in offsprings by disrupting critical pathways involved in fibrosis, metabolism, immunity, and apoptosis. The combination of transcriptomic and orthogonal data underscores the need for balanced maternal nutrition during pregnancy to promote long-term respiratory health in offsprings. These results provide new insights into the sex-specific vulnerabilities to lung disease arising from maternal diet.
Collapse
Affiliation(s)
- Shuangyi Zhao
- Department of Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhimin Chen
- Department of Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huina Liu
- Department of Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinyan Wang
- Department of Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiuru Zhang
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Huirong Shi
- Department of Gynaecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
2
|
Endrias EE, Geta T, Desalegn B, Ataro BA, Israel E, Kebede C, Belayneh M, Ahmed B, Moloro AH, Nigussie G. Exploring experiences and perspectives of patients on hypertension management in Southern Ethiopia: a phenomenological study. BMC Health Serv Res 2024; 24:1625. [PMID: 39702319 DOI: 10.1186/s12913-024-12111-8] [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: 10/20/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Hypertension is a significant public health challenge, particularly in low- and middle-income countries, where its management is often inadequate. Understanding patients' experiences and perspectives is crucial for improving hypertension care. Despite a growing body of literature on hypertension in Ethiopia, most studies focus on quantitative data, leaving a significant gap in qualitative insights into patient experiences and perspectives. This study uniquely contributes to the field by exploring the lived experiences of patients in Southern Ethiopia, providing context-specific evidence to inform patient-centered hypertension management strategies. OBJECTIVE This qualitative study aimed to explore the experiences and perspectives of patients with hypertension regarding their management and care at a comprehensive hospital in Southern Ethiopia. METHODS A phenomenological approach was employed to capture the experiences and perspectives of participants. In-depth interviews were conducted with 14 patients with hypertension, selected through purposive sampling. The interviews were audio-recorded, transcribed verbatim, and analyzed using OpenCode version 4.02 software to facilitate systematic coding and thematic analysis. Then, key themes and subthemes were identified from the data related to patient experiences and perspectives providing a relevant framework for understanding the complexities of hypertension management from the patients' perspectives. RESULTS The six key themes emerged, depicting participants' challenges, coping strategies, and interactions with the healthcare system. The themes identified are: adherence to treatment and self-care, awareness of perceived risk and health literacy, experience with the diagnosis, family support in hypertension management, stress awareness and management, and healthcare system and proximity to facilities. Participants noted obstacles to adherence, including long waiting times, forgetting medications, and stress related to their condition. CONCLUSION AND RECOMMENDATION: The findings highlight the need for specific patient-centered strategies that address the challenges faced by hypertensive patients in Southern Ethiopia, such as reducing wait times, enhancing family involvement in care, increasing adherence, and addressing stress. Strengthening healthcare services is essential for sustainable hypertension management.
Collapse
Affiliation(s)
- Eshetu Elfios Endrias
- School of Nursing, College of Health Science and Medicine, Wolaita Sodo University, Wolaita Sodo, Ethiopia.
| | - Temesgen Geta
- School of Nursing, College of Health Science and Medicine, Wolaita Sodo University, Wolaita Sodo, Ethiopia
| | - Bemnet Desalegn
- School of Public Health, College of Health Science and Medicine, Wolaita Sodo University, Wolaita Sodo, Ethiopia
| | - Bizuayehu Atinafu Ataro
- School of Nursing, College of Health Science and Medicine, Wolaita Sodo University, Wolaita Sodo, Ethiopia
| | - Eskinder Israel
- School of Public Health, College of Health Science and Medicine, Wolaita Sodo University, Wolaita Sodo, Ethiopia
| | - Christian Kebede
- School of Nursing, College of Health Science and Medicine, Wolaita Sodo University, Wolaita Sodo, Ethiopia
| | - Melesse Belayneh
- Department of Public Health, School of Public Health, College of Medicine and Health Science, Bahir Dar University, Bahir Dar, Ethiopia
| | - Beker Ahmed
- Department of Midwifery, College of Health Science and Medicine, Arsi University, Asela, Ethiopia
| | | | - Getachew Nigussie
- School of Nursing, College of Health Science and Medicine, Wolaita Sodo University, Wolaita Sodo, Ethiopia
| |
Collapse
|
3
|
Bakinowska E, Stańska W, Kiełbowski K, Szwedkowicz A, Boboryko D, Pawlik A. Gut Dysbiosis and Dietary Interventions in Rheumatoid Arthritis-A Narrative Review. Nutrients 2024; 16:3215. [PMID: 39339815 PMCID: PMC11435214 DOI: 10.3390/nu16183215] [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: 08/11/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic and progressive autoimmune disease. The pathogenesis of RA is complex and involves interactions between articular cells, such as fibroblast-like synoviocytes, and immune cells. These cells secrete pro-inflammatory cytokines, chemokines, metalloproteinases and other molecules that together participate in joint degradation. The current evidence suggests the important immunoregulatory role of the gut microbiome, which can affect susceptibility to diseases and infections. An altered microbiome, a phenomenon known as gut dysbiosis, is associated with the development of inflammatory diseases. Importantly, the profile of the gut microbiome depends on dietary habits. Therefore, dietary elements and interventions can indirectly impact the progression of diseases. This review summarises the evidence on the involvement of gut dysbiosis and diet in the pathogenesis of RA.
Collapse
Affiliation(s)
- Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Wiktoria Stańska
- Department of Medical Biology, Medical University of Warsaw, 00-575 Warsaw, Poland
| | - Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Agata Szwedkowicz
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Dominika Boboryko
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
4
|
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
|
5
|
Hamaya R, Sun Q, Li J, Yun H, Wang F, Curhan GC, Huang T, Manson JE, Willett WC, Rimm EB, Clish C, Liang L, Hu FB, Ma Y. 24-h urinary sodium and potassium excretions, plasma metabolomic profiles, and cardiometabolic biomarkers in the United States adults: a cross-sectional study. Am J Clin Nutr 2024; 120:153-161. [PMID: 38762185 PMCID: PMC11251214 DOI: 10.1016/j.ajcnut.2024.05.010] [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: 01/30/2024] [Revised: 04/23/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024] Open
Abstract
BACKGROUND High-sodium and low-potassium intakes are associated with a higher risk of hypertension and cardiovascular disease, but there are limited data on the circulating metabolomics profiles of 24-h urinary sodium and potassium excretions in free-living individuals. OBJECTIVES We aimed to characterize the metabolomics signatures of a high-sodium and low-potassium diet in a cross-sectional study. METHODS In 1028 healthy older adults from the Women's and Men's Lifestyle Validation Studies, we investigated the association of habitual sodium and potassium intakes measured by 2 to 4 24-h urine samples with plasma metabolites (quantified using liquid chromatography-tandem mass spectrometry) and metabolomic pathways. Our primary exposures were energy-adjusted 24-h urinary sodium excretion, potassium excretion, and sodium-to-potassium ratio, calculated based on energy expenditure derived from the doubly labeled water method. We then assessed the partial correlations of their metabolomics scores, derived from elastic net regressions, with cardiometabolic biomarkers. RESULTS Higher sodium excretion was associated with 38 metabolites including higher piperine, phosphatidylethanolamine, and C5:1 carnitine. In pathway analysis, higher sodium excretion was associated with enhanced biotin and propanoate metabolism and enhanced degradation of lysine and branched-chain amino acids (BCAAs). Metabolites associated with higher potassium and lower sodium-to-potassium ratio included quinic acid and proline-betaine. After adjusting for confounding factors, the metabolomics score for sodium-to-potassium ratio positively correlated with fasting insulin (Spearman's rank correlation coefficient ρ = 0.27), C-peptide (ρ = 0.30), and triglyceride (ρ = 0.46), and negatively with adiponectin (ρ = -0.40), and high-density lipoprotein cholesterol (ρ = -0.42). CONCLUSIONS We discovered metabolites and metabolomics pathways associated with a high-sodium diet, including metabolites related to biotin, propanoate, lysine, and BCAA pathways. The metabolomics signature for a higher sodium low-potassium diet is associated with multiple components of elevated cardiometabolic risk.
Collapse
Affiliation(s)
- Rikuta Hamaya
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Qi Sun
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Jun Li
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Huan Yun
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Fenglei Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Gary C Curhan
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States; Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - Tianyi Huang
- Division of Women's Health, Department of Medicine, Connors Center for Women's Health and Gender Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - JoAnn E Manson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States; Mary Horrigan Connors Center for Women's Health and Gender Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Walter C Willett
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Eric B Rimm
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Clary Clish
- Metabolomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Liming Liang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Frank B Hu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States.
| | - Yuan Ma
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States.
| |
Collapse
|
6
|
Vallianou NG, Kounatidis D, Panagopoulos F, Evangelopoulos A, Stamatopoulos V, Papagiorgos A, Geladari E, Dalamaga M. Gut Microbiota and Its Role in the Brain-Gut-Kidney Axis in Hypertension. Curr Hypertens Rep 2023; 25:367-376. [PMID: 37632662 DOI: 10.1007/s11906-023-01263-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2023] [Indexed: 08/28/2023]
Abstract
PURPOSE OF REVIEW The role of the gut microbiota in modulating blood pressure is increasingly being recognized, currently. The purpose of this review is to summarize recent findings about the mechanisms involved in hypertension with regard to the phenomenon of "gut dysbiosis." RECENT FINDINGS Gut dysbiosis, i.e., the imbalance between the gut microbiota and the host, is characterized by a disruption of the tight junction proteins, such as occludins, claudins, and JAMs (junctional adhesion molecules), resulting in increased gut permeability or the so called "leaky gut." Due to the influence of genetic as well as environmental factors, various metabolites produced by the gut microbiota, such as indole and p-cresol, are increased. Thereby, uremic toxins, such as indoxyl sulfates and p-cresol sulfates, accumulate in the blood and the urine, causing damage in the podocytes and the tubular cells. In addition, immunological mechanisms are implicated as well. In particular, a switch from M2 macrophages to M1 macrophages, which produce pro-inflammatory cytokines, occurs. Moreover, a higher level of Th17 cells, releasing large amounts of interleukin-17 (IL-17), has been reported, when a diet rich in salt is consumed. Therefore, apart from the aggravation of uremic toxins, which may account for direct harmful effects on the kidney, there is inflammation not only in the gut, but in the kidneys as well. This crosstalk between the gut and the kidney is suggested to play a crucial role in hypertension. Notably, the brain is also implicated, with an increasing sympathetic output. The brain-gut-kidney axis seems to be deeply involved in the development of hypertension and chronic kidney disease (CKD). The notion that, by modulating the gut microbiota, we could regulate blood pressure is strongly supported by the current evidence. A healthy diet, low in animal protein and fat, and low in salt, together with the utilization of probiotics, prebiotics, synbiotics, or postbiotics, may contribute to our fight against hypertension.
Collapse
Affiliation(s)
| | | | - Fotis Panagopoulos
- Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | | | | | | | - Eleni Geladari
- Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias str, Athens, Greece
| |
Collapse
|
7
|
O'Connor LE, Hall KD, Herrick KA, Reedy J, Chung ST, Stagliano M, Courville AB, Sinha R, Freedman ND, Hong HG, Albert PS, Loftfield E. Metabolomic Profiling of an Ultraprocessed Dietary Pattern in a Domiciled Randomized Controlled Crossover Feeding Trial. J Nutr 2023; 153:2181-2192. [PMID: 37276937 PMCID: PMC10447619 DOI: 10.1016/j.tjnut.2023.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND Objective markers of ultraprocessed foods (UPF) may improve the assessment of UPF intake and provide insight into how UPF influences health. OBJECTIVES To identify metabolites that differed between dietary patterns (DPs) high in or void of UPF according to Nova classification. METHODS In a randomized, crossover, controlled-feeding trial (clinicaltrials.govNCT03407053), 20 domiciled healthy participants (mean ± standard deviation: age 31 ± 7 y, body mass index [kg/m2] 22 ± 11.6) consumed ad libitum a UPF-DP (80% UPF) and an unprocessed DP (UN-DP; 0% UPF) for 2 wk each. Metabolites were measured using liquid chromatography with tandem mass spectrometry in ethylenediaminetetraacetic acid plasma, collected at week 2 and 24-h, and spot urine, collected at weeks 1 and 2, of each DP. Linear mixed models, adjusted for energy intake, were used to identify metabolites that differed between DPs. RESULTS After multiple comparisons correction, 257 out of 993 plasma and 606 out of 1279 24-h urine metabolites differed between UPF-DP and UN-DP. Overall, 21 known and 9 unknown metabolites differed between DPs across all time points and biospecimen types. Six metabolites were higher (4-hydroxy-L-glutamic acid, N-acetylaminooctanoic acid, 2-methoxyhydroquinone sulfate, 4-ethylphenylsulfate, 4-vinylphenol sulfate, and acesulfame) and 14 were lower following the UPF-DP; pimelic acid, was lower in plasma but higher in urine following the UPF-DP. CONCLUSIONS Consuming a DP high in, compared with 1 void of, UPF has a measurable impact on the short-term human metabolome. Observed differential metabolites could serve as candidate biomarkers of UPF intake or metabolic response in larger samples with varying UPF-DPs. This trial was registered at clinicaltrials.gov as NCT03407053 and NCT03878108.
