1
|
Chen H, He X, Fan J, Mi Y, Li F. The relationship between dietary niacin intake and the incidence of all-cause and cardiovascular mortality among chronic kidney disease patients. Am J Med Sci 2024:S0002-9629(24)01520-9. [PMID: 39665972 DOI: 10.1016/j.amjms.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/18/2024] [Accepted: 11/15/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Chronic kidney disease (CKD) is a major cause of human mortality and cardiovascular disease (CVD)-related death. Niacin can treat dyslipidemia and can lower overall cardiovascular event incidence and mortality rates. The present study was designed to clarify the link between dietary consumption of niacin and cardiovascular mortality. METHODS This study enrolled subjects ≥18 years of age from the National Health and Nutrition Examination Survey 2009-2014 and excluded any individuals for whom data regarding their CKD status, dietary niacin intake, or other covariate information was unavailable. Relationships between dietary niacin intake levels and overall or CVD-related mortality among these CKD patients were assessed using univariate and multivariate Cox regression analyses. RESULTS The study included 1,798 subjects and recorded 514 and 186 instances of all-cause and cardiovascular death, respectively. Males comprised 51.8% of the study cohort, and the mean age of these subjects was 65. Cox proportional hazard model analyses revealed no relationship between dietary niacin intake and all-cause or cardiovascular death risk among the overall CKD patient population (P > 0.05). However, in age-stratified analyses, those subjects <60 years of age exhibiting the highest levels of dietary niacin consumption (≥ 38 mg/day) were found to face a significantly higher risk of all-cause mortality, and this association remained intact in sensitivity analyses. CONCLUSION These results do not support any link between the dietary intake of niacin and all-cause or cardiovascular mortality risk among patients with CKD. Age and niacin intake exhibited a significant interaction related to all-cause mortality.
Collapse
Affiliation(s)
- Hongxi Chen
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, PR China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, PR China
| | - Xu He
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, PR China; Department of Experiment Teaching Center of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, , PR China
| | - Junming Fan
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, PR China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, PR China
| | - Yongjie Mi
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, PR China; Department of Experiment Teaching Center of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, , PR China.
| | - Feiyan Li
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, PR China; Department of Experiment Teaching Center of Clinical Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, , PR China; Collaborative Innovation Center of Sichuan for Elderly Care and Health, Chengdu Medical College, Chengdu, PR China.
| |
Collapse
|
2
|
Zhou Z, Yao X. Dietary niacin intake and mortality among chronic kidney disease patients. Front Nutr 2024; 11:1435297. [PMID: 39639940 PMCID: PMC11617148 DOI: 10.3389/fnut.2024.1435297] [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: 05/20/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Background The relationship between niacin and the risk of mortality in chronic kidney disease (CKD) patients remains unclear. This study aims to investigate the potential correlation. Methods This cohort study utilized data from the 2003-2018 National Health and Nutrition Examination Survey (NHANES). The study included 6,110 patients with CKD aged 18 years or older. Weighted Cox proportional hazards models and restricted cubic splines (RCS) were employed to estimate hazard ratios for all-cause mortality and cardiovascular disease (CVD) mortality. Niacin intake was estimated using the 24 h dietary recall method, based on the type and amount of food consumed. All-cause mortality and cardiac mortality rates were determined using National Death Index (NDI) mortality data (as of 31 December 2018). Results The median niacin intake was 20.89 mg/day, with an interquartile range of 15.67-27.99 mg/day. During the follow-up period (median of 87 months), there were 1,984 all-cause deaths, including 714 CVD deaths. Compared with low niacin intake, the multivariate-adjusted hazard ratio for dietary intake of 22 mg or higher was 0.71 (95% CI, 0.57-0.88) for all-cause mortality and 0.75 (95% CI, 0.57, 0.98) for CVD mortality. Conclusion Dietary niacin intake is associated with a reduction in all-cause and cardiac mortality among CKD patients.
Collapse
Affiliation(s)
- Zhengxi Zhou
- Department of Urology, Ningbo Mingzhou Hospital, Zhejiang, China
| | - Xiaotian Yao
- Department of Urology, Ningbo Mingzhou Hospital, Zhejiang, China
- The Division of Nephrology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Zhang C, Cheng Q, Yang X, Zhao W, Luo K, Qin Y. Association of dietary niacin intake with all-cause and cardiovascular mortality of adult patients with chronic kidney disease in the United States: results from NHANES 1999-2018. Front Nutr 2024; 11:1436836. [PMID: 39279901 PMCID: PMC11392853 DOI: 10.3389/fnut.2024.1436836] [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: 05/22/2024] [Accepted: 08/20/2024] [Indexed: 09/18/2024] Open
Abstract
Objective The relationship between dietary niacin intake (DNI) and mortality rates among patients afflicted with chronic kidney disease (CKD) is a subject of debate. Utilizing data derived from the National Health and Nutrition Examination Survey (NHANES), this study adopts a retrospective cohort design with an aim to investigate the association in the American adult patients with CKD. Methods A cohort study was conducted in the National Health and Nutrition Examination Survey (NHANES) between 2009 and 2018 that enrolled 6,191 CKD patients aged 20 years and above. We collected data on mortality through 31 December 2018. DNI was measured using a 24-h recall method. The relationship between DNI levels and mortality from all causes and cardiovascular causes was analyzed using weighted Cox proportional hazards models. The Kaplan-Meier (K-M) survival curve was plotted to illustrate these associations. Results Following a median monitoring period of 85 months, it was observed that 2,419 individuals (33.08%) succumbed to all causes, whereas cardiovascular-related deaths were recorded for 746 participants (10.45%). When controlling for confounders, an inverse relationship was established between DNI and mortality rates. Specifically, a marginal increase of 1 mg/day in DNI corresponded to a reduced Hazard Ratios (HRs) of 0.993 (0.987, 0.999; p = 0.027) for all-cause mortality and 0.980 (0.969, 0.991; p < 0.001) for cardiovascular mortality. A further stratified analysis by quartiles of DNI, with the lowest quartile serving as the reference, revealed that the highest quartile was associated with HRs of 0.820 (0.697, 0.966) for all-cause mortality and 0.663 (0.465, 0.944) for cardiovascular mortality, both displaying a significant trend (p < 0.001). However, a subdivision of CKD patients by age showed that the protective effects of higher DNI were only confined to individuals aged 60 years and above but not to those under 60 years of age. Conclusion A negative correlation between DNI and mortality due to all causes and cardiovascular issues among CKD patients aged 60 and above was revealed based on the datasets; however, this association was not observed in younger individuals under 60. Consequently, enhancing DNI might serve as a beneficial therapeutic strategy specifically for the older CKD demographic.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Nephrology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China
| | - Qi Cheng
- Outpatient Department, Bethune International Peace Hospital, Shijiazhuang, Hebei, China
| | - Xinjun Yang
- Department of Nephrology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China
| | - Wei Zhao
- Department of Nephrology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China
| | - Kaifa Luo
- Department of Nephrology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China
| | - Yunlong Qin
- Department of Nephrology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
4
|
Tian X, Chen S, Xia X, Xu Q, Zhang Y, Zhang X, Wang P, Wu S, Wang A. Causal Association of Arterial Stiffness With the Risk of Chronic Kidney Disease. JACC. ASIA 2024; 4:444-453. [PMID: 39100705 PMCID: PMC11291385 DOI: 10.1016/j.jacasi.2023.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/10/2023] [Accepted: 10/21/2023] [Indexed: 08/06/2024]
Abstract
Background Previous studies on the direction of the association between arterial stiffness (AS) and chronic kidney disease (CKD) were inconsistent, leaving a knowledge gap in understanding the temporal sequence of the association. Objectives This study sought to assess the temporal and longitudinal relationship between AS and CKD. Methods The temporal relationship between AS measured by brachial ankle pulse wave velocity and CKD measured by estimated glomerular filtration rate (eGFR) was analyzed among 7,753 participants with repeated examinations in the Kailuan study using cross-lagged panel analysis. The longitudinal associations of AS status and vascular aging (VA) phenotype with incident CKD were analyzed among 10,535 participants. Results The adjusted cross-lagged path coefficient (β 1 = -0.03; 95% CI: -0.06 to -0.01; P < 0.0001) from baseline brachial ankle pulse wave velocity to follow-up eGFR was significantly greater than the path coefficient (β 2 = -0.01; 95% CI: -0.02 to 0.01; P = 0.6202) from baseline eGFR to follow-up brachial ankle pulse wave velocity (P < 0.0001 for the difference). During a median follow-up of 8.48 years, 953 cases of incident CKD (9.05%) occurred. After adjustment for confounders, borderline (HR: 1.17; 95% CI: 1.08-1.38) and elevated AS (HR: 1.39; 95% CI: 1.12-1.72) was associated a higher risk of CKD, compared with normal AS. Consistently, supernormal VA (HR: 0.76; 95% CI: 0.66-0.86) was associated with a decreased and early VA (HR: 1.36; 95% CI: 1.29-1.43) was associated with an increased risk of CKD, compared with normal VA. Conclusions AS appeared to precede the decrease in eGFR. Additionally, increased AS and early VA were associated with an increased risk of incident CKD.
Collapse
Affiliation(s)
- Xue Tian
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Shuohua Chen
- Department of Cardiology, Kailuan Hospital, North China University of Science and Technology, Tangshan, China
| | - Xue Xia
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qin Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yijun Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Xiaoli Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Penglian Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shouling Wu
- Department of Cardiology, Kailuan Hospital, North China University of Science and Technology, Tangshan, China
| | - Anxin Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
- Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Garofalo C, Borrelli S, Liberti ME, Chiodini P, Peccarino L, Pennino L, Polese L, De Gregorio I, Scognamiglio M, Ruotolo C, Provenzano M, Conte G, Minutolo R, De Nicola L. Secular Trend in GFR Decline in Non-Dialysis CKD Based on Observational Data From Standard of Care Arms of Trials. Am J Kidney Dis 2024; 83:435-444.e1. [PMID: 37956953 DOI: 10.1053/j.ajkd.2023.09.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 08/24/2023] [Accepted: 09/07/2023] [Indexed: 11/21/2023]
Abstract
RATIONALE & OBJECTIVE The standard of care (SoC) group of randomized controlled trials (RCTs) is a useful setting to explore the secular trends in kidney disease progression because implementation of best clinical practices is pursued for all patients enrolled in trials. This meta-analysis evaluated the secular trend in the change of glomerular filtration rate (GFR) decline in the SoC arm of RCTs in chronic kidney disease (CKD) published in the last 30 years. STUDY DESIGN Systematic review and meta-analysis of the SoC arms of RCTs analyzed as an observational study. SETTING & STUDY POPULATIONS Adult patients with CKD enrolled in the SoC arm of RCTs. SELECTION CRITERIA FOR STUDIES Phase 3 RCTs evaluating GFR decline as an outcome in SoC arms. DATA EXTRACTION Two independent reviewers evaluated RCTs for eligibility and extracted relevant data. ANALYTICAL APPROACH The mean of GFR declines extracted in the SoC arm of selected RCTs were pooled by using a random effects model. Meta-regression analyses were performed to identify factors that may explain heterogeneity. RESULTS The SoC arms from 92 RCTs were included in the meta-analysis with a total of 32,202 patients. The overall mean GFR decline was-4.00 (95% CI, -4.55 to-3.44) mL/min/1.73m2 per year in the SoC arms with a high level of heterogeneity (I2, 98.4% [95% CI, 98.2-98.5], P<0.001). Meta-regression analysis showed an association between publication year (β estimate, 0.09 [95% CI, 0.032-0.148], P=0.003) and reduction in GFR over time. When evaluating publication decade categorically, GFR decline was-5.44 (95% CI, -7.15 to-3.73), -3.92 (95% CI, -4.82 to-3.02), and -3.20 (95% CI, -3.75 to -2.64) mL/min/1.73m2 per year during 1991-2000, 2001-2010, and 2011-2023, respectively. Using meta-regression, the heterogeneity of GFR decline was mainly explained by age and proteinuria. LIMITATIONS Different methods assessing GFR in selected trials and observational design of the study. CONCLUSIONS In the last 3 decades, GFR decline has decreased over time in patients enrolled in RCTs who received the standard of care. TRIAL REGISTRATION Registered at PROSPERO with record number CRD42022357704. PLAIN-LANGUAGE SUMMARY This study evaluated the secular trend in the change in glomerular filtration rate (GFR) decline in the placebo arms of randomized controlled trials (RCTs) that were studying approaches to protect the kidneys in the setting of chronic kidney disease. The placebo groups of RCTs are useful for examining whether the rate of progression of kidney disease has changed over time. We found an improvement in the slope of change in GFR over time. These findings suggest that adherence to standards of kidney care as implemented in clinical trials may be associated with improved clinical outcomes, and these data may inform the design of future RCTs in nephrology.
