1
|
Lee H, Feranil JB, Jose PA. An Overview on Renal and Central Regulation of Blood Pressure by Neuropeptide FF and Its Receptors. Int J Mol Sci 2024; 25:13284. [PMID: 39769048 PMCID: PMC11675822 DOI: 10.3390/ijms252413284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Neuropeptide FF (NPFF) is an endogenous octapeptide that was originally isolated from the bovine brain. It belongs to the RFamide family of peptides that has a wide range of physiological functions and pathophysiological effects. NPFF and its receptors, NPFFR1 and NPFFR2, abundantly expressed in rodent and human brains, participate in cardiovascular regulation. However, the expressions of NPFF and its receptors are not restricted within the central nervous system but are also found in peripheral organs, including the kidneys. Both NPFFR1 and NPFFR2 mainly couple to Gαi/o, which inhibits cyclic adenosine monophosphate (cAMP) production. NPFF also weakly binds to other RFamide receptors and the Mas receptor. Relevant published articles were searched in PubMed, Google Scholar, Web of Science, and Scopus. Herein, we review evidence for the role of NPFF in the regulation of blood pressure, in the central nervous system, particularly within the hypothalamic paraventricular nucleus and the brainstem, and the kidneys. NPFF is a potential target in the treatment of hypertension.
Collapse
Affiliation(s)
- Hewang Lee
- Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (J.B.F.); (P.A.J.)
| | - Jun B. Feranil
- Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (J.B.F.); (P.A.J.)
| | - Pedro A. Jose
- Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (J.B.F.); (P.A.J.)
- Department of Pharmacology & Physiology, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA
| |
Collapse
|
2
|
Albritton CF, Demirci M, Neikirk K, Ertuglu LA, Ishimwe JA, Mutchler AL, Sheng Q, Laffer CL, Wanjalla CN, Ahmed T, Haynes AP, Saleem M, Beasley HK, Marshall AG, Vue Z, Ikizler AT, Kleyman TR, Kon V, Hinton A, Kirabo A. Myeloid Cell Glucocorticoid, Not Mineralocorticoid Receptor Signaling, Contributes to Salt-Sensitive Hypertension in Humans via Cortisol. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598374. [PMID: 38915603 PMCID: PMC11195113 DOI: 10.1101/2024.06.10.598374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
BACKGROUND Salt sensitivity of blood pressure (SSBP) is an independent risk factor for cardiovascular morbidity and mortality, yet the etiology is poorly understood. We previously found that serum/glucocorticoid-regulated kinase 1 (SGK1) and epoxyeicosatrienoic acids (EETs) regulate epithelial sodium channel (ENaC)-dependent sodium entry into monocyte-derived antigen-presenting cells (APCs) and activation of NADPH oxidase, leading to the formation of isolevuglandins (IsoLGs) in SSBP. Whereas aldosterone via the mineralocorticoid receptor (MR) activates SGK1 leading to hypertension, our past findings indicate that levels of plasma aldosterone do not correlate with SSBP, and there is little to no MR expression in APCs. Thus, we hypothesized that cortisol acting via the glucocorticoid receptor (GR), not the MR in APCs mediates SGK1 actions to induce SSBP. METHODS We performed cellular indexing of transcriptomes and epitopes by sequencing (CITE-Seq) analysis on peripheral blood mononuclear cells of humans rigorously phenotyped for SSBP using an inpatient salt loading/depletion protocol to determine expression of MR, GR, and SGK1 in immune cells. In additional experiments, we performed bulk transcriptomic analysis on isolated human monocytes following in vitro treatment with high salt from a separate cohort. We then measured urine and plasma cortisol, cortisone, renin, and aldosterone. Subsequently, we measured the association of these hormones with changes in systolic, diastolic, mean arterial pressure and pulse pressure as well as immune cell activation via IsoLG formation. RESULTS We found that myeloid APCs predominantly express the GR and SGK1 with no expression of the MR. Expression of the GR in APCs increased after salt loading and decreased with salt depletion in salt-sensitive but not salt-resistant people and was associated with increased expression of SGK1. Moreover, we found that plasma and urine cortisol/cortisone but not aldosterone/renin correlated with SSBP and APCs activation via IsoLGs. We also found that cortisol negatively correlates with EETs. CONCLUSION Our findings suggest that renal cortisol signaling via the GR but not the MR in APCs contributes to SSBP via cortisol. Urine and plasma cortisol may provide an important currently unavailable feasible diagnostic tool for SSBP. Moreover, cortisol-GR-SGK1-ENaC signaling pathway may provide treatment options for SSBP.
Collapse
Affiliation(s)
- Claude F. Albritton
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208-3501, USA
| | - Mert Demirci
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kit Neikirk
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Lale A. Ertuglu
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jeanne A Ishimwe
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Ashley L Mutchler
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Cheryl L Laffer
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Celestine N. Wanjalla
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Taseer Ahmed
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
- Department of Pharmacology, College of Pharmacy, University of Sargodha, University Road, Sargodha, Punjab, 40100, Pakistan
| | - Alexandria Porcia Haynes
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Mohammad Saleem
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
| | - Heather K. Beasley
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Andrea G. Marshall
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Zer Vue
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Alp T Ikizler
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Thomas R. Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Valentina Kon
- Division of Nephrology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Antentor Hinton
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37212-8802, USA
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Immunology and Inflammation
- Vanderbilt Institute for Global Health
| |
Collapse
|
3
|
Maamor NH, Ismail J, Malek KA, Yusoff K, Boon-Peng H. AGT, CYP11B2 & ADRB2 gene polymorphism & essential hypertension (HT): A meta-analysis. Indian J Med Res 2024; 159:619-626. [PMID: 39382462 PMCID: PMC11463865 DOI: 10.25259/ijmr_520_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Indexed: 10/10/2024] Open
Abstract
Background & objectives The results of the genetic association studies between the selected candidate genes and hypertension (HT) contradicted across different populations. Majority of the meta-analyses carried out did not consider population genetic ancestry as a confounding factor. Therefore, this meta-analysis attempted to consolidate and re-evaluate the findings of the association between the selected candidate variants (AGT-rs699, CYP11B2-rs1799998, ADRB2-rs1042713 and rs1042714) and HT, by categorizing the genotyping data based on known genetic ancestry, and/or major geographical populations. Methods Publications were retrieved from PubMed, Cochrane and World of Science. The included articles were further divided into different populations based on their known genetic and/or geographical ancestry. Results AGTrs699-G was significantly associated with HT among Indians for (i) allele [P=0.03, Odds ratio (OR): 1.37, 95% Confidence Interval (CI): 1.03-1.82], and (ii) dominant mode of inheritance (P=0.009, OR:1.45, 95% CI: 1.09-1.91). CYP11B2rs1799998-G was significantly associated with HT in Europeans for (i) allele (P=6.9 × 10-5, OR: 0.82, 95% CI: 0.74-0.9), (ii) recessive (P=6.38 × 10-5, OR: 0.7, 95% CI: 0.59-0.83) and (iii) dominant mode of inheritance (P=0.008, OR: 0.81, 95% CI: 0.7-0.94). ADRB2-rs1042713-G was significantly associated with HT in east Asians for (i) allele (P=0.01, OR: 1.26, 95% CI: 1.05-1.51), and (ii) recessive mode of inheritance (P=0.04, OR: 1.36, 95% CI: 1.01-1.83). Interpretation & conclusions Different genotype and allele frequencies in diverse populations result in different genetic associations with HT across populations. This meta-analysis finding provides an update and summary of the genetic association between the selected simple nucleotide polymorphism (SNPs) and HT across different populations and essential insights into selecting appropriate pharmacogenetic marker(s) for effective HT management in populations of different ancestries.
Collapse
Affiliation(s)
- Nur Hasnah Maamor
- Faculty of Medicine & Health Sciences, UCSI University, UCSI Hospital, Negeri Sembilan, Malaysia
- Sector for Evidence-Based Healthcare, National Institute of Health, Ministry of Health Setia Alam, Malaysia
| | - Johanrizwal Ismail
- UiTM Private Specialist Centre, Jalan Hospital, Selangor, Malaysia
- Cardiology Unit, Prince Court Medical Centre, Kuala Lumpur, Malaysia
- MAA Medicare Cardiac Diagnostic Centre, Kuala Lumpur, Malaysia
| | - Khasnur Abd Malek
- Faculty of Medicine, Universiti Teknologi MARA, Jalan Hospital, Selangor, Malaysia
| | - Khalid Yusoff
- Faculty of Medicine & Health Sciences, UCSI University, UCSI Hospital, Negeri Sembilan, Malaysia
| | - Hoh Boon-Peng
- Faculty of Medicine & Health Sciences, UCSI University, UCSI Hospital, Negeri Sembilan, Malaysia
| |
Collapse
|
4
|
Abstract
The year 2024 marks the centennial of the initiation of the American Heart Association. Over the past 100 years, the American Heart Association has led groundbreaking discoveries in cardiovascular disease including salt sensitivity of blood pressure, which has been studied since the mid-1900s. Salt sensitivity of blood pressure is an important risk factor for cardiovascular events, but the phenotype remains unclear because of insufficient understanding of the underlying mechanisms and lack of feasible diagnostic tools. In honor of this centennial, we commemorate the initial discovery of salt sensitivity of blood pressure and chronicle the subsequent scientific discoveries and efforts to mitigate salt-induced cardiovascular disease with American Heart Association leading the way. We also highlight determinants of the pathophysiology of salt sensitivity of blood pressure in humans and recent developments in diagnostic methods and future prospects.
Collapse
Affiliation(s)
- Jeremiah Afolabi
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.A., C.L.L., A.K.)
| | - Cheryl L Laffer
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.A., C.L.L., A.K.)
| | - Heather K Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN (H.K.B., A.H.)
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN (H.K.B., A.H.)
| | - Sepiso K Masenga
- HAND Research Group (Hypertension, HIV/AIDS, Nutrition, Diabetes and Dyslipidemia Research Group), School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia (S.K.M.)
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.A., C.L.L., A.K.)
- Vanderbilt Center for Immunobiology, Nashville, TN (A.K.)
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN (A.K.)
- Vanderbilt Institute for Global Health, Nashville, TN (A.K.)
| |
Collapse
|
5
|
Fagunwa O, Davies K, Bradbury J. The Human Gut and Dietary Salt: The Bacteroides/ Prevotella Ratio as a Potential Marker of Sodium Intake and Beyond. Nutrients 2024; 16:942. [PMID: 38612976 PMCID: PMC11013828 DOI: 10.3390/nu16070942] [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: 01/23/2024] [Revised: 03/09/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
The gut microbiota is a dynamic ecosystem that plays a pivotal role in maintaining host health. The perturbation of these microbes has been linked to several health conditions. Hence, they have emerged as promising targets for understanding and promoting good health. Despite the growing body of research on the role of sodium in health, its effects on the human gut microbiome remain under-explored. Here, using nutrition and metagenomics methods, we investigate the influence of dietary sodium intake and alterations of the human gut microbiota. We found that a high-sodium diet (HSD) altered the gut microbiota composition with a significant reduction in Bacteroides and inverse increase in Prevotella compared to a low-sodium diet (LSD). However, there is no clear distinction in the Firmicutes/Bacteroidetes (F/B) ratio between the two diet types. Metabolic pathway reconstruction revealed the presence of sodium reabsorption genes in the HSD, but not LSD. Since it is currently difficult in microbiome studies to confidently associate the F/B ratio with what is considered healthy (e.g., low sodium) or unhealthy (e.g., high sodium), we suggest that the use of a genus-based ratio such as the Bacteroides/Prevotella (B/P) ratio may be more beneficial for the application of microbiome studies in health.
Collapse
Affiliation(s)
- Omololu Fagunwa
- Institute for Global Food Security, School of Biological Sciences, Queen’s University Belfast, Belfast BT9 5DL, UK
| | - Kirsty Davies
- School of Applied Sciences, University of Huddersfield, Huddersfield HD1 3DH, UK;
| | - Jane Bradbury
- School of Medicine, Edge Hill University, Ormskirk L39 4QP, UK;
| |
Collapse
|
6
|
Mitsuno R, Uchiyama K, Nakayama T, Takahashi R, Yoshimoto N, Yamaguchi S, Washida N, Kanda T, Hayashi K, Itoh H. Comparison of the effects of angiotensin receptor-neprilysin inhibitors and thiazide diuretic/renin-angiotensin system inhibitor combination therapy in hypertensive patients: a retrospective cohort study. J Hum Hypertens 2023; 37:1049-1055. [PMID: 37488277 DOI: 10.1038/s41371-023-00851-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/08/2023] [Accepted: 07/17/2023] [Indexed: 07/26/2023]
Abstract
Angiotensin receptor-neprilysin inhibitors (ARNIs) have been approved as antihypertensive agents in Japan, and thiazide diuretics (TZDs) are widely used concomitantly with renin-angiotensin system inhibitors (RASIs) for hypertension. This retrospective study included patients with hypertension who switched from RASI to ARNI therapy (ARNI group) and those who were prescribed TZDs with RASIs (TZD/RASI group). Drug-related changes in the estimated glomerular filtration rate (eGFR), blood pressure (BP), body weight (BW), serum electrolytes, uric acid (UA), and triglyceride levels were compared between the two groups. Overall, 70 participants (31 and 39 in the ARNI and TZD/RASI groups, respectively) were enrolled and observed for a median of 2 months. According to linear mixed models, compared with the TZD/RASI group, the ARNI group exhibited a significant change in mean eGFR of 3.71 mL/min/1.73 m2 [95% confidence interval (CI), 0.57-6.84; P = 0.02] from the time of switching drug to the next outpatient visit. Further, compared with the TZD/RASI group, the ARNI group exhibited significant changes in mean serum UA (-1.27; 95% CI, -1.66 to -0.88), sodium (1.22; 95% CI, 0.12 to -2.32), chloride (2.14; 95% CI, 0.75-3.52), and triglyceride (-52.1; 95% CI, -100.9 to -3.29) levels. Conversely, serum potassium levels, BW, and systolic and diastolic BP did not differ significantly between the two groups (P = 0.69, 0.44, 0.49, and 0.66, respectively). Compared with the combination therapy of TZD and RASI, ARNI therapy causes less renal dysfunction, hyperuricemia, and hypertriglyceridemia with fewer electrolyte abnormalities and no significant difference in antihypertensive effects.
