1
|
Billany RE, Bishop NC, Castle EM, Graham-Brown MPM, Greenwood SA, Lightfoot CJ, Wilkinson TJ. Physical activity interventions in adult kidney transplant recipients: an updated systematic review and meta-analysis of randomized controlled trials. Ren Fail 2025; 47:2480246. [PMID: 40148080 PMCID: PMC11951324 DOI: 10.1080/0886022x.2025.2480246] [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: 11/11/2024] [Revised: 02/15/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Kidney transplant recipients (KTRs) exhibit a high prevalence of cardiovascular disease (CVD) and adverse changes in physical fitness and body composition. Post-transplant management recommends being physically active and evidence in this field is growing. The aim of this review was to update our previous systematic review and meta-analysis of randomized controlled trials (RCTs) assessing the effects of physical activity and exercise training interventions in KTRs. METHODS A comprehensive literature search between March 2021 and September 2024 identified seven additional RCTs. Therefore, this updated review and meta-analysis includes 23 RCTs. Outcomes included cardiorespiratory fitness (CRF), strength, blood pressure, body composition, heart rate, markers of dyslipidemia and kidney function, and health-related quality of life. RESULTS Twenty-three RCTs, including 1,139 KTRs, were included. The median intervention length was 12 weeks with participants exercising between 2 and 7×/week. Most studies used a mixture of aerobic and resistance training but reporting and intervention content was highly varied. Significant improvements were observed in CRF (V̇O2peak; +3.87 mL/kg/min, p = .0004), physical function (sit-to-stand-60; +7.72 repetitions, p = .0001), and high-density lipoprotein (HDL; +0.13 mmol/L, p = .02). Isolated studies reported improvements in strength, bone health, lean mass, and quality of life (QoL). All studies were found to have a high or moderate risk of bias. CONCLUSIONS Exercise training or increasing physical activity may confer several benefits in adult KTRs, especially through the improvements in CRF and HDL which have been linked to CVD risk. Despite new literature, there is still a need for long-term larger sampled RCTs and more detailed reporting of intervention details and program adherence.
Collapse
Affiliation(s)
- Roseanne E. Billany
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Leicester, UK
| | - Nicolette C. Bishop
- School of Sport, Exercise, and Health Sciences, Loughborough University, Loughborough, UK
| | - Ellen M. Castle
- Faculty of Health Sciences, Curtin School of Allied Health, Curtin University, Perth, Australia
- Physiotherapy Division, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| | | | - Sharlene A. Greenwood
- Department of Renal Medicine, King’s College Hospital NHS Trust, London, UK
- Renal Sciences, Faculty of Life Sciences and Medicine, Kings College London, London, UK
| | - Courtney J. Lightfoot
- NIHR Leicester Biomedical Research Centre, Leicester, UK
- Department of Population Health Sciences, University of Leicester, Leicester, UK
| | - Thomas J. Wilkinson
- NIHR Leicester Biomedical Research Centre, Leicester, UK
- Leicester Diabetes Centre, University of Leicester, Leicester, UK
| |
Collapse
|
2
|
Garcia-Fernandez H, Alcala-Diaz JF, Quintana-Navarro GM, Lopez-Moreno J, Luque-Cordoba D, Ruiz-Diaz Narvaez E, Arenas-de Larriva AP, Gutierrez-Mariscal FM, Torres-Peña JD, Rodriguez-Cano D, Luque RM, Priego-Capote F, Lopez-Miranda J, Camargo A. Trimethylamine Oxidation into the Proatherogenic Trimethylamine N-Oxide Is Higher in Coronary Heart Disease Men: From the CORDIOPREV Study. World J Mens Health 2025; 43:249-258. [PMID: 39344118 PMCID: PMC11704170 DOI: 10.5534/wjmh.230366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 10/01/2024] Open
Abstract
PURPOSE Cardiovascular disease (CVD) is more prevalent in men than women, but the mechanisms responsible for this are not fully understood. We aimed to evaluate differences in trimethylamine (TMA), a microbial metabolite and its oxidized form, trimethylamine N-oxide (TMAO), which is thought to promote atherosclerosis, between men and women with coronary heart disease (CHD), using as a reference a non-CVD population. MATERIALS AND METHODS This study was carried out within the framework of the CORDIOPREV study (NCT00924937; June 19, 2009), a clinical trial which included 827 men and 175 women with CHD, with a non-CVD population of 375 individuals (270 men and 105 women) as a reference group. Plasma TMA and TMAO were measured by HPLC-MS/MS. The carotid study was ultrasonically assessed bilaterally by the quantification of intima-media thickness of both common carotid arteries (IMT-CC). RESULTS We found higher TMAO levels and TMAO/TMA ratio in CHD men than CHD women (p=0.034 and p=0.026, respectively). No TMA sex differences were found in CHD patients. The TMA and TMAO levels and TMAO/TMA ratio were lower, and no differences between sexes were found in the non-CVD population. TMAO levels in CHD patients were consistent with higher IMT-CC and more carotid plaques (p=0.032 and p=0.037, respectively) and lower cholesterol efflux in CHD men than CHD women (p<0.001). CONCLUSIONS Our results suggest that CHD men have augmented TMAO levels compared with CHD women, presumably as a consequence of higher rate of TMA to TMAO oxidation, which could be associated with CVD, as these sex differences are not observed in a non-CVD population.