Collapse
Affiliation(s)
- Carlo Garofalo
- Division of Nephrology, University of Campania "Luigi Vanvitelli," Naples, Italy.
| | - Silvio Borrelli
- Division of Nephrology, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Maria Elena Liberti
- Division of Nephrology, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Paolo Chiodini
- Medical Statistics Unit, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Laura Peccarino
- Division of Nephrology, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Luigi Pennino
- Division of Nephrology, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Lucio Polese
- Division of Nephrology, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Ilaria De Gregorio
- Division of Nephrology, University of Campania "Luigi Vanvitelli," Naples, Italy
| | | | - Chiara Ruotolo
- Division of Nephrology, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Michele Provenzano
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Giuseppe Conte
- Division of Nephrology, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Roberto Minutolo
- Division of Nephrology, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Luca De Nicola
- Division of Nephrology, University of Campania "Luigi Vanvitelli," Naples, Italy
| |
Collapse
|
6
|
Wang AYM, Elsurer Afsar R, Sussman-Dabach EJ, White JA, MacLaughlin H, Ikizler TA. Vitamin Supplement Use in Patients With CKD: Worth the Pill Burden? Am J Kidney Dis 2024; 83:370-385. [PMID: 37879527 DOI: 10.1053/j.ajkd.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 08/29/2023] [Accepted: 09/03/2023] [Indexed: 10/27/2023]
Abstract
All vitamins play essential roles in various aspects of body function and systems. Patients with chronic kidney disease (CKD), including those receiving dialysis, may be at increased risk of developing vitamin deficiencies due to anorexia, poor dietary intake, protein energy wasting, restricted diet, dialysis loss, or inadequate sun exposure for vitamin D. However, clinical manifestations of most vitamin deficiencies are usually subtle or undetected in this population. Testing for circulating levels is not undertaken for most vitamins except folate, B12, and 25-hydroxyvitamin D because assays may not be available or may be costly to perform and do not always correlate with body stores. The last systematic review through 2016 was performed for the Kidney Disease Outcome Quality Initiative (KDOQI) 2020 Nutrition Guideline update, so this article summarizes the more recent evidence. We review the use of vitamins supplementation in the CKD population. To date there have been no randomized trials to support the benefits of any vitamin supplementation for kidney, cardiovascular, or patient-centered outcomes. The decision to supplement water-soluble vitamins should be individualized, taking account the patient's dietary intake, nutritional status, risk of vitamins deficiency/insufficiency, CKD stage, comorbid status, and dialysis loss. Nutritional vitamin D deficiency should be corrected, but the supplementation dose and formulation need to be personalized, taking into consideration the degree of 25-hydroxyvitamin D deficiency, parathyroid hormone levels, CKD stage, and local formulation. Routine supplementation of vitamins A and E is not supported due to potential toxicity. Although more trial data are required to elucidate the roles of vitamin supplementation, all patients with CKD should undergo periodic assessment of dietary intake and aim to receive various vitamins through natural food sources and a healthy eating pattern that includes vitamin-dense foods.
Collapse
Affiliation(s)
- Angela Yee-Moon Wang
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, People's Republic of China.
| | - Rengin Elsurer Afsar
- Department of Nephrology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey; Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Jennifer A White
- California State University at Northridge, Northridge, California
| | - Helen MacLaughlin
- School of Exercise and Nutrition Sciences, Queensland University of Technology, Brisbane, Australia; Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - T Alp Ikizler
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt O'Brien Kidney Center, Nashville, Tennessee; Tennessee Valley Healthcare System, Nashville VA Medical Center, Nashville, Tennessee
| |
Collapse
|
7
|
Chazot C, Steiber A, Kopple JD. Vitamin Needs and Treatment for Chronic Kidney Disease Patients. J Ren Nutr 2023; 33:S21-S29. [PMID: 36182060 DOI: 10.1053/j.jrn.2022.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/20/2022] [Accepted: 09/21/2022] [Indexed: 11/11/2022] Open
Abstract
This paper summarizes the biochemistry, metabolism, and dietary needs of vitamins in patients with chronic kidney disease (CKD) and kidney transplant recipients. Evidence indicates that the dietary intake, in vivo synthesis, urinary excretion or metabolism of different vitamins may be substantially altered in kidney failure. There are discrepancies in vitamin status assessment depending on whether the assay is functional or measuring the blood vitamin level. Whether vitamin supplements should be routinely prescribed for patients with CKD is controversial. Because low dietary intake and compounds that interfere with vitamin activity are not uncommon in patients with CKD, and water-soluble vitamin supplements appear safe and not costly, the authors recommend that supplements of the water-soluble vitamins should be routinely offered to these individuals. More research is needed to assess vitamin nutrition and function and to determine the daily vitamin needs for all patients with CKD.
Collapse
Affiliation(s)
- Charles Chazot
- AURA Paris, Ivry sur Seine, France; INI-CRCT Network (Investigation Network Initiative-Cardiovascular and Renal Clinical Trialists), Nancy, France.
| | - Alison Steiber
- Academy of Nutrition and Dietetics Research, International and Scientific Affairs, Chicago, Illinois
| | - Joel D Kopple
- Division of Nephrology and Hypertension, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California; David Geffen School of Medicine at UCLA, UCLA Fielding School of Public Health, Los Angeles, California
| |
Collapse
|
8
|
Kintu C, Soremekun O, Kamiza AB, Kalungi A, Mayanja R, Kalyesubula R, Bagaya S B, Jjingo D, Fabian J, Gill D, Nyirenda M, Nitsch D, Chikowore T, Fatumo S. The causal effects of lipid traits on kidney function in Africans: bidirectional and multivariable Mendelian-randomization study. EBioMedicine 2023; 90:104537. [PMID: 37001235 PMCID: PMC10070509 DOI: 10.1016/j.ebiom.2023.104537] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 03/08/2023] [Accepted: 03/08/2023] [Indexed: 03/31/2023] Open
Abstract
BACKGROUND Observational studies have investigated the effect of serum lipids on kidney function, but these findings are limited by confounding, reverse causation and have reported conflicting results. Mendelian randomization (MR) studies address this confounding problem. However, they have been conducted mostly in European ancestry individuals. We, therefore, set out to investigate the effect of lipid traits on the estimated glomerular filtration rate (eGFR) based on serum creatinine in individuals of African ancestry. METHODS We used the two-sample and multivariable Mendelian randomization (MVMR) approaches; in which instrument variables (IV's) for the predictor (lipid traits) were derived from summary-level data of a meta-analyzed African lipid GWAS (MALG, n = 24,215) from the African Partnership for Chronic Disease Research (APCDR) (n = 13,612) & the Africa Wits-IN-DEPTH partnership for Genomics studies (AWI-Gen) dataset (n = 10,603). The outcome IV's were computed from the eGFR summary-level data of African-ancestry individuals within the Million Veteran Program (n = 57,336). A random-effects inverse variance method was used in our primary analysis, and pleiotropy was adjusted for using robust and penalized sensitivity testing. The lipid predictors for the MVMR were high-density lipoprotein (HDL) cholesterol, low-density lipoprotein (LDL) cholesterol, and triglycerides (TG). FINDINGS We found a significant causal association between genetically predicted low-density lipoprotein (LDL) cholesterol and eGFR in African ancestry individuals β = 1.1 (95% CI [0.411-1.788]; p = 0.002). Similarly, total cholesterol (TC) showed a significant causal effect on eGFR β = 1.619 (95% CI [0.412-2.826]; p = 0.009). However, the IVW estimate showed that genetically predicted HDL-C β = -0.164, (95% CI = [-1.329 to 1.00]; p = 0.782), and TG β = -0.934 (CI = [-2.815 to 0.947]; p = 0.33) were not significantly causally associated with the risk of eGFR. In the multivariable analysis inverse-variance weighted (MVIVW) method, there was evidence for a causal association between LDL and eGFR β = 1.228 (CI = [0.477-1.979]; p = 0.001). A significant causal effect of Triglycerides (TG) on eGFR in the MVIVW analysis β = -1.3 ([-2.533 to -0.067]; p = 0.039) was observed as well. All the causal estimates reported reflect a unit change in the outcome per a 1 SD increase in the exposure. HDL showed no evidence of a significant causal association with eGFR in the MVIVW method (β = -0.117 (95% CI [-1.252 to 0.018]; p = 0.840)). We found no evidence of a reverse causal impact of eGFR on serum lipids. All our sensitivity analyses indicated no strong evidence of pleiotropy or heterogeneity between our instrumental variables for both the forward and reverse MR analysis. INTERPRETATION In this African ancestry population, genetically predicted higher LDL-C and TC are causally associated with higher eGFR levels, which may suggest that the relationship between LDL, TC and kidney function may be U-shaped. And as such, lowering LDL_C does not necessarily improve risk of kidney disease. This may also imply the reason why LDL_C is seen to be a poorer predictor of kidney function compared to HDL. In addition, this further supports that more work is warranted to confirm the potential association between lipid traits and risk of kidney disease in individuals of African Ancestry. FUNDING Wellcome (220740/Z/20/Z).
Collapse
Affiliation(s)
- Christopher Kintu
- The African Computational Genomics (TACG) Research Group, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda; Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda; MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - Opeyemi Soremekun
- The African Computational Genomics (TACG) Research Group, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda; MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - Abram B Kamiza
- The African Computational Genomics (TACG) Research Group, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - Allan Kalungi
- The African Computational Genomics (TACG) Research Group, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda; MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - Richard Mayanja
- The African Computational Genomics (TACG) Research Group, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda; MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - Robert Kalyesubula
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda; MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - Bernard Bagaya S
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Daudi Jjingo
- African Center of Excellence in Bioinformatics (ACE-B), Makerere University, Kampala 10101, Uganda
| | - June Fabian
- Medical Research Council/Wits University Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Wits Donald Gordon Medical Centre, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK; Chief Scientific Advisor Office, Research and Early Development, Novo Nordisk, Copenhagen, Denmark
| | - Moffat Nyirenda
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda; Department of Non-Communicable Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Dorothea Nitsch
- Department of Non-Communicable Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Tinashe Chikowore
- MRC/Wits Developmental Pathways for Health Research Unit, Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Segun Fatumo
- The African Computational Genomics (TACG) Research Group, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda; MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda; Department of Non-Communicable Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
9
|
Jeong SY, Wee CC, Kovell LC, Plante TB, Miller ER, Appel LJ, Mukamal KJ, Juraschek SP. Effects of Diet on 10-Year Atherosclerotic Cardiovascular Disease Risk (from the DASH Trial). Am J Cardiol 2023; 187:10-17. [PMID: 36459731 PMCID: PMC10122756 DOI: 10.1016/j.amjcard.2022.10.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/29/2022] [Accepted: 10/06/2022] [Indexed: 12/02/2022]
Abstract
Although modern risk estimators, such as the American College of Cardiology/American Heart Association Pooled Cohort Equation, play a central role in the decisions of patients to start pharmacologic therapy to prevent atherosclerotic cardiovascular disease (ASCVD), there is limited evidence to inform expectations for 10-year ASCVD risk reduction from established lifestyle interventions. Using data from the original DASH (Dietary Approaches to Stop Hypertension) trial, we determined the effects of adopting the DASH diet on 10-year ASCVD risk compared with adopting a control or a fruits and vegetables (F/V) diet. The DASH trial included 459 adults aged 22 to 75 years without CVD and not taking antihypertensive or diabetes mellitus medications, who were randomized to controlled feeding of a control diet, an F/V diet, or the DASH diet for 8 weeks. We determined 10-year ASCVD risk with the American College of Cardiology/American Heart Association Pooled Cohort Equation based on blood pressure and lipids measured before and after the 8-week intervention. Compared with the control diet, the DASH and F/V diets changed 10-year ASCVD risk by -10.3% (95% confidence interval [CI] -14.4 to -5.9) and -9.9% (95% CI -14.0 to -5.5) respectively; these effects were more pronounced in women and Black adults. There was no difference between the DASH and F/V diets (-0.4%, 95% CI -6.9 to 6.5). ASCVD reductions attributable to the difference in systolic blood pressure alone were -14.6% (-17.3 to -11.7) with the DASH diet and -7.9% (-10.9 to -4.8) with the F/V diet, a net relative advantage of 7.2% greater relative reduction from DASH compared with F/V. This was offset by the effects on high-density lipoprotein of the DASH diet, which increased 10-year ASCVD by 8.8% (5.5 to 12.3) compared with the more neutral effect of the F/V diet of -1.9% (-5.0 to 1.2). In conclusion, compared with a typical American diet, the DASH and F/V diets reduced 10-year ASCVD risk scores by about 10% over 8 weeks. These findings are informative for counseling patients on both choices of diet and expectations for 10-year ASCVD risk reduction.
Collapse
Affiliation(s)
- Sun Young Jeong
- Department of Medicine, American College of Physicians, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Christina C Wee
- Department of Medicine, American College of Physicians, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Lara C Kovell
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts
| | - Timothy B Plante
- Department of Medicine, Larner College of Medicine at the University of Vermont, Burlington, Vermont
| | - Edgar R Miller
- Department of Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Lawrence J Appel
- Department of Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Kenneth J Mukamal
- Department of Medicine, American College of Physicians, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Stephen P Juraschek
- Department of Medicine, American College of Physicians, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
10
|
Zhong O, Wang J, Tan Y, Lei X, Tang Z. Effects of NAD+ precursor supplementation on glucose and lipid metabolism in humans: a meta-analysis. Nutr Metab (Lond) 2022; 19:20. [PMID: 35303905 PMCID: PMC8932245 DOI: 10.1186/s12986-022-00653-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/21/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This meta-analysis was performed to investigate the effects of nicotinamide adenine dinucleotide (NAD+) precursor supplementation on glucose and lipid metabolism in human body. METHODS PubMed, Embase, CENTRAL, Web of Science, Scopus databases were searched to collect clinical studies related to the supplement of NAD+ precursor from inception to February 2021. Then the retrieved documents were screened, the content of the documents that met the requirements was extracted. Meta-analysis and quality evaluation was performed detection were performed using RevMan5.4 software. Stata16 software was used to detect publication bias, Egger and Begg methods were mainly used. The main research terms of NAD+ precursors were Nicotinamide Riboside (NR), Nicotinamide Mononucleotide (NMN), Nicotinic Acid (NA), Nicotinamide (NAM). The changes in the levels of triglyceride (TG), total cholesterol (TC), low-density lipoprotein (LDL), high-density lipoprotein (HDL), and fasting blood glucose were mainly concerned. RESULTS A total of 40 articles were included in the meta-analysis, with a sample of 14,750 cases, including 7406 cases in the drug group and 7344 cases in the control group. The results of meta-analysis showed that: NAD+ precursor can significantly reduce TG level (SMD = - 0.35, 95% CI (- 0.52, - 0.18), P < 0.0001), and TC (SMD = - 0.33, 95% CI (- 0.51, - 0.14), P = 0.0005), and LDL (SMD = - 0.38, 95% CI (- 0.50, - 0.27), P < 0.00001), increase HDL level (SMD = 0.66, 95% CI (0.56, 0.76), P < 0.00001), and plasma glucose level in the patients (SMD = 0.27, 95% CI (0.12, 0.42), P = 0.0004). Subgroup analysis showed that supplementation of NA had the most significant effect on the levels of TG, TC, LDL, HDL and plasma glucose. CONCLUSIONS In this study, a meta-analysis based on currently published clinical trials with NAD+ precursors showed that supplementation with NAD+ precursors improved TG, TC, LDL, and HDL levels in humans, but resulted in hyperglycemia, compared with placebo or no treatment. Among them, NA has the most significant effect on improving lipid metabolism. In addition, although NR and NAM supplementation had no significant effect on improving human lipid metabolism, the role of NR and NAM could not be directly denied due to the few relevant studies at present. Based on subgroup analysis, we found that the supplement of NAD+ precursors seems to have little effect on healthy people, but it has a significant beneficial effect on patients with cardiovascular disease and dyslipidemia. Due to the limitation of the number and quality of included studies, the above conclusions need to be verified by more high-quality studies.