Collapse
Affiliation(s)
- Ryunosuke Mitsuno
- Department of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kiyotaka Uchiyama
- Department of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
- Department of Nephrology, International University of Health and Welfare Narita Hospital, 852 Hatakeda, Narita, Chiba, 286-8520, Japan.
| | - Takashin Nakayama
- Department of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Rina Takahashi
- Department of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Norifumi Yoshimoto
- Department of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shintaro Yamaguchi
- Department of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Naoki Washida
- Department of Nephrology, International University of Health and Welfare Narita Hospital, 852 Hatakeda, Narita, Chiba, 286-8520, Japan
| | - Takeshi Kanda
- Department of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kaori Hayashi
- Department of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroshi Itoh
- Department of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
7
|
Barreto J, Campos-Staffico AM, Nadruz W, Quinaglia T, Sposito AC. The role of SGLT2i in attenuating residual cardiovascular risk through blood pressure-lowering: mechanistic insights and perspectives. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2023; 4:1243530. [PMID: 37822556 PMCID: PMC10562622 DOI: 10.3389/fcdhc.2023.1243530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/11/2023] [Indexed: 10/13/2023]
Abstract
Sodium glucose cotransporter 2 inhibitors (SGLT2) have been increasingly pursued as a promising target for addressing residual cardiovascular risk. Prior trials demonstrated that SGLT2i not only promotes glucose-lowering, but also improves endothelial dysfunction, adiposity, fluid overload, and insulin sensitivity thus contributing to hemodynamic changes implicated in its cardiorenal benefits. The mechanisms in the effect of SGLT2i on blood pressure and their potential role in preventing cardiovascular events are hereby revised.
Collapse
Affiliation(s)
- Joaquim Barreto
- Laboratory of Atherosclerosis and Vascular Biology, University of Campinas (Unicamp), Campinas, Sao Paulo, Brazil
| | | | - Wilson Nadruz
- Cardiology Division, Clinics Hospital, Unicamp, Campinas, Sao Paulo, Brazil
| | - Thiago Quinaglia
- Massachussets General Hospital, Harvard University, Boston, MA, United States
| | - Andrei C. Sposito
- Laboratory of Atherosclerosis and Vascular Biology, University of Campinas (Unicamp), Campinas, Sao Paulo, Brazil
- Cardiology Division, Clinics Hospital, Unicamp, Campinas, Sao Paulo, Brazil
| |
Collapse
|
8
|
Simon Y, Jacques D, Bkaily G. High salt-induced morphological and glycocalyx remodeling of human vascular smooth muscle cells is reversible but induces a high sodium salt-like sensitive memory. Can J Physiol Pharmacol 2023; 101:437-446. [PMID: 37290125 DOI: 10.1139/cjpp-2023-0087] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Our recent work showed that short-term treatment (1-2 days) with high sodium salt had no effect on the morphology of human vascular smooth muscle cells (hVSMCs). However, chronic (long-term treatment, 6-16 days) high sodium salt (CHSS) induced hypertrophy and decreased the relative density of the glycocalyx in hVSMCs. Whether this CHSS effect is reversible at both the morphological and the intracellular calcium and sodium levels is unknown. In the present study, we tested the hypothesis that the effect of CHSS on the morphological and functional levels of hVSMCs is reversible. However, it induced an irreversible increase in the sensitivity of the cells following short-term treatment with high extracellular Na+. We tested the effects of the removal of CHSS treatment on the morphology and intracellular sodium and calcium of hVSMCs. Our results showed that restoring average sodium concentration (145 mM) modeled back the relative density of the glycocalyx, the intracellular resting calcium and sodium levels, and the whole cell and nuclear volumes of hVSMCs. In addition, it induced a permanent remodeling of hVSMCs' response to a short-term increase in the extracellular level of sodium salt by developing spontaneous cytosolic and nuclear calcium waves. Our results showed that CHSS is reversible at both the morphological and basal intracellular ionic levels. However, it maintained a high sensitivity to short-term elevation of extracellular sodium. These results suggest that even if chronic high salt is corrected, it induces a high sodium salt-like sensitive memory.
Collapse
Affiliation(s)
- Yanick Simon
- Department of immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Danielle Jacques
- Department of immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Ghassan Bkaily
- Department of immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| |
Collapse
|
9
|
Jung CY, Kim Y, Kim HW, Han SH, Yoo TH, Kang SW, Park JT. Effectiveness of a Smartphone Application for Dietary Sodium Intake Measurement. Nutrients 2023; 15:3590. [PMID: 37630780 PMCID: PMC10459655 DOI: 10.3390/nu15163590] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Accurate estimation of sodium intake is a key requirement for evaluating the efficacy of interventional strategies to reduce salt intake. The effectiveness of a smartphone application in measuring dietary sodium intake was assessed. This study included 46 participants who consented to register in Noom's food-logging program. All participants were followed up for six months from the day of enrollment. The mean age of the participants was 40.2 ± 12.3 years, and 22 (48%) participants were male. The average number of times/weeks the meals were logged was 16.2 ± 10.3. At baseline, the mean 24-h urine sodium was 124.3 mmol/24 h. The mean sodium intake measured by the smartphone application and calculated using the 24-h urine sodium was 2020.9 mg/24 h and 2857.6 mg/24 h, respectively. During the second visit, the mean 24-h urine sodium was 117.4 mmol/24 h. The mean sodium intake measured by the smartphone application and calculated using the 24-h urine sodium was 1456.0 mg/24 h and 2698.3 mg/24 h, respectively. Sodium intake measured using the smartphone application positively correlated with that calculated using the 24-h urine sodium at baseline (r = 0.464; p < 0.001) and follow-up (r = 0.334; p= 0.023). Dietary sodium intake measured using a smartphone application correlated well with that estimated using 24-h urine sodium level.
Collapse
Affiliation(s)
- Chan-Young Jung
- Department of Internal Medicine, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea; (C.-Y.J.)
- Division of Nephrology, Department of Internal Medicine, Asan Medical Center, Seoul 05505, Republic of Korea
| | | | - Hyung Woo Kim
- Department of Internal Medicine, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea; (C.-Y.J.)
| | - Seung Hyeok Han
- Department of Internal Medicine, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea; (C.-Y.J.)
- Institute of Kidney Disease Research, Yonsei University, Seoul 03722, Republic of Korea
| | - Tae-Hyun Yoo
- Department of Internal Medicine, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea; (C.-Y.J.)
- Institute of Kidney Disease Research, Yonsei University, Seoul 03722, Republic of Korea
| | - Shin-Wook Kang
- Department of Internal Medicine, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea; (C.-Y.J.)
- Institute of Kidney Disease Research, Yonsei University, Seoul 03722, Republic of Korea
| | - Jung Tak Park
- Department of Internal Medicine, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea; (C.-Y.J.)
- Institute of Kidney Disease Research, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
10
|
Shimada S, Hoffmann BR, Yang C, Kurth T, Greene AS, Liang M, Dash RK, Cowley AW. Metabolic Responses of Normal Rat Kidneys to a High Salt Intake. FUNCTION 2023; 4:zqad031. [PMID: 37575482 PMCID: PMC10413938 DOI: 10.1093/function/zqad031] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/06/2023] [Accepted: 06/12/2023] [Indexed: 08/15/2023] Open
Abstract
In this study, novel methods were developed, which allowed continuous (24/7) measurement of arterial blood pressure and renal blood flow in freely moving rats and the intermittent collection of arterial and renal venous blood to estimate kidney metabolic fluxes of O2 and metabolites. Specifically, the study determined the effects of a high salt (HS; 4.0% NaCl) diet upon whole kidney O2 consumption and arterial and renal venous plasma metabolomic profiles of normal Sprague-Dawley rats. A separate group of rats was studied to determine changes in the cortex and outer medulla tissue metabolomic and mRNAseq profiles before and following the switch from a 0.4% to 4.0% NaCl diet. In addition, targeted mRNA expression analysis of cortical segments was performed. Significant changes in the metabolomic and transcriptomic profiles occurred with feeding of the HS diet. A progressive increase of kidney O2 consumption was found despite a reduction in expression of most of the mRNA encoding enzymes of TCA cycle. A novel finding was the increased expression of glycolysis-related genes in Cx and isolated proximal tubular segments in response to an HS diet, consistent with increased release of pyruvate and lactate from the kidney to the renal venous blood. Data suggests that aerobic glycolysis (eg, Warburg effect) may contribute to energy production under these circumstances. The study provides evidence that kidney metabolism responds to an HS diet enabling enhanced energy production while protecting from oxidative stress and injury. Metabolomic and transcriptomic analysis of kidneys of Sprague-Dawley rats fed a high salt diet.
Collapse
Affiliation(s)
- Satoshi Shimada
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Brian R Hoffmann
- Mass Spectrometry and Protein Chemistry, Protein Sciences, The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Chun Yang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Theresa Kurth
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Andrew S Greene
- Mass Spectrometry and Protein Chemistry, Protein Sciences, The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Mingyu Liang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ranjan K Dash
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI 53226, USA
| | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
11
|
Shimada S, Hoffmann BR, Yang C, Kurth T, Greene AS, Liang M, Dash RK, Cowley AW. Metabolic responses of normal rat kidneys to a high salt intake. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.18.524636. [PMID: 36711564 PMCID: PMC9882299 DOI: 10.1101/2023.01.18.524636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In the present study, novel methods were developed which allowed continuous (24/7) measurement of blood pressure (BP) and renal blood flow (RBF) in freely moving rats and the intermittent collection of arterial and renal venous blood to estimate kidney metabolic fluxes of O 2 and metabolites. The study determined the effects of a high salt (HS) diet upon whole kidney O 2 consumption and the metabolomic profiles of normal Sprague Dawley (SD) rats. A separate group of rats was studied to determine changes in the cortex (Cx) and outer medulla (OM) tissue metabolomic and mRNAseq profiles before and following the switch from a 0.4% to a 4.0% NaCl diet. Significant changes in the metabolomic and transcriptomic profiles occurred with feeding of the HS diet. A progressive increase of kidney O 2 consumption was found despite a reduction in expression of most of the mRNA encoding enzymes of TCA cycle. Increased glycolysis was evident with the elevation of mRNA expression encoding key glycolytic enzymes and release of pyruvate and lactate from the kidney in the renal venous blood. Glycolytic production of NADH is used in either the production of lactate or oxidized via the malate aspartate shuttle. Aerobic glycolysis (e.g., Warburg-effect) may account for the needed increase in cellular energy. The study provides evidence that kidney metabolism responds to a HS diet enabling enhanced energy production while protecting from oxidate stress and injury.
Collapse
|
12
|
Ashraf UM, Atari E, Alasmari F, Waghulde H, Kumar V, Sari Y, Najjar SM, Jose PA, Kumarasamy S. Intrarenal Dopaminergic System Is Dysregulated in SS- Resp18mutant Rats. Biomedicines 2023; 11:111. [PMID: 36672619 PMCID: PMC9855394 DOI: 10.3390/biomedicines11010111] [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] [Received: 12/04/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
The genetic and molecular basis of developing high blood pressure and renal disease are not well known. Resp18mutant Dahl salt-sensitive (SS-Resp18mutant) rats fed a 2% NaCl diet for six weeks have high blood pressure, increased renal fibrosis, and decreased mean survival time. Impairment of the dopaminergic system also leads to hypertension that involves renal and non-renal mechanisms. Deletion of any of the five dopamine receptors may lead to salt-sensitive hypertension. Therefore, we investigated the interaction between Resp18 and renal dopamine in SS-Resp18mutant and Dahl salt-sensitive (SS) rats. We found that SS-Resp18mutant rats had vascular dysfunction, as evidenced by a decrease in vasorelaxation in response to sodium nitroprusside. The pressure-natriuresis curve in SS-Resp18mutant rats was shifted down and to the right of SS rats. SS-Resp18mutant rats had decreased glomerular filtration rate and dopamine receptor subtypes, D1R and D5R. Renal dopamine levels were decreased, but urinary dopamine levels were increased, which may be the consequence of increased renal dopamine production, followed by secretion into the tubular lumen. The increased renal dopamine production in SS-Resp18mutant rats in vivo was substantiated by the increased dopamine production in renal proximal tubule cells treated with L-DOPA. Overall, our study provides evidence that targeted disruption of the Resp18 locus in the SS rat dysregulates the renal dopaminergic system.