Collapse
Affiliation(s)
- Helena Garcia-Fernandez
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan F Alcala-Diaz
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Gracia M Quintana-Navarro
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Lopez-Moreno
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Diego Luque-Cordoba
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- Department of Analytical Chemistry and Nanochemistry University Institute, University of Cordoba, Cordoba, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Eugenia Ruiz-Diaz Narvaez
- Department of Clinical Nutrition and Diet Therapy, Clinics Hospital, Faculty of Medical Sciences, National University of Asuncion, San Lorenzo, Paraguay
| | - Antonio P Arenas-de Larriva
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco M Gutierrez-Mariscal
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose D Torres-Peña
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Raul M Luque
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
| | - Feliciano Priego-Capote
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- Department of Analytical Chemistry and Nanochemistry University Institute, University of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
| | - Jose Lopez-Miranda
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| | - Antonio Camargo
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofía University Hospital, Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
3
|
Kang H, Song J, Cheng Y. HDL regulates the risk of cardiometabolic and inflammatory-related diseases: Focusing on cholesterol efflux capacity. Int Immunopharmacol 2024; 138:112622. [PMID: 38971111 DOI: 10.1016/j.intimp.2024.112622] [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: 06/08/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Dyslipidemia, characterized by higher serum concentrations of low-density lipoprotein cholesterol (LDL-C), very low-density lipoprotein cholesterol (VLDL-C), triglyceride (TG), and lower serum concentrations of high-density lipoprotein cholesterol (HDL-C), is confirmed as a hallmark of cardiovascular diseases (CVD), posing serious risks to the future health of humans. Aside from the role of HDL-C concentrations, the capacity of cholesterol efflux to HDL is being identified as an enssential messurement for the dyslipidemic morbidity. Through inducing the progression of reverse cholesterol transport (RCT), the HDL-related cholesterol efflux plays a vital role in atherosclerotic plaque formation. In addition, increasing results demonstrated that the relationships between cholesterol efflux and cardiovascular events might be influenced by multiple factors, such as atherosclerosis, diabetes, and, inflammatory diseases. These risk factors could affect the intracellular composition of HDL, which might subsqently influence the cholesterol efflux process induced by HDL particle. In the present comprehensive article, we summarize the latest findings which described the modulatory roles of HDL in cardiometabolic disorders and inflammatory related diseases, focusing on its capacity in mediating cholesterol efflux. Moreover, the potential mechanisms whereby HDL regulate the risk of cardiometabolic disorders or inflammatory related diseases, at least partly, via cholesterol efflux pathway, are also well-listed.