Collapse
Affiliation(s)
- Ou Zhong
- Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jinyuan Wang
- Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yongpeng Tan
- Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xiaocan Lei
- Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Zhihan Tang
- Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
11
|
Chu X, Raju RP. Regulation of NAD + metabolism in aging and disease. Metabolism 2022; 126:154923. [PMID: 34743990 PMCID: PMC8649045 DOI: 10.1016/j.metabol.2021.154923] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/17/2021] [Accepted: 10/25/2021] [Indexed: 01/03/2023]
Abstract
More than a century after discovering NAD+, information is still evolving on the role of this molecule in health and diseases. The biological functions of NAD+ and NAD+ precursors encompass pathways in cellular energetics, inflammation, metabolism, and cell survival. Several metabolic and neurological diseases exhibit reduced tissue NAD+ levels. Significantly reduced levels of NAD+ are also associated with aging, and enhancing NAD+ levels improved healthspan and lifespan in animal models. Recent studies suggest a causal link between senescence, age-associated reduction in tissue NAD+ and enzymatic degradation of NAD+. Furthermore, the discovery of transporters and receptors involved in NAD+ precursor (nicotinic acid, or niacin, nicotinamide, and nicotinamide riboside) metabolism allowed for a better understanding of their role in cellular homeostasis including signaling functions that are independent of their functions in redox reactions. We also review studies that demonstrate that the functional effect of niacin is partially due to the activation of its cell surface receptor, GPR109a. Based on the recent progress in understanding the mechanism and function of NAD+ and NAD+ precursors in cell metabolism, new strategies are evolving to exploit these molecules' pharmacological potential in the maintenance of metabolic balance.
Collapse
Affiliation(s)
- Xiaogang Chu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America
| | - Raghavan Pillai Raju
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States of America.
| |
Collapse
|
12
|
de Araújo Lira AL, de Fátima Mello Santana M, de Souza Pinto R, Minanni CA, Iborra RT, de Lima AMS, Correa-Giannella ML, Passarelli M, Queiroz MS. Serum albumin modified by carbamoylation impairs macrophage cholesterol efflux in diabetic kidney disease. J Diabetes Complications 2021; 35:107969. [PMID: 34183248 DOI: 10.1016/j.jdiacomp.2021.107969] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 05/05/2021] [Accepted: 05/23/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND AIMS Abnormalities in lipid metabolism, accumulation of uremic toxins and advanced glycation end products may contribute to worsening atherosclerosis. This study characterized the glycation and carbamoylation profile of serum albumin isolated from individuals with diabetic kidney disease and its influence on cholesterol efflux. MATERIAL AND METHODS 49 patients with type 2 diabetes (T2DM) and different eGFR evaluated glycation and carbamoylation profile by measurement of carboxymethyl lysine (CML) and carbamoylated proteins (CBL) in plasma by ELISA, homocitrulline (HCit) in plasma by colorimetry. In the isolated albumins, we quantified CBL (ELISA) and total AGE and pentosidine by fluorescence. Macrophages were treated with albumin isolated, and 14C-Cholesterol efflux mediated by HDL2 or HDL3 was measured. Kruskal-Wallis test, Jonckheere-Terpstra test and Brunner's posttest were used for comparisons among groups. RESULTS Determination of CML, HCit, CBL in plasma, as total AGE and pentosidine in albumins, did not differ between groups; however, CBL in the isolated albumins was higher in the more advanced stages of CKD (p=0.0414). There was reduction in the 14C-cholesterol efflux after treatment for 18h with albumin isolated from patients with eGFR<60mL/min/1.73m2 compared with control group mediated by HDL2 (p=0.0288) and HDL3 (p<0.0001), as well as when compared with eGFR ≥60mL/min/1.73m2 per HDL2 (p=0.0001) and HDL3 (p<0.0001). Treatment for 48h showed that eGFR<15mL/min/1.73m2 had a lower percentage of 14C-cholesterol efflux mediated by HDL2 compared to control and other CKD groups (p=0.0274). CONCLUSIONS Albumins isolated from individuals with T2DM and eGFR<60mL/min/1.73m2 suffer greater carbamoylation, and they impair the cholesterol efflux mediated by HDL2 and HDL3. In turn, this could promote lipids accumulation in macrophages and disorders in reverse cholesterol transport.
Collapse
Affiliation(s)
| | | | - Raphael de Souza Pinto
- Lipids Laboratory (LIM 10), Faculty of Medical Sciences, University of Sao Paulo, Brazil
| | - Carlos André Minanni
- Lipids Laboratory (LIM 10), Faculty of Medical Sciences, University of Sao Paulo, Brazil
| | - Rodrigo Tallada Iborra
- Lipids Laboratory (LIM 10), Faculty of Medical Sciences, University of Sao Paulo, Brazil; Sao Judas Tadeu University, Sao Paulo, Brazil
| | - Adriana Machado Saldiba de Lima
- Lipids Laboratory (LIM 10), Faculty of Medical Sciences, University of Sao Paulo, Brazil; Sao Judas Tadeu University, Sao Paulo, Brazil
| | - Maria Lúcia Correa-Giannella
- Laboratory of Carbohydrates and Radioimuneassays (LIM 18), Clinical Hospital, Medical School, University of Sao Paulo, Sao Paulo, Brazil; Department of Graduation in Medicine, Nove de Julho University (Uninove), Sao Paulo, Brazil
| | - Marisa Passarelli
- Lipids Laboratory (LIM 10), Faculty of Medical Sciences, University of Sao Paulo, Brazil; Department of Graduation in Medicine, Nove de Julho University (Uninove), Sao Paulo, Brazil
| | - Márcia Silva Queiroz
- Endocrinology Division, Internal Medicine Department, University of Sao Paulo Medical School, Sao Paulo, Brazil; Department of Graduation in Medicine, Nove de Julho University (Uninove), Sao Paulo, Brazil.
| |
Collapse
|
13
|
Rasheed H, Zheng J, Rees J, Sanderson E, Thomas L, Richardson TG, Fang S, Bekkevold OJ, Stovner EB, Gabrielsen ME, Skogholt AH, Romundstad S, Brumpton B, Hallan S, Willer C, Burgess S, Hveem K, Davey Smith G, Gaunt TR, Åsvold BO. The causal effects of serum lipids and apolipoproteins on kidney function: multivariable and bidirectional Mendelian-randomization analyses. Int J Epidemiol 2021; 50:1569-1579. [PMID: 34151951 PMCID: PMC8580277 DOI: 10.1093/ije/dyab014] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2021] [Indexed: 12/03/2022] Open
Abstract
Background The causal nature of the observed associations between serum lipids and apolipoproteins and kidney function are unclear. Methods Using two-sample and multivariable Mendelian randomization (MR), we examined the causal effects of serum lipids and apolipoproteins on kidney function, indicated by the glomerular-filtration rate estimated using creatinine (eGFRcrea) or cystatin C (eGFRcys) and the urinary albumin-to-creatinine ratio (UACR). We obtained lipid- and apolipoprotein-associated genetic variants from the Global Lipids Genetics Consortium (n = 331 368) and UK Biobank (n = 441 016), respectively, and kidney-function markers from the Trøndelag Health Study (HUNT; n = 69 736) and UK Biobank (n = 464 207). The reverse causal direction was examined using variants associated with kidney-function markers selected from recent genome-wide association studies. Results There were no strong associations between genetically predicted lipid and apolipoprotein levels with kidney-function markers. Some, but inconsistent, evidence suggested a weak association of higher genetically predicted atherogenic lipid levels [indicated by low-density lipoprotein cholesterol (LDL-C), triglycerides and apolipoprotein B] with increased eGFR and UACR. For high-density lipoprotein cholesterol (HDL-C), results differed between eGFRcrea and eGFRcys, but neither analysis suggested substantial effects. We found no clear evidence of a reverse causal effect of eGFR on lipid or apolipoprotein traits, but higher UACR was associated with higher LDL-C, triglyceride and apolipoprotein B levels. Conclusion Our MR estimates suggest that serum lipid and apolipoprotein levels do not cause substantial changes in kidney function. A possible weak effect of higher atherogenic lipids on increased eGFR and UACR warrants further investigation. Processes leading to higher UACR may lead to more atherogenic lipid levels.
Collapse
Affiliation(s)
- Humaira Rasheed
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Department of Chemistry, University of Engineering and Technology, Lahore, Pakistan
- Corresponding author. K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway. E-mail:
| | - Jie Zheng
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Jessica Rees
- Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Eleanor Sanderson
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Laurent Thomas
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Tom G Richardson
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Si Fang
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ole-Jørgen Bekkevold
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Endre Bakken Stovner
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Maiken Elvestad Gabrielsen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anne Heidi Skogholt
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Solfrid Romundstad
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Internal Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Ben Brumpton
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Department of Thoracic Medicine, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Stein Hallan
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Nephrology, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Cristen Willer
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Stephen Burgess
- Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, UK
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, University of Bristol, Bristol, UK
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, University of Bristol, Bristol, UK
| | - Bjørn Olav Åsvold
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, Clinic of Medicine, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
14
|
Mathew RO, Rosenson RS, Lyubarova R, Chaudhry R, Costa SP, Bangalore S, Sidhu MS. Concepts and Controversies: Lipid Management in Patients with Chronic Kidney Disease. Cardiovasc Drugs Ther 2021; 35:479-489. [PMID: 32556851 DOI: 10.1007/s10557-020-07020-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains an important contributor of morbidity and mortality in patients with chronic kidney disease (CKD). CKD is recognized as an important risk enhancer that identifies patients as candidates for more intensive low-density lipoprotein (LDL) cholesterol lowering. However, there is controversy regarding the efficacy of lipid-lowering therapy, especially in patients on dialysis. Among patients with CKD, not yet on dialysis, there is clinical trial evidence for the use of statins with or without ezetimibe to reduce ASCVD events. Newer cholesterol lowering agents have been introduced for the management of hyperlipidemia to reduce ASCVD, but these therapies have not been tested in the CKD population except in secondary analyses of patients with primarily CKD stage 3. This review summarizes the role of hyperlipidemia in ASCVD and treatment strategies for hyperlipidemia in the CKD population.
Collapse
Affiliation(s)
- Roy O Mathew
- Columbia V.A. Health Care System, 6439 Garners Ferry Road, Columbia, SC, 29209, USA.
- University of South Carolina School of Medicine, Columbia, SC, USA.
| | | | | | | | | | | | - Mandeep S Sidhu
- Albany Medical College and Albany Medical Center, Albany, NY, USA
| |
Collapse
|
15
|
Lee SM, Son YK, Kim SE, Kim YH, Park Y, An WS. Effect of pravastatin on erythrocyte membrane fatty acid contents in patients with chronic kidney disease. Kidney Res Clin Pract 2021; 40:392-400. [PMID: 34078022 PMCID: PMC8476301 DOI: 10.23876/j.krcp.20.247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/21/2021] [Indexed: 12/03/2022] Open
Abstract
Background Statin treatment has decreased the risk of cardiovascular events in patients with chronic kidney disease (CKD). Erythrocyte membrane oleic acid level is higher in patients with acute coronary syndrome. This study aimed to evaluate the effect of pravastatin on the erythrocyte membrane fatty acid (FA) contents in patients with CKD. Methods Sixty-two patients were enrolled from January 2017 to March 2019 (NCT02992548). Pravastatin was initially administered at a dose of 20 mg for 24 weeks. The pravastatin dose was increased to 40 mg after 12 weeks if it was necessary to control dyslipidemia. The primary outcome was change in erythrocyte membrane FA, including oleic acid, after pravastatin treatment for 24 weeks. Results Forty-five patients finished this study, and there was no adverse effect related to pravastatin. Compared with baseline, total cholesterol and low-density lipoprotein cholesterol levels were significantly decreased after pravastatin treatment. Compared with baseline, saturated FA, oleic acid, and arachidonic acid levels were significantly increased and polyunsaturated FA and linoleic acid (LA) levels were significantly decreased after pravastatin treatment. There was also a decrease in eicosapentaenoic acid after pravastatin treatment in CKD patients with estimated glomerular filtration rate < 60 mL/min/1.73 m2. Conclusion Administration of pravastatin in patients with CKD leads to a decrease in FA known to be protective against the risk of CVD. Omega-3 FA or LA supplementation might be necessary to recover changes in erythrocyte membrane FA contents when pravastatin is used for treating dyslipidemia in patients with CKD.