Collapse
Affiliation(s)
- Usman M. Ashraf
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Ealla Atari
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Fawaz Alasmari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo College of Pharmacy & Pharmaceutical Sciences, Toledo, OH 43614, USA
| | - Harshal Waghulde
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Vikash Kumar
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo College of Pharmacy & Pharmaceutical Sciences, Toledo, OH 43614, USA
| | - Sonia M. Najjar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Pedro A. Jose
- Department of Medicine, Division of Kidney Diseases & Hypertension, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA
- Department of Pharmacology and Physiology, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA
| | - Sivarajan Kumarasamy
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
13
|
Fu Z, Zheng H, Kaewsaro K, Lambert J, Chen Y, Yang T. Mutagenesis of the cleavage site of (pro)renin receptor abrogates aldosterone-salt-induced hypertension and renal injury in mice. Am J Physiol Renal Physiol 2023; 324:F1-F11. [PMID: 36302140 PMCID: PMC9762973 DOI: 10.1152/ajprenal.00088.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 09/29/2022] [Accepted: 10/17/2022] [Indexed: 02/04/2023] Open
Abstract
Soluble (pro)renin receptor (sPRR), the extracellular domain of (pro)renin receptor (PRR), is primarily generated by site-1 protease and furin. It has been reported that sPRR functions as an important regulator of intrarenal renin contributing to angiotensin II (ANG II)-induced hypertension. Relatively, less is known for the function of sPRR in ANG II-independent hypertension such as mineralocorticoid excess. In the present study, we used a novel mouse model with mutagenesis of the cleavage site in PRR (termed as PRRR279V/L282V or mutant) to examine the phenotype during aldosterone (Aldo)-salt treatment. The hypertensive response of mutant mice to Aldo-salt treatment was blunted in parallel with the attenuated response of plasma volume expansion and renal medullary α-epithelial Na+ channel expression. Moreover, Aldo-salt-induced hypertrophy in the heart and kidney as well as proteinuria were improved, accompanied by blunted polydipsia and polyuria. Together, these results represent strong evidence favoring endogenous sPRR as a mediator of Aldo-salt-induced hypertension and renal injury.NEW & NOTEWORTHY We used a novel mouse model with mutagenesis of the cleavage site of PRR to support soluble PRR as an essential mediator of aldosterone-salt-induced hypertension and also as a potential therapeutic target for patients with mineralocorticoid excess. We firstly report that soluble PRR-dependent pathway medicates the Na+-retaining action of aldosterone in the distal nephron, which opens up a new area for a better understanding of the molecular basis of renal handling of Na+ balance and blood pressure.
Collapse
Affiliation(s)
- Ziwei Fu
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah
| | - Huaqing Zheng
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah
- Renal Section, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah
| | - Kannaree Kaewsaro
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah
| | - Jacob Lambert
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah
| | - Yanting Chen
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah
| | - Tianxin Yang
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah
- Renal Section, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah
| |
Collapse
|
14
|
Ou H, Liu D, Zhao G, Gong C, Li Y, Zhao Q. Association between AT1 receptor gene polymorphism and left ventricular hypertrophy and arterial stiffness in essential hypertension patients: a prospective cohort study. BMC Cardiovasc Disord 2022; 22:571. [PMID: 36577936 PMCID: PMC9795750 DOI: 10.1186/s12872-022-03024-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AT1 receptor gene (AGTR1) is related to essential hypertension (EH), and left ventricular hypertrophy (LVH) and arterial stiffness are common complications of EH. This study aimed to explore the association between AGTR1 genotype and LVH and arterial stiffness in EH patients. METHODS A total of 179 EH patients were recruited in this study. Oral exfoliated cells were collected from each patient, and the genetic polymorphism of AGTR1(rs4524238) was assessed using a gene sequencing platform. The outcomes were LVH and arterial stiffness. RESULTS Among 179 patients, 114 were with AGTR1 genotype of GG (57 males, aged 59.54 ± 13.49 years) and 65 were with AGTR1 genotype of GA or AA (36 males, aged 61.28 ± 12.79 years). Patients with AGTR1 genotype of GG were more likely to have LVH (47 [41.23%] vs. 14 [21.54%], P = 0.006) and arterial stiffness (30 [26.32%] vs. 8 [12.31%], P = 0.036). The AGTR1 polymorphism frequency was in accordance with Hardy-Weinberg equilibrium (P = 0.291). The multivariate logistic regression showed that AGTR1 genotype of GA or AA was independently associated with lower risk of LVH (OR = 0.344, 95%CI 160~0.696, P = 0.003) and arterial stiffness (OR = 0.371, 95%CI 0.155~0.885, P = 0.025) after adjusting for gender, age, and diabetes. CONCLUSION EH patients with the AGTR1 genotype of GA or AA were at lower risk for LVH and arterial stiffness than those with the GG genotype.
Collapse
Affiliation(s)
- Hangjun Ou
- grid.452244.1Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou China
| | - Danan Liu
- grid.452244.1Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou China
| | - Guangjian Zhao
- grid.452244.1Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou China
| | - Caiwei Gong
- grid.452244.1Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou China
| | - Yunyun Li
- grid.452244.1Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou China
| | - Quanwei Zhao
- grid.452244.1Department of Cardiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou China
| |
Collapse
|
15
|
Maaliki D, Itani MM, Itani HA. Pathophysiology and genetics of salt-sensitive hypertension. Front Physiol 2022; 13:1001434. [PMID: 36176775 PMCID: PMC9513236 DOI: 10.3389/fphys.2022.1001434] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Most hypertensive cases are primary and heavily associated with modifiable risk factors like salt intake. Evidence suggests that even small reductions in salt consumption reduce blood pressure in all age groups. In that regard, the ACC/AHA described a distinct set of individuals who exhibit salt-sensitivity, regardless of their hypertensive status. Data has shown that salt-sensitivity is an independent risk factor for cardiovascular events and mortality. However, despite extensive research, the pathogenesis of salt-sensitive hypertension is still unclear and tremendously challenged by its multifactorial etiology, complicated genetic influences, and the unavailability of a diagnostic tool. So far, the important roles of the renin-angiotensin-aldosterone system, sympathetic nervous system, and immune system in the pathogenesis of salt-sensitive hypertension have been studied. In the first part of this review, we focus on how the systems mentioned above are aberrantly regulated in salt-sensitive hypertension. We follow this with an emphasis on genetic variants in those systems that are associated with and/or increase predisposition to salt-sensitivity in humans.
Collapse
Affiliation(s)
- Dina Maaliki
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Maha M. Itani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hana A. Itani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
16
|
Vesnina A, Prosekov A, Atuchin V, Minina V, Ponasenko A. Tackling Atherosclerosis via Selected Nutrition. Int J Mol Sci 2022; 23:8233. [PMID: 35897799 PMCID: PMC9368664 DOI: 10.3390/ijms23158233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 12/02/2022] Open
Abstract
The development and pathogenesis of atherosclerosis are significantly influenced by lifestyle, particularly nutrition. The modern level of science and technology development promote personalized nutrition as an efficient preventive measure against atherosclerosis. In this survey, the factors were revealed that contribute to the formation of an individual approach to nutrition: genetic characteristics, the state of the microbiota of the gastrointestinal tract (GIT) and environmental factors (diets, bioactive components, cardioprotectors, etc.). In the course of the work, it was found that in order to analyze the predisposition to atherosclerosis associated with nutrition, genetic features affecting the metabolism of nutrients are significant. The genetic features include the presence of single nucleotide polymorphisms (SNP) of genes and epigenetic factors. The influence of telomere length on the pathogenesis of atherosclerosis and circadian rhythms was also considered. Relatively new is the study of the relationship between chrono-nutrition and the development of metabolic diseases. That is, to obtain the relationship between nutrition and atherosclerosis, a large number of genetic markers should be considered. In this relation, the question arises: "How many genetic features need to be analyzed in order to form a personalized diet for the consumer?" Basically, companies engaged in nutrigenetic research and choosing a diet for the prevention of a number of metabolic diseases use SNP analysis of genes that accounts for lipid metabolism, vitamins, the body's antioxidant defense system, taste characteristics, etc. There is no set number of genetic markers. The main diets effective against the development of atherosclerosis were considered, and the most popular were the ketogenic, Mediterranean, and DASH-diets. The advantage of these diets is the content of foods with a low amount of carbohydrates, a high amount of vegetables, fruits and berries, as well as foods rich in antioxidants. However, due to the restrictions associated with climatic, geographical, material features, these diets are not available for a number of consumers. The way out is the use of functional products, dietary supplements. In this approach, the promising biologically active substances (BAS) that exhibit anti-atherosclerotic potential are: baicalin, resveratrol, curcumin, quercetin and other plant metabolites. Among the substances, those of animal origin are popular: squalene, coenzyme Q10, omega-3. For the prevention of atherosclerosis through personalized nutrition, it is necessary to analyze the genetic characteristics (SNP) associated with the metabolism of nutrients, to assess the state of the microbiota of the GIT. Based on the data obtained and food preferences, as well as the individual capabilities of the consumer, the optimal diet can be selected. It is topical to exclude nutrients of which their excess consumption stimulates the occurrence and pathogenesis of atherosclerosis and to enrich the diet with functional foods (FF), BAS containing the necessary anti-atherosclerotic, and stimulating microbiota of the GIT nutrients. Personalized nutrition is a topical preventive measure and there are a number of problems hindering the active use of this approach among consumers. The key factors include weak evidence of the influence of a number of genetic features, the high cost of the approach, and difficulties in the interpretation of the results. Eliminating these deficiencies will contribute to the maintenance of a healthy state of the population through nutrition.
Collapse
Affiliation(s)
- Anna Vesnina
- Laboratory of Natural Nutraceuticals Biotesting, Research Department, Kemerovo State University, 650043 Kemerovo, Russia;
| | - Alexander Prosekov
- Laboratory of Biocatalysis, Kemerovo State University, 650043 Kemerovo, Russia;
| | - Victor Atuchin
- Laboratory of Optical Materials and Structures, Institute of Semiconductor Physics, 630090 Novosibirsk, Russia
- Research and Development Department, Kemerovo State University, 650000 Kemerovo, Russia
- Laboratory of Applied Physics, Novosibirsk State University, 630090 Novosibirsk, Russia
- Department of Industrial Machinery Design, Novosibirsk State Technical University, 630073 Novosibirsk, Russia
- R&D Center “Advanced Electronic Technologies”, Tomsk State University, 634034 Tomsk, Russia
| | - Varvara Minina
- Department of Genetic and Fundamental Medicine, Kemerovo State University, 650000 Kemerovo, Russia;
| | - Anastasia Ponasenko
- Laboratory of Genome Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 650002 Kemerovo, Russia;
| |
Collapse
|
17
|
Association of Circulating Biomarkers of lnc-IGSF3-1:1, SCOC-AS1, and SLC8A1-AS1 with Salt Sensitivity of Blood Pressure in Chinese Population. J Cardiovasc Transl Res 2021; 15:906-917. [PMID: 34855149 DOI: 10.1007/s12265-021-10190-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/16/2021] [Indexed: 01/24/2023]
Abstract
Accumulating evidence suggested that long non-coding RNAs (lncRNAs) could play biological roles in cardiovascular diseases. We investigated whether lncRNAs can serve as biomarkers for salt sensitivity of blood pressure (SSBP). Participants were divided into salt-sensitive (SS) and salt-resistant (SR) ones by oral saline test. LncRNAs were tested by microarray (N = 20) and two-stage qRT-PCR (N = 89 and 228). We identified five differently expressed lncRNAs (lnc-IGSF3-1:1, SCOC-AS1, SLC8A1-AS1, KCNQ1OT1, and lnc-GNG-10-3:1) between SS and SR. In single-lncRNA analyses, lnc-IGSF3-1:1 displayed better diagnostic performance in hypertensive patients (AUC = 0.840), while SCOC-AS1 in normotensive (AUC = 0.810). In multi-lncRNA analyses, lnc-IGSF3-1:1 + SCOC-AS1 + SLC8A1-AS1 combination showed the best diagnostic performance in hypertensive (AUC = 0.853) and normotensive groups (AUC = 0.873). We constructed a lncRNA-mRNA-GO-KEGG-disease network by bioinformatic analysis; lnc-IGSF3-1:1 and SLC8A1-AS1 were identified as hub biomarkers. Our findings suggest that lnc-IGSF3-1:1, SCOC-AS1, and SLC8A1-AS1 may represent as genetic susceptible biomarkers for SSBP, and had different SS diagnostic performance in hypertensive patients and normotensive individuals.
Collapse
|
18
|
Kolahdouz-Mohammadi R, Soltani S, Clayton ZS, Salehi-Abargouei A. Sodium status is associated with type 2 diabetes mellitus: a systematic review and meta-analysis of observational studies. Eur J Nutr 2021; 60:3543-3565. [PMID: 34052916 DOI: 10.1007/s00394-021-02595-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 05/21/2021] [Indexed: 11/30/2022]
Abstract
PURPOSE The relationship between sodium intake and the risk of developing type 2 diabetes mellitus (T2DM) is inconsistent. We, therefore, aimed to summarize the current evidence by conducting a systematic review and meta-analysis of observational studies. METHODS We retrieved studies which compared any marker of sodium status between individuals with T2DM and those without diabetes published in any language by searching online databases from inception up to June 2019. Summary effects were derived using random-effects model. RESULTS A total of 44 studies with 503,830 participants from 25 countries were included in this study. Sodium status was significantly different between individuals with and without T2DM (Hedges' g = 0.21; 95% CI 0.02, 0.40; P = 0.029). Individuals with T2DM had higher sodium intake compared to non-diabetic controls (WMD = 621.79 mg/day; 95% CI 321.53, 922.06; P < 0.001) and 24-h urinary excretion was associated with likelihood of developing T2DM (OR = 1.27, 95% CI 1.15, 1.41; P < 0.001). Furthermore, salivary, hair, and platelet sodium were higher in patients with T2DM compared to controls (P < 0.05). CONCLUSION The findings of the current meta-analysis suggest that sodium levels are higher in patients with T2DM compared to non-diabetic controls; however, given that these studies are observational, it is not possible to infer causality.