Collapse
Affiliation(s)
- Lauren E O'Connor
- Food Components and Health Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, USDA, Beltsville, MD, USA; Division of Cancer Control and Population Sciences, Risk Factor Assessment Branch, NCI, Bethesda, MD, USA
| | - Kevin D Hall
- Laboratory of Biological Modeling, NIDDK, Bethesda, MD, USA
| | - Kirsten A Herrick
- Division of Cancer Control and Population Sciences, Risk Factor Assessment Branch, NCI, Bethesda, MD, USA
| | - Jill Reedy
- Division of Cancer Control and Population Sciences, Risk Factor Assessment Branch, NCI, Bethesda, MD, USA
| | - Stephanie T Chung
- Diabetes, Endocrinology, and Obesity Branch, NIDDK, Bethesda, MD, USA
| | - Michael Stagliano
- Diabetes, Endocrinology, and Obesity Branch, NIDDK, Bethesda, MD, USA
| | - Amber B Courville
- Diabetes, Endocrinology, and Obesity Branch, NIDDK, Bethesda, MD, USA
| | - Rashmi Sinha
- Division of Cancer Epidemiology and Genetics, Metabolic Epidemiology Branch, NCI, Bethesda, MD, USA
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, Metabolic Epidemiology Branch, NCI, Bethesda, MD, USA
| | - Hyokyoung G Hong
- Division of Cancer Epidemiology and Genetics, Biostatistics Branch, NCI, Bethesda, MD, USA
| | - Paul S Albert
- Division of Cancer Epidemiology and Genetics, Biostatistics Branch, NCI, Bethesda, MD, USA
| | - Erikka Loftfield
- Division of Cancer Epidemiology and Genetics, Metabolic Epidemiology Branch, NCI, Bethesda, MD, USA.
| |
Collapse
|
8
|
Ishimwe JA, Ferguson JF, Kirabo A. Sex Differences in Fatty Acid Metabolism and Blood Pressure Response to Dietary Salt in Humans. CARDIOGENETICS 2023; 13:33-46. [PMID: 38605973 PMCID: PMC11008634 DOI: 10.3390/cardiogenetics13010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024] Open
Abstract
Salt sensitivity is a trait in which high dietary sodium (Na+) intake causes an increase in blood pressure (BP). We previously demonstrated that in the gut, elevated dietary Na+ causes dysbiosis. The mechanistic interplay between excess dietary Na+-induced alteration in the gut microbiome and sex differences is less understood. The goal of this study was to identify novel metabolites in sex differences and blood pressure in response to a high dietary Na+ intake. We performed stool and plasma metabolomics analysis and measured the BP of human volunteers with salt intake above or below the American Heart Association recommendations. We also performed RNA sequencing on human monocytes treated with high salt in vitro. The relationship between BP and dietary Na+ intake was different in women and men. Network analysis revealed that fatty acids as top subnetworks differentially changed with salt intake. We found that women with high dietary Na+ intake have high levels of arachidonic acid related metabolism, suggesting a role in sex differences of the blood pressure response to Na+. The exposure of monocytes to high salt in vitro upregulates the transcription of fatty acid receptors and arachidonic acid-related genes. These findings provide potentially novel insights into metabolic changes underlying gut dysbiosis and inflammation in salt sensitivity of BP.
Collapse
Affiliation(s)
- Jeanne A. Ishimwe
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA
| | - Jane F. Ferguson
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Nashville, TN 37235, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA
- Medical Center, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
9
|
McCormick JA, Topf J, Tomacruz ID, Grimm PR. A New Understanding of Potassium's Influence Upon Human Health and Renal Physiology. ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:137-147. [PMID: 36868729 DOI: 10.1053/j.akdh.2023.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 03/05/2023]
Abstract
Potassium channels are expressed in virtually all cell types, and their activity is the dominant determinant of cellular membrane potential. As such, potassium flux is a key regulator of many cellular processes including the regulation of action potentials in excitable cells. Subtle changes in extracellular potassium can initiate signaling processes vital for survival (insulin signaling) while more extreme and chronic changes may lead to pathological states (acid-base disturbances and cardiac arrhythmia). While many factors acutely influence extracellular potassium levels, it is principally the role of the kidneys to maintain potassium balance by matching urinary excretion with dietary intake. When this balance is disrupted, human health is negatively impacted. In this review, we discuss evolving views of dietary potassium intake as means of preventing and mitigating diseases. We also provide an update on a molecular pathway called the potassium switch, a mechanism by which extracellular potassium regulates distal nephron sodium reabsorption. Finally, we review recent literature describing how several popular therapeutics influence potassium homeostasis.
Collapse
Affiliation(s)
- James A McCormick
- Division of Nephrology and Hypertension, Oregon Health and Science University, Portland, OR
| | - Joel Topf
- Department of Medicine, Oakland University William Beaumont School of Medicine, Rochester, MI
| | | | - P Richard Grimm
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
10
|
Ko J, Wang J, Chung ML, Sharma K. Examining the Individual Response to a Low-Sodium Diet in Patients with Hypertension: Protocol for a Pilot Randomized Controlled Trial. JMIR Res Protoc 2023; 12:e39058. [PMID: 36780210 PMCID: PMC9972206 DOI: 10.2196/39058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 12/13/2022] [Accepted: 01/03/2023] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Excessive dietary sodium intake is an independent risk factor for hypertension and cardiovascular disease (CVD). Despite the large body of evidence concerning the effects of dietary interventions on blood pressure (BP) and CVD outcomes, trials have often reported low adherence to decreased sodium intake, likely due in part to heterogeneous BP responses. To address the challenges, recent clinical findings suggested a precise and personalized dietary approach that seeks to deliver more preventive and practical dietary advice than the "one-size-fits-all" guidelines and weighs the personal risk of developing specific diseases. OBJECTIVE The purpose of this pilot randomized controlled trial was to test the feasibility and preliminary efficacy of integrating the use of mobile technology and metabolomics with a low-sodium diet intervention in patients with hypertension to develop personalized low-sodium diet programs. Additionally, the study will examine the associations of urine metabolites with urinary sodium levels and BP control based on the hypothesis that targeted urine metabolites. In this report, we describe the design and protocol of the pilot trial. METHODS A total of 40 patients with hypertension will be randomly assigned to either a 8-week low-sodium diet group (n=20) or a standard care group (n=20). Each week, intervention participants went through individual sessions with an interventionist via videoconferencing to discuss low-sodium diet regimens, patients' food choices, and BP tracks on mobile apps. The control group followed their usual care for hypertension management. All participants in both groups monitored diet and BP using mobile apps for 8 weeks. A 24-hour urinary sodium excretion for the estimation of dietary sodium intake, systolic, and diastolic BPs were measured at the baseline and at 8 weeks. The primary outcomes of this study include the feasibility of conducting a randomized controlled trial (RCT) by reporting recruitment, retention, and completion statistics. The preliminary effects of intervention will be tested by a generalized estimating equation model. RESULTS This pilot RCT study was approved by the institutional review board at the University of Texas Health San Antonio in January 2021. The first participant was enrolled in April 2021, and currently, 26 participants were enrolled. All data collection is expected to conclude by March 2023, with data analysis and study results ready for reporting by December 2023. Findings from this pilot RCT will further guide the team in planning a future large-scale study. CONCLUSIONS The findings of this proposed study will establish a comprehensive knowledge base for future research and development of personalized dietary interventions to promote adherence to dietary strategies and self-management of chronic disease using the Precision Health approach for millions of Americans who are struggling with uncontrolled hypertension. TRIAL REGISTRATION ClinicalTrials.gov NCT04764253; https://clinicaltrials.gov/ct2/show/NCT04764253. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/39058.
Collapse
Affiliation(s)
- Jisook Ko
- School of Nursing, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Jing Wang
- College of Nursing, Florida State University, Tallahassee, FL, United States
| | - Misook L Chung
- College of Nursing, University of Kentucky, Lexington, KY, United States
| | - Kumar Sharma
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
11
|
Jeong S, Jang HB, Kim HJ, Lee HJ. Identification of Biomarkers Related to Metabolically Unhealthy Obesity in Korean Obese Adolescents: A Cross-Sectional Study. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10020322. [PMID: 36832451 PMCID: PMC9955165 DOI: 10.3390/children10020322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/25/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
BACKGROUND The current study aimed to screen for relationships and different potential metabolic biomarkers involved between metabolically healthy obesity (MHO) and metabolically unhealthy obesity (MUO) in adolescents. METHODS The study included 148 obese adolescents aged between 14 and 16. The study participants were divided into MUO and MHO groups based on the age-specific adolescent metabolic syndrome (MetS) criteria of the International Diabetes Federation. The current study was conducted to investigate the clinical and metabolic differences between the MHO and MUO groups. Multivariate analyses were conducted to investigate the metabolites as independent predictors for the odds ratio and the presence of the MetS. RESULTS There were significant differences in the three acylcarnitines, five amino acids, glutamine/glutamate ratio, three biogenic amines, two glycerophospholipids, and the triglyceride-glucose index between the MUO group and those in the MHO group. Moreover, several metabolites were associated with the prevalence of MUO. Additionally, several metabolites were inversely correlated with MHO in the MUO group. CONCLUSIONS In this study, the biomarkers found in this study have the potential to reflect the clinical outcomes of the MUO group. These biomarkers will lead to a better understanding of MetS in obese adolescents.
Collapse
Affiliation(s)
| | | | | | - Hye-Ja Lee
- Correspondence: ; Tel.: +82-43-719-8452; Fax: +82-43-719-8709
| |
Collapse
|
12
|
Bhat MA, Mishra AK, Tantray JA, Alatawi HA, Saeed M, Rahman S, Jan AT. Gut Microbiota and Cardiovascular System: An Intricate Balance of Health and the Diseased State. Life (Basel) 2022; 12:1986. [PMID: 36556351 PMCID: PMC9780831 DOI: 10.3390/life12121986] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/13/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022] Open
Abstract
Gut microbiota encompasses the resident microflora of the gut. Having an intricate relationship with the host, it plays an important role in regulating physiology and in the maintenance of balance between health and disease. Though dietary habits and the environment play a critical role in shaping the gut, an imbalance (referred to as dysbiosis) serves as a driving factor in the occurrence of different diseases, including cardiovascular disease (CVD). With risk factors of hypertension, diabetes, dyslipidemia, etc., CVD accounts for a large number of deaths among men (32%) and women (35%) worldwide. As gut microbiota is reported to have a direct influence on the risk factors associated with CVDs, this opens up new avenues in exploring the possible role of gut microbiota in regulating the gross physiological aspects along the gut-heart axis. The present study elaborates on different aspects of the gut microbiota and possible interaction with the host towards maintaining a balance between health and the occurrence of CVDs. As the gut microbiota makes regulatory checks for these risk factors, it has a possible role in shaping the gut and, as such, in decreasing the chances of the occurrence of CVDs. With special emphasis on the risk factors for CVDs, this paper includes information on the prominent bacterial species (Firmicutes, Bacteriodetes and others) towards an advance in our understanding of the etiology of CVDs and an exploration of the best possible therapeutic modules for implementation in the treatment of different CVDs along the gut-heart axis.