Collapse
Affiliation(s)
- Su Mi Lee
- Department of Internal Medicine, Dong-A University, Busan, Republic of Korea
| | - Young Ki Son
- Department of Internal Medicine, Dong-A University, Busan, Republic of Korea
| | - Seong Eun Kim
- Department of Internal Medicine, Dong-A University, Busan, Republic of Korea
| | - Yeong Hoon Kim
- Department of Internal Medicine, Inje University Busan Paik Hospital, Busan, Republic of Korea
| | - Yongsoon Park
- Department of Food and Nutrition, Hanyang University, Seoul, Republic of Korea
| | - Won Suk An
- Department of Internal Medicine, Dong-A University, Busan, Republic of Korea
| |
Collapse
|
16
|
Qi YY, Yan L, Wang ZM, Wang X, Meng H, Li WB, Chen DC, Li M, Liu J, An ST. Comparative efficacy of pharmacological agents on reducing the risk of major adverse cardiovascular events in the hypertriglyceridemia population: a network meta-analysis. Diabetol Metab Syndr 2021; 13:15. [PMID: 33514420 PMCID: PMC7845128 DOI: 10.1186/s13098-021-00626-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/06/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Hypertriglyceridemia (HTG) is considered an independent risk factor for major adverse cardiovascular events (MACE). METHODS This study analyzed the effects of various agents on MACE risk reduction in HTG (serum triglyceride ≥ 150 mg/dl) populations by performing a network meta-analysis. We performed a frequentist network meta-analysis to conduct direct and indirect comparisons of interventions. PubMed, EMBASE, and the Cochrane library were searched for trials until Jul 6, 2020. Randomized controlled trials that reported MACE associated with agents in entire HTG populations or in subgroups were included. The primary outcome was MACE. RESULTS Of the 2005 articles screened, 21 trials including 56,471 patients were included in the analysis. The network meta-analysis results for MACE risk based on frequency data showed that eicosapentaenoic acid (EPA) (OR: 1.32; 95% CI 1.19-1.46), gemfibrozil (OR: 1.53; 95% CI 1.20-1.95), niacin plus clofibrate (OR: 2.00; 95% CI 1.23-3.25), pravastatin (OR: 1.32; 95% CI 1.15-1.52), simvastatin (OR: 2.38; 95% CI 1.55-3.66), and atorvastatin (OR: 0.55; 95% CI 0.37-0.82) significantly reduced the risk of MACE compared to the control conditions. In the subgroup analysis of HTG patients with triglycerides ≥ 200 mg/dL, bezafibrate (OR: 0.56; 95% CI 0.33-0.94), EPA (OR: 0.72; 95% CI 0.62-0.82), and pravastatin (OR: 1.33; 95% CI 1.01-1.75) significantly reduced the MACE risk. CONCLUSIONS Simvastatin had a clear advantage in reducing the risk of MACE in the entire HTG population analyzed in this meta-analysis. EPA, but not omega-3 fatty acid, was considered an effective HTG intervention. Among fibrates, gemfibrozil was most effective, though bezafibrate may significantly reduce the risk of MACE in populations with triglyceride levels of 200-300 mg/dL. Trial registration retrospectively registered in PROSPERO (CRD42020213705).
Collapse
Affiliation(s)
- Yan-Yan Qi
- Department of Cardiology, People's Hospital of Zhengzhou University No, 7, Weiwu Road, Zhengzhou, 450003, China
| | - Li Yan
- Department of Cardiology, Hongxing hospital, Hami, 839000, China
| | - Zhong-Min Wang
- Department of Cardiology, People's Hospital of Zhengzhou University No, 7, Weiwu Road, Zhengzhou, 450003, China
| | - Xi Wang
- Department of Cardiology, People's Hospital of Zhengzhou University No, 7, Weiwu Road, Zhengzhou, 450003, China
| | - Hua Meng
- Department of Cardiology, People's Hospital of Zhengzhou University No, 7, Weiwu Road, Zhengzhou, 450003, China
| | - Wen-Bo Li
- Department of Cardiology, People's Hospital of Zhengzhou University No, 7, Weiwu Road, Zhengzhou, 450003, China
| | - Dong-Chang Chen
- Department of Cardiology, People's Hospital of Zhengzhou University No, 7, Weiwu Road, Zhengzhou, 450003, China
| | - Meng Li
- Department of Cardiology, People's Hospital of Zhengzhou University No, 7, Weiwu Road, Zhengzhou, 450003, China
| | - Jun Liu
- Department of Cardiology, People's Hospital of Zhengzhou University No, 7, Weiwu Road, Zhengzhou, 450003, China
| | - Song-Tao An
- Department of Cardiology, People's Hospital of Zhengzhou University No, 7, Weiwu Road, Zhengzhou, 450003, China.
| |
Collapse
|
17
|
Schiano E, Annunziata G, Ciampaglia R, Iannuzzo F, Maisto M, Tenore GC, Novellino E. Bioactive Compounds for the Management of Hypertriglyceridemia: Evidence From Clinical Trials and Putative Action Targets. Front Nutr 2020; 7:586178. [PMID: 33330588 PMCID: PMC7734325 DOI: 10.3389/fnut.2020.586178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/26/2020] [Indexed: 01/22/2023] Open
Abstract
Hypertriglyceridemia refers to the presence of elevated concentrations of triglycerides (TG) in the bloodstream (TG >200 mg/dL). This lipid alteration is known to be associated with an increased risk of atherosclerosis, contributing overall to the onset of atherosclerotic cardiovascular disease (CVD). Guidelines for the management of hypertriglyceridemia are based on both lifestyle intervention and pharmacological treatment, but poor adherence, medication-related costs and side effects can limit the success of these interventions. For this reason, the search for natural alternative approaches to reduce plasma TG levels currently represents a hot research field. This review article summarizes the most relevant clinical trials reporting the TG-reducing effect of different food-derived bioactive compounds. Furthermore, based on the evidence obtained from in vitro studies, we provide a description and classification of putative targets of action through which several bioactive compounds can exert a TG-lowering effect. Future research may lead to investigations of the efficacy of novel nutraceutical formulations consisting in a combination of bioactive compounds which contribute to the management of plasma TG levels through different action targets.
Collapse
Affiliation(s)
| | | | | | - Fortuna Iannuzzo
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Maria Maisto
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Gian Carlo Tenore
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Ettore Novellino
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
18
|
Madsen CM, Varbo A, Nordestgaard BG. Novel Insights From Human Studies on the Role of High-Density Lipoprotein in Mortality and Noncardiovascular Disease. Arterioscler Thromb Vasc Biol 2020; 41:128-140. [PMID: 33232200 DOI: 10.1161/atvbaha.120.314050] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The vast majority of research about HDL (high-density lipoprotein) has for decades revolved around the possible role of HDL in atherosclerosis and its therapeutic potential within cardiovascular disease prevention; however, failures with therapies aimed at increasing HDL cholesterol has left questions as to what the role and function of HDL in human health and disease is. Recent observational studies have further shown that extreme high HDL cholesterol is associated with high mortality leading to speculations that HDL could in some instances be harmful. In addition, evidence from observational, and to a lesser extent genetic studies has emerged indicating that HDL might be associated with the development of other major noncardiovascular diseases, such as infectious disease, autoimmune disease, cancer, type 2 diabetes, kidney disease, and lung disease. In this review, we discuss (1) the association between extreme high HDL cholesterol and mortality and (2) the emerging human evidence linking HDL to several major diseases outside the realm of cardiovascular disease.
Collapse
Affiliation(s)
- Christian M Madsen
- Department of Clinical Biochemistry (C.M.M., A.V., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark.,The Copenhagen General Population Study (C.M.M., A.V., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (C.M.M., A.V., B.G.N.)
| | - Anette Varbo
- Department of Clinical Biochemistry (C.M.M., A.V., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark.,The Copenhagen General Population Study (C.M.M., A.V., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (C.M.M., A.V., B.G.N.)
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry (C.M.M., A.V., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark.,The Copenhagen General Population Study (C.M.M., A.V., B.G.N.), Herlev and Gentofte Hospital, Copenhagen University Hospital, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Denmark (C.M.M., A.V., B.G.N.).,The Copenhagen City Heart Study, Frederiksberg Hospital, Copenhagen University Hospital, Denmark (B.G.N.)
| |
Collapse
|
19
|
Lunney M, Ruospo M, Natale P, Quinn RR, Ronksley PE, Konstantinidis I, Palmer SC, Tonelli M, Strippoli GF, Ravani P. Pharmacological interventions for heart failure in people with chronic kidney disease. Cochrane Database Syst Rev 2020; 2:CD012466. [PMID: 32103487 PMCID: PMC7044419 DOI: 10.1002/14651858.cd012466.pub2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Approximately half of people with heart failure have chronic kidney disease (CKD). Pharmacological interventions for heart failure in people with CKD have the potential to reduce death (any cause) or hospitalisations for decompensated heart failure. However, these interventions are of uncertain benefit and may increase the risk of harm, such as hypotension and electrolyte abnormalities, in those with CKD. OBJECTIVES This review aims to look at the benefits and harms of pharmacological interventions for HF (i.e., antihypertensive agents, inotropes, and agents that may improve the heart performance indirectly) in people with HF and CKD. SEARCH METHODS We searched the Cochrane Kidney and Transplant Register of Studies through 12 September 2019 in consultation with an Information Specialist and using search terms relevant to this review. Studies in the Register are identified through searches of CENTRAL, MEDLINE, and EMBASE, conference proceedings, the International Clinical Trials Register (ICTRP) Search Portal and ClinicalTrials.gov. SELECTION CRITERIA We included randomised controlled trials of any pharmacological intervention for acute or chronic heart failure, among people of any age with chronic kidney disease of at least three months duration. DATA COLLECTION AND ANALYSIS Two authors independently screened the records to identify eligible studies and extracted data on the following dichotomous outcomes: death, hospitalisations, worsening heart failure, worsening kidney function, hyperkalaemia, and hypotension. We used random effects meta-analysis to estimate treatment effects, which we expressed as a risk ratio (RR) with 95% confidence intervals (CI). We assessed the risk of bias using the Cochrane tool. We applied the GRADE methodology to rate the certainty of evidence. MAIN RESULTS One hundred and twelve studies met our selection criteria: 15 were studies of adults with CKD; 16 studies were conducted in the general population but provided subgroup data for people with CKD; and 81 studies included individuals with CKD, however, data for this subgroup were not provided. The risk of bias in all 112 studies was frequently high or unclear. Of the 31 studies (23,762 participants) with data on CKD patients, follow-up ranged from three months to five years, and study size ranged from 16 to 2916 participants. In total, 26 studies (19,612 participants) reported disaggregated and extractable data on at least one outcome of interest for our review and were included in our meta-analyses. In acute heart failure, the effects of adenosine A1-receptor antagonists, dopamine, nesiritide, or serelaxin on death, hospitalisations, worsening heart failure or kidney function, hyperkalaemia, hypotension or quality of life were uncertain due to sparse data or were not reported. In chronic heart failure, the effects of angiotensin-converting enzyme inhibitors (ACEi) or angiotensin receptor blockers (ARB) (4 studies, 5003 participants: RR 0.85, 95% CI 0.70 to 1.02; I2 = 78%; low certainty evidence), aldosterone antagonists (2 studies, 34 participants: RR 0.61 95% CI 0.06 to 6.59; very low certainty evidence), and vasopressin receptor antagonists (RR 1.26, 95% CI 0.55 to 2.89; 2 studies, 1840 participants; low certainty evidence) on death (any cause) were uncertain. Treatment with beta-blockers may reduce the risk of death (any cause) (4 studies, 3136 participants: RR 0.69, 95% CI 0.60 to 0.79; I2 = 0%; moderate certainty evidence). Treatment with ACEi or ARB (2 studies, 1368 participants: RR 0.90, 95% CI 0.43 to 1.90; I2 = 97%; very low certainty evidence) had uncertain effects on hospitalisation for heart failure, as treatment estimates were consistent with either benefit or harm. Treatment with beta-blockers may decrease hospitalisation for heart failure (3 studies, 2287 participants: RR 0.67, 95% CI 0.43 to 1.05; I2 = 87%; low certainty evidence). Aldosterone antagonists may increase the risk of hyperkalaemia compared to placebo or no treatment (3 studies, 826 participants: RR 2.91, 95% CI 2.03 to 4.17; I2 = 0%; low certainty evidence). Renin inhibitors had uncertain risks of hyperkalaemia (2 studies, 142 participants: RR 0.86, 95% CI 0.49 to 1.49; I2 = 0%; very low certainty). We were unable to estimate whether treatment with sinus node inhibitors affects the risk of hyperkalaemia, as there were few studies and meta-analysis was not possible. Hyperkalaemia was not reported for the CKD subgroup in studies investigating other therapies. The effects of ACEi or ARB, or aldosterone antagonists on worsening heart failure or kidney function, hypotension, or quality of life were uncertain due to sparse data or were not reported. Effects of anti-arrhythmic agents, digoxin, phosphodiesterase inhibitors, renin inhibitors, sinus node inhibitors, vasodilators, and vasopressin receptor antagonists were very uncertain due to the paucity of studies. AUTHORS' CONCLUSIONS The effects of pharmacological interventions for heart failure in people with CKD are uncertain and there is insufficient evidence to inform clinical practice. Study data for treatment outcomes in patients with heart failure and CKD are sparse despite the potential impact of kidney impairment on the benefits and harms of treatment. Future research aimed at analysing existing data in general population HF studies to explore the effect in subgroups of patients with CKD, considering stage of disease, may yield valuable insights for the management of people with HF and CKD.