Collapse
Affiliation(s)
- Roya Kolahdouz-Mohammadi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Sepideh Soltani
- Yazd Cardiovascular Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Amin Salehi-Abargouei
- Nutrition and Food Security Research Center, Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
19
|
Association between physiological stress load and diet quality patterns differs between male and female adults. Physiol Behav 2021; 240:113538. [PMID: 34314759 DOI: 10.1016/j.physbeh.2021.113538] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 10/20/2022]
Abstract
A promising, yet relatively unexplored factor that may influence a person's stress response, is diet. Diet can affect the physiological response to stress, but relationships between diet quality and the chronic stress marker allostatic load (AL) are insufficiently studied. Furthermore, sex, age, and BMI may interact with diet quality to influence AL. 358 adults were recruited across predetermined sex, age, and BMI ranges. Cluster analysis of 13 Healthy Eating Index (HEI) sub-scores across all participants revealed six distinct diet quality patterns (HEI-P). We found sex and HEI-P interacted (PHEIxSex = 0.0232) to affect AL, reflecting a significantly different AL between women and men consuming a diet more closely aligned with the Dietary Guidelines for Americans for dairy, refined grains, and sodium consumption, but less aligned for added sugar, saturated fat, and fruits/vegetables intake. Sex and HEI-P also interacted to affect cholesterol (PHEIxSex = 0.0157), norepinephrine (PHEIxSex = 0.0315), epinephrine (PHEIxSex = 0.0204), and systolic blood pressure (PHEIxSex = 0.0457) but, compared to total allostatic load, no individual component of this biomarker explained the entire array of sex by HEI-P interactions. Our results suggest that differences in HEI-P and sex interact to influence physiological stress load which, in turn, may help resolve discrepancies in diet and sex-related disease risk.
Collapse
|
20
|
Allu AS, Tiriveedhi V. Cancer Salt Nostalgia. Cells 2021; 10:cells10061285. [PMID: 34064273 PMCID: PMC8224381 DOI: 10.3390/cells10061285] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022] Open
Abstract
High-salt (sodium chloride) diets have been strongly associated with disease states and poor health outcomes. Traditionally, the impact of salt intake is primarily studied in cardiovascular diseases, hypertension and renal diseases; however, recently there has been increasing evidence demonstrating the role of salt in autoimmune diseases. Salt has been shown to modulate the inflammatory activation of immune cells leading to chronic inflammation-related ailments. To date, there is minimal evidence showing a direct correlation of salt with cancer incidence and/or cancer-related adverse clinical outcomes. In this review article, we will discuss the recent understanding of the molecular role of salt, and elucidate the apparent double-edged sword nature of the relationship between salt and cancer progression.
Collapse
Affiliation(s)
- Aashish S. Allu
- Department of Sciences, Lafayette High School, Wildwood, MO 63011, USA;
| | - Venkataswarup Tiriveedhi
- Department of Biological Sciences, Tennessee State University, 3500 John A Merritt Blvd, Nashville, TN 37209, USA
- Division of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
- Correspondence: ; Tel.: +1-615-963-5779; Fax: +1-615-963-5747
| |
Collapse
|
21
|
Kim HY, Choi HS, Kim CS, Bae EH, Ma SK, Sung SA, Han SH, Oh KH, Ahn C, Kim SW. Effect of urinary angiotensinogen and high-salt diet on blood pressure in patients with chronic kidney disease: results from the Korean Cohort Study for Outcome in Patients with Chronic Kidney Disease (KNOW-CKD). Korean J Intern Med 2021; 36:659-667. [PMID: 33028070 PMCID: PMC8137406 DOI: 10.3904/kjim.2020.077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/23/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND/AIMS This study aimed to investigate whether urinary angiotensinogen (UAGT) excretion was associated with elevated blood pressure in patients with chronic kidney disease (CKD) and to evaluate the relationship among blood pressure, intra-renal renin-angiotensin system (RAS) activity, and dietary sodium in patients with CKD. METHODS Participants from the Korean Cohort Study for Outcome in Patients with Chronic Kidney Disease (KNOW-CKD) were included. Of the total cohort of 2,238 individuals with CKD, we included 1,955 participants who underwent complete 24-hour urinary sodium (24-hour UNa) analysis. They were categorized into three groups according to three tertiles of their 24-hour UNa, reflecting daily salt intake. To measure intra-renal RAS activity, the UAGT excretion was assayed with an enzyme-linked immunosorbent assay. RESULTS Elevated 24-hour UNa levels, logarithm of UAGT-to-creatinine ratio (UAGT/Cr), increased waist-to-hip ratio, and decreased estimated glomerular filtration rate were the risk factors for increased systolic blood pressure. Systolic blood pressure showed a positive correlation with 24-hour UNa levels and logarithm of UAGT/Cr. CONCLUSION UAGT and urinary sodium excretion are independent determinants of systolic blood pressure in patients with CKD. These findings suggest that increased systolic blood pressure in CKD patients is associated with both increased dietary sodium levels and intra-renal RAS activity. The risk of elevated systolic blood pressure in the 3rd tertile of both the UAGT/Cr and 24-hour UNa groups was about 2.3 times higher than that in the reference group.
Collapse
Affiliation(s)
- Ha Yeon Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Hong Sang Choi
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Chang Seong Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Eun Hui Bae
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Seong Kwon Ma
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Su-Ah Sung
- Department of Internal Medicine, Nowon Eulji Medical Center, Eulji University, Seoul, Korea
| | - Seung Hyeok Han
- Department of Internal Medicine and Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Curie Ahn
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Soo Wan Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
- Correspondence to Soo Wan Kim, M.D. Department of Internal Medicine, Chonnam National University Medical School, 42 Jebong-ro, Dong-gu, Gwangju 61469, Korea Tel: +82-62-220-6271, Fax: +82-62-225-8578 E-mail:
| |
Collapse
|
22
|
Łabno-Kirszniok K, Kujawa-Szewieczek A, Wiecek A, Piecha G. The Effects of Short-Term Changes in Sodium Intake on Plasma Marinobufagenin Levels in Patients with Primary Salt-Sensitive and Salt-Insensitive Hypertension. Nutrients 2021; 13:nu13051502. [PMID: 33946894 PMCID: PMC8147121 DOI: 10.3390/nu13051502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 11/25/2022] Open
Abstract
Increased marinobufagenin (MBG) synthesis has been suggested in response to high dietary salt intake. The aim of this study was to determine the effects of short-term changes in sodium intake on plasma MBG levels in patients with primary salt-sensitive and salt-insensitive hypertension. In total, 51 patients with primary hypertension were evaluated during acute sodium restriction and sodium loading. Plasma or serum concentrations of MBG, natriuretic pro-peptides, aldosterone, sodium, potassium, as well as hematocrit (Hct) value, plasma renin activity (PRA) and urinary sodium and potassium excretion were measured. Ambulatory blood pressure monitoring (ABPM) and echocardiography were performed at baseline. In salt-sensitive patients with primary hypertension plasma MBG correlated positively with diastolic blood pressure (ABPM) and serum NT-proANP concentration at baseline and with serum NT-proANP concentration after dietary sodium restriction. In this subgroup plasma MBG concentration decreased during sodium restriction, and a parallel increase of PRA was observed. Acute salt loading further decreased plasma MBG concentration in salt-sensitive subjects in contrast to salt insensitive patients. No correlation was found between plasma MBG concentration and left ventricular mass index. In conclusion, in salt-sensitive hypertensive patients plasma MBG concentration correlates with 24-h diastolic blood pressure and dietary sodium restriction reduces plasma MBG levels. Decreased MBG secretion in response to acute salt loading may play an important role in the pathogenesis of salt sensitivity.
Collapse
Affiliation(s)
| | | | | | - Grzegorz Piecha
- Correspondence: ; Tel.: +48-322-591-429; Fax: +48-322-553-726
| |
Collapse
|
23
|
The Role of the Renal Dopaminergic System and Oxidative Stress in the Pathogenesis of Hypertension. Biomedicines 2021; 9:biomedicines9020139. [PMID: 33535566 PMCID: PMC7912729 DOI: 10.3390/biomedicines9020139] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 01/11/2023] Open
Abstract
The kidney is critical in the long-term regulation of blood pressure. Oxidative stress is one of the many factors that is accountable for the development of hypertension. The five dopamine receptor subtypes (D1R–D5R) have important roles in the regulation of blood pressure through several mechanisms, such as inhibition of oxidative stress. Dopamine receptors, including those expressed in the kidney, reduce oxidative stress by inhibiting the expression or action of receptors that increase oxidative stress. In addition, dopamine receptors stimulate the expression or action of receptors that decrease oxidative stress. This article examines the importance and relationship between the renal dopaminergic system and oxidative stress in the regulation of renal sodium handling and blood pressure. It discusses the current information on renal dopamine receptor-mediated antioxidative network, which includes the production of reactive oxygen species and abnormalities of renal dopamine receptors. Recognizing the mechanisms by which renal dopamine receptors regulate oxidative stress and their degree of influence on the pathogenesis of hypertension would further advance the understanding of the pathophysiology of hypertension.
Collapse
|
24
|
Wang F, Sun Y, Luo R, Lu X, Yang B, Yang T. COX-2-independent activation of renal (pro)renin receptor contributes to DOCA-salt hypertension in rats. Am J Physiol Renal Physiol 2020; 319:F647-F653. [PMID: 32799674 PMCID: PMC7642891 DOI: 10.1152/ajprenal.00112.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 12/21/2022] Open
Abstract
It has been shown that cyclooxygenase (COX)-2-dependent activation of renal (pro)renin receptor (PRR) contributes to angiotensin II (ANG II)-induced hypertension. However, less is known about the involvement of this mechanism in ANG II-independent hypertension. The goal of the present study was to test whether or not COX-2-dependent upregulation of PRR serves as a universal mechanism contributing to ANG II-dependent and -independent hypertension. Here, we examined the association between renal COX-2 and PRR during deoxycorticosterone acetate (DOCA)-salt hypertension in rats. By immunoblot analysis and immunofluorescence, renal protein expression of PRR was remarkably upregulated by DOCA-salt treatment. Surprisingly, this upregulation of renal PRR expression was unaffected by a COX-2 inhibitor, celecoxib. To address the role of renal PRR to the pathogenesis of DOCA-salt hypertension, a decoy PRR inhibitor, PRO20, was infused to the renal medulla of uninephrectomized Sprague-Dawley rats for 14 days. Radiotelemetry demonstrated effective attenuation of DOCA-salt hypertension by intramedullary infusion of a PRR inhibitor, PRO20. In parallel, DOCA-salt-induced hypertrophy in the heart and kidney as well as proteinuria were improved, accompanied with blunted polydipsia and polyuria. In contrast, intravenous infusion of PRO20 was less effective in attenuating DOCA-salt hypertension and cardiorenal injury. Together, these results suggest that COX-2-independent activation of renal PRR contributes to DOCA-salt hypertension.
Collapse
Affiliation(s)
- Fei Wang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Ying Sun
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Renfei Luo
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Xiaohan Lu
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Baoxue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| |
Collapse
|
25
|
Saber-Ayad M, Hammoudeh S, Radwan H, Manzoor S, Jabbar H, Wardeh R, Ashraf A, Habib P, Alsamman AM, Hamoudi R. The FGF-21 genetic variants rs838133 and rs838145 are associated with high salt intake in the Emirati population. J Adv Res 2020; 24:485-494. [PMID: 32566284 PMCID: PMC7296188 DOI: 10.1016/j.jare.2020.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/21/2020] [Accepted: 05/17/2020] [Indexed: 12/17/2022] Open
Abstract
Food predilection is linked to variants in the hepatokine "Fibroblast Growth Factor-21" gene (FGF21); with rs838133 linked to the sweet tooth in Caucasians. The effect of FGF21 variants on food intake is still unclear in other populations. A cohort of 196 healthy Emirati subjects was investigated [age: 30.34 ± 9.75yrs (44.4% males)]. The FGF21 rs838133 and rs838145 were genotyped. The daily intake was calculated based on a 61-item food frequency questionnaire. Multivariate analysis was performed using in house R script that implements two-way unsupervised hierarchical clustering to detect the association of the studied single-nucleotide polymorphisms (SNPs) and related SNPs in linkage disequilibrium, using data from the 1000 genome project. Both SNPs were in Hardy-Weinberg Equilaribium (HWE). BMI positively correlated with age (p = 0.002), but not with caloric intake. Salt intake was significantly higher in subjects homozygous (A: rs838133) and (G:rs838145),(p = 0.03 and 0.01, respectively). An interaction was observed between both SNPs; significantly associated with high salt intake. Using publicly available data, both SNPs fall within a region transmitted in Iberians which has a profile closely similar to Caucasians, but far from Chinese population. In conclusion, the minor alleles of FGF21 rs838145 and rs838133 are associated with high salt intake in Emiratis and may suggest neuro-metabolic link to dietary preference across different populations.
Collapse
Affiliation(s)
- Maha Saber-Ayad
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, Cairo University, Cairo, Egypt
| | - Sarah Hammoudeh
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Hadia Radwan
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Clinical Nutrition and Dietetics Department, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Shaista Manzoor
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Hussein Jabbar
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Rahaf Wardeh
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Ahmed Ashraf
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Peter Habib
- International Center for Agricultural Research in the Dry Areas (ICARDA), Giza, Egypt
| | - Alsamman M. Alsamman
- Genome Mapping Department, Agricultural Genetic Engineering Research Institute (AGERI), Giza, Egypt
| | - Rifat Hamoudi
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
26
|
Sabir JSM, El Omri A, Banaganapalli B, Aljuaid N, Omar AMS, Altaf A, Hajrah NH, Zrelli H, Arfaoui L, Elango R, Alharbi MG, Alhebshi AM, Jansen RK, Shaik NA, Khan M. Unraveling the role of salt-sensitivity genes in obesity with integrated network biology and co-expression analysis. PLoS One 2020; 15:e0228400. [PMID: 32027667 PMCID: PMC7004317 DOI: 10.1371/journal.pone.0228400] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/14/2020] [Indexed: 02/07/2023] Open
Abstract
Obesity is a multifactorial disease caused by complex interactions between genes and dietary factors. Salt-rich diet is related to the development and progression of several chronic diseases including obesity. However, the molecular basis of how salt sensitivity genes (SSG) contribute to adiposity in obesity patients remains unexplored. In this study, we used the microarray expression data of visceral adipose tissue samples and constructed a complex protein-interaction network of salt sensitivity genes and their co-expressed genes to trace the molecular pathways connected to obesity. The Salt Sensitivity Protein Interaction Network (SSPIN) of 2691 differentially expressed genes and their 15474 interactions has shown that adipose tissues are enriched with the expression of 23 SSGs, 16 hubs and 84 bottlenecks (p = 2.52 x 10-16) involved in diverse molecular pathways connected to adiposity. Fifteen of these 23 SSGs along with 8 other SSGs showed a co-expression with enriched obesity-related genes (r ≥ 0.8). These SSGs and their co-expression partners are involved in diverse metabolic pathways including adipogenesis, adipocytokine signaling pathway, renin-angiotensin system, etc. This study concludes that SSGs could act as molecular signatures for tracing the basis of adipogenesis among obese patients. Integrated network centered methods may accelerate the identification of new molecular targets from the complex obesity genomics data.