Collapse
Affiliation(s)
- Mujtaba Aamir Bhat
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, Jammu and Kashmir, India
| | - Awdhesh Kumar Mishra
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Javeed Ahmad Tantray
- Department of Zoology, Central University of Kashmir, Ganderbal 191131, Jammu and Kashmir, India
| | - Hanan Ali Alatawi
- Department of Biological Sciences, University College of Haqel, University of Tabuk, Tabuk 47512, Saudi Arabia
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Hail, Hail 55476, Saudi Arabia
| | - Safikur Rahman
- Department of Botany, MS College, BR Ambedkar Bihar University, Muzaffarpur 842001, Bihar, India
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, Jammu and Kashmir, India
| |
Collapse
|
13
|
Dhillon J, Jacobs AG, Ortiz S, Diaz Rios LK. A Systematic Review of Literature on the Representation of Racial and Ethnic Minority Groups in Clinical Nutrition Interventions. Adv Nutr 2022; 13:1505-1528. [PMID: 35108358 PMCID: PMC9526835 DOI: 10.1093/advances/nmac002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/29/2021] [Accepted: 01/10/2022] [Indexed: 02/04/2023] Open
Abstract
The racial and ethnic disparities in diet-related chronic diseases are major concerns. This systematic review examines the extent to which diet-induced changes in health outcomes, such as cardiometabolic, inflammation, cancer, bone health, and kidney function outcomes, etc., have been reported and discussed by race or ethnicity in randomized trials with 2 or more diet arms that recruited both minority and non-Hispanic White groups. Databases (i.e., PubMed, Cochrane Library, and Web of Science) were searched up to August 2021. Thirty-four studies that discussed effects of defined dietary interventions on health outcomes by racial or ethnic minority group compared with non-Hispanic Whites were included in the systematic review (PROSPERO registration number: CRD42021229256). Acute trials and those with 1 diet arm that accounted for race or ethnicity in their analyses and studies that focused on a single racial or ethnic group were discussed separately. Most studies were conducted in Black compared with White adults testing effects of energy restriction, macronutrient modification, sodium reduction, or variations of the Dietary Approaches to Stop Hypertension (DASH) diet on cardiometabolic outcomes. There was limited focus on other minority groups. Evidence suggests greater blood pressure reduction for Black adults compared with Whites particularly with DASH (or similar) diets. Overall, there was limited consideration for group-specific eating patterns and diet acceptability. Overall risk of bias was low. With emerging precision nutrition initiatives that aim to optimize metabolic responses in population subgroups through tailored approaches, it is imperative to ensure adequate representation of racial and ethnic subgroups for addressing health disparities. Factors that help explain variability in responses such as socioecological context should be included and adequately powered. Given the racial and ethnic disparities in chronic diseases, studying the adoption, maintenance, and effectiveness of dietary interventions on health outcomes among different groups is critical for developing approaches that can mitigate diet-related health disparities.
Collapse
Affiliation(s)
- Jaapna Dhillon
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
- Department of Molecular & Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA, USA
| | | | - Sigry Ortiz
- Department of Molecular & Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA, USA
| | - L Karina Diaz Rios
- Division of Agriculture and Natural Resources, University of California Merced, Merced, CA, USA
| |
Collapse
|
14
|
Kelly RS, Stewart ID, Bayne H, Kachroo P, Spiro A, Vokonas P, Sparrow D, Weiss ST, Knihtilä HM, Litonjua AA, Wareham NJ, Langenberg C, Lasky-Su JA. Metabolomic differences in lung function metrics: evidence from two cohorts. Thorax 2022; 77:919-928. [PMID: 34650005 PMCID: PMC9008068 DOI: 10.1136/thoraxjnl-2020-216639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 09/18/2021] [Indexed: 12/20/2022]
Abstract
RATIONALE The biochemical mechanisms underlying lung function are incompletely understood. OBJECTIVES To identify and validate the plasma metabolome of lung function using two independent adult cohorts: discovery-the European Prospective Investigation into Cancer-Norfolk (EPIC-Norfolk, n=10 460) and validation-the VA Normative Aging Study (NAS) metabolomic cohort (n=437). METHODS We ran linear regression models for 693 metabolites to identify associations with forced expiratory volume in one second (FEV1) and the ratio of FEV1 to forced vital capacity (FEV1/FVC), in EPIC-Norfolk then validated significant findings in NAS. Significance in EPIC-Norfolk was denoted using an effective number of tests threshold of 95%; a metabolite was considered validated in NAS if the direction of effect was consistent and p<0.05. MEASUREMENTS AND MAIN RESULTS Of 156 metabolites that associated with FEV1 in EPIC-Norfolk after adjustment for age, sex, body mass index, height, smoking and asthma status, 34 (21.8%) validated in NAS, including several metabolites involved in oxidative stress. When restricting the discovery sample to men only, a similar percentage, 18 of 79 significant metabolites (22.8%) were validated. A smaller number of metabolites were validated for FEV1/FVC, 6 of 65 (9.2%) when including all EPIC-Norfolk as the discovery population, and 2 of 34 (5.9%) when restricting to men. These metabolites were characterised by involvement in respiratory track secretants. Interestingly, no metabolites were validated for both FEV1 and FEV1/FVC. CONCLUSIONS The validation of metabolites associated with respiratory function can help to better understand mechanisms of lung health and may assist the development of biomarkers.
Collapse
Affiliation(s)
- Rachel S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | - Haley Bayne
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Priyadarshini Kachroo
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Avron Spiro
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), 150 South Huntington Avenue, Boston, MA 02130, USA, VA Boston Healthcare System, Boston, MA 02130, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA 02118, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Pantel Vokonas
- VA Normative Aging Study, Boston University School of Medicine, Boston, MA 02118, USA
| | - David Sparrow
- VA Normative Aging Study, Boston University School of Medicine, Boston, MA 02118, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Hanna M Knihtilä
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Augusto A Litonjua
- Division of Pediatric Pulmonary Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | | | | | - Jessica A Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Chen X, Wu H, Huang S. Excessive Sodium Intake Leads to Cardiovascular Disease by Promoting Sex-Specific Dysfunction of Murine Heart. Front Nutr 2022; 9:830738. [PMID: 35845798 PMCID: PMC9285006 DOI: 10.3389/fnut.2022.830738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 05/16/2022] [Indexed: 11/17/2022] Open
Abstract
Background Globally, a high-salt diet (HSD) has become a threat to human health as it can lead to a high risk of cardiac damage. Although some studies investigating HSD have been carried out, the majority has been conducted in males, and there are few female-specific studies, thereby ignoring any effects of sex-specific damage on the heart. In this study, we determined how HSD induces different pathways of cardiovascular diseases through sex-specific effects on cardiac damage in mice. Methods An HSD murine model of male and female C57BL/6J mice was fed with sodium-rich chow (4% NaCl). After 8 weeks, cardiac tissues were collected, and the whole gene transcriptome of the hearts of male and female mice was characterized and analyzed using high-throughput RNA sequencing. Immunohistochemistry staining was used to further assess the harmful effects of HSD on protein expression of genes associated with immunity, fibrosis, and apoptosis in male and female mice. Results HSD drastically altered the cardiac transcriptome compared to that of the normal heart in both male and female mice and had a sex-specific effect on the cardiac composition in the transcriptome. HSD produced various differentially expressed genes and affected different KEGG pathways of the transcriptome in male and female mice. Furthermore, we found that HSD induced different pathways of cardiovascular disease in the male mice and female mice. The pathway of hypertrophic cardiomyopathy is significantly enriched in HSD-treated male mice, while the pathway of dilated cardiomyopathy is significantly enriched in HSD-treated female mice. Finally, metabolism, immunity, fibrosis, and apoptosis in the mouse heart showed sex-specific changes predicting cardiac damage. Conclusion Our results demonstrate that HSD adversely impacts cardiac structure and function by affecting the metabolism, immunity, fibrosis, and apoptosis in the murine heart and induces the mouse to suffer from sex-specific cardiovascular disease. This study provides a new perspective and basis for the differences in the pharmacology and interventional treatment of sex-specific cardiovascular diseases induced by HSD in men and women.
Collapse
Affiliation(s)
- Xiuli Chen
- Obstetrical Department, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Haiying Wu
- Obstetrical Department, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Shenzhen Huang
| | - Shenzhen Huang
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Henan University, People's Hospital of Zhengzhou University, Zhengzhou, China
- Haiying Wu
| |
Collapse
|
16
|
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
|
17
|
Zou M, Chen Y, Zheng Z, Sheng S, Jia Y, Wang X, Ren S, Yang Y, Li X, Dong W, Guan M, Zhang Q, Xue Y. High-Salt Attenuates the Efficacy of Dapagliflozin in Tubular Protection by Impairing Fatty Acid Metabolism in Diabetic Kidney Disease. Front Pharmacol 2022; 12:741087. [PMID: 34987387 PMCID: PMC8720966 DOI: 10.3389/fphar.2021.741087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 11/23/2021] [Indexed: 11/19/2022] Open
Abstract
High-salt intake leads to kidney damage and even limits the effectiveness of drugs. However, it is unclear whether excessive intake of salt affects renal tubular energy metabolism and the efficacy of dapagliflozin on renal function in diabetic kidney disease (DKD). In this study, we enrolled 350 DKD patients and examined the correlation between sodium level and renal function, and analyzed influencing factors. The results demonstrated that patients with macroalbuminuria have higher 24 h urinary sodium levels. After establishment of type 2 diabetes mellitus model, the animals received a high-salt diet or normal-salt diet. In the presence of high-salt diet, the renal fibrosis was aggravated with fatty acid metabolism dysregulation. Furthermore, Na+/K+-ATPase expression was up-regulated in the renal tubules of diabetic mice, while the fatty acid metabolism was improved by inhibiting Na+/K+-ATPase of renal tubular epithelial cells. Of note, the administration with dapagliflozin improved renal fibrosis and enhanced fatty acid metabolism. But high salt weakened the above-mentioned renal protective effects of dapagliflozin in DKD. Similar results were recapitulated in vitro after incubating proximal tubular epithelial cells in high-glucose and high-salt medium. In conclusion, our results indicate that high salt can lead to fatty acid metabolism disorders by increasing Na+/K+-ATPase expression in the renal tubules of DKD. High salt intake diminishes the reno-protective effect of dapagliflozin in DKD.
Collapse
Affiliation(s)
- Meina Zou
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanrong Chen
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zongji Zheng
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuyue Sheng
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yijie Jia
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiangyu Wang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shijing Ren
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanling Yang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaomin Li
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenhui Dong
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Meiping Guan
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yaoming Xue
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
18
|
Itoh N, Tsuya A, Togashi H, Kimura H, Konta T, Nemoto K, Yamashita H, Kayama T. Increased salt intake is associated with diabetes and characteristic dietary habits: a community-based cross-sectional study in Japan. J Clin Biochem Nutr 2022; 71:143-150. [PMID: 36213786 PMCID: PMC9519413 DOI: 10.3164/jcbn.21-153] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/29/2022] [Indexed: 11/22/2022] Open
Abstract
We investigated the association of salt intake with lifestyle-related diseases and also the association of habitually consumed foods with salt intake. A cross-sectional study was conducted using data from a baseline survey of 2,129 residents of Yonezawa city (980 males and 1,149 females), Yamagata prefecture. The residents were divided into three groups based on their estimated daily salt intake: low, medium, and high. In both genders, the prevalence of hypertension and diabetes increased in the order of high > medium > low salt intake (trend p<0.001). Similar trends were observed in the prevalence of hyperlipidemia in females and metabolic syndrome in males. The prevalence of diabetes in the high salt intake group was significantly higher than that in the control group (matched from the low and medium salt intake groups), even when confounding factors were excluded by propensity score matching (p<0.01). Network analysis showed that the low salt intake group had a greater tendency to habitually consume various vegetables than the high salt intake group. Our findings reveal that the prevalence of lifestyle-related diseases increased with higher salt intake. We speculate that a dietary shift to multiple vegetable consumption could have salt-lowering effects.
Collapse
Affiliation(s)
- Nanami Itoh
- Yamagata University Health Administration Center
| | - Atsushi Tsuya
- Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine
| | | | - Hirohito Kimura
- Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine
| | - Tsuneo Konta
- Department of Public Health and Hygiene, Yamagata University Faculty of Medicine
| | - Kenji Nemoto
- Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine
| | - Hidetoshi Yamashita
- Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine
| | - Takamasa Kayama
- Institute for Promotion of Medical Science Research, Yamagata University Faculty of Medicine
| |
Collapse
|
19
|
Dietary Management of Heart Failure: DASH Diet and Precision Nutrition Perspectives. Nutrients 2021; 13:nu13124424. [PMID: 34959976 PMCID: PMC8708696 DOI: 10.3390/nu13124424] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/06/2021] [Accepted: 12/06/2021] [Indexed: 12/14/2022] Open
Abstract
Heart failure (HF) is a major health care burden increasing in prevalence over time. Effective, evidence-based interventions for HF prevention and management are needed to improve patient longevity, symptom control, and quality of life. Dietary Approaches to Stop Hypertension (DASH) diet interventions can have a positive impact for HF patients. However, the absence of a consensus for comprehensive dietary guidelines and for pragmatic evidence limits the ability of health care providers to implement clinical recommendations. The refinement of medical nutrition therapy through precision nutrition approaches has the potential to reduce the burden of HF, improve clinical care, and meet the needs of diverse patients. The aim of this review is to summarize current evidence related to HF dietary recommendations including DASH diet nutritional interventions and to develop initial recommendations for DASH diet implementation in outpatient HF management. Articles involving human studies were obtained using the following search terms: Dietary Approaches to Stop Hypertension (DASH diet), diet pattern, diet, metabolism, and heart failure. Only full-text articles written in English were included in this review. As DASH nutritional interventions have been proposed, limitations of these studies are the small sample size and non-randomization of interventions, leading to less reliable evidence. Randomized controlled interventions are needed to offer definitive evidence related to the use of the DASH diet in HF management.