Collapse
Affiliation(s)
- Meaghan Lunney
- University of Calgary, Department of Community Health Sciences, 3330 Hospital Drive NW, Calgary, Alberta, Canada, T2N 4N1
| | - Marinella Ruospo
- The University of Sydney, Sydney School of Public Health, Sydney, Australia
- University of Bari, Department of Emergency and Organ Transplantation, Bari, Italy
| | - Patrizia Natale
- The University of Sydney, Sydney School of Public Health, Sydney, Australia
- University of Bari, Department of Emergency and Organ Transplantation, Bari, Italy
| | - Robert R Quinn
- University of Calgary, Department of Community Health Sciences, 3330 Hospital Drive NW, Calgary, Alberta, Canada, T2N 4N1
- Cumming School of Medicine, University of Calgary, Department of Medicine, Calgary, Canada
| | - Paul E Ronksley
- University of Calgary, Department of Community Health Sciences, 3330 Hospital Drive NW, Calgary, Alberta, Canada, T2N 4N1
| | - Ioannis Konstantinidis
- University of Pittsburgh Medical Center, Department of Medicine, 3459 Fifth Avenue, Pittsburgh, PA, USA, 15213
| | - Suetonia C Palmer
- Christchurch Hospital, University of Otago, Department of Medicine, Nephrologist, Christchurch, New Zealand
| | - Marcello Tonelli
- Cumming School of Medicine, University of Calgary, Department of Medicine, Calgary, Canada
| | - Giovanni Fm Strippoli
- The University of Sydney, Sydney School of Public Health, Sydney, Australia
- University of Bari, Department of Emergency and Organ Transplantation, Bari, Italy
- The Children's Hospital at Westmead, Cochrane Kidney and Transplant, Centre for Kidney Research, Westmead, NSW, Australia, 2145
| | - Pietro Ravani
- University of Calgary, Department of Community Health Sciences, 3330 Hospital Drive NW, Calgary, Alberta, Canada, T2N 4N1
- Cumming School of Medicine, University of Calgary, Department of Medicine, Calgary, Canada
| |
Collapse
|
20
|
Heine GH, Eller K, Stadler JT, Rogacev KS, Marsche G. Lipid-modifying therapy in chronic kidney disease: Pathophysiological and clinical considerations. Pharmacol Ther 2019; 207:107459. [PMID: 31863818 DOI: 10.1016/j.pharmthera.2019.107459] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/09/2019] [Indexed: 12/29/2022]
Abstract
Chronic kidney disease (CKD), which affects >10% of the population worldwide, is associated with a dramatically increased rate of cardiovascular disease (CVD). More people with CKD will die from CVD than develop end-stage renal disease with dialysis-dependency. However, the contribution of classical atherosclerotic cardiovascular risk factors is less evident than in the general population. Particularly, the relationship between dyslipidemia and CVD morbidity and mortality in CKD patients is not as evident as in the general population. While LDL cholesterol-lowering drugs such as statins significantly reduce the rate of cardiovascular events in the general population, their role in patients with end-stage renal disease has been questioned. This could be caused by a shift from atherosclerotic to non-atherosclerotic CVD in patients with advanced CKD, which cannot be effectively prevented by lipid-lowering drugs. In addition, many lines of evidence suggest that impaired renal function directly affects the metabolism, composition and functionality of lipoproteins, which may affect their responsiveness to pharmacological interventions. In this review, we highlight the challenges for the therapeutic application of lipid-lowering treatment strategies in CKD and discuss why treatment strategies used in the general population cannot be applied uncritically to CKD patients.
Collapse
Affiliation(s)
- Gunnar H Heine
- Agaplesion Markus Krankenhaus, Frankfurt, Germany; Saarland University Faculty of Medicine, Homburg, Germany.
| | - Kathrin Eller
- Department of Internal Medicine, Clinical Division of Nephrology, Medical University of Graz, Austria
| | - Julia T Stadler
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Austria
| | - Kyrill S Rogacev
- Internal Medicine II/Cardiology, Sana HANSE-Klinikum Wismar, Germany; Nephrology/Lipidology, B Braun - ViaMedis, MVZ Schwerin West, Germany
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Austria.
| |
Collapse
|
21
|
Lamprea-Montealegre JA, Staplin N, Herrington WG, Haynes R, Emberson J, Baigent C, de Boer IH. Apolipoprotein B, Triglyceride-Rich Lipoproteins, and Risk of Cardiovascular Events in Persons with CKD. Clin J Am Soc Nephrol 2019; 15:47-60. [PMID: 31831577 PMCID: PMC6946066 DOI: 10.2215/cjn.07320619] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 10/18/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND OBJECTIVES Triglyceride-rich lipoproteins may contribute to the high cardiovascular risk of patients with CKD. This study evaluated associations of apo-B and markers of triglyceride-rich lipoproteins with cardiovascular events in people with CKD. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Analyses were conducted in 9270 participants with CKD in the Study of Heart and Renal Protection (SHARP): 6245 not on dialysis (mean eGFR 26.5 ml/min per 1.73 m2), and 3025 on dialysis when recruited. Cox regression methods were used to evaluate associations of lipids with incident atherosclerotic and nonatherosclerotic vascular events, adjusting for demographics and clinical characteristics. Hazard ratios (HRs) were calculated per 1 SD higher level for apo-B, HDL cholesterol, LDL cholesterol, triglyceride-rich lipoprotein cholesterol (i.e., total cholesterol minus LDL cholesterol minus HDL cholesterol), non-HDL cholesterol, log triglyceride, and log ratio of triglyceride to HDL cholesterol. RESULTS During a median follow-up of 4.9 years (interquartile range, 4.0-5.5 years), 1406 participants experienced at least one atherosclerotic vascular event. In multivariable adjusted models, positive associations with atherosclerotic vascular events were observed for apo-B (HR per 1 SD, 1.19; 95% confidence interval, 1.12 to 1.27), triglycerides (1.06; 1.00 to 1.13), the ratio of triglyceride to HDL cholesterol (1.10; 1.03 to 1.18), and triglyceride-rich lipoprotein cholesterol (1.14; 1.05 to 1.25). By contrast, inverse associations with nonatherosclerotic vascular events were observed for each of these lipid markers: apo-B (HR per 1 SD, 0.92; 0.85 to 0.98), triglycerides (0.86; 0.81 to 0.92), the ratio of triglyceride to HDL cholesterol (0.88; 0.82 to 0.94), and triglyceride-rich lipoprotein cholesterol (0.85; 0.77 to 0.94). CONCLUSIONS Higher apo-B, triglycerides, ratio of triglyceride to HDL cholesterol, and triglyceride-rich lipoprotein cholesterol concentrations were associated with increased risk of atherosclerotic vascular events in CKD. Reducing triglyceride-rich lipoproteins using novel therapeutic agents could potentially lower the risk of atherosclerotic cardiovascular disease risk in the CKD population.
Collapse
Affiliation(s)
| | - Natalie Staplin
- Medical Research Council Population Health Research Unit and.,Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - William G Herrington
- Medical Research Council Population Health Research Unit and.,Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Richard Haynes
- Medical Research Council Population Health Research Unit and.,Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Jonathan Emberson
- Medical Research Council Population Health Research Unit and.,Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Colin Baigent
- Medical Research Council Population Health Research Unit and.,Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Ian H de Boer
- Kidney Research Institute.,Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| | | |
Collapse
|
22
|
Holmes MV, Davey Smith G. Challenges in Interpreting Multivariable Mendelian Randomization: Might "Good Cholesterol" Be Good After All? Am J Kidney Dis 2019; 71:149-153. [PMID: 29389382 DOI: 10.1053/j.ajkd.2017.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 10/11/2017] [Indexed: 01/29/2023]
Affiliation(s)
- Michael V Holmes
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, United Kingdom; Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; National Institute for Health Research, Oxford Biomedical Research Centre, Oxford University Hospital, Oxford, United Kingdom; Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom.
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Population Health Sciences, University of Bristol, Bristol, United Kingdom.
| |
Collapse
|
23
|
Bajaj A, Xie D, Cedillo-Couvert E, Charleston J, Chen J, Deo R, Feldman HI, Go AS, He J, Horwitz E, Kallem R, Rahman M, Weir MR, Anderson AH, Rader DJ. Lipids, Apolipoproteins, and Risk of Atherosclerotic Cardiovascular Disease in Persons With CKD. Am J Kidney Dis 2019; 73:827-836. [PMID: 30686529 PMCID: PMC6615056 DOI: 10.1053/j.ajkd.2018.11.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 11/26/2018] [Indexed: 01/06/2023]
Abstract
RATIONALE & OBJECTIVE A large residual risk for atherosclerotic cardiovascular disease (ASCVD) remains in the setting of chronic kidney disease (CKD) despite treatment with statins. We sought to evaluate the associations of lipid and apolipoprotein levels with risk for ASCVD in individuals with CKD. STUDY DESIGN Prospective cohort study. SETTINGS & PARTICIPANTS Adults aged 21 to 74 years with non-dialysis-dependent CKD at baseline enrolled in the Chronic Renal Insufficiency Cohort (CRIC) Study in 7 clinical study centers in the United States. PREDICTOR Baseline total cholesterol, non-high-density lipoprotein cholesterol (non-HDL-C), very low-density lipoprotein cholesterol (VLDL-C), triglycerides, low-density lipoprotein cholesterol (LDL-C), apolipoprotein B (Apo-B), HDL-C, and apolipoprotein AI (Apo-AI) values stratified into tertiles. OUTCOME A composite ASCVD event of myocardial infarction or ischemic stroke. ANALYTIC APPROACH Multivariable Cox proportional hazards regression to estimate the risk for ASCVD for each tertile of lipoprotein predictor. RESULTS Among 3,811 CRIC participants (mean age, 57.7 years; 41.8% white), there were 451 ASCVD events during a median follow-up of 7.9 years. There was increased ASCVD risk among participants with VLDL-C levels in the highest tertile (HR, 1.28; 95% CI, 1.01-1.64), Apo-B levels in the middle tertile (HR, 1.30; 95% CI, 1.03-1.64), HDL-C levels in the middle and lowest tertiles (HRs of 1.40 [95% CI, 1.08-1.83] and 1.77 [95% CI, 1.35-2.33], respectively), and Apo-AI levels in the middle and lowest tertiles (HRs of 1.77 [95% CI, 1.02-1.74] and 1.77 [95% CI, 1.36-2.32], respectively). LDL-C level was not significantly associated with the ASCVD outcome in this population (HR, 1.00 [95% CI, 0.77-1.30] for the highest tertile). LIMITATIONS Associations based on observational data do not permit inferences about causal associations. CONCLUSIONS Higher VLDL-C and Apo-B levels, as well as lower HDL-C and Apo-AI levels, are associated with increased risk for ASCVD. These findings support future investigations into pharmacologic targeting of lipoproteins beyond LDL-C, such as triglyceride-rich lipoproteins, to reduce residual risk for ASCVD among individuals with CKD.
Collapse
Affiliation(s)
- Archna Bajaj
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.
| | - Dawei Xie
- Department of Biostatistics and Epidemiology and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Esteban Cedillo-Couvert
- Division of Nephrology, Department of Medicine, University of Illinois College of Medicine at Chicago, Chicago, IL
| | - Jeanne Charleston
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Jing Chen
- Division of Nephrology and Hypertension, Department of Medicine, Tulane University School of Medicine, New Orleans, LA
| | - Rajat Deo
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Harold I Feldman
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Department of Biostatistics and Epidemiology and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Alan S Go
- Division of Research, Kaiser Permanente Northern California, Oakland, CA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - Edward Horwitz
- Division of Nephrology, MetroHealth Medical Center, Case Western Reserve University, Cleveland, OH
| | - Radhakrishna Kallem
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Mahboob Rahman
- Division of Nephrology and Hypertension, Case Western Reserve University, University Hospitals Case Medical Center, Cleveland, OH
| | - Matthew R Weir
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Amanda H Anderson
- Department of Biostatistics and Epidemiology and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - Daniel J Rader
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; The Penn Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Institute for Translational Medicine and Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
24
|
Streja E, Streja DA, Soohoo M, Kleine CE, Hsiung JT, Park C, Moradi H. Precision Medicine and Personalized Management of Lipoprotein and Lipid Disorders in Chronic and End-Stage Kidney Disease. Semin Nephrol 2019; 38:369-382. [PMID: 30082057 DOI: 10.1016/j.semnephrol.2018.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Precision medicine is an emerging field that calls for individualization of treatment strategies based on characteristics unique to each patient. In lipid management, current guidelines are driven mainly by clinical trial results that presently indicate that patients with non-dialysis-dependent chronic kidney disease (CKD) should be treated with a β-hydroxy β-methylglutaryl-CoA reductase inhibitor, also known as statin therapy. For patients with end-stage kidney disease (ESKD) being treated with hemodialysis, statin therapy has not been shown to successfully reduce poor outcomes in trials and therefore is not recommended. The two major guidelines dissent on whether statin therapy should be of moderate or high intensity in non-dialysis-dependent CKD patients, but often leave the prescribing clinician to make that decision. These decisions often are complicated by the increased concerns for adverse events such as myopathies in patients with advanced kidney disease and ESKD. In the future, there may be an opportunity to further identify CKD and ESKD patients who are more likely to benefit from lipid-modifying therapy as opposed to those who likely will suffer from its side effects using precision medicine tools. For now, data from genetics studies and subgroup analyses may provide insight for future research directions in this field and we review some of the work that has been published in this regard.