Collapse
Affiliation(s)
- Jamal Sabir M. Sabir
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdelfatteh El Omri
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Babajan Banaganapalli
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nada Aljuaid
- Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulkader M. Shaikh Omar
- Biology, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulmalik Altaf
- Department of Surgery, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nahid H. Hajrah
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Houda Zrelli
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Leila Arfaoui
- Clinical Nutrition Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ramu Elango
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mona G. Alharbi
- Biology, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Alawiah M. Alhebshi
- Biology, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Robert K. Jansen
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, United States of America
| | - Noor A. Shaik
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muhummadh Khan
- Center of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Genomics and Biotechnology Section and Research Group, Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- * E-mail:
| |
Collapse
|
27
|
Zhang Y, Shi F, Yu Z, Yang A, Zeng M, Wang J, Yin H, Zhang B, Ma X. A cross-sectional study on factors associated with hypertension and genetic polymorphisms of renin-angiotensin-aldosterone system in Chinese hui pilgrims to hajj. BMC Public Health 2019; 19:1223. [PMID: 31484569 PMCID: PMC6727391 DOI: 10.1186/s12889-019-7357-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/22/2019] [Indexed: 01/11/2023] Open
Abstract
Background Hypertension is the leading risk factor for cardiovascular disease (CVD), however, the studies on lifestyle and genetic risks in Chinese pilgrims to Hajj was limited. The aim of this study is to examine the prevalence and associated lifestyle and genetic risks for hypertension among Hui Hajj pilgrims in China. Methods We performed a cross-sectional analysis of data in 1,465 participants aged 30–70 years who participated in a medical examination for Hui Hajj pilgrims from Gansu province, China in 2017. Multiple logistic regression was used to evaluate the association of potential risk factors with hypertension. Deoxyribonucleic acid (DNA) polymorphism was examined at sites in the renin-angiotensin-aldosterone system (RAAS). Results The prevalence of hypertension was 47% among this population. Lifestyle factors such as fried food preference (like vs. dislike: odds ratio [OR]: =1.53, 95% confidence interval [CI]: 1.13–2.09) and barbecued food preference (like vs. dislike: OR = 1.45, 95% CI: 1.06–1.97) were associated with elevated risk of hypertension among Hui pilgrims. Comparing with Angiotensin converting enzyme (ACE) rs4425 AA genotype, TT genotype was associated with hypertension risk (OR = 2.16, 95% CI: 1.17–4.00). Similar results were also observed for ACE rs4437 CC genotype (OR = 1.95, 95% CI: 1.07–3.55), Angiotensin II receptor (ATR) rs129876 AA genotype (OR = 4.10, 95% CI: 2.30–7.32) and Aldosterone synthase (CYP11B2) rs1912 TT genotype (OR = 2.82, 95% CI: 1.57–5.06) genotypes. Conclusions Unhealthy lifestyle and genetic factors were associated with the prevalence of hypertension in Chinese Hui pilgrims and their interactions were also observed. Electronic supplementary material The online version of this article (10.1186/s12889-019-7357-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yinxia Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610000, China.,Northwest Minzu University, Lanzhou, 730030, China
| | - Fangfang Shi
- Center for Disease Control and Prevention, Kongtong District, Pingliang, Gansu, China
| | - Zhanbiao Yu
- Qingyang People's Hospital, Qingyang, 745000, China
| | - Aimin Yang
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Kowloon, Hong Kong SAR
| | - Maolan Zeng
- Northwest Minzu University, Lanzhou, 730030, China
| | - Jiaoyue Wang
- Gansu International Travel Healthcare Center, Lanzhou, 730000, China
| | - Haiping Yin
- Gansu International Travel Healthcare Center, Lanzhou, 730000, China
| | - Benzhong Zhang
- School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Xiao Ma
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610000, China.
| |
Collapse
|
28
|
Capolongo G, Suzumoto Y, D'Acierno M, Simeoni M, Capasso G, Zacchia M. ERK1,2 Signalling Pathway along the Nephron and Its Role in Acid-base and Electrolytes Balance. Int J Mol Sci 2019; 20:E4153. [PMID: 31450703 PMCID: PMC6747339 DOI: 10.3390/ijms20174153] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/14/2019] [Accepted: 08/22/2019] [Indexed: 12/17/2022] Open
Abstract
Mitogen-activated protein kinases (MAPKs) are intracellular molecules regulating a wide range of cellular functions, including proliferation, differentiation, apoptosis, cytoskeleton remodeling and cytokine production. MAPK activity has been shown in normal kidney, and its over-activation has been demonstrated in several renal diseases. The extracellular signal-regulated protein kinases (ERK 1,2) signalling pathway is the first described MAPK signaling. Intensive investigations have demonstrated that it participates in the regulation of ureteric bud branching, a fundamental process in establishing final nephron number; in addition, it is also involved in the differentiation of the nephrogenic mesenchyme, indicating a key role in mammalian kidney embryonic development. In the present manuscript, we show that ERK1,2 signalling mediates several cellular functions also in mature kidney, describing its role along the nephron and demonstrating whether it contributes to the regulation of ion channels and transporters implicated in acid-base and electrolytes homeostasis.
Collapse
Affiliation(s)
- Giovanna Capolongo
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
| | | | | | - Mariadelina Simeoni
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
| | - Giovambattista Capasso
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy
- Biogem Scarl, 83031 Ariano Irpino, Italy
| | - Miriam Zacchia
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", 80131 Naples, Italy.
| |
Collapse
|
29
|
Masenga SK, Hamooya BM, Nzala S, Kwenda G, Heimburger DC, Mutale W, Koethe JR, Kirabo A, Munsaka SM. HIV, immune activation and salt-sensitive hypertension (HISH): a research proposal. BMC Res Notes 2019; 12:424. [PMID: 31311574 PMCID: PMC6636142 DOI: 10.1186/s13104-019-4470-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 07/11/2019] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE The objective of this study is to quantify and compare the effect of excess dietary salt on immune cell activation and blood pressure in HIV versus HIV negative individuals. RESULTS Salt-sensitivity is associated with increased immune cell activation in animal studies. This concept has not been tested in people living with HIV. This study will therefore add more information in elucidating the interaction between HIV infection and/or anti-retroviral therapy (ART), immune-activation/inflammation and hypertension.
Collapse
Affiliation(s)
- Sepiso K. Masenga
- School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka, Zambia
- Vanderbilt Institute for Global Health, Nashville, TN USA
| | - Benson M. Hamooya
- School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia
- Department of Epidemiology and Biostatistics, School of Public Health, University of Zambia, Lusaka, Zambia
| | - Selestine Nzala
- Department of Medical Education Development, University of Zambia, Lusaka, Zambia
| | - Geoffrey Kwenda
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka, Zambia
| | | | - Wilbroad Mutale
- Department of Health Policy and Management, School of Public Health, University of Zambia, Lusaka, Zambia
| | - John R. Koethe
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN USA
| | - Sody M. Munsaka
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka, Zambia
| |
Collapse
|
30
|
Snyder EM, Kelley EF, Sprissler R, Olson TP. The importance and challenges of developing a pharmacogenetics test for hypertension. Pharmacogenomics 2019; 20:563-566. [PMID: 31136254 DOI: 10.2217/pgs-2019-0056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
| | - Eli F Kelley
- School of Kinesiology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ryan Sprissler
- Geneticure, Inc., Rochester, MN 55902, USA.,University of Arizona Genomics Core, Tucson, Arizona, AZ 85721 USA
| | - Thomas P Olson
- Geneticure, Inc., Rochester, MN 55902, USA.,Division of Cardiovascular Diseases, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| |
Collapse
|
31
|
Adachi T, Kamiya K, Takagi D, Ashikawa H, Hori M, Kondo T, Yamada S. Combined effects of obesity and objectively-measured daily physical activity on the risk of hypertension in middle-aged Japanese men: A 4-year prospective cohort study. Obes Res Clin Pract 2019; 13:365-370. [PMID: 31076264 DOI: 10.1016/j.orcp.2019.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND The combined effects of physical inactivity and obesity on hypertension have been recognized; however, previous studies evaluated physical activity using questionnaires. We aimed to examine the effects of physical activity, measured using an accelerometer, and obesity on hypertension onset. METHODS At baseline, 426 middle-aged Japanese men who were not on antihypertensive medications were included. Physical activity was measured for 7 consecutive days using an accelerometer. Mean daily moderate to vigorous physical activity (MVPA) and step count (SC) were calculated. Low MVPA and low SC were each defined as the first tertile. Obesity was defined as ≥25 kg/m2 of body mass index. The onset of hypertension was defined as receiving antihypertensive agents during the 4-year follow-up. The combined effects of obesity and physical inactivity on hypertension were examined using Cox regression analysis. Potential confounders included age, smoking, alcohol consumption, daily salt intake, dyslipidemia, diabetes mellitus, and systolic and diastolic blood pressures. RESULTS Cox regression analysis revealed that both obesity and low MVPA predicted hypertension in patients, independent of confounders (hazard ratio [HR]: 2.64, 95% confidence interval [CI]: 1.08-6.42, p = 0.033), unlike obesity alone (HR: 1.50, 95% CI: 0.50-3.26, p = 0.590). Stratification by obesity and SC revealed similar hypertension risks among the two groups (Obesity with low SC [HR: 2.10, 95% CI 0.88-5.24, p = 0.089]; Obesity without low SC [HR: 1.72, 95% CI 0.93-4.01, p = 0.082]). CONCLUSIONS Here, findings suggest that the coexistence of obesity and decreased MVPA may increase the risk of hypertension onset.
Collapse
Affiliation(s)
- Takuji Adachi
- Program in Physical and Occupational Therapy, Nagoya University Graduate School of Medicine, 1-1-20, Daiko-minami, Higashi-ku, Nagoya 461-8673, Japan
| | - Kuniyasu Kamiya
- Program in Physical and Occupational Therapy, Nagoya University Graduate School of Medicine, 1-1-20, Daiko-minami, Higashi-ku, Nagoya 461-8673, Japan
| | - Daichi Takagi
- Program in Physical and Occupational Therapy, Nagoya University Graduate School of Medicine, 1-1-20, Daiko-minami, Higashi-ku, Nagoya 461-8673, Japan
| | - Hironobu Ashikawa
- Program in Physical and Occupational Therapy, Nagoya University Graduate School of Medicine, 1-1-20, Daiko-minami, Higashi-ku, Nagoya 461-8673, Japan
| | - Masaya Hori
- Program in Physical and Occupational Therapy, Nagoya University Graduate School of Medicine, 1-1-20, Daiko-minami, Higashi-ku, Nagoya 461-8673, Japan
| | - Takaaki Kondo
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, 1-1-20, Daiko-minami, Higashi-ku, Nagoya 461-8673, Japan
| | - Sumio Yamada
- Department of Health Sciences, Nagoya University Graduate School of Medicine, 1-1-20, Daiko-minami, Higashi-ku, Nagoya 461-8673, Japan.
| |
Collapse
|
32
|
Souza LAC, Worker CJ, Li W, Trebak F, Watkins T, Gayban AJB, Yamasaki E, Cooper SG, Drumm BT, Feng Y. (Pro)renin receptor knockdown in the paraventricular nucleus of the hypothalamus attenuates hypertension development and AT 1 receptor-mediated calcium events. Am J Physiol Heart Circ Physiol 2019; 316:H1389-H1405. [PMID: 30925093 DOI: 10.1152/ajpheart.00780.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Activation of the brain renin-angiotensin system (RAS) is a pivotal step in the pathogenesis of hypertension. The paraventricular nucleus (PVN) of the hypothalamus is a critical part of the angiotensinergic sympatho-excitatory neuronal network involved in neural control of blood pressure and hypertension. However, the importance of the PVN (pro)renin receptor (PVN-PRR)-a key component of the brain RAS-in hypertension development has not been examined. In this study, we investigated the involvement and mechanisms of the PVN-PRR in DOCA-salt-induced hypertension, a mouse model of hypertension. Using nanoinjection of adeno-associated virus-mediated Cre recombinase expression to knock down the PRR specifically in the PVN, we report here that PVN-PRR knockdown attenuated the enhanced blood pressure and sympathetic tone associated with hypertension. Mechanistically, we found that PVN-PRR knockdown was associated with reduced activation of ERK (extracellular signal-regulated kinase)-1/2 in the PVN and rostral ventrolateral medulla during hypertension. In addition, using the genetically encoded Ca2+ biosensor GCaMP6 to monitor Ca2+-signaling events in the neurons of PVN brain slices, we identified a reduction in angiotensin II type 1 receptor-mediated Ca2+ activity as part of the mechanism by which PVN-PRR knockdown attenuates hypertension. Our study demonstrates an essential role of the PRR in PVN neurons in hypertension through regulation of ERK1/2 activation and angiotensin II type 1 receptor-mediated Ca2+ activity. NEW & NOTEWORTHY PRR knockdown in PVN neurons attenuates the development of DOCA-salt hypertension and autonomic dysfunction through a decrease in ERK1/2 activation in the PVN and RVLM during hypertension. In addition, PRR knockdown reduced AT1aR expression and AT1R-mediated calcium activity during hypertension. Furthermore, we characterized the neuronal targeting specificity of AAV serotype 2 in the mouse PVN and validated the advantages of the genetically encoded calcium biosensor GCaMP6 in visualizing neuronal calcium activity in the PVN.