Collapse
Affiliation(s)
- Huiyuan Kang
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China
| | - Jingjin Song
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China
| | - Ye Cheng
- Department of Cardiology, the Xiamen Cardiovascular Hospital of Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
4
|
Mayer MS, Portella AF, Maçalai C, Zambra AL, Mori NC, Kessler Nunes VC, Bortolotto JW, Azzolin GB, Parisi MM. Yerba Mate as a Protectant against Lipoproteins Oxidation. Chem Biodivers 2024; 21:e202301770. [PMID: 38330241 DOI: 10.1002/cbdv.202301770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/10/2024]
Abstract
Oxidative modification of low-density lipoproteins (LDL) and high-density lipoproteins (HDL) are important factors determining cardiovascular risk. This study investigated the antioxidant mechanisms and potential protective effect of a hydroethanolic extract of yerba mate (Ilex paraguaiensis; EHEM) on the in vitro oxidation of LDL and HDL. EHEM was found to possess ferric reducing power, DPPH free radical scavenging capacity, metal chelating activity, and NO radical scavenging activity. In addition, EHEM reduced the lipoperoxidation induced by α,α'-Azodiisobutyramidine dihydrochloride (AAPH) in HDL and LDL at all tested concentrations. In this study, we demonstrate the antioxidant properties of yerba mate and its phytochemical compounds. These properties may effectively prevent the in vitro oxidation of LDL and HDL molecules, a phenomenon linked to the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Mariana Spanamberg Mayer
- Group of Integral Attention to Health, Center for Health and Rural Sciences, University of Cruz Alta, Brazil
- Postgraduate Program in Integral Attention to Health (PPGAIS) Unicruz, URI-Erechim, Unijuí, University of Cruz Alta, Brazil
| | - Amanda Felipe Portella
- Group of Integral Attention to Health, Center for Health and Rural Sciences, University of Cruz Alta, Brazil
| | - Camila Maçalai
- Group of Integral Attention to Health, Center for Health and Rural Sciences, University of Cruz Alta, Brazil
| | - Andressa Leal Zambra
- Group of Integral Attention to Health, Center for Health and Rural Sciences, University of Cruz Alta, Brazil
- Postgraduate Program in Integral Attention to Health (PPGAIS) Unicruz, URI-Erechim, Unijuí, University of Cruz Alta, Brazil
| | - Natacha Cossettin Mori
- Group of Integral Attention to Health, Center for Health and Rural Sciences, University of Cruz Alta, Brazil
| | - Viviane Cecília Kessler Nunes
- Group of Integral Attention to Health, Center for Health and Rural Sciences, University of Cruz Alta, Brazil
- Postgraduate Program in Integral Attention to Health (PPGAIS) Unicruz, URI-Erechim, Unijuí, University of Cruz Alta, Brazil
| | - Josiane Woutheres Bortolotto
- Group of Integral Attention to Health, Center for Health and Rural Sciences, University of Cruz Alta, Brazil
- Postgraduate Program in Integral Attention to Health (PPGAIS) Unicruz, URI-Erechim, Unijuí, University of Cruz Alta, Brazil
| | - Gabriela Bonfanti Azzolin
- Group of Integral Attention to Health, Center for Health and Rural Sciences, University of Cruz Alta, Brazil
- Postgraduate Program in Integral Attention to Health (PPGAIS) Unicruz, URI-Erechim, Unijuí, University of Cruz Alta, Brazil
| | - Mariana Migliorini Parisi
- Group of Integral Attention to Health, Center for Health and Rural Sciences, University of Cruz Alta, Brazil
- Postgraduate Program in Integral Attention to Health (PPGAIS) Unicruz, URI-Erechim, Unijuí, University of Cruz Alta, Brazil
- Center for Health and Rural Sciences, University of Cruz Alta, Rodovia Municipal Jacob Della Mea, s/n km 5,6 - Parada Benito, Cruz Alta, RS-98020-290, Brazil
| |
Collapse
|
5
|
Liu RX, Koyawala N, Thiessen-Philbrook HR, Doshi MD, Reese PP, Hall IE, Mohan S, Parikh CR. Untargeted metabolomics of perfusate and their association with hypothermic machine perfusion and allograft failure. Kidney Int 2023; 103:762-771. [PMID: 36549364 DOI: 10.1016/j.kint.2022.11.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/28/2022] [Accepted: 11/14/2022] [Indexed: 12/24/2022]
Abstract
Although hypothermic machine perfusion (HMP) is associated with improved kidney graft viability and function, the underlying biological mechanisms are unknown. Untargeted metabolomic profiling may identify potential metabolites and pathways that can help assess allograft viability and contribute to organ preservation. Therefore, in this multicenter study, we measured all detectable metabolites in perfusate collected at the beginning and end of deceased-donor kidney perfusion and evaluated their associations with graft failure. In our cohort of 190 kidney transplants, 33 (17%) had death-censored graft failure over a median follow-up of 5.0 years (IQR 3.0-6.1 years). We identified 553 known metabolites in perfusate and characterized their experimental and biological consistency through duplicate samples and unsupervised clustering. After perfusion-time adjustment and false discovery correction, six metabolites in post-HMP perfusate were significantly associated with death-censored graft failure, including alpha-ketoglutarate, 3-carboxy-4-methyl-5-propyl-2-furanpropanoate, 1-carboxyethylphenylalanine, and three glycerol-phosphatidylcholines. All six metabolites were associated with an increased risk of graft failure (Hazard Ratio per median absolute deviation range 1.04-1.45). Four of six metabolites also demonstrated significant interaction with donation after cardiac death with notably greater risk in the donation after cardiac death group (Hazard Ratios up to 1.69). Discarded kidneys did not have significantly different levels of any death-censored graft failure-associated metabolites. On interrogation of pathway analysis, production of reactive oxygen species and increased metabolism of fatty acids were upregulated in kidneys that subsequently developed death-censored graft failure. Thus, further understanding the role of these metabolites may inform the HMP process and help improve the objective evaluation of allograft offers, thereby reducing the discard of potentially viable organs.