Collapse
Affiliation(s)
- Elani Streja
- Harold Simmons Center for Kidney Disease Research and Epidemiology, Division of Nephrology and Hypertension, University of California Irvine Medical Center, Orange, CA.; Nephrology Section, Tibor Rubin Veterans Affairs Medical Center, Long Beach, CA..
| | - Dan A Streja
- Division of Endocrinology, Diabetes and Metabolism, West Los Angeles VA Medical Center, Greater Los Angeles VA Healthcare System, Los Angeles, CA
| | - Melissa Soohoo
- Harold Simmons Center for Kidney Disease Research and Epidemiology, Division of Nephrology and Hypertension, University of California Irvine Medical Center, Orange, CA.; Nephrology Section, Tibor Rubin Veterans Affairs Medical Center, Long Beach, CA
| | - Carola-Ellen Kleine
- Harold Simmons Center for Kidney Disease Research and Epidemiology, Division of Nephrology and Hypertension, University of California Irvine Medical Center, Orange, CA.; Nephrology Section, Tibor Rubin Veterans Affairs Medical Center, Long Beach, CA
| | - Jui-Ting Hsiung
- Harold Simmons Center for Kidney Disease Research and Epidemiology, Division of Nephrology and Hypertension, University of California Irvine Medical Center, Orange, CA.; Nephrology Section, Tibor Rubin Veterans Affairs Medical Center, Long Beach, CA
| | - Christina Park
- Harold Simmons Center for Kidney Disease Research and Epidemiology, Division of Nephrology and Hypertension, University of California Irvine Medical Center, Orange, CA.; Nephrology Section, Tibor Rubin Veterans Affairs Medical Center, Long Beach, CA
| | - Hamid Moradi
- Harold Simmons Center for Kidney Disease Research and Epidemiology, Division of Nephrology and Hypertension, University of California Irvine Medical Center, Orange, CA.; Nephrology Section, Tibor Rubin Veterans Affairs Medical Center, Long Beach, CA
| |
Collapse
|
25
|
Zhang X, Wang B, Yang J, Wang J, Yu Y, Jiang C, Xie L, Song Y, Zhong B, Li Y, Liang M, Wang G, Li J, Zhang Y, Huo Y, Xu X, Qin X. Serum Lipids and Risk of Rapid Renal Function Decline in Treated Hypertensive Adults With Normal Renal Function. Am J Hypertens 2019; 32:393-401. [PMID: 30615058 DOI: 10.1093/ajh/hpz001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/16/2018] [Accepted: 01/03/2019] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND We aim to evaluate the effect of different lipids parameters, including triglycerides (TG), high-density lipoprotein cholesterol (HDL-C), the TG to HDL-C (TG:HDL-C) ratio, total cholesterol (TC), and low-density lipoprotein cholesterol (LDL-C), on the risk of rapid renal function decline and examine any possible effect modifiers in general hypertensive patients with normal renal function. METHODS A total of 12,549 hypertensive patients with estimated glomerular filtration rate (eGFR) ≥60 ml/min/1.73 m2 in the renal sub-study of the China Stroke Primary Prevention Trial were included in the analyses. The primary outcome was rapid renal function decline, defined as an average decline in eGFR ≥ 5 ml/min/1.73 m2 per year. RESULTS The median treatment duration was 4.4 years. After the full adjustment for TC, TG, HDL-C, and other major covariates, a significantly higher risk of rapid renal function decline was found in participants with higher TG [≥150 vs. <150 mg/dl, 7.7% vs. 5.5%; odds ratios (OR): 1.27; 95% confidence interval (CI): 1.06-1.51], higher TG:HDL-C ratio [≥2.7 (median) vs. <2.7, 7.7% vs. 5.0%; OR: 1.39; 95% CI: 1.14-1.71), lower TC (≥200 vs. <200 mg/dl, 6.0% vs. 7.0%; OR: 0.79; 95% CI: 0.67-0.93), or lower LDL-C levels (≥130 vs. <130 mg/dl, 6.1% vs. 7.0%; OR: 0.79; 95% CI: 0.67-0.94). Moreover, the increased risk of the primary outcome associated with elevated TG was particularly evident among individuals with lower total homocysteine levels [<12.4 (median) vs. ≥ 12.4 μmol/l, P interaction = 0.036]. CONCLUSIONS Higher TG and TG:HDL-C ratio were independent risk factors for rapid renal function decline in hypertensive adults with normal renal function.
Collapse
Affiliation(s)
- Xianglin Zhang
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Binyan Wang
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Institute for Biomedicine, Anhui Medical University, Hefei, China
| | - Juan Yang
- Department of Nephrology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jiancheng Wang
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yaren Yu
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chongfei Jiang
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liling Xie
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yun Song
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Biyan Zhong
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youbao Li
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Min Liang
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guobao Wang
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Yan Zhang
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Yong Huo
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Xiping Xu
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xianhui Qin
- Division of Nephrology, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
The effect of chronic kidney disease on lipid metabolism. Int Urol Nephrol 2018; 51:265-277. [DOI: 10.1007/s11255-018-2047-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/27/2018] [Indexed: 12/26/2022]
|
27
|
Probstfield JL, Boden WE, Anderson T, Branch K, Kashyap M, Fleg JL, Desvigne-Nickens P, McBride R, McGovern M. Cardiovascular outcomes during extended follow-up of the AIM-HIGH trial cohort. J Clin Lipidol 2018; 12:1413-1419. [DOI: 10.1016/j.jacl.2018.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 07/05/2018] [Accepted: 07/17/2018] [Indexed: 10/28/2022]
|
28
|
Abstract
Chronic kidney disease mineral and bone disorder (MBD) encompasses changes in mineral ion and vitamin D metabolism that are widespread in the setting of chronic kidney disease and end-stage renal disease. MBD components associate with cardiovascular disease in many epidemiologic studies. Through impacts on hypertension, activation of the renin-angiotensin-aldosterone system, vascular calcification, endothelial function, and cardiac remodeling and conduction, MBD may be a direct and targetable cause of cardiovascular disease. However, assessment and treatment of MBD is rife with challenges owing to biological tensions between its many components, such as calcium and phosphorus with their regulatory hormones fibroblast growth factor 23 and parathyroid hormone; fibroblast growth factor 23 with its co-receptor klotho; and vitamin D with control of calcium and phosphorus. These complex interactions between MBD components hinder the simple translation to clinical trials, which ultimately are needed to prove the benefits of treating MBD. Deeper investigation using precision medicine tools and principles, including genomics and individualized risk assessment and therapy, may help move the field closer toward clinical applications. This review provides a high-level overview of conventional and precision epidemiology in MBD, potential mechanisms of cardiovascular disease pathogenesis, and guiding therapeutic principles for established and emerging treatments.
Collapse
Affiliation(s)
- Joseph Lunyera
- Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Julia J Scialla
- Department of Medicine, Duke University School of Medicine, Durham, NC; Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC; Department of Medicine, Durham Veterans Affairs Medical Center, Durham, NC.
| |
Collapse
|
29
|
Navaneethan SD, Schold JD, Walther CP, Arrigain S, Jolly SE, Virani SS, Winkelmayer WC, Nally JV. High-density lipoprotein cholesterol and causes of death in chronic kidney disease. J Clin Lipidol 2018; 12:1061-1071.e7. [PMID: 29699917 DOI: 10.1016/j.jacl.2018.03.085] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 03/21/2018] [Accepted: 03/27/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND Recent data suggest a U-shaped association between high-density lipoprotein cholesterol (HDL-c) and death in chronic kidney disease (CKD). However, whether the increased mortality in patients with extreme levels is explained by specific causes of death remains unclear. OBJECTIVES We studied the associations between HDL-c and cause-specific deaths in CKD. METHODS We included 38,377 patients with estimated glomerular filtration rate 15-59 mL/min/1.73 m2. We classified deaths into 3 major categories: (1) cardiovascular; (2) malignant; and (3) noncardiovascular/nonmalignant causes. We fitted Cox regression models for overall mortality and separate competing risk models for each major cause of death category to evaluate their respective associations with categories of HDL-c (≤30, 31-40, 41-50 [referent], 51-60, >60 mg/dL). Separate analyses were conducted for men and women. RESULTS During a median follow-up of 4.5 years, 9665 patients died. After adjusting for relevant covariates, in both sexes, HDL-c 31 to 40 mg/dL and ≤30 mg/dL were associated with higher risk of all-cause mortality, cardiovascular mortality, malignancy-related deaths, and noncardiovascular/nonmalignancy-related deaths. HDL-c >60 mg/dL was associated with lower all-cause (hazard ratio: 0.75, 95% confidence interval: 0.69, 0.81), cardiovascular, malignancy-related, and noncardiovascular/nonmalignancy-related deaths among women but not in men. Similar results were noted when HDL-c was examined as a continuous measure. CONCLUSIONS In a non-dialysis-dependent CKD population, HDL-c ≤40 mg/dL was associated with risk of higher all-cause, cardiovascular, malignant, and noncardiovascular/nonmalignant mortality in men and women. HDL >60 mg/dL was associated with lower risk of all-cause, cardiovascular, malignant, and noncardiovascular/nonmalignant mortality in women but not in men.
Collapse
Affiliation(s)
- Sankar D Navaneethan
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Section of Nephrology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.
| | - Jesse D Schold
- Department of Nephrology and Hypertension, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Carl P Walther
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Susana Arrigain
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Stacey E Jolly
- Department of General Internal Medicine, Medicine Institute, Cleveland Clinic, Cleveland, OH, USA; Lerner College of Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Salim S Virani
- The Health Policy, Quality & Informatics Program, Michael E. DeBakey Veterans Affairs Medical Center Health Services Research and Development Center for Innovations & Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Wolfgang C Winkelmayer
- Selzman Institute for Kidney Health, Section of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Joseph V Nally
- Department of Nephrology and Hypertension, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA; Lerner College of Medicine, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
30
|
Malhotra R, Katz R, Hoofnagle A, Bostom A, Rifkin DE, Mcbride R, Probstfield J, Block G, Ix JH. The Effect of Extended Release Niacin on Markers of Mineral Metabolism in CKD. Clin J Am Soc Nephrol 2018; 13:36-44. [PMID: 29208626 PMCID: PMC5753310 DOI: 10.2215/cjn.05440517] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 10/03/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND OBJECTIVES Niacin downregulates intestinal sodium-dependent phosphate transporter 2b expression and reduces intestinal phosphate transport. Short-term studies have suggested that niacin lowers serum phosphate concentrations in patients with CKD and ESRD. However, the long-term effects of niacin on serum phosphate and other mineral markers are unknown. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS The Atherothrombosis Intervention in Metabolic Syndrome with Low HDL/High Triglycerides: Impact on Global Health Trial was a randomized, double-blind, placebo-controlled trial testing extended release niacin in persons with prevalent cardiovascular disease. We examined the effect of randomized treatment with niacin (1500 or 2000 mg) or placebo on temporal changes in markers of mineral metabolism in 352 participants with eGFR<60 ml/min per 1.73 m2 over 3 years. Changes in each marker were compared over time between the niacin and placebo arms using linear mixed effects models. RESULTS Randomization to niacin led to 0.08 mg/dl lower plasma phosphate concentrations per year of treatment compared with placebo (P<0.01) and 0.25 mg/dl lower mean phosphate 3 years after baseline (3.32 versus 3.57 mg/dl; P=0.03). In contrast, randomization to niacin was not associated with statistically significant changes in plasma intact fibroblast growth factor 23, parathyroid hormone, calcium, or vitamin D metabolites over 3 years. CONCLUSIONS The use of niacin over 3 years lowered serum phosphorous concentrations but did not affect other markers of mineral metabolism in participants with CKD.