Collapse
Affiliation(s)
- Lucas A C Souza
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Caleb J Worker
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Wencheng Li
- Department of Pathology, Wake Forest University , Winston-Salem, North Carolina
| | - Fatima Trebak
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Trevor Watkins
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Ariana Julia B Gayban
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Evan Yamasaki
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Silvana G Cooper
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Yumei Feng
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
| |
Collapse
|
33
|
Hoh BP, Abdul Rahman T, Yusoff K. Natural selection and local adaptation of blood pressure regulation and their perspectives on precision medicine in hypertension. Hereditas 2019; 156:1. [PMID: 30636949 PMCID: PMC6323824 DOI: 10.1186/s41065-019-0080-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 01/01/2019] [Indexed: 01/09/2023] Open
Abstract
Prevalence of hypertension (HTN) varies substantially across different populations. HTN is not only common - affecting at least one third of the world's adult population - but is also the most important driver for cardiovascular diseases. Yet up to a third of hypertensive patients are resistant to therapy, contributed by secondary hypertension but more commonly the hitherto inability to precisely predict response to specific antihypertensive agents. Population and individual genomics information could be useful in guiding the selection and predicting the response to treatment - an approach known as precision medicine. However this cannot be achieved without the knowledge of genetic variations that influence blood pressure (BP). A number of evolutionary factors including population demographics and forces of natural selection may be involved. This article explores some ideas on how natural selection influences BP regulation in ethnically and geographically diverse populations that could lead to them being susceptible to HTN. We explore how such evolutionary factors could impact the implementation of precision medicine in HTN. Finally, in order to ensure the success of precision medicine in HTN, we call for more initiatives to understand the genetic architecture within and between diverse populations with ancestry from different parts of the world, and to precisely classify the intermediate phenotypes of HTN.
Collapse
Affiliation(s)
- Boon-Peng Hoh
- 1Faculty of Medicine and Health Sciences, UCSI University, Cheras, 56000 Kuala Lumpur, Malaysia.,2Chinese Academy of Sciences Key Laboratory of Computational Biology, Max Planck Independent Research Group on Population Genomics, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, CAS, Shanghai, 200031 China
| | - Thuhairah Abdul Rahman
- 3Clinical Pathology Diagnostic Centre Research Laboratory, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, 47000 Sungai Buloh, Selangor Malaysia
| | - Khalid Yusoff
- 1Faculty of Medicine and Health Sciences, UCSI University, Cheras, 56000 Kuala Lumpur, Malaysia
| |
Collapse
|
34
|
Whelton PK, Carey RM, Aronow WS, Casey DE, Collins KJ, Dennison Himmelfarb C, DePalma SM, Gidding S, Jamerson KA, Jones DW, MacLaughlin EJ, Muntner P, Ovbiagele B, Smith SC, Spencer CC, Stafford RS, Taler SJ, Thomas RJ, Williams KA, Williamson JD, Wright JT. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the Prevention, Detection, Evaluation, and Management of High Blood Pressure in Adults: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2018; 138:e484-e594. [PMID: 30354654 DOI: 10.1161/cir.0000000000000596] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Paul K Whelton
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Robert M Carey
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Wilbert S Aronow
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Donald E Casey
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Karen J Collins
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Cheryl Dennison Himmelfarb
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Sondra M DePalma
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Samuel Gidding
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Kenneth A Jamerson
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Daniel W Jones
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Eric J MacLaughlin
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Paul Muntner
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Bruce Ovbiagele
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Sidney C Smith
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Crystal C Spencer
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Randall S Stafford
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Sandra J Taler
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Randal J Thomas
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Kim A Williams
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Jeff D Williamson
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| | - Jackson T Wright
- American Society for Preventive Cardiology Representative. †ACC/AHA Representative. ‡Lay Volunteer/Patient Representative. §Preventive Cardiovascular Nurses Association Representative. ‖American Academy of Physician Assistants Representative. ¶Task Force Liaison. #Association of Black Cardiologists Representative. **American Pharmacists Association Representative. ††ACC/AHA Prevention Subcommittee Liaison. ‡‡American College of Preventive Medicine Representative. §§American Society of Hypertension Representative. ‖‖Task Force on Performance Measures Liaison. ¶¶American Geriatrics Society Representative. ##National Medical Association Representative
| |
Collapse
|
35
|
Negishi E, Fukuda N, Otsuki T, Katakawa M, Komatsu K, Chen L, Tanaka S, Kobayashi H, Hatanaka Y, Ueno T, Endo M, Mashimo T, Nishiyama A, Abe M. Involvement of complement 3 in the salt-sensitive hypertension by activation of renal renin-angiotensin system in spontaneously hypertensive rats. Am J Physiol Renal Physiol 2018; 315:F1747-F1758. [PMID: 30256128 DOI: 10.1152/ajprenal.00370.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
We previously showed that complement 3 (C3) is highly expressed in mesenchymal tissues in spontaneously hypertensive rats (SHR). We targeted C3 gene by zinc-finger nuclease (ZFN) gene-editing technology and investigated blood pressure and phenotype in SHR. Blood pressure was measured by tail-cuff and telemetry methods. Histology and expression of liver X receptor α (LXRα), renin, Krüppel-like factor 5 (KLF5), and E-cadherin were evaluated in kidneys. Mesangial cells (MCs) were removed from glomeruli from three strains, and we evaluated the phenotype in vitro. SHR showed the salt-sensitive hypertension that was abolished in C3 knockout (KO) SHR. Proliferation of MCs from SHR was higher than that from Wistar-Kyoto (WKY) rats and showed a synthetic phenotype. Renal injury scores were higher in SHR than in WKY rats and C3 KO SHR. Expression of E-cadherin was lower, and expression of renin was higher in the nephrotubulus from SHR than WKY rats and C3 KO SHR. Expression of C3 α-chain protein and α-smooth muscle actin protein was significantly higher in renal medulla from SHR than from WKY rats. Expression of angiotensinogen, LXRα, renin, and KLF5 mRNA was increased in kidney from SHR compared with C3 KO SHR. Intrarenal angiotensin II levels were significantly higher in kidney from SHR than WKY rats and C3 KO SHR. Urinary epinephrine and norepinephrine excretions were significantly higher in SHR than in WKY rats and C3 KO SHR. These findings showed that increased C3 induces salt-sensitive hypertension with increases in urinary catecholamine excretion and intrarenal activation of the renin-angiotensin system by the dedifferentiation of mesenchymal tissues in kidney from SHR.
Collapse
Affiliation(s)
- Eriko Negishi
- Division of Nephrology, Hypertension, and Endocrinology, Department of Medicine, Nihon University School of Medicine , Tokyo , Japan
| | - Noboru Fukuda
- Division of Nephrology, Hypertension, and Endocrinology, Department of Medicine, Nihon University School of Medicine , Tokyo , Japan.,Research Center, Nihon University , Tokyo , Japan
| | - Tomoyasu Otsuki
- Division of Nephrology, Hypertension, and Endocrinology, Department of Medicine, Nihon University School of Medicine , Tokyo , Japan
| | - Mayumi Katakawa
- Division of Cell Regeneration and Transplantation, Department of Functional Morphology, Nihon University School of Medicine , Tokyo , Japan
| | - Kazutoshi Komatsu
- Division of Nephrology, Hypertension, and Endocrinology, Department of Medicine, Nihon University School of Medicine , Tokyo , Japan
| | - Lan Chen
- Division of Cell Regeneration and Transplantation, Department of Functional Morphology, Nihon University School of Medicine , Tokyo , Japan
| | - Sho Tanaka
- Division of Nephrology, Hypertension, and Endocrinology, Department of Medicine, Nihon University School of Medicine , Tokyo , Japan
| | - Hiroki Kobayashi
- Division of Nephrology, Hypertension, and Endocrinology, Department of Medicine, Nihon University School of Medicine , Tokyo , Japan
| | - Yoshinari Hatanaka
- Division of Nephrology, Hypertension, and Endocrinology, Department of Medicine, Nihon University School of Medicine , Tokyo , Japan
| | - Takahiro Ueno
- Division of Nephrology, Hypertension, and Endocrinology, Department of Medicine, Nihon University School of Medicine , Tokyo , Japan
| | - Morito Endo
- Faculty of Human Health Science, Hachinohe Gakuin University, Hachinohe, Aomori, Japan
| | - Tomoji Mashimo
- Institute of Experimental Animal Sciences, Graduate School of Medicine, Osaka University , Osaka , Japan
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University School of Medicine , Takamatsu, Kagawa , Japan
| | - Masanori Abe
- Division of Nephrology, Hypertension, and Endocrinology, Department of Medicine, Nihon University School of Medicine , Tokyo , Japan
| |
Collapse
|
36
|
Hachiya T, Narita A, Ohmomo H, Sutoh Y, Komaki S, Tanno K, Satoh M, Sakata K, Hitomi J, Nakamura M, Ogasawara K, Yamamoto M, Sasaki M, Hozawa A, Shimizu A. Genome-wide analysis of polymorphism × sodium interaction effect on blood pressure identifies a novel 3'-BCL11B gene desert locus. Sci Rep 2018; 8:14162. [PMID: 30242241 PMCID: PMC6155053 DOI: 10.1038/s41598-018-32074-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 08/24/2018] [Indexed: 12/19/2022] Open
Abstract
Excessive sodium intake is a global risk factor for hypertension. Sodium effects on blood pressure vary from person to person; hence, high-risk group targeting based on personal genetic information can play a complementary role to ongoing population preventive approaches to reduce sodium consumption. To identify genetic factors that modulate sodium effects on blood pressure, we conducted a population-based genome-wide interaction analysis in 8,768 Japanese subjects, which was >3 times larger than a similar previous study. We tested 7,135,436 polymorphisms in the discovery cohort, and loci that met suggestive significance were further examined in an independent replication cohort. We found that an interaction between a novel 3'-BCL11B gene desert locus and daily sodium consumption was significantly associated with systolic blood pressure in both discovery and replication cohorts under the recessive model. Further statistical analysis of rs8022678, the sentinel variant of the 3'-BCL11B gene desert locus, showed that differences in mean systolic blood pressure between high and low sodium consumption subgroups were 5.9 mm Hg (P = 8.8 × 10-12) in rs8022678 A carriers and -0.3 mm Hg (P = 0.27) in rs8022678 A non-carriers, suggesting that the rs8022678 genotype can classify persons into sodium-sensitive (A carriers) and sodium-insensitive (A non-carriers) subgroups. Our results implied that rs8022678 A carriers may receive a greater benefit from sodium-lowering interventions than non-carriers.
Collapse
Affiliation(s)
- Tsuyoshi Hachiya
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Shiwa, Japan
| | - Akira Narita
- Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Hideki Ohmomo
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Shiwa, Japan
| | - Yoichi Sutoh
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Shiwa, Japan
| | - Shohei Komaki
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Shiwa, Japan
| | - Kozo Tanno
- Division of Clinical Research and Epidemiology, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Shiwa, Japan
- Department of Hygiene and Preventive Medicine, School of Medicine, Iwate Medical University, Morioka, Japan
| | - Mamoru Satoh
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Shiwa, Japan
- Division of Biomedical Information Analysis, Institute for Biomedical Sciences, Iwate Medical University, Shiwa, Japan
| | - Kiyomi Sakata
- Division of Clinical Research and Epidemiology, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Shiwa, Japan
- Department of Hygiene and Preventive Medicine, School of Medicine, Iwate Medical University, Morioka, Japan
| | - Jiro Hitomi
- Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Shiwa, Japan
- Department of Anatomy, School of Medicine, Iwate Medical University, Shiwa, Japan
| | - Motoyuki Nakamura
- Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Shiwa, Japan
- Department of Internal Medicine, School of Medicine, Iwate Medical University, Morioka, Japan
| | - Kuniaki Ogasawara
- Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Shiwa, Japan
- Department of Neurosurgery, School of Medicine, Iwate Medical University, Morioka, Japan
| | - Masayuki Yamamoto
- Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, 980-8573, Japan
| | - Makoto Sasaki
- Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Shiwa, Japan
- Division of Ultrahigh Field MRI, Institute for Biomedical Sciences, Iwate Medical University, Shiwa, Japan
| | - Atsushi Hozawa
- Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan.
| | - Atsushi Shimizu
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Shiwa, Japan.