Collapse
|
20
|
Ghaseminasabparizi M, Nazarinia MA, Akhlaghi M. Adherence to the dietary approaches to stop hypertension dietary pattern and rheumatoid arthritis in Iranian adults. Public Health Nutr 2021; 24:6085-6093. [PMID: 34412722 PMCID: PMC11148601 DOI: 10.1017/s1368980021003608] [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: 11/07/2022]
Abstract
OBJECTIVE To examine the hypothesis that rheumatoid arthritis (RA) patients are less likely than healthy individuals to adhere to the dietary approaches to stop hypertension (DASH) dietary pattern. DESIGN A multi-centre cross-sectional study involving a total of 300 eligible Iranian adults (aged >19 years; 93·0 % female) recruited during 2019-2020. Participants' actual dietary intakes were measured via self-administered 3-d dietary records. The DASH score was computed based on the energy-adjusted intakes of eight major dietary components usually emphasised (i.e. fruits, vegetables, nuts and legumes, low-fat dairy products and whole grains) or minimised (i.e. sweets, red or processed meats and sodium) in the DASH diet. The higher the DASH score of subjects, the greater their adherence to the DASH pattern. SETTING The outpatient clinics of major general hospitals in Shiraz, Iran. PARTICIPANTS 100 incident cases with definite RA according to the 2010 American College of Rheumatology/European League Against Rheumatism Classification Criteria for RA and 200 apparently healthy controls frequency-matched by gender and age. RESULTS After adjusting for several potential covariates in the binary logistic regression analysis, RA cases were less likely than controls to have high adherence to the DASH pattern (OR = 0·08; 95 % CI 0·03, 0·20; P = 0·001). CONCLUSIONS Our findings in a sample of Iranian adults revealed that RA patients are less likely than healthy individuals to adhere to the DASH dietary pattern. However, the potential causal association of greater adherence to the DASH pattern and lower risk of RA needs to be confirmed by prospective studies of high methodological quality.
Collapse
Affiliation(s)
- Maryam Ghaseminasabparizi
- Department of Clinical Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Masoumeh Akhlaghi
- Department of Community Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
21
|
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
|
22
|
An exploratory analysis of comparative plasma metabolomic and lipidomic profiling in salt-sensitive and salt-resistant individuals from The Dietary Approaches to Stop Hypertension Sodium Trial. J Hypertens 2021; 39:1972-1981. [PMID: 34001808 PMCID: PMC8429079 DOI: 10.1097/hjh.0000000000002904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Objective: This study conducted exploratory metabolomic and lipidomic profiling of plasma samples from the DASH (Dietary Approaches to Stop Hypertension) Sodium Trial to identify unique plasma biomarkers to identify salt-sensitive versus salt-resistant participants. Methods: Utilizing plasma samples from the DASH-Sodium Trial, we conducted untargeted metabolomic and lipidomic profiling on plasma from salt-sensitive and salt-resistant DASH-Sodium Trial participants. Study 1 analyzed plasma from 106 salt-sensitive and 85 salt-resistant participants obtained during screening when participants consumed their regular diet. Study 2 examined paired within-participant plasma samples in 20 salt-sensitive and 20 salt-resistant participants during a high-salt and low-salt dietary intervention. To investigate differences in metabolites or lipidomes that could discriminate between salt-sensitive and salt-resistant participants or the response to a dietary sodium intervention Principal Component Analysis and Orthogonal Partial Least Square Discriminant Analysis was conducted. Differential expression analysis was performed to validate observed variance and to determine the statistical significance. Results: Differential expression analysis between salt-sensitive and salt-resistant participants at screening revealed no difference in plasma metabolites or lipidomes. In contrast, three annotated plasma metabolites, tocopherol alpha, 2-ketoisocaproic acid, and citramalic acid, differed significantly between high-sodium and low-sodium dietary interventions in salt-sensitive participants. Conclusion: In DASH-Sodium Trial participants on a regular diet, plasma metabolomic or lipidomic signatures were not different between salt-sensitive and salt-resistant participants. High-sodium intake was associated with changes in specific circulating metabolites in salt-sensitive participants. Further studies are needed to validate the identified metabolites as potential biomarkers that are associated with the salt sensitivity of blood pressure.
Collapse
|
23
|
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
|
24
|
Yang T, Chakraborty S, Mandal J, Mei X, Joe B. Microbiota and Metabolites as Factors Influencing Blood Pressure Regulation. Compr Physiol 2021; 11:1731-1757. [PMID: 33792901 DOI: 10.1002/cphy.c200009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The study of microbes has rapidly expanded in recent years due to a surge in our understanding that humans host a plethora of commensal microbes, which reside in their bodies and depending upon their composition, contribute to either normal physiology or pathophysiology. This article provides a general foundation for learning about host-commensal microbial interactions as an emerging area of research. The article is divided into two sections. The first section is dedicated to introducing commensal microbiota and its known effects on the host. The second section is on metabolites, which are biochemicals that the host and the microbes use for bi-directional communication with each other. Together, the sections review what is known about how microbes interact with the host to impact cardiovascular physiology, especially blood pressure regulation. © 2021 American Physiological Society. Compr Physiol 11:1731-1757, 2021.
Collapse
Affiliation(s)
- Tao Yang
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Saroj Chakraborty
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Juthika Mandal
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Xue Mei
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Bina Joe
- Center for Hypertension and Precision Medicine and Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| |
Collapse
|
25
|
Nassir CMNCM, Ghazali MM, Hashim S, Idris NS, Yuen LS, Hui WJ, Norman HH, Gau CH, Jayabalan N, Na Y, Feng L, Ong LK, Abdul Hamid H, Ahamed HN, Mustapha M. Diets and Cellular-Derived Microparticles: Weighing a Plausible Link With Cerebral Small Vessel Disease. Front Cardiovasc Med 2021; 8:632131. [PMID: 33718454 PMCID: PMC7943466 DOI: 10.3389/fcvm.2021.632131] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/19/2021] [Indexed: 12/24/2022] Open
Abstract
Cerebral small vessel disease (CSVD) represents a spectrum of pathological processes of various etiologies affecting the brain microcirculation that can trigger neuroinflammation and the subsequent neurodegenerative cascade. Prevalent with aging, CSVD is a recognized risk factor for stroke, vascular dementia, Alzheimer disease, and Parkinson disease. Despite being the most common neurodegenerative condition with cerebrocardiovascular axis, understanding about it remains poor. Interestingly, modifiable risk factors such as unhealthy diet including high intake of processed food, high-fat foods, and animal by-products are known to influence the non-neural peripheral events, such as in the gastrointestinal tract and cardiovascular stress through cellular inflammation and oxidation. One key outcome from such events, among others, includes the cellular activations that lead to elevated levels of endogenous cellular-derived circulating microparticles (MPs). MPs can be produced from various cellular origins including leukocytes, platelets, endothelial cells, microbiota, and microglia. MPs could act as microthrombogenic procoagulant that served as a plausible culprit for the vulnerable end-artery microcirculation in the brain as the end-organ leading to CSVD manifestations. However, little attention has been paid on the potential role of MPs in the onset and progression of CSVD spectrum. Corroboratively, the formation of MPs is known to be influenced by diet-induced cellular stress. Thus, this review aims to appraise the body of evidence on the dietary-related impacts on circulating MPs from non-neural peripheral origins that could serve as a plausible microthrombosis in CSVD manifestation as a precursor of neurodegeneration. Here, we elaborate on the pathomechanical features of MPs in health and disease states; relevance of dietary patterns on MP release; preclinical studies pertaining to diet-based MPs contribution to disease; MP level as putative surrogates for early disease biomarkers; and lastly, the potential of MPs manipulation with diet-based approach as a novel preventive measure for CSVD in an aging society worldwide.
Collapse
Affiliation(s)
| | - Mazira Mohamad Ghazali
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Sabarisah Hashim
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Nur Suhaila Idris
- Department of Family Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Lee Si Yuen
- Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Wong Jia Hui
- Neurobiology of Aging and Disease Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Haziq Hazman Norman
- Anatomy Unit, International Medical School (IMS), Management and Science University (MSU), Shah Alam, Malaysia
| | - Chuang Huei Gau
- Department of Psychology and Counselling, Faculty of Arts and Social Science, Universiti Tunku Abdul Rahman (UTAR), Kampar, Malaysia
| | - Nanthini Jayabalan
- Translational Neuroscience Lab, University of Queensland (UQ), Centre for Clinical Research, The University of Queensland, Herston, QLD, Australia
| | - Yuri Na
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Linqing Feng
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Lin Kooi Ong
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Malaysia
- School of Biomedical Sciences and Pharmacy, Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, National Health and Medical Research Council (NHMRC), Heidelberg, VIC, Australia
| | - Hafizah Abdul Hamid
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Haja Nazeer Ahamed
- Crescent School of Pharmacy, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, India
| | - Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
- Hospital Universiti Sains Malaysia, Jalan Raja Perempuan Zainab II, Kubang Kerian, Malaysia
| |
Collapse
|
26
|
Kim H, Lichtenstein AH, Wong KE, Appel LJ, Coresh J, Rebholz CM. Urine Metabolites Associated with the Dietary Approaches to Stop Hypertension (DASH) Diet: Results from the DASH-Sodium Trial. Mol Nutr Food Res 2021; 65:e2000695. [PMID: 33300290 PMCID: PMC7967699 DOI: 10.1002/mnfr.202000695] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/11/2020] [Indexed: 12/25/2022]
Abstract
SCOPE Serum metabolomic markers of the Dietary Approaches to Stop Hypertension (DASH) diet are previously reported. In an independent study, the similarity of urine metabolomic markers are investigated. METHODS AND RESULTS In the DASH-Sodium trial, participants are randomly assigned to the DASH diet or control diet, and received three sodium interventions (high, intermediate, low) within each randomized diet group in random order for 30 days each. Urine samples are collected at the end of each intervention period and analyzed for 938 metabolites. Two comparisons are conducted: 1) DASH-high sodium (n = 199) versus control-high sodium (n = 193), and 2) DASH-low sodium (n = 196) versus control-high sodium. Significant metabolites identified using multivariable linear regression are compared and the top 10 influential metabolites identified using partial least-squares discriminant analysis to the results from the DASH trial. Nine out of 10 predictive metabolites of the DASH-high sodium and DASH-low sodium diets are identical. Most candidate biomarkers from the DASH trial replicated. N-methylproline, chiro-inositol, stachydrine, and theobromine replicated as influential metabolites of DASH diets. CONCLUSIONS Candidate biomarkers of the DASH diet identified in serum replicated in urine. Replicated influential metabolites are likely to be objective biomarkers of the DASH diet.
Collapse
Affiliation(s)
- Hyunju Kim
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Alice H. Lichtenstein
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, Massachusetts, USA
| | - Kari E. Wong
- Metabolon, Research Triangle Park, Morrisville, North Carolina, USA
| | - Lawrence J. Appel
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Casey M. Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
27
|
Associations between untargeted plasma metabolomic signatures and gut microbiota composition in the Milieu Intérieur population of healthy adults. Br J Nutr 2020; 126:982-992. [PMID: 33298217 DOI: 10.1017/s0007114520004870] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Host-microbial co-metabolism products are being increasingly recognised to play important roles in physiological processes. However, studies undertaking a comprehensive approach to consider host-microbial metabolic relationships remain scarce. Metabolomic analysis yielding detailed information regarding metabolites found in a given biological compartment holds promise for such an approach. This work aimed to explore the associations between host plasma metabolomic signatures and gut microbiota composition in healthy adults of the Milieu Intérieur study. For 846 subjects, gut microbiota composition was profiled through sequencing of the 16S rRNA gene in stools. Metabolomic signatures were generated through proton NMR analysis of plasma. The associations between metabolomic variables and α- and β-diversity indexes and relative taxa abundances were tested using multi-adjusted partial Spearman correlations, permutational ANOVA and multivariate associations with linear models, respectively. A multiple testing correction was applied (Benjamini-Hochberg, 10 % false discovery rate). Microbial richness was negatively associated with lipid-related signals and positively associated with amino acids, choline, creatinine, glucose and citrate (-0·133 ≤ Spearman's ρ ≤ 0·126). Specific associations between metabolomic signals and abundances of taxa were detected (twenty-five at the genus level and nineteen at the species level): notably, numerous associations were observed for creatinine (positively associated with eleven species and negatively associated with Faecalibacterium prausnitzii). This large-scale population-based study highlights metabolites associated with gut microbial features and provides new insights into the understanding of complex host-gut microbiota metabolic relationships. In particular, our results support the implication of a 'gut-kidney axis'. More studies providing a detailed exploration of these complex interactions and their implications for host health are needed.