Collapse
Affiliation(s)
- Rakesh Malhotra
- Division of Nephrology-Hypertension, Department of Medicine and
- Imperial Valley Family Care Medical Group, El Centro, California
| | - Ronit Katz
- Division of Nephrology, Department of Medicine
| | | | - Andrew Bostom
- Division of Hypertension and Kidney Diseases, Rhode Island Hospital, Providence, Rhode Island
| | - Dena E. Rifkin
- Division of Nephrology-Hypertension, Department of Medicine and
- Division of Preventive Medicine, Department of Family Medicine and Public Health, University of California, San Diego, California
- Nephrology Section, Veterans Affairs San Diego Healthcare System, San Diego, California
| | | | - Jeffrey Probstfield
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, Washington
| | | | - Joachim H. Ix
- Division of Nephrology-Hypertension, Department of Medicine and
- Division of Preventive Medicine, Department of Family Medicine and Public Health, University of California, San Diego, California
- Nephrology Section, Veterans Affairs San Diego Healthcare System, San Diego, California
| |
Collapse
|
31
|
|
32
|
Yin P, Zhou Y, Li B, Hong L, Chen W, Yu X. Effect of low and high HDL-C levels on the prognosis of lupus nephritis patients: a prospective cohort study. Lipids Health Dis 2017; 16:232. [PMID: 29212518 PMCID: PMC5719733 DOI: 10.1186/s12944-017-0622-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 11/22/2017] [Indexed: 11/16/2022] Open
Abstract
Background Few data has been available on the effect of serum HDL-C levels on the prognosis of lupus nephritis (LN) patients. The present study therefore aimed to explore the effect of serum HDL-C levels on LN patients. Methods We included 775 patients with follow-up information registered in an LN database between 1 January 2006 and 31 December 2011. The patients were divided into groups with low, intermediate and high HDL-C, according to NCEP ATPIII criteria. Cox regression analyses were used to explore the effects of HDL-C levels on end-stage renal disease (ESRD), all-cause mortality and cardiovascular disease (CVD) mortality. Results During a median follow-up of 56 months (3–206 months), 71 (9.2%) had ESRD. 84 (10.8%) deaths occurred, 17 (20.2%) of which were due to CVD. There was no statistically significant association of HDL-C category or continuous HDL-C levels with ESRD in the total cohort, but in subgroup analyses by eGFR, with each 0.1 mmol/L increase in HDL-C level, adjusted HRs for ESRD were 0.92 (95% CI: 0.83–1.04, P = 0.173) for eGFR ≥60 ml/min/1.73m2 and 1.11 (95% CI: 1.01–1.23, P = 0.036) for eGFR <60 ml/min/1.73m2. The effect of the interaction between eGFR category and serum HDL-C level on ESRD was statistically significant (β = −1.738, P = 0.005). Low HDL-C was associated with all-cause mortality (HR = 2.16, 95% CI: 1.06–4.40, P = 0.033) with intermediate HDL-C as reference category after adjusting for several variables. Conclusions Our results demonstrate that high HDL-C levels were associated with increased risk of ESRD in LN patients with advanced renal dysfunction. While low HDL-C levels were associated with increased risk of all-cause mortality in LN patients. Trial registration ClinicalTrials.gov Identifier: NCT03001973, 22 December 2016 retrospectively registered. Electronic supplementary material The online version of this article (10.1186/s12944-017-0622-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peiran Yin
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Key Laboratory of Nephrology, Ministry of Health and Guangdong Province, Guangzhou, Guangdong, 510080, China
| | - Ying Zhou
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Key Laboratory of Nephrology, Ministry of Health and Guangdong Province, Guangzhou, Guangdong, 510080, China
| | - Bin Li
- Clinical Research Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Lingyao Hong
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Key Laboratory of Nephrology, Ministry of Health and Guangdong Province, Guangzhou, Guangdong, 510080, China
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China. .,Key Laboratory of Nephrology, Ministry of Health and Guangdong Province, Guangzhou, Guangdong, 510080, China.
| | - Xueqing Yu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.,Key Laboratory of Nephrology, Ministry of Health and Guangdong Province, Guangzhou, Guangdong, 510080, China.,Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| |
Collapse
|
33
|
Lanktree MB, Thériault S, Walsh M, Paré G. HDL Cholesterol, LDL Cholesterol, and Triglycerides as Risk Factors for CKD: A Mendelian Randomization Study. Am J Kidney Dis 2017; 71:166-172. [PMID: 28754456 DOI: 10.1053/j.ajkd.2017.06.011] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 06/02/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND High-density lipoprotein (HDL) cholesterol, low-density lipoprotein (LDL) cholesterol, and triglyceride concentrations are heritable risk factors for vascular disease, but their role in the progression of chronic kidney disease (CKD) is unclear. STUDY DESIGN 2-sample Mendelian randomization analysis of data derived from the largest published lipid and CKD studies. SETTING & PARTICIPANTS Effect of independent genetic variants significantly associated with lipid concentrations was obtained from the Global Lipids Genetics Consortium (n=188,577), and the effect of these same variants on estimated glomerular filtration rate (eGFR), CKD (defined as eGFR<60mL/min/1.73m2), and albuminuria was obtained from the CKD Genetics Consortium (n=133,814). FACTOR Using conventional, multivariable, and Egger Mendelian randomization approaches, we assessed the causal association between genetically determined lipid concentrations and kidney traits. OUTCOME eGFR, dichotomous eGFR<60mL/min/1.73m2, and albuminuria. RESULTS In multivariable analysis, a 17-mg/dL higher HDL cholesterol concentration was associated with an 0.8% higher eGFR (95% CI, 0.4%-1.3%; P=0.004) and lower risk for eGFR<60mL/min/1.73m2 (OR, 0.85; 95% CI, 0.77-0.93; P<0.001), while Egger analysis showed no evidence of pleiotropy. There was no evidence for a causal relationship between LDL cholesterol concentration and any kidney disease measure. Genetically higher triglyceride concentrations appeared associated with higher eGFRs, but this finding was driven by a single pleiotropic variant in the glucokinase regulator gene (GCKR). After exclusion, genetically higher triglyceride concentration was not associated with any kidney trait. LIMITATIONS Individual patient-level phenotype and genotype information were unavailable. CONCLUSIONS 2-sample Mendelian randomization analysis of data from the largest lipid and CKD cohorts supports genetically higher HDL cholesterol concentration as causally associated with better kidney function. There was no association between genetically altered LDL cholesterol or triglyceride concentration and kidney function. Further analysis of CKD outcomes in HDL cholesterol intervention trials is warranted.
Collapse
Affiliation(s)
- Matthew B Lanktree
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| | - Sébastien Thériault
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Michael Walsh
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada; Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, Ontario, Canada
| | - Guillaume Paré
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Department of Clinical Epidemiology & Biostatistics, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
34
|
Schandelmaier S, Briel M, Saccilotto R, Olu KK, Arpagaus A, Hemkens LG, Nordmann AJ. Niacin for primary and secondary prevention of cardiovascular events. Cochrane Database Syst Rev 2017; 6:CD009744. [PMID: 28616955 PMCID: PMC6481694 DOI: 10.1002/14651858.cd009744.pub2] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Nicotinic acid (niacin) is known to decrease LDL-cholesterol, and triglycerides, and increase HDL-cholesterol levels. The evidence of benefits with niacin monotherapy or add-on to statin-based therapy is controversial. OBJECTIVES To assess the effectiveness of niacin therapy versus placebo, administered as monotherapy or add-on to statin-based therapy in people with or at risk of cardiovascular disease (CVD) in terms of mortality, CVD events, and side effects. SEARCH METHODS Two reviewers independently and in duplicate screened records and potentially eligible full texts identified through electronic searches of CENTRAL, MEDLINE, Embase, Web of Science, two trial registries, and reference lists of relevant articles (latest search in August 2016). SELECTION CRITERIA We included all randomised controlled trials (RCTs) that either compared niacin monotherapy to placebo/usual care or niacin in combination with other component versus other component alone. We considered RCTs that administered niacin for at least six months, reported a clinical outcome, and included adults with or without established CVD. DATA COLLECTION AND ANALYSIS Two reviewers used pre-piloted forms to independently and in duplicate extract trials characteristics, risk of bias items, and outcomes data. Disagreements were resolved by consensus or third party arbitration. We conducted random-effects meta-analyses, sensitivity analyses based on risk of bias and different assumptions for missing data, and used meta-regression analyses to investigate potential relationships between treatment effects and duration of treatment, proportion of participants with established coronary heart disease and proportion of participants receiving background statin therapy. We used GRADE to assess the quality of evidence. MAIN RESULTS We included 23 RCTs that were published between 1968 and 2015 and included 39,195 participants in total. The mean age ranged from 33 to 71 years. The median duration of treatment was 11.5 months, and the median dose of niacin was 2 g/day. The proportion of participants with prior myocardial infarction ranged from 0% (4 trials) to 100% (2 trials, median proportion 48%); the proportion of participants taking statin ranged from 0% (4 trials) to 100% (12 trials, median proportion 100%).Using available cases, niacin did not reduce overall mortality (risk ratio (RR) 1.05, 95% confidence interval (CI) 0.97 to 1.12; participants = 35,543; studies = 12; I2 = 0%; high-quality evidence), cardiovascular mortality (RR 1.02, 95% CI 0.93 to 1.12; participants = 32,966; studies = 5; I2 = 0%; moderate-quality evidence), non-cardiovascular mortality (RR 1.12, 95% CI 0.98 to 1.28; participants = 32,966; studies = 5; I2 = 0%; high-quality evidence), the number of fatal or non-fatal myocardial infarctions (RR 0.93, 95% CI 0.87 to 1.00; participants = 34,829; studies = 9; I2 = 0%; moderate-quality evidence), nor the number of fatal or non-fatal strokes (RR 0.95, 95% CI 0.74 to 1.22; participants = 33,661; studies = 7; I2 = 42%; low-quality evidence). Participants randomised to niacin were more likely to discontinue treatment due to side effects than participants randomised to control group (RR 2.17, 95% CI 1.70 to 2.77; participants = 33,539; studies = 17; I2 = 77%; moderate-quality evidence). The results were robust to sensitivity analyses using different assumptions for missing data. AUTHORS' CONCLUSIONS Moderate- to high-quality evidence suggests that niacin does not reduce mortality, cardiovascular mortality, non-cardiovascular mortality, the number of fatal or non-fatal myocardial infarctions, nor the number of fatal or non-fatal strokes but is associated with side effects. Benefits from niacin therapy in the prevention of cardiovascular disease events are unlikely.
Collapse
Affiliation(s)
- Stefan Schandelmaier
- McMaster UniversityDepartment of Health Research Methods, Evidence, and Impact1280 Main Street WestHamiltonONCanadaL8S4L8
| | - Matthias Briel
- University of BaselBasel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical ResearchBaselSwitzerland
| | - Ramon Saccilotto
- University of BaselBasel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical ResearchBaselSwitzerland
| | - Kelechi K Olu
- University of BaselBasel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical ResearchBaselSwitzerland
| | - Armon Arpagaus
- University of BaselBasel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical ResearchBaselSwitzerland
| | - Lars G Hemkens
- University of BaselBasel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical ResearchBaselSwitzerland
| | - Alain J Nordmann
- University of BaselBasel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical ResearchBaselSwitzerland
| | | |
Collapse
|
35
|
Papademetriou V, Lovato L, Tsioufis C, Cushman W, Applegate WB, Mottle A, Punthakee Z, Nylen E, Doumas M. Effects of High Density Lipoprotein Raising Therapies on Cardiovascular Outcomes in Patients with Type 2 Diabetes Mellitus, with or without Renal Impairment: The Action to Control Cardiovascular Risk in Diabetes Study. Am J Nephrol 2016; 45:136-145. [PMID: 27992863 DOI: 10.1159/000453626] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 11/14/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND The role of high density lipoprotein-raising interventions in addition to statin therapy in patients with diabetes remains controversial. Chronic kidney disease (CKD) is a strong modifier of cardiovascular (CV) outcomes. We therefore investigated the impact of CKD status at baseline on outcomes in patients with diabetes randomized to standard statin or statin plus fenofibrate treatment in the Action to Control Cardiovascular Risk in Diabetes (ACCORD) lipid trial. METHODS Among 5,464 participants in the ACCORD lipid trial, 3,554 (65%) were free of CKD at baseline, while 1,910 (35%) had mild to moderate CKD. Differences in CV outcomes during follow-up between CKD and non-CKD subgroups were examined. In addition, the effect of fenofibrate as compared to placebo on CV outcomes was examined for both subgroups. RESULTS All CV outcomes were 1.4-3 times higher among patients with CKD as compared to non-CKD patients. In patients with CKD, the addition of fenofibrate had no effect on any of the primary or secondary outcomes. In patients without CKD, however, the addition of fenofibrate was associated with a significant 36% reduction of CV mortality (hazards ratio [HR] 0.64; 95% CI 0.42-0.97; p value for treatment interaction <0.05) and 44% lower rate of fatal or non-fatal congestive heart failure (CHF; HR 0.56; 95% CI 0.37-0.84; p value treatment interaction <0.03). CONCLUSIONS For patients with type 2 diabetes at high CV risk but no CKD, fenofibrate therapy added to statin reduced the CV mortality and the rate of fatal and non-fatal CHF.
Collapse
|
36
|
Pascual V, Serrano A, Pedro-Botet J, Ascaso J, Barrios V, Millán J, Pintó X, Cases A. [Chronic kidney disease and dyslipidaemia]. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2016; 29:22-35. [PMID: 27863896 DOI: 10.1016/j.arteri.2016.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 07/18/2016] [Indexed: 12/18/2022]
Abstract
Chronic kidney disease (CKD) has to be considered as a high, or even very high risk cardiovascular risk condition, since it leads to an increase in cardiovascular mortality that continues to increase as the disease progresses. An early diagnosis of CKD is required, together with an adequate identification of the risk factors, in order to slow down its progression to more severe states, prevent complications, and to delay, whenever possible, the need for renal replacement therapy. Dyslipidaemia is a factor of the progression of CKD that increases the risk in developing atherosclerosis and its complications. Its proper control contributes to reducing the elevated cardiovascular morbidity and mortality presented by these patients. In this review, an assessment is made of the lipid-lowering therapeutic measures required to achieve to recommended objectives, by adjusting the treatment to the progression of the disease and to the characteristics of the patient. In CKD, it seems that an early and intensive intervention of the dyslipidaemia is a priority before there is a significant decrease in kidney function. Treatment with statins has been shown to be safe and effective in decreasing LDL-Cholesterol, and in the reduction of cardiovascular events in individuals with CKD, or after renal transplant, although there is less evidence in the case of dialysed patients.