| |
Collapse
|
37
|
Dong OM. Excessive dietary sodium intake and elevated blood pressure: a review of current prevention and management strategies and the emerging role of pharmaconutrigenetics. BMJ Nutr Prev Health 2018; 1:7-16. [PMID: 33235949 PMCID: PMC7678480 DOI: 10.1136/bmjnph-2018-000004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/25/2022] Open
Affiliation(s)
- Olivia M Dong
- Center for Pharmacogenomics and Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
38
|
Whelton PK, Carey RM, Aronow WS, Casey DE, Collins KJ, Dennison Himmelfarb C, DePalma SM, Gidding S, Jamerson KA, Jones DW, MacLaughlin EJ, Muntner P, Ovbiagele B, Smith SC, Spencer CC, Stafford RS, Taler SJ, Thomas RJ, Williams KA, Williamson JD, Wright JT. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the Prevention, Detection, Evaluation, and Management of High Blood Pressure in Adults: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Hypertension 2018; 71:e13-e115. [PMID: 29133356 DOI: 10.1161/hyp.0000000000000065] [Citation(s) in RCA: 1749] [Impact Index Per Article: 249.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
39
|
Whelton PK, Carey RM, Aronow WS, Casey DE, Collins KJ, Dennison Himmelfarb C, DePalma SM, Gidding S, Jamerson KA, Jones DW, MacLaughlin EJ, Muntner P, Ovbiagele B, Smith SC, Spencer CC, Stafford RS, Taler SJ, Thomas RJ, Williams KA, Williamson JD, Wright JT. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the Prevention, Detection, Evaluation, and Management of High Blood Pressure in Adults: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Hypertension 2018. [DOI: 10.1161/hyp.0000000000000065 10.1016/j.jacc.2017.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
40
|
Whelton PK, Carey RM, Aronow WS, Casey DE, Collins KJ, Dennison Himmelfarb C, DePalma SM, Gidding S, Jamerson KA, Jones DW, MacLaughlin EJ, Muntner P, Ovbiagele B, Smith SC, Spencer CC, Stafford RS, Taler SJ, Thomas RJ, Williams KA, Williamson JD, Wright JT. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the Prevention, Detection, Evaluation, and Management of High Blood Pressure in Adults: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol 2018; 71:e127-e248. [PMID: 29146535 DOI: 10.1016/j.jacc.2017.11.006] [Citation(s) in RCA: 3397] [Impact Index Per Article: 485.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
41
|
Douma LG, Gumz ML. Circadian clock-mediated regulation of blood pressure. Free Radic Biol Med 2018; 119:108-114. [PMID: 29198725 PMCID: PMC5910276 DOI: 10.1016/j.freeradbiomed.2017.11.024] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 11/21/2017] [Accepted: 11/28/2017] [Indexed: 12/24/2022]
Abstract
Most bodily functions vary over the course of a 24h day. Circadian rhythms in body temperature, sleep-wake cycles, metabolism, and blood pressure (BP) are just a few examples. These circadian rhythms are controlled by the central clock in the suprachiasmatic nucleus (SCN) of the hypothalamus and peripheral clocks located throughout the body. Light and food cues entrain these clocks to the time of day and this synchronicity contributes to the regulation of a variety of physiological processes with effects on overall health. The kidney, brain, nervous system, vasculature, and heart have been identified through the use of mouse models and clinical trials as peripheral clock regulators of BP. The dysregulation of this circadian pattern of BP, with or without hypertension, is associated with increased risk for cardiovascular disease. The mechanism of this dysregulation is unknown and is a growing area of research. In this review, we highlight research of human and mouse circadian models that has provided insight into the roles of these molecular clocks and their effects on physiological functions. Additional tissue-specific studies of the molecular clock mechanism are needed, as well as clinical studies including more diverse populations (different races, female patients, etc.), which will be critical to fully understand the mechanism of circadian regulation of BP. Understanding how these molecular clocks regulate the circadian rhythm of BP is critical in the treatment of circadian BP dysregulation and hypertension.
Collapse
Affiliation(s)
- Lauren G Douma
- Department of Medicine, Division of Nephrology, Hypertension and Renal Transplantation, University of Florida, Gainesville, FL 32610, United States; Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, United States
| | - Michelle L Gumz
- Department of Medicine, Division of Nephrology, Hypertension and Renal Transplantation, University of Florida, Gainesville, FL 32610, United States; Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, United States.
| |
Collapse
|
42
|
Liu K, Liu Z, Qi H, Liu B, Wu J, Liu Y, Zhang J, Cao H, Yan Y, He Y, Zhang L. Genetic Variation in SLC8A1 Gene Involved in Blood Pressure Responses to Acute Salt Loading. Am J Hypertens 2018; 31:415-421. [PMID: 29182730 DOI: 10.1093/ajh/hpx179] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 11/02/2017] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Salt sensitivity of blood pressure (SSBP) increases the risk of cardiovascular complications, and the heritability of SSBP is about 50% in Chinese population. However, studies identifying genes involved in BP responses to acute sodium loading and diuresis shrinkage are still limited. METHOD A total of 342 essential hypertensives from Beijing were recruited in our study. A modified Sullivan's acute oral saline load and diuresis shrinkage test was conducted to each individual. Medical history and lifestyle risk factors were obtained by questionnaire. Generalized linear model was used to examine the associations of 29 single-nucleotide polymorphisms (SNPs) with SSBP and false discovery rate (FDR) was used to correct P values for multiple testing. RESULTS In the process of acute sodium loading, after adjusting for age and 24-hour urinary sodium concentration, SNPs in CYP11B2, PRKG1, SLC8A1 genes were significantly associated with systolic BP (SBP) rising in the additive and recessive model; SNPs in CYP4A11, PRKG1, SLC8A1, and ADRB2 genes were significantly associated with diastolic BP (DBP) rising. In the process of diuresis shrinkage, SNPs of CLCNKA, eNOS, PRKG1 gene were associated with SBP and DBP decreasing. After FDR correction, rs434082 in SLC8A1 gene was still significantly associated with blood pressure rising during salt load. In the additive model, A allele increased DBP of 2.8 mm Hg (FDR_q = 0.029) and MAP of 3.1 mm Hg (FDR_q = 0.029) after adjusting for age and 24-hour urinary sodium concentration. CONCLUSION SLC8A1 gene may contribute to BP change in the process of acute sodium loading in a Han Chinese population.
Collapse
Affiliation(s)
- Kuo Liu
- Department of Epidemiology and Health statistics, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Zheng Liu
- Department of Epidemiology and Health statistics, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Han Qi
- Department of Epidemiology and Health statistics, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Bin Liu
- Department of Epidemiology and Health statistics, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Jingjing Wu
- Department of Epidemiology and Health statistics, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yezhou Liu
- Department of Epidemiology and Health statistics, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
| | - Jie Zhang
- Department of Epidemiology and Health statistics, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Han Cao
- Department of Epidemiology and Health statistics, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yuxiang Yan
- Department of Epidemiology and Health statistics, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yan He
- Department of Epidemiology and Health statistics, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Ling Zhang
- Department of Epidemiology and Health statistics, School of Public Health, Capital Medical University, Beijing, People’s Republic of China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| |
Collapse
|
43
|
Chen YL, Li TJ, Hao Y, Wu BG, Li H, Geng N, Sun ZQ, Zheng LQ, Sun YX. Association of rs2271037 and rs3749585 polymorphisms in CORIN with susceptibility to hypertension in a Chinese Han population: A case-control study. Gene 2018; 651:79-85. [PMID: 29391274 DOI: 10.1016/j.gene.2018.01.080] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 01/12/2018] [Accepted: 01/24/2018] [Indexed: 01/01/2023]
Abstract
Corins are membrane-bound protease that regulates blood pressure by activating the natriuretic peptides. These pro-atrial natriuretic peptide convertases are essential for sodium homeostasis and normal blood pressure. CORIN variants have been identified in humans and other animals, but no studies of CORIN polymorphisms have been conducted in northeastern China. This study aims to investigate the association of 2 single nucleotide polymorphisms (SNPs) in CORIN (rs2271037 and rs3749585) with hypertension, as well as their potential interactions with some risk factors of hypertension in a Han population of northeastern China. A case-control study, including 402 patients with hypertension and 406 participants with normal blood pressure, was conducted in Liaoning province. SNP genotyping was carried out by high resolution melting (HRM) after polymerase chain reaction amplifications. Since rs3749585 is located in 3' untranslated region (UTR) of CORIN, in silico analysis was used to predict target micro RNAs on TargetScan, miRanda, and DIANA-microT. As a result, mutant T allele in rs2271037 (odds ratio [OR], 1.693; 95% confidence [CI], 1.528-1.877; p < 0.001) and C allele in rs3749585 (OR, 1.114; 95% CI 1.011-1.227; p = 0.029) increased the risk of hypertension, comparing with wild G allele and T allele, respectively. Patients with genotype TT (OR, 10.209; 95% CI, 6.414-16.250; p < 0.001) and GT (OR, 1.730; 95% CI, 1.226-2.443; p = 0.002) have higher risk of hypertension than those with genotype GG. SNP rs2271037 was significantly associated with susceptibility to hypertension in all genetic models (dominant model: OR, 2.879; 95% CI, 2.080-3.986; p < 0.001; recessive model: OR, 7.159; 95% CI, 4.779-10.724; p < 0.001; additive model: OR, 1.535; 95% CI, 1.163-2.027; p = 0.002). SNP rs3749585 was significantly correlated with hypertension susceptibility only in dominant model (OR, 1.533; 95% CI, 1.073-2.189; p = 0.019), but not in recessive model (OR, 1.220; 95% CI, 0.906-1.644; p = 0.191) or additive model (OR, 0.915; 95% CI, 0.694-1.205; p = 0.527). After adjusting for age, gender, body mass index (BMI), smoking, low-density lipoprotein cholesterol, and serum sodium level in logistic models, the same statistical results were obtained. Interaction study showed the association between CORIN polymorphisms and hypertension could be changed by overweight (BMI ≥ 25 kg/m2). In silico analyses implicated hsa-miR-495 as a target miRNA that potentially interacts with the 3' UTR of CORIN. In conclusion, polymorphisms of rs2271037 and rs3749585 in CORIN were significantly associated with hypertension in a Han population of northeastern China. The mutant-type T allele of rs2271037 and C allele of rs3749585 might increase the susceptibility to hypertension in this population.
Collapse
Affiliation(s)
- Yan-Li Chen
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Tie-Jun Li
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Ying Hao
- Department of Geriatrics, Jinqiu Hospital, Shenyang, Liaoning, PR China
| | - Bao-Gang Wu
- Department of Geriatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Hong Li
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Ning Geng
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Zhao-Qing Sun
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Li-Qiang Zheng
- Department of Clinical Epidemiology, Library, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Ying-Xian Sun
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
44
|
Jensen PN, Bao TQ, Huong TTT, Heckbert SR, Fitzpatrick AL, LoGerfo JP, Ngoc TLV, Mokdad AH. The association of estimated salt intake with blood pressure in a Viet Nam national survey. PLoS One 2018; 13:e0191437. [PMID: 29346423 PMCID: PMC5773206 DOI: 10.1371/journal.pone.0191437] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 01/04/2018] [Indexed: 12/03/2022] Open
Abstract
OBJECTIVE To evaluate the association of salt consumption with blood pressure in Viet Nam, a developing country with a high level of salt consumption. DESIGN AND SETTING Analysis of a nationally representative sample of Vietnamese adults 25-65 years of age who were surveyed using the World Health Organization STEPwise approach to Surveillance protocol. Participants who reported acute illness, pregnancy, or current use of antihypertensive medications were excluded. Daily salt consumption was estimated from fasting mid-morning spot urine samples. Associations of salt consumption with systolic blood pressure and prevalent hypertension were assessed using adjusted linear and generalized linear models. Interaction terms were tested to assess differences by age, smoking, alcohol consumption, and rural/urban status. RESULTS The analysis included 2,333 participants (mean age: 37 years, 46% male, 33% urban). The average estimated salt consumption was 10g/day. No associations of salt consumption with blood pressure or prevalent hypertension were observed at a national scale in men or women. The associations did not differ in subgroups defined by age, smoking, or alcohol consumption; however, associations differed between urban and rural participants (p-value for interaction of urban/rural status with salt consumption, p = 0.02), suggesting that higher salt consumption may be associated with higher systolic blood pressure in urban residents but lower systolic blood pressure in rural residents. CONCLUSIONS Although there was no evidence of an association at a national level, associations of salt consumption with blood pressure differed between urban and rural residents in Viet Nam. The reasons for this differential association are not clear, and given the large rate of rural to urban migration experienced in Viet Nam, this topic warrants further investigation.
Collapse
Affiliation(s)
- Paul N. Jensen
- Department of Epidemiology, University of Washington, Seattle, WA, United States of America
| | - Tran Quoc Bao
- Department of Preventive Medicine, Viet Nam Ministry of Health, Hanoi, Viet Nam
| | - Tran Thi Thanh Huong
- Department of Ethics and Social Medicine, Hanoi Medical University, Hanoi, Viet Nam
| | - Susan R. Heckbert
- Department of Epidemiology, University of Washington, Seattle, WA, United States of America
| | - Annette L. Fitzpatrick
- Department of Epidemiology, University of Washington, Seattle, WA, United States of America
- Department of Global Health, University of Washington, Seattle, WA, United States of America
| | - James P. LoGerfo
- Department of Global Health, University of Washington, Seattle, WA, United States of America
- Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Truong Le Van Ngoc
- Department of Preventive Medicine, Viet Nam Ministry of Health, Hanoi, Viet Nam
| | - Ali H. Mokdad
- Department of Epidemiology, University of Washington, Seattle, WA, United States of America
- Department of Global Health, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
45
|
Estapé ES, González-Sepúlveda L, Wei W, Rodríguez-Rivera I, Torres-Negrón I. Low to Normal Plasma Levels of Marinobufagenin 24 Hours or More after an Ischemic Stroke: A Pilot Study. ACTA ACUST UNITED AC 2018; 4. [PMID: 30957100 DOI: 10.23937/2572-4142.1510006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Background Numerous studies have demonstrated a strong relationship between circulating levels of marinobufagenin (MBG) and salt-sensitivity. Since salt-sensitive hypertensives have increased plasma levels of MBG and are known to be at a higher risk of having cardiovascular events, stroke and increased mortality, we evaluated the possibility of an association between MBG and ischemic stroke. In this pilot study, we determined plasma MBG levels in patients after surviving an ischemic stroke compared to similar age and gender groups of treated hypertensives and normotensive controls. Methods We measured plasma MBG levels in a total of 40 participants subdivided into three groups: After an ischemic stroke STR (n = 13), participants with a diagnosis of hypertension receiving blood pressure medication HT (n = 14) and normotensive control subjects CTL (n = 13). We used inferential statistics (parametric or non-parametric) and ordered logistic regression models (unadjusted and adjusted) and all statistical analyses were performed using Stata 14. Results We did not include a subject from the CTL group because of a diagnosis of glucose-6-phosphate dehydrogenase deficiency and an extreme plasma MBG value of 2,246 pmol/L. Participants' mean age was 60.4 ± 11.5 years; 56% were male. There was no significant difference between study groups (p > 0.05) for gender, age, and body mass index. HbA1c levels were significantly higher in the STR as compared to the CTL p < 0.05). In the STR group MBG levels were below the normal range (< 200 pmol/L) in three (23%), eight (61%) were in the normal range (200-400 pmol/L), while two (16%) had increased MBG values (> 400 pmol/L). Also, among the STR, the plasma MBG levels did not differ between those receiving and not receiving thrombolytic therapy (p > 0.05). From the 14 HT participants, six (43%) had MBG plasma levels within the normal range, and eight (57%) had high concentrations (> 400 pmol/L). Four (29%) of the treated hypertensives had extreme MBG levels (> 1,000 pmol/L) and normal values of blood pressure. Conclusion There was no significant elevation of plasma MBG in survivors 24 h or more after an ischemic stroke. The extreme values of plasma MBG in 29% of the treated hypertensives suggests the presence of salt-sensitivity and a possible side effect of a specific combination of medications. Both of these findings contribute new knowledge to the design of studies to define if there is an MBG molecular mechanism underlying the complex associations among salt-sensitivity, hypertension, and ischemic stroke.