Collapse
Affiliation(s)
- Richard X Liu
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Neel Koyawala
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Mona D Doshi
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Peter P Reese
- Renal-Electrolyte and Hypertension Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Isaac E Hall
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Sumit Mohan
- Department of Medicine, Vagelos College of Physicians and Surgeons, New York, New York, USA; Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Chirag R Parikh
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
6
|
Schachtl-Riess JF, Schönherr S, Lamina C, Forer L, Coassin S, Streiter G, Kheirkhah A, Li Y, Meiselbach H, Di Maio S, Eckardt KU, Köttgen A, Kronenberg F. KLKB1 and CLSTN2 are associated with HDL-mediated cholesterol efflux capacity in a genome-wide association study. Atherosclerosis 2023; 368:1-11. [PMID: 36812656 DOI: 10.1016/j.atherosclerosis.2023.01.022] [Citation(s) in RCA: 2] [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: 08/25/2022] [Revised: 01/06/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIMS HDL-mediated cholesterol efflux capacity (CEC) may protect from cardiovascular disease. Thus, we aimed to identify its genetic and non-genetic determinants. METHODS We measured CEC to 2% apolipoprotein B-depleted serum using BODIPY-cholesterol and cAMP-stimulated J774A.1 macrophages using serum samples from 4,981 participants in the German Chronic Kidney Disease (GCKD) study. Variance of CEC explained by clinical and biochemical parameters in a multivariable linear regression model was calculated by proportional marginal variance decomposition. A genome-wide association study with 7,746,917 variants was performed based on an additive genetic model. The main model was adjusted for age, sex and principal components 1-10. Further models were selected for sensitivity analysis and to reduce residual variance by known CEC pathways. RESULTS Variables that explained 1% and more of the variance of CEC were concentrations of triglycerides (12.9%), HDL-cholesterol (11.8%), LDL-cholesterol (3.0%), apolipoprotein A-IV (2.8%), PCSK9 (1.0%), and eGFR (1.0%). The KLKB1 (chr4) and APOE/C1 (chr19) loci were genome-wide significantly (p < 5x10-8) associated with CEC in our main model (p = 8.8x10-10 and p = 3.3x10-10, respectively). KLKB1 remained significantly associated after additional adjustment for either kidney parameters, HDL-cholesterol, triglycerides or apolipoprotein A-IV concentrations, while the APOE/C1 locus was not significantly associated anymore after adjustment for triglycerides. Adjustment for triglycerides also revealed an association with the CLSTN2 locus (chr3; p = 6.0x10-9). CONCLUSIONS We identified HDL-cholesterol and triglycerides as the main determinants of CEC. Furthermore, we newly found a significant association of CEC with the KLKB1 and the CLSTN2 locus and confirmed the association with the APOE/C1 locus, likely mediated by triglycerides.