Collapse
Affiliation(s)
| | - Adalberto Serrano
- Centro de Salud de Repelega, Osakidetza, Portugalete, Bizkaia, España
| | - Juan Pedro-Botet
- Unidad de Lípidos y Riesgo Vascular, Servicio de Endocrinología y Nutrición, Hospital del Mar, Universitat Autònoma de Barcelona, Barcelona, España
| | - Juan Ascaso
- Servicio de Endocrinología, Hospital Clínico Universitario, Universitat de València, Valencia, España
| | - Vivencio Barrios
- Servicio de Cardiología, Hospital Universitario Ramón y Cajal, Universidad de Alcalá de Henares, Madrid, España
| | - Jesús Millán
- Unidad de Lípidos, Servicio de Medicina Interna, Hospital Universitario de Bellvitge, Universitat de Barcelona, CIBERobn-ISCIII, Barcelona, España
| | - Xavier Pintó
- Servicio de Medicina Interna, Hospital Universitario Gregorio Marañón, Universidad Complutense de Madrid, Madrid, España
| | - Aleix Cases
- Servicio de Nefrología, Hospital Clínic, Universitat de Barcelona, Red de Investigación Cardiovascular (RIC), Barcelona, España
| |
Collapse
|
37
|
Bowe B, Xie Y, Xian H, Balasubramanian S, Zayed MA, Al-Aly Z. High Density Lipoprotein Cholesterol and the Risk of All-Cause Mortality among U.S. Veterans. Clin J Am Soc Nephrol 2016; 11:1784-1793. [PMID: 27515591 PMCID: PMC5053782 DOI: 10.2215/cjn.00730116] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/08/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND OBJECTIVES The relationship between HDL cholesterol and all-cause mortality in patients with kidney disease is not clear. We sought to characterize the relationship of HDL cholesterol and risk of death and examine the association by eGFR levels. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We built a cohort of 1,764,986 men who were United States veterans with at least one eGFR between October of 2003 and September of 2004 and followed them until September of 2013 or death. RESULTS Patients with low HDL cholesterol and low eGFR had a higher burden of comorbid illnesses. Over a median of 9.1 years (interquartile range, 7.7-9.4 years), 26,247 (40.1%), 109,222 (32.3%), 152,625 (29.2%), 113,785 (28.5%), and 139,803 (31.8%) participants with HDL cholesterol ≤25, >25 to <34, ≥34 to ≤42, >42 to <50, and ≥50 mg/dl died. In adjusted survival models, compared with the referent group of patients with low HDL cholesterol (≤25 mg/dl), intermediate HDL cholesterol levels (>25 to <34, ≥34 to ≤42, and >42 to <50 mg/dl) were associated with lower risk of death across all levels of eGFR. The lower risk was partially abrogated in those with high HDL cholesterol (≥50 mg/dl), and the risk of death was similar to the referent category among those with eGFR<30 or ≥90 ml/min per 1.73 m2. Analysis by HDL cholesterol deciles and spline analyses suggest that the relationship between HDL cholesterol and death follows a U-shaped curve. There was a significant interaction between eGFR and HDL cholesterol in that lower eGFR attenuated the salutary association of HDL cholesterol and risk of death (P for interaction <0.01). Presence of coronary artery disease attenuated the lower risk of high HDL cholesterol and all-cause mortality in those with eGFR≥60 ml/min per 1.73 m2 (P for interaction <0.05). CONCLUSIONS Our results show a U-shaped relationship between HDL cholesterol and risk of all-cause mortality across all eGFR categories. The risk is modified by eGFR and cardiovascular disease.
Collapse
Affiliation(s)
- Benjamin Bowe
- Clinical Epidemiology Center, Research and Education Service
| | - Yan Xie
- Clinical Epidemiology Center, Research and Education Service
| | - Hong Xian
- Clinical Epidemiology Center, Research and Education Service
- Department of Biostatistics, College for Public Health and Social Justice, St. Louis University, St. Louis, Missouri; and
| | | | - Mohamed A. Zayed
- Clinical Epidemiology Center, Research and Education Service
- Section of Vascular Surgery, Surgery Service, and
- Division of Vascular and Endovascular Surgery, Department of Surgery and
| | - Ziyad Al-Aly
- Clinical Epidemiology Center, Research and Education Service
- Division of Nephrology, Department of Medicine, Veterans Affairs St. Louis Health Care System, St. Louis, Missouri
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
38
|
Bowe B, Xie Y, Xian H, Balasubramanian S, Al-Aly Z. Low levels of high-density lipoprotein cholesterol increase the risk of incident kidney disease and its progression. Kidney Int 2016; 89:886-96. [PMID: 26924057 DOI: 10.1016/j.kint.2015.12.034] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 11/14/2015] [Accepted: 12/03/2015] [Indexed: 01/11/2023]
Abstract
Available experimental evidence suggests a role for high-density lipoprotein cholesterol (HDL-C) in incident chronic kidney disease (CKD) and its progression. However, clinical studies are inconsistent. We therefore built a cohort of 1,943,682 male US veterans and used survival models to examine the association between HDL-C and risks of incident CKD or CKD progression (doubling of serum creatinine, eGFR decline of 30% or more), or a composite outcome of ESRD, dialysis, or renal transplantation. Models were adjusted for demographics, comorbid conditions, eGFR, body mass index, lipid parameters, and statin use over a median follow-up of 9 years. Compared to those with HDL-C of 40 mg/dl or more, low HDL-C (under 30 mg/dl) was associated with increased risk of incident eGFR under 60 ml/min/1.73 m(2) (hazard ratio: 1.18; confidence interval: 1.17-1.19) and risk of incident CKD (1.20; 1.18-1.22). Adjusted models demonstrate an association between low HDL-C and doubling of serum creatinine (1.14; 1.12-1.15), eGFR decline of 30% or more (1.13; 1.12-1.14), and the composite renal end point (1.08; 1.06-1.11). Cubic spline analyses of the relationship between HDL-C levels and renal outcomes showed a U-shaped relationship, where risk was increased in lowest and highest deciles of HDL-C. Thus, a significant association exists between low HDL-C levels and risks of incident CKD and CKD progression. Further studies are needed to explain the increased risk of adverse renal outcomes in patients with high HDL-C.
Collapse
Affiliation(s)
- Benjamin Bowe
- Clinical Epidemiology Center, VA Saint Louis Health Care System, St. Louis, Missouri, USA
| | - Yan Xie
- Clinical Epidemiology Center, VA Saint Louis Health Care System, St. Louis, Missouri, USA
| | - Hong Xian
- Clinical Epidemiology Center, VA Saint Louis Health Care System, St. Louis, Missouri, USA; Department of Biostatistics, College for Public Health and Social Justice, Saint Louis University, St. Louis, Missouri, USA
| | | | - Ziyad Al-Aly
- Clinical Epidemiology Center, VA Saint Louis Health Care System, St. Louis, Missouri, USA; Department of Medicine, Division of Nephrology, VA Saint Louis Health Care System, St. Louis, Missouri, USA; Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.
| |
Collapse
|
39
|
Beckett RD, Wilhite AL, Robinson N, Rosene A. Drugs That Affect Lipid Metabolism. SIDE EFFECTS OF DRUGS ANNUAL 2016:469-477. [DOI: 10.1016/bs.seda.2016.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
40
|
Reiss AB, Voloshyna I, De Leon J, Miyawaki N, Mattana J. Cholesterol Metabolism in CKD. Am J Kidney Dis 2015; 66:1071-82. [PMID: 26337134 DOI: 10.1053/j.ajkd.2015.06.028] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 06/16/2015] [Indexed: 02/07/2023]
Abstract
Patients with chronic kidney disease (CKD) have a substantial risk of developing coronary artery disease. Traditional cardiovascular disease (CVD) risk factors such as hypertension and hyperlipidemia do not adequately explain the high prevalence of CVD in CKD. Both CVD and CKD are inflammatory states and inflammation adversely affects lipid balance. Dyslipidemia in CKD is characterized by elevated triglyceride levels and high-density lipoprotein levels that are both decreased and dysfunctional. This dysfunctional high-density lipoprotein becomes proinflammatory and loses its atheroprotective ability to promote cholesterol efflux from cells, including lipid-overloaded macrophages in the arterial wall. Elevated triglyceride levels result primarily from defective clearance. The weak association between low-density lipoprotein cholesterol level and coronary risk in CKD has led to controversy over the usefulness of statin therapy. This review examines disrupted cholesterol transport in CKD, presenting both clinical and preclinical evidence of the effect of the uremic environment on vascular lipid accumulation. Preventative and treatment strategies are explored.
Collapse
Affiliation(s)
- Allison B Reiss
- Department of Medicine and Winthrop Research Institute, Winthrop University Hospital, Mineola, NY.
| | - Iryna Voloshyna
- Department of Medicine and Winthrop Research Institute, Winthrop University Hospital, Mineola, NY
| | - Joshua De Leon
- Department of Medicine and Winthrop Research Institute, Winthrop University Hospital, Mineola, NY
| | - Nobuyuki Miyawaki
- Department of Medicine and Winthrop Research Institute, Winthrop University Hospital, Mineola, NY
| | - Joseph Mattana
- Department of Medicine and Winthrop Research Institute, Winthrop University Hospital, Mineola, NY
| |
Collapse
|
41
|
Athyros VG, Tziomalos K, Karagiannis A. Treatment options for dyslipidemia in chronic kidney disease and for protection from contrast-induced nephropathy. Expert Rev Cardiovasc Ther 2015. [PMID: 26206619 DOI: 10.1586/14779072.2015.1072047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chronic kidney disease (CKD) is highly prevalent worldwide and represents a major cardiovascular risk factor. Dyslipidemia is present in most patients with CKD and further worsens CKD, creating a vicious cycle. The treatment of CKD-related dyslipidemia has been a controversial topic. The use of statins is recommended in all stages of CKD, but it appears to reduce cardiovascular and renal events only in the early CKD stages, up to stage 3. The use of atorvastatin has proven very beneficial; thus, the earliest we start statin treatment, the better for the patient. Atorvastatin and pitavastatin do not need dose adjustments at any level of renal function. Fibrates can be administered in mixed hyperlipidemia, combined with statins in early CKD stages. Omega-3 fatty acids are useful for treating hypertriglyceridemia in CKD. Antibodies against proprotein convertase subtilisin/kexin type 9 hold promise for a better control of dyslipidemia and a greater reduction of cardiovascular risk.
Collapse
Affiliation(s)
- Vasilios G Athyros
- a 1 Second Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Hippocration Hospital, Thessaloniki, Greece
| | | | | |
Collapse
|
42
|
Croyal M, Ouguerram K, Passard M, Ferchaud-Roucher V, Chétiveaux M, Billon-Crossouard S, de Gouville AC, Lambert G, Krempf M, Nobécourt E. Effects of Extended-Release Nicotinic Acid on Apolipoprotein (a) Kinetics in Hypertriglyceridemic Patients. Arterioscler Thromb Vasc Biol 2015; 35:2042-7. [PMID: 26160958 DOI: 10.1161/atvbaha.115.305835] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/24/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To determine the mechanisms by which extended-release nicotinic acid reduces circulating lipoprotein (a) concentrations in hypertriglyceridemic patients. APPROACH AND RESULTS Eight nondiabetic, obese male subjects (aged 48±12 years; body mass index, 31.2±1.8 kg/m(2)) with hypertriglyceridemia (triglycerides, 226±78 mg/dL) were enrolled in an 8 week, double blind, placebo-controlled cross-over study. At the end of each treatment phase, fasted subjects received a 10 µmol/L per kg bolus injection of [5,5,5-(2)H3]-l-Leucine immediately followed by constant infusion of [5,5,5-(2)H3]-l-Leucine (10 µmol L(-1) kg(-1) h(-1)) for 14 hours, and blood samples were collected. A liquid chromatography-tandem mass spectrometry method was used to study apolipoprotein (a) (Apo(a)) kinetics. The fractional catabolic rate of Apo(a) was calculated with a single compartmental model using the apolipoprotein B100 (ApoB100) containing very low density lipoprotein tracer enrichment as a precursor pool. Extended-release nicotinic acid decreased plasma triglycerides (-46%; P=0.023), raised high-density lipoprotein cholesterol (+20%; P=0.008), and decreased Apo(a) plasma concentrations (-20%; P=0.008). Extended-release nicotinic acid also decreased ApoB100 (22%; P=0.008) and proprotein convertase subtilisin/kexin type 9 (PCSK9, -29%; P=0.008) plasma concentrations. Apo(a) fractional catabolic rate and production rates were decreased by 37% (0.58±0.28 versus 0.36±0.19 pool/d; P=0.008) and 50% (1.4±0.8 versus 0.7±0.4 nmol/kg per day; P=0.008), respectively. CONCLUSIONS Extended-release nicotinic acid treatment decreased Apo(a) plasma concentrations by 20%, production rates by 50%, and catabolism by 37%. ApoB100 and PCSK9 concentrations were also decreased by treatment, but no correlation was found with Apo(a) kinetic parameters.
Collapse
Affiliation(s)
- Mikaël Croyal
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| | - Khadija Ouguerram
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| | - Maxime Passard
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| | - Véronique Ferchaud-Roucher
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| | - Maud Chétiveaux
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| | - Stéphanie Billon-Crossouard
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| | - Anne-Charlotte de Gouville
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| | - Gilles Lambert
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| | - Michel Krempf
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.).
| | - Estelle Nobécourt
- From the CRNH, West Human Nutrition Research Center, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K., E.N.); UMR 1280 PhAN Laboratory, National Institute of Agronomic Research, INRA, CHU Hôtel Dieu, HNB1, Nantes, France (M.C., K.O., M.P., V.F.-R., S.B.-C., G.L., M.K.); University of Nantes and Medical School, Nantes, France (M.C., K.O., M.P., M.C., S.B.-C., G.L., M.K., E.N.); GlaxoSmithKline, Les Ulis, France (A.-C.d.G.); and Endocrinology and Nutrition Department, G and R Laennec Hospital, Bd Jacques Monod, Nantes, France (M.K., E.N.)
| |
Collapse
|