Collapse
|
28
|
Kim H, Hu EA, E Wong K, Yu B, Steffen LM, Seidelmann SB, Boerwinkle E, Coresh J, Rebholz CM. Serum Metabolites Associated with Healthy Diets in African Americans and European Americans. J Nutr 2020; 151:40-49. [PMID: 33244610 PMCID: PMC7779213 DOI: 10.1093/jn/nxaa338] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/28/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND High diet quality is associated with a lower risk of chronic diseases. Metabolomics can be used to identify objective biomarkers of diet quality. OBJECTIVES We used metabolomics to identify serum metabolites associated with 4 diet indices and the components within these indices in 2 samples from African Americans and European Americans. METHODS We studied cross-sectional associations between known metabolites and Healthy Eating Index (HEI)-2015, Alternative Healthy Eating Index (AHEI)-2010, the Dietary Approaches to Stop Hypertension Trial (DASH) diet, alternate Mediterranean diet (aMED), and their components using untargeted metabolomics in 2 samples (n1 = 1,806, n2 = 2,056) of the Atherosclerosis Risk in Communities study (aged 45-64 y at baseline). Dietary intakes were assessed using an FFQ. We used multivariable linear regression models to examine associations between diet indices and serum metabolites in each sample, adjusting for participant characteristics. Metabolites significantly associated with diet indices were meta-analyzed across 2 samples. C-statistics were calculated to examine if these candidate biomarkers improved prediction of individuals in the highest compared with lowest quintile of diet scores beyond participant characteristics. RESULTS Seventeen unique metabolites (HEI: n = 6; AHEI: n = 5; DASH: n = 14; aMED: n = 2) were significantly associated with higher diet scores after Bonferroni correction in sample 1 and sample 2. Six of 17 significant metabolites [glycerate, N-methylproline, stachydrine, threonate, pyridoxate, 3-(4-hydroxyphenyl)lactate)] were associated with ≥1 dietary pattern. Candidate biomarkers of HEI, AHEI, and DASH distinguished individuals with highest compared with lowest quintile of diet scores beyond participant characteristics in samples 1 and 2 (P value for difference in C-statistics <0.02 for all 3 diet indices). Candidate biomarkers of aMED did not improve C-statistics beyond participant characteristics (P value = 0.930). CONCLUSIONS A considerable overlap of metabolites associated with HEI, AHEI, DASH, and aMED reflects the similar food components and similar metabolic pathways involved in the metabolism of healthy diets in African Americans and European Americans.
Collapse
Affiliation(s)
- Hyunju Kim
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA,Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, USA
| | - Emily A Hu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA,Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, USA
| | - Kari E Wong
- Metabolon, Research Triangle Park, Morrisville, NC, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics & Environmental Sciences, University of Texas Health Sciences Center at Houston School of Public Health, Houston, TX, USA
| | - Lyn M Steffen
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | | | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics & Environmental Sciences, University of Texas Health Sciences Center at Houston School of Public Health, Houston, TX, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA,Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
29
|
Wang Y, Wang H, Howard AG, Tsilimigras MCB, Avery CL, Meyer KA, Sha W, Sun S, Zhang J, Su C, Wang Z, Zhang B, Fodor AA, Gordon-Larsen P. Associations of sodium and potassium consumption with the gut microbiota and host metabolites in a population-based study in Chinese adults. Am J Clin Nutr 2020; 112:1599-1612. [PMID: 33022700 PMCID: PMC7727480 DOI: 10.1093/ajcn/nqaa263] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 08/24/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND There is increasing evidence that sodium consumption alters the gut microbiota and host metabolome in murine models and small studies in humans. However, there is a lack of population-based studies that capture large variations in sodium consumption as well as potassium consumption. OBJECTIVE We examined the associations of energy-adjusted dietary sodium (milligrams/kilocalorie), potassium, and sodium-to-potassium (Na/K) ratio with the microbiota and plasma metabolome in a well-characterized Chinese cohort with habitual excessive sodium and deficient potassium consumption. METHODS We estimated dietary intakes from 3 consecutive validated 24-h recalls and household inventories. In 2833 adults (18-80 y old, 51.2% females), we analyzed microbial (genus-level 16S ribosomal RNA) between-person diversity, using distance-based redundancy analysis (dbRDA), and within-person diversity and taxa abundance using linear regression, accounting for geographic variation in both. In a subsample (n = 392), we analyzed the overall metabolome (dbRDA) and individual metabolites (linear regression). P values for specific taxa and metabolites were false discovery rate adjusted (q-value). RESULTS Sodium, potassium, and Na/K ratio were associated with microbial between-person diversity (dbRDA P < 0.01) and several specific taxa with large geographic variation, including pathogenic Staphylococcus and Moraxellaceae, and SCFA-producing Phascolarctobacterium and Lachnospiraceae (q-value < 0.05). For example, sodium and Na/K ratio were positively associated with Staphylococcus and Moraxellaceae in Liaoning, whereas potassium was positively associated with 2 genera from Lachnospiraceae in Shanghai. Additionally, sodium, potassium, and Na/K ratio were associated with the overall metabolome (dbRDA P ≤ 0.01) and several individual metabolites, including butyrate/isobutyrate and gut-derived phenolics such as 1,2,3-benzenetriol sulfate, which was negatively associated with sodium in Guizhou (q-value < 0.05). CONCLUSIONS Our findings suggest that sodium and potassium consumption is associated with taxa and metabolites that have been implicated in cardiometabolic health, providing insights into the potential roles of gut microbiota and host metabolites in the pathogenesis of sodium- and potassium-associated diseases. More studies are needed to confirm our results.
Collapse
Affiliation(s)
- Yiqing Wang
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill (UNC-Chapel Hill), Chapel Hill, NC, USA
| | - Huijun Wang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Annie Green Howard
- Department of Biostatistics, Gillings School of Global Public Health, UNC-Chapel Hill, Chapel Hill, NC, USA,Carolina Population Center, UNC-Chapel Hill, Chapel Hill, NC, USA
| | - Matthew C B Tsilimigras
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill (UNC-Chapel Hill), Chapel Hill, NC, USA,Carolina Population Center, UNC-Chapel Hill, Chapel Hill, NC, USA,Department of Epidemiology, Gillings School of Global Public Health, UNC-Chapel Hill, Chapel Hill, NC, USA
| | - Christy L Avery
- Carolina Population Center, UNC-Chapel Hill, Chapel Hill, NC, USA,Department of Epidemiology, Gillings School of Global Public Health, UNC-Chapel Hill, Chapel Hill, NC, USA
| | - Katie A Meyer
- Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill (UNC-Chapel Hill), Chapel Hill, NC, USA,Nutrition Research Institute, UNC-Chapel Hill, Kannapolis, NC, USA
| | - Wei Sha
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA,Department of Cancer Biostatistics, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Shan Sun
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Jiguo Zhang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chang Su
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhihong Wang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Bing Zhang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Anthony A Fodor
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | | |
Collapse
|
30
|
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: 296] [Impact Index Per Article: 59.2] [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
|
31
|
Vallianou NG, Geladari E, Kounatidis D. Microbiome and hypertension: where are we now? J Cardiovasc Med (Hagerstown) 2020; 21:83-88. [PMID: 31809283 DOI: 10.2459/jcm.0000000000000900] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Hypertension is the leading risk factor for cardiovascular disease and accounts for approximately 9.4 million deaths globally every year. Hypertension is a complex entity, which is influenced by genetic and environmental factors, such as physical inactivity, obesity, alcohol consumption, tobacco use, stress, diet and why not the microbiome. METHODS We searched PubMed using the words 'microbiome', 'microbiota' and 'hypertension' until December 2018. We found information regarding the role of the brain-gut--bone marrow axis, the brain-gut--kidney axis, the high-salt diet, short-chain fatty acids (SCFAs), neurotransitters, such as serotonin, dopamine and norepinephrine, nitric oxide, endothelin and steroids in modulating gut microbiota and in contributing to the pathogenesis of hypertension. The brain--gut--bone marrow axis refers to the hypothesis that hematopoietic stem cells might migrate to the brain or to the gut, and thus, contribute to local inflammation and several immune responses. This migration may further enhance the sympathetic activity and contribute to blood pressure elevation. On the other hand, SCFAs, such as acetate and butyrate, have been shown to exert anti-inflammatory effects on myeloid and intestinal epithelial cells. Also, researchers have noted diminution in microbial richness and diversity in hypertensive patients as well as marked differences in circulating inflammatory cells in hypertensive patients, when compared with controls. In addition, activation of renal sympathetic nerve activity might directly influence renal physiology, by altering body fluid balance and plasma metabolite secretion and retention. These events culminate in the development of chronic kidney disease and hypertension. CONCLUSION There is a long way ahead regarding the role of gut microbiota in the pathogenesis and as an adjunctive treatment of hypertension. Treatment of dysbiosis could be a useful therapeutic approach to add to traditional antihypertensive therapy. Manipulating gut microbiota using prebiotics and probiotics might prove a valuable tool to traditional antihypertensives.
Collapse
Affiliation(s)
- Natalia G Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, Athens, Greece
| | | | | |
Collapse
|
32
|
Overwyk KJ, Quader ZS, Maalouf J, Bates M, Webster J, George MG, Merritt RK, Cogswell ME. Dietary Sodium Intake and Health Indicators: A Systematic Review of Published Literature between January 2015 and December 2019. Adv Nutr 2020; 11:1174-1200. [PMID: 32449929 PMCID: PMC7490163 DOI: 10.1093/advances/nmaa049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/26/2020] [Accepted: 04/01/2020] [Indexed: 12/29/2022] Open
Abstract
As the science surrounding population sodium reduction evolves, monitoring and evaluating new studies on intake and health can help increase our understanding of the associated benefits and risks. Here we describe a systematic review of recent studies on sodium intake and health, examine the risk of bias (ROB) of selected studies, and provide direction for future research. Seven online databases were searched monthly from January 2015 to December 2019. We selected human studies that met specified population, intervention, comparison, outcome, time, setting/study design (PICOTS) criteria and abstracted attributes related to the study population, design, intervention, exposure, and outcomes, and evaluated ROB for the subset of studies on sodium intake and cardiovascular disease risks or indicators. Of 41,601 abstracts reviewed, 231 studies were identified that met the PICOTS criteria and ROB was assessed for 54 studies. One hundred and fifty-seven (68%) studies were observational and 161 (70%) focused on the general population. Five types of sodium interventions and a variety of urinary and dietary measurement methods were used to establish and quantify sodium intake. Five observational studies used multiple 24-h urine collections to assess sodium intake. Evidence mainly focused on cardiovascular-related indicators (48%) but encompassed an assortment of outcomes. Studies varied in ROB domains and 87% of studies evaluated were missing information on ≥1 domains. Two or more studies on each of 12 outcomes (e.g., cognition) not previously included in systematic reviews and 9 new studies at low ROB suggest the need for ongoing or updated systematic reviews of evidence on sodium intake and health. Summarizing evidence from assessments on sodium and health outcomes was limited by the various methods used to measure sodium intake and outcomes, as well as lack of details related to study design and conduct. In line with research recommendations identified by the National Academies of Science, future research is needed to identify and standardize methods for measuring sodium intake.