Collapse
Affiliation(s)
- Johanna F Schachtl-Riess
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Sebastian Schönherr
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudia Lamina
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas Forer
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Coassin
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Gertraud Streiter
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Azin Kheirkhah
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Yong Li
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Heike Meiselbach
- Department of Nephrology and Hypertension, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Silvia Di Maio
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany; Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
7
|
Zanotti I. High-Density Lipoproteins in Non-Cardiovascular Diseases. Int J Mol Sci 2022; 23:ijms23169413. [PMID: 36012681 PMCID: PMC9408873 DOI: 10.3390/ijms23169413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/12/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Ilaria Zanotti
- Dipartimento di Scienze Degli Alimenti e del Farmaco, Università di Parma, 42124 Parma, Italy
| |
Collapse
|
8
|
Baker LA, March DS, Wilkinson TJ, Billany RE, Bishop NC, Castle EM, Chilcot J, Davies MD, Graham-Brown MPM, Greenwood SA, Junglee NA, Kanavaki AM, Lightfoot CJ, Macdonald JH, Rossetti GMK, Smith AC, Burton JO. Clinical practice guideline exercise and lifestyle in chronic kidney disease. BMC Nephrol 2022; 23:75. [PMID: 35193515 PMCID: PMC8862368 DOI: 10.1186/s12882-021-02618-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 11/22/2021] [Indexed: 12/13/2022] Open
Affiliation(s)
| | | | | | | | | | | | | | - Mark D. Davies
- Betsi Cadwaladr University Health Board and Bangor University, Bangor, UK
| | | | | | | | | | | | - Jamie H. Macdonald
- School of Sport, Health and Exercise Sciences, Bangor University, Bangor, UK
| | | | | | - James O. Burton
- University of Leicester and Leicester Hospitals NHS Trust, Leicester, UK
| |
Collapse
|
9
|
HDL and Surgery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1377:189-195. [DOI: 10.1007/978-981-19-1592-5_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
10
|
Lee JJ, Chi G, Fitzgerald C, Kazmi SHA, Kalayci A, Korjian S, Duffy D, Shaunik A, Kingwell B, Yeh RW, Bhatt DL, Gibson CM. Cholesterol Efflux Capacity and Its Association With Adverse Cardiovascular Events: A Systematic Review and Meta-Analysis. Front Cardiovasc Med 2021; 8:774418. [PMID: 34966797 PMCID: PMC8710716 DOI: 10.3389/fcvm.2021.774418] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Serum high-density lipoprotein cholesterol (HDL-C) levels are inversely associated with cardiovascular disease events. Yet, emerging evidence suggests that it is the functional properties of HDL, in particular, reverse cholesterol transport, which is a key protective mechanism mediating cholesterol removal from macrophage cells and reducing plaque lipid content. Cholesterol efflux capacity (CEC) measures the capacity of HDL to perform this function. A systematic review and meta-analysis were conducted to explore the association of CEC and adverse cardiovascular events. Methods: A comprehensive literature review of Embase, PubMed, and Web of Science Core Collection from inception to September 2019 was performed for all studies that examined the association between CEC and cardiovascular outcomes. The primary outcome was adverse cardiovascular events, which were inclusive of atherosclerotic cardiovascular disease (ASCVD) or mortality. Results: A total of 20 trials were included. Compared with low CEC levels, high CEC levels were associated with a 37% lower risk of adverse cardiovascular events (crude RR = 0.63; 95% CI, 0.52–0.76; P < 0.00001). Every SD increase of CEC was associated with a 20% lower risk of adverse cardiovascular events (HR = 0.80; 95% CI, 0.66–0.97; P = 0.02). The association remained significant after adjusting for cardiovascular risk factors, medications, and HDL-C levels (HR = 0.76; 95% CI, 0.63–0.91; P = 0.004). A significant CEC-endpoint relationship was observed (P = 0.024) such that for every 0.1 unit increase in CEC, there was a 5% reduced risk for adverse cardiovascular events (RR = 0.95; 95% CI, 0.91–0.99). Conclusions: Higher CEC is associated with lower adverse cardiovascular outcomes. These findings warrant further research on whether CEC is merely a biomarker or a mechanism that could be targeted as a pharmacologic intervention for improving clinical outcomes. PROSPERO Registration Number: CRD42020146681; https://www.crd.york.ac.uk/prospero/.