Collapse
Affiliation(s)
- Estela S Estapé
- University of Puerto Rico, Medical Sciences Campus, Puerto Rico.,San Juan Bautista School of Medicine, Caguas, Puerto Rico
| | | | - Wen Wei
- National Institute of Aging, National Institutes of Health, Baltimore, MD, USA
| | | | - Ivette Torres-Negrón
- Universidad del Turabo, Ana G Méndez University System, Puerto Rico.,Universidad Metropolitana, Ana G Méndez University System, Puerto Rico
| |
Collapse
|
46
|
Mishra S, Ingole S, Jain R. Salt sensitivity and its implication in clinical practice. Indian Heart J 2017; 70:556-564. [PMID: 30170653 PMCID: PMC6116721 DOI: 10.1016/j.ihj.2017.10.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/01/2017] [Accepted: 10/10/2017] [Indexed: 02/06/2023] Open
Abstract
Hypertension (HTN) is a complex multi-factorial disease and is considered one of the foremost modifiable risk factors for stroke, heart failure, ischemic heart disease and renal dysfunction. Over the past century, salt and its linkage to HTN and cardiovascular (CV) mortality has been the subject of intense scientific scrutiny. There is now consensus that different individuals have different susceptibilities to blood pressure (BP)-raising effects of salt and this susceptiveness is called as salt sensitivity. Several renal and extra-renal mechanisms are believed to play a role. Blunted activity of the renin–angiotensin–aldosterone system (RAAS), adrenal Rac1-MR-Sgk1-NCC/ENaC pathway, renal SNS-GR-WNK4-NCC pathway, defect of membrane ion transportation, inflammation and abnormalities of Na+/Ca2+ exchange have all been implicated as pathophysiological basis for salt sensitive HTN. While salt restriction is definitely beneficial recent observation suggests that treatment with Azilsartan may improve salt sensitivity by selectively reducing renal proximal tubule Na+/H+ exchange. This encourages the future potential benefits of recognizing and therapeutically addressing the salt sensitive phenotype in humans.
Collapse
|
47
|
Simeoni M, Damiano S, Capolongo G, Trepiccione F, Zacchia M, Fuiano G, Capasso G. Rare Renal Diseases Can Be Used as Tools to Investigate Common Kidney Disorders. KIDNEY DISEASES (BASEL, SWITZERLAND) 2017; 3:43-49. [PMID: 28868291 PMCID: PMC5566759 DOI: 10.1159/000475841] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/15/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND The prevention and slowing of chronic kidney disease still represent major challenges in nephrology. To this end, a major contribution may come from the extensive knowledge on the molecular pathways involved in the pathogenesis of rare kidney diseases, since it is now possible to shed light on several aspects of these pathologies thanks to the introduction of new technologies, including next-generation sequencing. SUMMARY In steroid-resistant nephrotic patients, a genetic background has been demonstrated in both children and adults; individualized mutations have been correlated with glomerular filtration barrier alterations. In addition, studies on genetic tubulopathies expressing hypertensive phenotypes can provide useful information for a correct diagnostic and therapeutic approach in patients with essential hypertension and a poor responsiveness to therapy. KEY MESSAGE This review deals with the pathogenesis of rare glomerular diseases and tubulopathies associated with hypertension, highlighting the importance of the study of rare diseases to better understand the molecular basis of more common and complex disorders leading to end-stage renal disease.
Collapse
Affiliation(s)
- Mariadelina Simeoni
- Department of Nephrology, Magna Graecia University Hospital, Catanzaro, Italy
| | - Sara Damiano
- Department of Nephrology, University of Campania “Luigi Vanvitelli,” Naples, Italy
| | - Giovanna Capolongo
- Department of Nephrology, University of Campania “Luigi Vanvitelli,” Naples, Italy
| | | | - Miriam Zacchia
- Department of Nephrology, University of Campania “Luigi Vanvitelli,” Naples, Italy
| | - Giorgio Fuiano
- Department of Nephrology, Magna Graecia University Hospital, Catanzaro, Italy
| | | |
Collapse
|
48
|
Imbrici P, Tricarico D, Mangiatordi GF, Nicolotti O, Lograno MD, Conte D, Liantonio A. Pharmacovigilance database search discloses ClC-K channels as a novel target of the AT 1 receptor blockers valsartan and olmesartan. Br J Pharmacol 2017; 174:1972-1983. [PMID: 28334417 DOI: 10.1111/bph.13794] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 03/03/2017] [Accepted: 03/04/2017] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Human ClC-K chloride channels are highly attractive targets for drug discovery as they have a variety of important physiological functions and are associated with genetic disorders. These channels are crucial in the kidney as they control chloride reabsorption and water diuresis. In addition, loss-of-function mutations of CLCNKB and BSND genes cause Bartter's syndrome (BS), whereas CLCNKA and CLCNKB gain-of-function polymorphisms predispose to a rare form of salt sensitive hypertension. Both disorders lack a personalized therapy that is in most cases only symptomatic. The aim of this study was to identify novel ClC-K ligands from drugs already on the market, by exploiting the pharmacological side activity of drug molecules available from the FDA Adverse Effects Reporting System database. EXPERIMENTAL APPROACH We searched for drugs having a Bartter-like syndrome as a reported side effect, with the assumption that BS could be causatively related to the block of ClC-K channels. The ability of the selected BS-causing drugs to bind and block ClC-K channels was then validated through an integrated experimental and computational approach based on patch clamp electrophysiology in HEK293 cells and molecular docking simulations. KEY RESULTS Valsartan and olmesartan were able to block ClC-Ka channels and the molecular requirements for effective inhibition of these channels have been identified. CONCLUSION AND IMPLICATIONS These results suggest additional mechanisms of action for these sartans further to their primary AT1 receptor antagonism and propose these compounds as leads for designing new potent ClC-K ligands.
Collapse
Affiliation(s)
- Paola Imbrici
- Department of Pharmacy - Drug Sciences, University of Bari 'Aldo Moro', Bari, Italy
| | - Domenico Tricarico
- Department of Pharmacy - Drug Sciences, University of Bari 'Aldo Moro', Bari, Italy
| | | | - Orazio Nicolotti
- Department of Pharmacy - Drug Sciences, University of Bari 'Aldo Moro', Bari, Italy
| | | | - Diana Conte
- Department of Pharmacy - Drug Sciences, University of Bari 'Aldo Moro', Bari, Italy
| | - Antonella Liantonio
- Department of Pharmacy - Drug Sciences, University of Bari 'Aldo Moro', Bari, Italy
| |
Collapse
|
49
|
Wang TD, Tan RS, Lee HY, Ihm SH, Rhee MY, Tomlinson B, Pal P, Yang F, Hirschhorn E, Prescott MF, Hinder M, Langenickel TH. Effects of Sacubitril/Valsartan (LCZ696) on Natriuresis, Diuresis, Blood Pressures, and NT-proBNP in Salt-Sensitive Hypertension. Hypertension 2017; 69:32-41. [DOI: 10.1161/hypertensionaha.116.08484] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 09/26/2016] [Accepted: 10/20/2016] [Indexed: 11/16/2022]
Abstract
Salt-sensitive hypertension (SSH) is characterized by impaired sodium excretion and subnormal vasodilatory response to salt loading. Sacubitril/valsartan (LCZ696) was hypothesized to increase natriuresis and diuresis and result in superior blood pressure control compared with valsartan in Asian patients with SSH. In this randomized, double-blind, crossover study, 72 patients with SSH received sacubitril/valsartan 400 mg and valsartan 320 mg once daily for 4 weeks each. SSH was diagnosed if the mean arterial pressure increased by ≥10% when patients switched from low (50 mmol/d) to high (320 mmol/d) sodium diet. The primary outcome was cumulative 6- and 24-hour sodium excretion after first dose administration. Compared with valsartan, sacubitril/valsartan was associated with a significant increase in natriuresis (adjusted treatment difference: 24.5 mmol/6 hours, 50.3 mmol/24 hours, both
P
<0.001) and diuresis (adjusted treatment difference: 291.2 mL/6 hours,
P
<0.001; 356.4 mL/24 hours,
P
=0.002) on day 1, but not on day 28, and greater reductions in office and ambulatory blood pressure on day 28. Despite morning dosing of both drugs, ambulatory blood pressure reductions were more pronounced at nighttime than at daytime or the 24-hour average. Compared with valsartan, sacubitril/valsartan significantly reduced N-terminal pro B-type natriuretic peptide levels on day 28 (adjusted treatment difference: −20%;
P
=0.001). Sacubitril/valsartan and valsartan were safe and well tolerated with no significant changes in body weight or serum sodium and potassium levels with either treatments. In conclusion, sacubitril/valsartan compared with valsartan was associated with short-term increases in natriuresis and diuresis, superior office and ambulatory blood pressure control, and significantly reduced N-terminal pro B-type natriuretic peptide levels in Asian patients with SSH.
Clinical Trial Registration—
URL:
http://www.clinicaltrials.gov
. Unique identifier: NCT01681576.
Collapse
Affiliation(s)
- Tzung-Dau Wang
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Ru-San Tan
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Hae-Young Lee
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Sang-Hyun Ihm
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Moo-Yong Rhee
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Brian Tomlinson
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Parasar Pal
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Fan Yang
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Elizabeth Hirschhorn
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Margaret F. Prescott
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Markus Hinder
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| | - Thomas H. Langenickel
- From the Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan (T.-D.W.); National Heart Centre, Department of Cardiology, Singapore (R.-S.T.); Seoul National University Hospital, Department of Internal Medicine, South Korea (H.-Y.L.); The Catholic University of Korea Bucheon, St Mary’s Hospital, Department of Cardiology, Bucheon-si, Gyeonggi-do, South Korea (S.-H.I.); Dongguk University Ilsan Hospital,
| |
Collapse
|
50
|
Lee M, Kwon DY, Park J. The impacts of the interaction of genetic variation, CYP11β2 and NEDD4L, with sodium intake on pediatric obesity with gender difference: a 3-year panel study. Int J Obes (Lond) 2016; 41:542-550. [PMID: 28017963 DOI: 10.1038/ijo.2016.232] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/01/2016] [Accepted: 11/29/2016] [Indexed: 12/22/2022]
Abstract
Backgrounds/Objectives:This panel study was to predict the incidences of pediatric obesity by the interaction of sodium (Na) intake and nine single-nucleotide polymorphisms (SNPs) of salt-sensitive genes (SSGs), ACE(angiotensin-converting enzyme), ADD1 G460W,AGT M235T,CYP11β2 (cytochrome P450 family 11-subfamily β-2, -aldosterone synthase),GNB3 C285T,GRK4(A142V)(G-protein-coupled receptor kinases type 4),GRK4 (A486V),NEDD4L (neural precursor cell expressed developmentally downregulated 4 like; rs2288774) and SLC12A3 (solute carrier family 12 (Na/Cl transporters)-member 3), selected from genome-wide association study. SUBJECTS/METHODS Non-obese (non-OB) Korean children of 9 years old were recruited from eight elementary schools in Seoul in 2007 and 2009, each. Follow-up subjects (total=798) in 2010 and 2012 were final participants. Participants were classified as OB group for those whose body mass index were over the 85th percentile using the 'Korean National Growth Charts', and others were classified as non-OB. With nine SNPs typing, the genetic interaction with the variation of Na intake for 3 years was evaluated as an obesity risk. RESULTS The obesity incidence rate for non-OB children at baseline after 3 years was 10.31%. Na intake in non-OB after 3 years was significantly decreased compared with the baseline, whereas Na intake reduction was undetectable in OB. We found gender differences on association between the changes of Na intake and the obesity incidence for 3 years by the SSG variation. Odds ratio for the obesity risk was 5.75 times higher in girls having hetero/mutant types of NEDD4L with higher Na intakes (Q2+Q3+Q4 in quartiles) compared with that in the wild type with the lowest Na intake (Q1). Girls with hetero/mutant of CYP11β2 tended to increase the obesity incidence as Na intake increased (Q1<Q2<Q3<Q4, P-value trend=0.047). The other seven SNPs of SSGs had no significance over Na intake. CONCLUSIONS From this panel study and the previous cross-sectional study, we found CYP11β2 as the common gene, powerful to explain the interaction between obesity incidence and Na intake, in particular, among girls. Girls with hetero/mutant allele of this gene should reduce their daily Na intake to prevent obesity.
Collapse
Affiliation(s)
- M Lee
- Department of Food and Nutrition, Sungshin Women's University, Food and Nutrition (A-658), Sungshin University, Seoul, Republic of Korea.,Division of Statistics, Research Institute of Obesity Sciences, Sugnshin Women's University, Seoul, Republic of Korea
| | - D Y Kwon
- Division of Nutrition and Metabolism Research, Korea Food Research Institute, Gyeonggi-do, Republic of Korea
| | - J Park
- Division of Statistics, Research Institute of Obesity Sciences, Sugnshin Women's University, Seoul, Republic of Korea
| |
Collapse
|