Collapse
Affiliation(s)
- Katherine J Overwyk
- Division for Heart Disease and Stroke Prevention, CDC, Atlanta, GA, USA
- IHRC, Inc. Atlanta, GA, USA
| | - Zerleen S Quader
- Division for Heart Disease and Stroke Prevention, CDC, Atlanta, GA, USA
- IHRC, Inc. Atlanta, GA, USA
| | - Joyce Maalouf
- Division for Heart Disease and Stroke Prevention, CDC, Atlanta, GA, USA
| | - Marlana Bates
- Division for Heart Disease and Stroke Prevention, CDC, Atlanta, GA, USA
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA
| | - Jacqui Webster
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Mary G George
- Division for Heart Disease and Stroke Prevention, CDC, Atlanta, GA, USA
| | - Robert K Merritt
- Division for Heart Disease and Stroke Prevention, CDC, Atlanta, GA, USA
| | - Mary E Cogswell
- Division for Heart Disease and Stroke Prevention, CDC, Atlanta, GA, USA
| |
Collapse
|
33
|
Nishiyama M, Nakamichi N, Yoshimura T, Masuo Y, Komori T, Ishimoto T, Matsuo JI, Kato Y. Homostachydrine is a Xenobiotic Substrate of OCTN1/SLC22A4 and Potentially Sensitizes Pentylenetetrazole-Induced Seizures in Mice. Neurochem Res 2020; 45:2664-2678. [PMID: 32844295 DOI: 10.1007/s11064-020-03118-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/10/2020] [Accepted: 08/15/2020] [Indexed: 12/23/2022]
Abstract
Understanding of the underlying mechanism of epilepsy is desired since some patients fail to control their seizures. The carnitine/organic cation transporter OCTN1/SLC22A4 is expressed in brain neurons and transports food-derived antioxidant ergothioneine (ERGO), L-carnitine, and spermine, all of which may be associated with epilepsy. This study aimed to clarify the possible association of this transporter with epileptic seizures. In both pentylenetetrazole (PTZ)-induced acute seizure and kindling models, ocnt1 gene knockout mice (octn1-/-) showed lower seizure scores compared with wild-type mice. Up-regulation of the epilepsy-related genes, c-fos and Arc, and the neurotrophic factor BDNF following PTZ administration was observed in the hippocampus of wild-type, but not octn1-/- mice. To find the OCTN1 substrate associated with the seizure, untargeted metabolomics analysis using liquid chromatography-quadrupole time-of-flight mass spectrometry was conducted on extracts from the hippocampus, frontal cortex, and plasma of both strains, leading to the identification of a plant alkaloid homostachydrine as a compound present in a lower concentration in octn1-/- mice. OCTN1-mediated uptake of deuterium-labeled homostachydrine was confirmed in OCTN1-transfected HEK293 cells, suggesting that this compound is a substrate of OCTN1. Homostachydrine administration increased PTZ-induced acute seizure scores and the expression of Arc in the hippocampus and that of Arc, Egr1, and BDNF in the frontal cortex. Conversely, administration of the OCTN1 substrate/inhibitor ERGO inhibited PTZ-induced kindling and reduced the plasma homostachydrine concentration. Thus, these results suggest that OCTN1 is at least partially associated with PTZ-induced seizures, which is potentially deteriorated by treatment with homostachydrine, a newly identified food-derived OCTN1 substrate.
Collapse
Affiliation(s)
- Misa Nishiyama
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Noritaka Nakamichi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan. .,Faculty of Pharmacy, Takasaki University of Health and Welfare, 60 Nakaorui-machi, Takasaki, Gunma, 370-0033, Japan.
| | - Tomoyuki Yoshimura
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Yusuke Masuo
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Tomoe Komori
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Takahiro Ishimoto
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Jun-Ichi Matsuo
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| |
Collapse
|
34
|
Derkach A, Moore SC, Boca SM, Sampson JN. Group testing in mediation analysis. Stat Med 2020; 39:2423-2436. [DOI: 10.1002/sim.8546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 11/01/2019] [Accepted: 03/05/2020] [Indexed: 11/09/2022]
Affiliation(s)
- Andriy Derkach
- Biostatistics Branch, Division of Cancer Epidemiology and GeneticsNational Cancer Institute Rockville Maryland USA
| | - Steven C. Moore
- Metabolomics Epidemiology Branch, Division of Cancer Epidemiology and GeneticsNational Cancer Institute Rockville Maryland USA
| | - Simina M. Boca
- Innovation Center for Biomedical Informatics, Department of Oncology and Biostatistics, Bioinformatics and BiomathematicsGeorgetown University Medical Center Washington District of Columbia USA
| | - Joshua N. Sampson
- Biostatistics Branch, Division of Cancer Epidemiology and GeneticsNational Cancer Institute Rockville Maryland USA
| |
Collapse
|
35
|
McGee EE, Kiblawi R, Playdon MC, Eliassen AH. Nutritional Metabolomics in Cancer Epidemiology: Current Trends, Challenges, and Future Directions. Curr Nutr Rep 2020; 8:187-201. [PMID: 31129888 DOI: 10.1007/s13668-019-00279-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Metabolomics offers several opportunities for advancement in nutritional cancer epidemiology; however, numerous research gaps and challenges remain. This narrative review summarizes current research, challenges, and future directions for epidemiologic studies of nutritional metabolomics and cancer. RECENT FINDINGS Although many studies have used metabolomics to investigate either dietary exposures or cancer, few studies have explicitly investigated diet-cancer relationships using metabolomics. Most studies have been relatively small (≤ ~ 250 cases) or have assessed a limited number of nutritional metabolites (e.g., coffee or alcohol-related metabolites). Nutritional metabolomic investigations of cancer face several challenges in study design; biospecimen selection, handling, and processing; diet and metabolite measurement; statistical analyses; and data sharing and synthesis. More metabolomics studies linking dietary exposures to cancer risk, prognosis, and survival are needed, as are biomarker validation studies, longitudinal analyses, and methodological studies. Despite the remaining challenges, metabolomics offers a promising avenue for future dietary cancer research.
Collapse
Affiliation(s)
- Emma E McGee
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Rama Kiblawi
- Division of Cancer Population Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Mary C Playdon
- Division of Cancer Population Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - A Heather Eliassen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
36
|
Louca P, Menni C, Padmanabhan S. Genomic Determinants of Hypertension With a Focus on Metabolomics and the Gut Microbiome. Am J Hypertens 2020; 33:473-481. [PMID: 32060494 DOI: 10.1093/ajh/hpaa022] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/28/2022] Open
Abstract
Epidemiologic and genomic studies have progressively improved our understanding of the causation of hypertension and the complex relationship with diet and environment. The majority of Mendelian forms of syndromic hypotension and hypertension (HTN) have all been linked to mutations in genes whose encoded proteins regulate salt-water balance in the kidney, supporting the primacy of the kidneys in blood pressure regulation. There are more than 1,477 single nucleotide polymorphisms associated with blood pressure and hypertension and the challenge is establishing a causal role for these variants. Hypertension is a complex multifactorial phenotype and it is likely to be influenced by multiple factors including interactions between diet and lifestyle factors, microbiome, and epigenetics. Given the finite genetic variability that is possible in humans, it is likely that incremental gains from single marker analyses have now plateaued and a greater leap in our understanding of the genetic basis of disease will come from integration of other omics and the interacting environmental factors. In this review, we focus on emerging results from the microbiome and metabolomics and discuss how leveraging these findings may facilitate a deeper understanding of the interrelationships between genomics, diet, and microbial ecology in humans in the causation of essential hypertension.
Collapse
Affiliation(s)
| | - Cristina Menni
- Department of Twin Research, King’s College London, London, UK
| | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
37
|
Xu H, Wang X, Feng W, Liu Q, Zhou S, Liu Q, Cai L. The gut microbiota and its interactions with cardiovascular disease. Microb Biotechnol 2020; 13:637-656. [PMID: 31984651 PMCID: PMC7111081 DOI: 10.1111/1751-7915.13524] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/22/2019] [Accepted: 11/23/2019] [Indexed: 12/13/2022] Open
Abstract
The intestine is colonized by a considerable community of microorganisms that cohabits within the host and plays a critical role in maintaining host homeostasis. Recently, accumulating evidence has revealed that the gut microbial ecology plays a pivotal role in the occurrence and development of cardiovascular disease (CVD). Moreover, the effects of imbalances in microbe-host interactions on homeostasis can lead to the progression of CVD. Alterations in the composition of gut flora and disruptions in gut microbial metabolism are implicated in the pathogenesis of CVD. Furthermore, the gut microbiota functions like an endocrine organ that produces bioactive metabolites, including trimethylamine/trimethylamine N-oxide, short-chain fatty acids and bile acids, which are also involved in host health and disease via numerous pathways. Thus, the gut microbiota and its metabolic pathways have attracted growing attention as a therapeutic target for CVD treatment. The fundamental purpose of this review was to summarize recent studies that have illustrated the complex interactions between the gut microbiota, their metabolites and the development of common CVD, as well as the effects of gut dysbiosis on CVD risk factors. Moreover, we systematically discuss the normal physiology of gut microbiota and potential therapeutic strategies targeting gut microbiota to prevent and treat CVD.
Collapse
Affiliation(s)
- Hui Xu
- Cardiovascular Centerthe First Hospital of Jilin UniversityChangchun130021China
- Pediatric Research InstituteDepartment of Pediatricsthe University of LouisvilleLouisvilleKY40202USA
| | - Xiang Wang
- Cardiovascular Centerthe First Hospital of Jilin UniversityChangchun130021China
| | - Wenke Feng
- Department of Pharmacology and Toxicologythe University of Louisville School of MedicineLouisvilleKY40202USA
- Division of Gastroenterology, Hepatology and NutritionDepartment of Medicinethe University of Louisville School of MedicineLouisvilleKY40202USA
| | - Qi Liu
- Department of Pharmacology and Toxicologythe University of Louisville School of MedicineLouisvilleKY40202USA
- Division of Gastroenterology, Hepatology and NutritionDepartment of Medicinethe University of Louisville School of MedicineLouisvilleKY40202USA
- The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou325035China
| | - Shanshan Zhou
- Cardiovascular Centerthe First Hospital of Jilin UniversityChangchun130021China
| | - Quan Liu
- Cardiovascular Centerthe First Hospital of Jilin UniversityChangchun130021China
| | - Lu Cai
- Pediatric Research InstituteDepartment of Pediatricsthe University of LouisvilleLouisvilleKY40202USA
- Department of Pharmacology and Toxicologythe University of Louisville School of MedicineLouisvilleKY40202USA
| |
Collapse
|
38
|
Song J, Tao C, Chen G, Chen S, Xu W, Du J, Yang Y, Huang Y. Reduced 24-h Sodium Excretion Is Associated With a Disturbed Plasma Acylcarnitine Profile in Vasovagal Syncope Children: A Pilot Study. Front Pediatr 2020; 8:98. [PMID: 32219086 PMCID: PMC7078237 DOI: 10.3389/fped.2020.00098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 02/26/2020] [Indexed: 11/13/2022] Open
Abstract
Objective: To investigate if the low sodium intake is associated with the plasma carnitine and acylcarnitine profile in children with vasovagal syncope (VVS). Materials and Methods: Twenty-six children suffering from VVS were recruited in the present study and divided into a group of low urinary sodium excretion or a group of normal urinary sodium excretion according to the excretion of 24-h urinary sodium <3 or 3-6 g, respectively. The excretion of 24-h urinary sodium was detected with ion-selective electrode approach. Plasma carnitine and acylcarnitine concentrations were measured with tandem mass spectrometry. Each participant completed the head-up tilt test. The demographics, clinical characteristics, hemodynamic parameters and plasma carnitine and acylcarnitine concentrations were compared between the two groups. A bivariate correlation between plasma acylcarnitine profiles and the excretion of 24-h urinary sodium was conducted with Spearman's correlation coefficients. Results: Of the enrolled VVS patients, 14 patients were assigned to the group of low urinary sodium excretion and the remaining 12 patients were assigned to the group of normal urinary sodium excretion. Symptoms of fatigue were more prevalent in the group of low urinary sodium excretion than in the group of normal urinary sodium excretion (p = 0.009). Aside from fatigue, no other differences in the demographics, clinical characteristics or hemodynamic parameters during the head-up tilt test were found between the two groups (p > 0.05). Concentrations of plasma tiglylcarnitine (C5:1), hydroxyhexadecanoylcarnitine (C16OH), hydroxyoctadecanoylcarnitine (C18OH), and carnitine C22 were significantly higher in the group of low urinary sodium excretion than in the group of normal urinary sodium excretion (all p-values = 0.048); moreover, they were all negatively correlated with 24-h urinary sodium levels (all p-values = 0.016). There were no differences between the two groups in other acylcarnitines or free carnitine. Conclusions: Reduced excretion of 24-h urinary sodium is associated with a disturbed plasma acylcarnitine profile in children with VVS. The findings suggest that restricted sodium intake-induced disturbance of plasma acylcarnitines and related cellular energy metabolism might be involved in the pathogenesis of VVS in children.