Collapse
Affiliation(s)
- Jane J Lee
- Baim Institute for Clinical Research, Boston, MA, United States
| | - Gerald Chi
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Clara Fitzgerald
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Syed Hassan A Kazmi
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Arzu Kalayci
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Serge Korjian
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | | | | | | | - Robert W Yeh
- Smith Center for Outcomes Research in Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Deepak L Bhatt
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - C Michael Gibson
- Baim Institute for Clinical Research, Boston, MA, United States.,Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
11
|
Wilkinson TJ, Bishop NC, Billany RE, Lightfoot CJ, Castle EM, Smith AC, Greenwood SA. The effect of exercise training interventions in adult kidney transplant recipients: a systematic review and meta-analysis of randomised control trials. PHYSICAL THERAPY REVIEWS 2021. [DOI: 10.1080/10833196.2021.2002641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Thomas J. Wilkinson
- Applied Research Collaboration East Midlands, Leicester Diabetes Centre, Leicester, UK
- Leicester Kidney Lifestyle Team, Department of Health Sciences, University of Leicester, Leicester, UK
| | - Nicolette C. Bishop
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Roseanne E. Billany
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Courtney J. Lightfoot
- Leicester Kidney Lifestyle Team, Department of Health Sciences, University of Leicester, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
| | - Ellen M. Castle
- Therapies Department, King’s College Hospital NHS Trust, London, UK
- Renal Medicine, School of Life Course Sciences, King’s College London, London, UK
| | - Alice C. Smith
- Leicester Kidney Lifestyle Team, Department of Health Sciences, University of Leicester, Leicester, UK
- Leicester NIHR Biomedical Research Centre, Leicester, UK
| | - Sharlene A. Greenwood
- Therapies Department, King’s College Hospital NHS Trust, London, UK
- Renal Medicine, School of Life Course Sciences, King’s College London, London, UK
| |
Collapse
|
12
|
Bem D, Sugrue D, Wilding B, Zile I, Butler K, Booth D, Tafesse E, McEwan P. The effect of hyperkalemia and long inter-dialytic interval on morbidity and mortality in patients receiving hemodialysis: a systematic review. Ren Fail 2021; 43:241-254. [PMID: 33478329 PMCID: PMC7833048 DOI: 10.1080/0886022x.2020.1871012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/18/2020] [Accepted: 12/26/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Patients with chronic kidney disease, especially those receiving hemodialysis (HD), are at risk of hyperkalemia (HK). This systematic review aimed to evaluate the prevalence of HK in patients with renal disease receiving HD and collate evidence on the effect of HK and differing HD patterns (i.e., long vs. short inter-dialytic intervals [LIDI and SIDI, respectively] in a thrice weekly schedule) on mortality. METHODS Comprehensive searches were conducted across six databases and selected conference proceedings by two independent reviewers up to September 2020. A hundred and two studies reporting frequency of HK, mortality, or cardiovascular (CV) outcomes in adult patients with acute, chronic or end-stage renal disease in receipt of HD were included. Narrative synthesis of results was undertaken with key findings presented in tables and figures. RESULTS Median prevalence of HK in patients with renal disease receiving HD was 21.6% and increased in patients receiving concomitant medications - mainly renin-angiotensin-aldosterone system inhibitors and potassium-sparing diuretics. Associations between elevated potassium levels and increased risk of both all-cause and CV mortality in the HD population were consistent across the included studies. In addition, there was a rise in all-cause and CV mortality on the day following LIDI compared with the day after the two SIDIs in patients on HD. CONCLUSIONS Evidence identified in this systematic review indicates a relationship between HK and LIDI with mortality in patients with renal disease receiving HD, emphasizing the need for effective monitoring and management to control potassium levels both in emergency and chronic HD settings.