Collapse
Affiliation(s)
- Jinqing Song
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Chunyan Tao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Guozhen Chen
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Department of Pediatrics, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Selena Chen
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Wenrui Xu
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Junbao Du
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yanling Yang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yaqian Huang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| |
Collapse
|
39
|
Calcium Oxalate Nephrolithiasis and Gut Microbiota: Not just a Gut-Kidney Axis. A Nutritional Perspective. Nutrients 2020; 12:nu12020548. [PMID: 32093202 PMCID: PMC7071363 DOI: 10.3390/nu12020548] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/14/2020] [Accepted: 02/18/2020] [Indexed: 12/13/2022] Open
Abstract
Recent studies have shown that patients with kidney stone disease, and particularly calcium oxalate nephrolithiasis, exhibit dysbiosis in their fecal and urinary microbiota compared with controls. The alterations of microbiota go far beyond the simple presence and representation of Oxalobacter formigenes, a well-known symbiont exhibiting a marked capacity of degrading dietary oxalate and stimulating oxalate secretion by the gut mucosa. Thus, alterations of the intestinal microbiota may be involved in the pathophysiology of calcium kidney stones. However, the role of nutrition in this gut-kidney axis is still unknown, even if nutritional imbalances, such as poor hydration, high salt, and animal protein intake and reduced fruit and vegetable intake, are well-known risk factors for kidney stones. In this narrative review, we provide an overview of the gut-kidney axis in nephrolithiasis from a nutritional perspective, summarizing the evidence supporting the role of nutrition in the modulation of microbiota composition, and their relevance for the modulation of lithogenic risk.
Collapse
|
40
|
Brown AW, Kaiser KA, Keitt A, Fontaine K, Gibson M, Gower BA, Shikany JM, Vorland CJ, Beitz DC, Bier DM, Brenna JT, Jacobs DR, Kris-Etherton P, Maki K, Miller M, St-Onge MP, Teran-Garcia M, Allison DB. Science dialogue mapping of knowledge and knowledge gaps related to the effects of dairy intake on human cardiovascular health and disease. Crit Rev Food Sci Nutr 2020; 61:179-195. [PMID: 32072820 DOI: 10.1080/10408398.2020.1722941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Dairy has been described as everything from a superfood to a poison; yet, arguments, assumptions, and data justifying these labels are not always clear. We used an issue-based information system, "dialogue mapping™," to summarize scientific points of a live panel discussion on the putative effects of dairy on cardiovascular diseases (CVD) from a day-long session among experts in nutrition and CVD. Dialogue mapping captures relations among ideas to explicitly, logically, and visually connect issues/questions, ideas, pro/con arguments, and agreements, even if discussed at different times. Experts discussed two propositions: for CVD risk, consumption of full-fat dairy products 1) should be minimized, in part because of their saturated fat content, or 2) need not be minimized, despite their saturated fat content. The panel discussed the dairy-CVD relation through blood lipids, diabetes, obesity, energy balance, blood pressure, dairy bioactives, biobehavioral components, and other putative causal pathways. Associations and effects reported in the literature have varied by fat content of dairy elements considered, study design, intake methods, and biomarker versus disease outcomes. Two conceptual topics emerged from the discussion: 1) individual variability: whether recommendations should be targeted only to those at high CVD risk; 2) quality of evidence: whether data on dairy-CVD relations are strong enough for reliable conclusions-positive, negative, or null. Future procedural improvements for science dialog mapping include using singular rather than competing propositions for discussion.
Collapse
Affiliation(s)
- Andrew W Brown
- Department of Applied Health Science, Indiana University School of Public Health-Bloomington, Bloomington, Indiana, USA
| | - Kathryn A Kaiser
- Department of Health Behavior, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Andrew Keitt
- Department of History, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kevin Fontaine
- Department of Health Behavior, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Madeline Gibson
- School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Barbara A Gower
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James M Shikany
- Division of Preventive Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Colby J Vorland
- Department of Applied Health Science, Indiana University School of Public Health-Bloomington, Bloomington, Indiana, USA
| | - Donald C Beitz
- Departments of Animal Science and Biochemistry, Biophysics, and Molecular Biology, Iowa State University, Ames, Iowa, USA
| | - Dennis M Bier
- Baylor College of Medicine, Department of Pediatrics, Children's Nutrition Research Center, Houston, Texas, USA
| | - J Thomas Brenna
- Dell Pediatric Research Institute, Deptartments of Pediatrics, of Chemistry, and of Nutrition, University of Texas at Austin, Austin, Texas, USA
| | - David R Jacobs
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Penny Kris-Etherton
- Distinguished Professor of Nutrition, Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Kevin Maki
- Department of Applied Health Science, Indiana University School of Public Health-Bloomington, Bloomington, Indiana, USA.,Midwest Biomedical Research/Center for Metabolic and Cardiovascular Health, Addison, Illinois, USA
| | - Michael Miller
- Epidemiology & Public Health, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Marie-Pierre St-Onge
- Division of Endocrinology and Sleep center of excellence, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Margarita Teran-Garcia
- Extension Specialist Hispanic Health Programs, Department of Human Development and Family Studies, Cooperative Extension, Division of Nutritional Sciences, Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - David B Allison
- Department of Epidemiology and Biostatistics, Indiana University School of Public Health-Bloomington, Bloomington, Indiana, USA
| |
Collapse
|
41
|
Long NP, Nghi TD, Kang YP, Anh NH, Kim HM, Park SK, Kwon SW. Toward a Standardized Strategy of Clinical Metabolomics for the Advancement of Precision Medicine. Metabolites 2020; 10:E51. [PMID: 32013105 PMCID: PMC7074059 DOI: 10.3390/metabo10020051] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/18/2022] Open
Abstract
Despite the tremendous success, pitfalls have been observed in every step of a clinical metabolomics workflow, which impedes the internal validity of the study. Furthermore, the demand for logistics, instrumentations, and computational resources for metabolic phenotyping studies has far exceeded our expectations. In this conceptual review, we will cover inclusive barriers of a metabolomics-based clinical study and suggest potential solutions in the hope of enhancing study robustness, usability, and transferability. The importance of quality assurance and quality control procedures is discussed, followed by a practical rule containing five phases, including two additional "pre-pre-" and "post-post-" analytical steps. Besides, we will elucidate the potential involvement of machine learning and demonstrate that the need for automated data mining algorithms to improve the quality of future research is undeniable. Consequently, we propose a comprehensive metabolomics framework, along with an appropriate checklist refined from current guidelines and our previously published assessment, in the attempt to accurately translate achievements in metabolomics into clinical and epidemiological research. Furthermore, the integration of multifaceted multi-omics approaches with metabolomics as the pillar member is in urgent need. When combining with other social or nutritional factors, we can gather complete omics profiles for a particular disease. Our discussion reflects the current obstacles and potential solutions toward the progressing trend of utilizing metabolomics in clinical research to create the next-generation healthcare system.
Collapse
Affiliation(s)
- Nguyen Phuoc Long
- College of Pharmacy, Seoul National University, Seoul 08826, Korea; (N.P.L.); (N.H.A.); (H.M.K.)
| | - Tran Diem Nghi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea; (T.D.N.); (S.K.P.)
| | - Yun Pyo Kang
- Department of Cancer Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA;
| | - Nguyen Hoang Anh
- College of Pharmacy, Seoul National University, Seoul 08826, Korea; (N.P.L.); (N.H.A.); (H.M.K.)
| | - Hyung Min Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Korea; (N.P.L.); (N.H.A.); (H.M.K.)
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea; (T.D.N.); (S.K.P.)
| | - Sung Won Kwon
- College of Pharmacy, Seoul National University, Seoul 08826, Korea; (N.P.L.); (N.H.A.); (H.M.K.)
| |
Collapse
|
42
|
Bartolomaeus H, Markó L, Wilck N, Luft FC, Forslund SK, Muller DN. Precarious Symbiosis Between Host and Microbiome in Cardiovascular Health. Hypertension 2019; 73:926-935. [PMID: 30905198 DOI: 10.1161/hypertensionaha.119.11786] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Hendrik Bartolomaeus
- From the Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., L.M., N.W., F.C.L., S.K.F., D.N.M.).,Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (H.B., L.M., N.W., F.C.L., D.N.M.).,Max Delbruck Center for Molecular Medicine, Berlin, Germany (H.B., F.C.L., S.K.F., D.N.M.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin (H.B., L.M., N.W., D.N.M., S.K.F.).,Berlin Institute of Health (BIH), Germany (H.B., L.M., N.W., S.K.F., D.N.M.)
| | - Lajos Markó
- From the Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., L.M., N.W., F.C.L., S.K.F., D.N.M.).,Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (H.B., L.M., N.W., F.C.L., D.N.M.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin (H.B., L.M., N.W., D.N.M., S.K.F.).,Berlin Institute of Health (BIH), Germany (H.B., L.M., N.W., S.K.F., D.N.M.)
| | - Nicola Wilck
- From the Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., L.M., N.W., F.C.L., S.K.F., D.N.M.).,Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (H.B., L.M., N.W., F.C.L., D.N.M.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin (H.B., L.M., N.W., D.N.M., S.K.F.).,Berlin Institute of Health (BIH), Germany (H.B., L.M., N.W., S.K.F., D.N.M.)
| | - Friedrich C Luft
- From the Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., L.M., N.W., F.C.L., S.K.F., D.N.M.).,Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (H.B., L.M., N.W., F.C.L., D.N.M.).,Max Delbruck Center for Molecular Medicine, Berlin, Germany (H.B., F.C.L., S.K.F., D.N.M.)
| | - Sofia K Forslund
- From the Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., L.M., N.W., F.C.L., S.K.F., D.N.M.).,Max Delbruck Center for Molecular Medicine, Berlin, Germany (H.B., F.C.L., S.K.F., D.N.M.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin (H.B., L.M., N.W., D.N.M., S.K.F.).,Berlin Institute of Health (BIH), Germany (H.B., L.M., N.W., S.K.F., D.N.M.)
| | - Dominik N Muller
- From the Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Germany (H.B., L.M., N.W., F.C.L., S.K.F., D.N.M.).,Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany (H.B., L.M., N.W., F.C.L., D.N.M.).,Max Delbruck Center for Molecular Medicine, Berlin, Germany (H.B., F.C.L., S.K.F., D.N.M.).,DZHK (German Centre for Cardiovascular Research), partner site Berlin (H.B., L.M., N.W., D.N.M., S.K.F.).,Berlin Institute of Health (BIH), Germany (H.B., L.M., N.W., S.K.F., D.N.M.)
| |
Collapse
|
43
|
Nealon NJ, Parker KD, Lahaie P, Ibrahim H, Maurya AK, Raina K, Ryan EP. Bifidobacterium longum-fermented rice bran and rice bran supplementation affects the gut microbiome and metabolome. Benef Microbes 2019; 10:823-839. [PMID: 31965839 DOI: 10.3920/bm2019.0017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This study investigated gut microbiota composition along with food, host, and microbial derived metabolites in the colon and systemic circulation of healthy mice following dietary rice bran and fermented rice bran intake. Adult male BALB/c mice were fed a control diet or one of two experimental diets containing 10% w/w rice bran fermented by Bifidobacterium longum or 10% w/w non-fermented rice bran for 15 weeks. Metabolomics was performed on the study diets (food), the murine colon and whole blood. These were analysed in concert with 16S rRNA amplicon sequencing of faeces, caecum, and colon microbiomes. Principal components analysis of murine microbiota composition displayed marked separation between control and experimental diets, and between faecal and tissue (caecum and colon) microbiomes. Colon and caecal microbiomes in both experimental diet groups showed enrichment of Roseburia, Lachnospiraceae, and Clostridiales related amplicon sequence variants compared to control. Bacterial composition was largely similar between experimental diets. Metabolite profiling revealed 530 small molecules comprising of 39% amino acids and 21% lipids that had differential abundances across food, colon, and blood matrices, and statistically significant between the control, rice bran, and fermented rice bran groups. The amino acid metabolite, N-delta-acetylornithine, was notably increased by B. longum rice bran fermentation when compared to non-fermented rice bran in food, colon, and blood. These findings support that dietary intake of rice bran fermented with B. longum modulates multiple metabolic pathways important to the gut and overall health.
Collapse
Affiliation(s)
- N J Nealon
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, USA.,Program in Cellular and Molecular Biology, Colorado State University, Fort Collins, 80521 CO, USA
| | - K D Parker
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, USA
| | - P Lahaie
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, USA
| | - H Ibrahim
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, USA.,Zagazig University, Department of Medical Biochemistry, Faculty of Medicine, 44511 Zagazig, Egypt
| | - A K Maurya
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - K Raina
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA.,Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - E P Ryan
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, USA.,Program in Cellular and Molecular Biology, Colorado State University, Fort Collins, 80521 CO, USA.,University of Colorado Cancer Center, Division of Cancer Control and Prevention, Aurora, CO 80045, USA
| |
Collapse
|
44
|
Protective Effects of Dietary MUFAs Mediating Metabolites against Hypertension Risk in the Korean Genome and Epidemiology Study. Nutrients 2019; 11:nu11081928. [PMID: 31426326 PMCID: PMC6722700 DOI: 10.3390/nu11081928] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 12/29/2022] Open
Abstract
Background and Aims: Metabolites related to dietary factors can be used to identify biological markers to prevent metabolic disease. However, most studies have been conducted in the United States and Europe, and those in the Asian region are limited. We investigated the effects of dietary monounsaturated fatty acids (MUFAs) and metabolites on new-onset hypertension in the Korean Genome and Epidemiology Study. Method and Results: A total of 1529 subjects without hypertension were divided into tertiles of dietary MUFAs intake. After a 4-year follow-up, 135 serum metabolites were measured using the AbsoluteIDQ p180 kit. During the 4-year follow-up period, 193 new-onset hypertension incidences were observed. The highest MUFAs intake group was inversely associated with the risk of hypertension compared with the lowest MUFAs intake group (odds ratio (OR) = 0.49, (95% confidence interval (CI) = 0.29–0.82)). Of the 135 metabolites, eight were significantly associated with MUFAs intake. Phosphatidylcholine-diacyl (PC aa) C 38:1 and hydroxysphingomyelin (SM OH) C 16:1 were associated with a decrease in hypertension risk (PC aa C 38:1, OR = 0.60 (95% CI = 0.37–0.96); SM OH C 16:1, OR = 0.42 (95% CI = 0.20–0.90)). The highest MUFAs intake group had a significantly decreased risk of hypertension, even considering PC aa C 38:1 and SM (OH) C 16:1 as a mediator. Conclusion: We confirmed that dietary MUFAs intake, and PC aa C 38:1 and SM (OH) C 16:1 had protective effects against hypertension. Furthermore, high MUFAs intake combined with PC aa C 38:1 and SM (OH) C 16:1 has the most significant effect on reducing the risk hypertension.
Collapse
|
45
|
Zhao X, Niu L, Clerici C, Russo R, Byrd M, Setchell KD. Data analysis of MS-based clinical lipidomics studies with crossover design: A tutorial mini-review of statistical methods. CLINICAL MASS SPECTROMETRY (DEL MAR, CALIF.) 2019; 13:5-17. [PMID: 34841080 PMCID: PMC8620525 DOI: 10.1016/j.clinms.2019.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 05/15/2019] [Accepted: 05/15/2019] [Indexed: 11/28/2022]
Abstract
Clinical lipidomics using mass spectrometry (MS) is important to support discovery of biomarkers for diagnosis and understanding the pathophysiology of diseases. Frequently, lipidomics data from clinical studies have large variations among individuals because the human metabolome/lipidome is strongly influenced by genotype, daily activity, diet and gut flora. This inter-personal variability makes data analysis more complex and normally requires a large cohort for robust statistical analysis. Crossover designed experiments treat each subject as his or her own control, thereby reducing the between-subject variability, such that the effects of exposure/treatment are more likely to be identified when using a relatively small number of subjects. This design repeatedly samples an individual when crossing over from one treatment/exposure to another during the course of the study. The acquired datasets have a distinct data structure resulting from repeated longitudinal measurements. A variety of statistical methods are used in published crossover studies, but many appear to ignore the data structure inherent in the experimental design. An appropriate data analysis approach is critical to discovering robust clinical biomarkers. Hereby, we summarize the statistical methodologies suitable for clinical lipidomics studies using crossover design. To help understand and apply these methods to practical cases, we focused on the general concepts of statistical models in the context of analysis of metabolomics data without spending too much effort on mathematical details. Importantly, we aim to evaluate these methods and provide suggestions for data analysis and biomarker discovery. We applied the discussed methods on a MS-based lipidomics dataset from a double-blind random crossover designed clinical dietary intervention study. The strength and potential pitfalls of each method are briefly discussed and a suggestion for analytic workflow proposed.
Collapse
Affiliation(s)
- Xueheng Zhao
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Liang Niu
- Division of Biostatistics and Bioinformatics, Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Carlo Clerici
- Clinica Di Gastroenterologia – Endoscopia – Epatologia, Policlinico, S. Maria Della Misericordia Azienda Ospedaliera Di Perugia, Italy
| | - Roberta Russo
- Clinica Di Gastroenterologia – Endoscopia – Epatologia, Policlinico, S. Maria Della Misericordia Azienda Ospedaliera Di Perugia, Italy
| | - Melissa Byrd
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Kenneth D.R. Setchell
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
46
|
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
|
47
|
Zubcevic J, Richards EM, Yang T, Kim S, Sumners C, Pepine CJ, Raizada MK. Impaired Autonomic Nervous System-Microbiome Circuit in Hypertension. Circ Res 2019; 125:104-116. [PMID: 31219753 PMCID: PMC6588177 DOI: 10.1161/circresaha.119.313965] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hypertension affects an estimated 103 million Americans, yet gaps in knowledge continue to limit its successful management. Rapidly emerging evidence is linking gut dysbiosis to many disorders and diseases including hypertension. The evolution of the -omics techniques has allowed determination of the abundance and potential function of gut bacterial species by next-generation bacterial sequencing, whereas metabolomics techniques report shifts in bacterial metabolites in the systemic circulation of hypertensive patients and rodent models of hypertension. The gut microbiome and host have evolved to exist in balance and cooperation, and there is extensive crosstalk between the 2 to maintain this balance, including during regulation of blood pressure. However, an understanding of the mechanisms of dysfunctional host-microbiome interactions in hypertension is still lacking. Here, we synthesize some of our recent data with published reports and present concepts and a rationale for our emerging hypothesis of a dysfunctional gut-brain axis in hypertension. Hopefully, this new information will improve the understanding of hypertension and help to address some of these knowledge gaps.
Collapse
Affiliation(s)
- Jasenka Zubcevic
- Department of Physiological Sciences, College of Veterinary Medicine; University of Florida, Gainesville FL32610
| | - Elaine M. Richards
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville FL32610
| | - Tao Yang
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville FL32610
| | - Seungbum Kim
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville FL32610
| | - Colin Sumners
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville FL32610
| | - Carl J Pepine
- Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville FL32610
| | - Mohan K Raizada
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville FL32610
| |
Collapse
|
48
|
Chen L, He FJ, Dong Y, Huang Y, Harshfield GA, Zhu H. Sodium Reduction, Metabolomic Profiling, and Cardiovascular Disease Risk in Untreated Black Hypertensives. Hypertension 2019; 74:194-200. [PMID: 31079530 PMCID: PMC9116731 DOI: 10.1161/hypertensionaha.119.12880] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dietary sodium restriction has multiple beneficial effects on cardiovascular health. The underlying mechanisms are not fully understood, and the roles of metabolomics have been rarely studied. We aimed to test the hypothesis that the reduction in dietary sodium intake would induce changes in metabolomic profiling among black hypertensives, and the changes would be associated with reduced blood pressure (BP) and improved skin capillary density. A total of 64 untreated black hypertensives were included from a randomized, double-blind, placebo-controlled cross-over trial of sodium reduction. The participants were given either 9 slow sodium tablets (10 mmol sodium per tablet) or placebo tablets daily for 6 weeks, they then crossed over to receive the other tablets for another 6 weeks, while on reduced sodium diet aiming at achieving daily sodium intake around 2.0 g. Untargeted metabolomic profiling was performed in paired serum samples, which were collected at the end of each period, so were BP and capillary density. Mixed-effects models were used. There were 34 metabolites identified with raw P's<0.05. Among those, 2 metabolites including β-hydroxyisovalerate and methionine sulfone were significantly increased with sodium reduction (false discovery rate =0.006 and 0.099, respectively). Increased β-hydroxyisovalerate was associated with reduced office systolic BP and ambulatory daytime systolic BP, whereas increased methionine sulfone was associated with reduced 24-hour diastolic BP, ambulatory nighttime diastolic BP, and increased skin capillary density. Our results suggest that dietary sodium reduction increases the circulating levels of β-hydroxyisovalerate and methionine sulfone. Further studies are warranted. Clinical Trial Registration- URL: http://www.clinicaltrials.gov . Unique identifier: NCT00152074.
Collapse
Affiliation(s)
- Li Chen
- From the Georgia Prevention Institute, Department of Population Health Sciences, Medical College of Georgia, Augusta University (L.C., Y.D., Y.H., G.A.H., H.Z.)
| | - Feng J He
- Center for Environmental and Preventive Medicine, Wolfson Institute of Preventive Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (F.J.H.)
| | - Yanbin Dong
- From the Georgia Prevention Institute, Department of Population Health Sciences, Medical College of Georgia, Augusta University (L.C., Y.D., Y.H., G.A.H., H.Z.)
| | - Ying Huang
- From the Georgia Prevention Institute, Department of Population Health Sciences, Medical College of Georgia, Augusta University (L.C., Y.D., Y.H., G.A.H., H.Z.)
| | - Gregory A Harshfield
- From the Georgia Prevention Institute, Department of Population Health Sciences, Medical College of Georgia, Augusta University (L.C., Y.D., Y.H., G.A.H., H.Z.)
| | - Haidong Zhu
- From the Georgia Prevention Institute, Department of Population Health Sciences, Medical College of Georgia, Augusta University (L.C., Y.D., Y.H., G.A.H., H.Z.)
| |
Collapse
|
49
|
Gordon-Dseagu VLZ, Derkach A, Xiao Q, Williams I, Sampson J, Stolzenberg-Solomon RZ. The association of sleep with metabolic pathways and metabolites: evidence from the Dietary Approaches to Stop Hypertension (DASH)-sodium feeding study. Metabolomics 2019; 15:48. [PMID: 30879189 PMCID: PMC8513072 DOI: 10.1007/s11306-019-1472-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 01/04/2019] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Sleep is increasingly being viewed as an issue of public health concern, yet few epidemiologic studies have explored associations between sleep habits and metabolomic profile. OBJECTIVES To assess the association between sleep and blood metabolites. METHODS We examined the association between sleep and 891 fasting plasma metabolites in a subgroup of 106 participants from the Dietary Approaches to Stop Hypertension (DASH)-Sodium feeding trial (1997-1999). We produced two sleep variables to analyze, sleep midpoint (median time between bedtime and waketime) and sleep duration, as well as bedtime and wake time. Metabolites were measured using liquid and gas chromatography, coupled with mass spectrometry. We assessed associations between sleep variables and log transformed metabolites using linear mixed-effects models. We combined the resulting p-values using Fisher's method to calculate associations between sleep and 38 metabolic pathways. RESULTS Sixteen pathways were associated (p < 0.05) with midpoint. Only the γ-glutamyl amino acid metabolism pathway reached Bonferroni-corrected threshold (0.0013). Eighty-three metabolites were associated with midpoint (FDR < 0.20). Similar associations were found for wake time. Neither bed time nor duration were strongly associated. The top metabolites (pathways given in brackets) associated with sleep were erythrulose (advanced glycation end-product) (positive association) and several γ-glutamyl pathway metabolites, including CMPF (fatty acid, dicarboxylate), isovalerate (valine, leucine and isoleucine and fatty acid metabolism) and HWESASXX (polypeptide) (inverse association). CONCLUSION Within our study, several metabolites that have previously been linked to inflammation and oxidative stress (processes involved in diseases such as cardiovascular disease and cancer) were found to be associated with sleep.
Collapse
Affiliation(s)
- Vanessa L Z Gordon-Dseagu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Bethesda, MD, 20850, USA.
| | - Andriy Derkach
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Bethesda, MD, 20850, USA
| | - Qian Xiao
- Department of Health and Human Physiology, College of Liberal Arts and Sciences, University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Ishmael Williams
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Bethesda, MD, 20850, USA
| | - Joshua Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Bethesda, MD, 20850, USA
| | - Rachael Z Stolzenberg-Solomon
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Bethesda, MD, 20850, USA
| |
Collapse
|
50
|
van Roekel EH, Loftfield E, Kelly RS, Zeleznik OA, Zanetti KA. Metabolomics in epidemiologic research: challenges and opportunities for early-career epidemiologists. Metabolomics 2019; 15:9. [PMID: 30830482 PMCID: PMC7844759 DOI: 10.1007/s11306-018-1468-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 12/21/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND The application of metabolomics to epidemiologic studies is increasing. AIM OF REVIEW Here, we describe the challenges and opportunities facing early-career epidemiologists aiming to apply metabolomics to their research. KEY SCIENTIFIC CONCEPTS OF REVIEW Many challenges inherent to metabolomics may provide early-career epidemiologists with the opportunity to play a pivotal role in answering critical methodological questions and moving the field forward. Although generating large-scale high-quality metabolomics data can be challenging, data can be accessed through public databases, collaboration with senior researchers or participation within interest groups. Such efforts may also assist with obtaining funding, provide knowledge on training resources, and help early-career epidemiologists to publish in the field of metabolomics.
Collapse
Affiliation(s)
- Eline H van Roekel
- Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, Netherlands.
| | - Erikka Loftfield
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, USA
| | - Rachel S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Oana A Zeleznik
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Krista A Zanetti
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Bethesda, USA
| |
Collapse
|