Collapse
Affiliation(s)
- Danai Bem
- Health Economics and Outcomes Research Ltd, Birmingham, UK
| | - Daniel Sugrue
- Health Economics and Outcomes Research Ltd, Cardiff, UK
| | - Ben Wilding
- Health Economics and Outcomes Research Ltd, Cardiff, UK
| | - Ina Zile
- Health Economics and Outcomes Research Ltd, Cardiff, UK
| | - Karin Butler
- Health Economics and Outcomes Research Ltd, Cardiff, UK
| | - David Booth
- Health Economics and Outcomes Research Ltd, Cardiff, UK
| | | | - Phil McEwan
- Health Economics and Outcomes Research Ltd, Cardiff, UK
- Swansea University, Swansea, UK
| |
Collapse
|
13
|
Schachtl-Riess JF, Coassin S, Lamina C, Demetz E, Streiter G, Hilbe R, Kronenberg F. Lysis reagents, cell numbers, and calculation method influence high-throughput measurement of HDL-mediated cholesterol efflux capacity. J Lipid Res 2021; 62:100125. [PMID: 34571016 PMCID: PMC8521207 DOI: 10.1016/j.jlr.2021.100125] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/09/2021] [Accepted: 09/15/2021] [Indexed: 11/23/2022] Open
Abstract
HDL-mediated cholesterol efflux capacity (CEC) may protect against cardiovascular disease. However, CEC assays are not standardized, hampering their application in large cohorts and comparison between studies. To improve standardization, we systematically investigated technical differences between existing protocols that influence assay performance that have not been previously addressed. CEC was measured in 96-well plates using J774A.1 macrophages labeled with BODIPY-cholesterol and incubated for 4 h with 2% apolipoprotein B-depleted human serum. The time zero method, which calculates CEC using control wells, and the per-well method, which calculates CEC based on the actual content of BODIPY-cholesterol in each well, were compared in 506 samples. We showed that the per-well method had a considerably lower sample rejection rate (4.74% vs. 13.44%) and intra-assay (4.48% vs. 5.28%) and interassay coefficients of variation (two controls: 7.85%, 9.86% vs. 13.58%, 15.29%) compared with the time zero method. Correction for plate-to-plate differences using four controls on each plate also improved assay performance of both methods. In addition, we observed that the lysis reagent used had a significant effect. Compared with cholic acid, lysis with sodium hydroxide results in higher (P = 0.0082) and Triton X-100 in lower (P = 0.0028) CEC values. Furthermore, large cell seeding errors (30% variation) greatly biased CEC for both referencing methods (P < 0.0001) as measured by a resazurin assay. In conclusion, lysis reagents, cell numbers, and assay setup greatly impact the quality and reliability of CEC quantification and should be considered when this method is newly established in a laboratory.
Collapse
Affiliation(s)
- Johanna F Schachtl-Riess
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Coassin
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudia Lamina
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Egon Demetz
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Gertraud Streiter
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Richard Hilbe
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Kronenberg
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
14
|
Clemens RK, Hunjadi M, Ritsch A, Rohrer L, Meier TO, Amann-Vesti B, von Eckardstein A, Annema W. Cholesterol Efflux Capacity Associates with the Ankle-Brachial Index but Not All-Cause Mortality in Patients with Peripheral Artery Disease. Diagnostics (Basel) 2021; 11:1407. [PMID: 34441341 PMCID: PMC8394478 DOI: 10.3390/diagnostics11081407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Cholesterol efflux is an important mechanism by which high-density lipoproteins (HDLs) protect against cardiovascular disease. As peripheral artery disease (PAD) is associated with high mortality rates, mainly due to cardiovascular causes, we investigated whether cholesterol efflux capacity (CEC) of apolipoprotein B (apoB)-depleted plasma, a widely used surrogate of HDL function, may serve as a predictive marker for mortality in this patient population. METHODS In this prospective single-center study (median follow-up time: 9.3 years), apoB-containing lipoproteins were precipitated from plasma of 95 patients with PAD and incubated with J744-macrophages, which were loaded with radiolabeled cholesterol. CEC was defined as the fractional radiolabel released during 4 h of incubation. RESULTS Baseline CEC was lower in PAD patients that currently smoked (p = 0.015) and had a history of myocardial infarction (p = 0.011). Moreover, CEC showed a significant correlation with HDL-cholesterol (p = 0.003) and apolipoprotein A-I levels (p = 0.001) as well as the ankle-brachial index (ABI, p = 0.018). However, CEC did not differ between survivors and non-survivors. Neither revealed Kaplan-Meier and Cox regression analyses any significant association of CEC with all-cause mortality rates. CONCLUSION Taken together, CEC is associated with ABI but does not predict all-cause mortality in patients with PAD.
Collapse
Affiliation(s)
- Robert K. Clemens
- Vascular Center, Cantonal Hospital Baden, University of Zurich, Im Ergel 1, CH-5404 Baden, Switzerland
| | - Monika Hunjadi
- Department of Internal Medicine, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.H.); (A.R.)
| | - Andreas Ritsch
- Department of Internal Medicine, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.H.); (A.R.)
| | - Lucia Rohrer
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland; (L.R.); (A.v.E.); (W.A.)
| | - Thomas O. Meier
- Clinic for Angiology, University Hospital Zurich, University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland;
| | | | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland; (L.R.); (A.v.E.); (W.A.)
| | - Wijtske Annema
- Institute of Clinical Chemistry, University Hospital Zurich, University of Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland; (L.R.); (A.v.E.); (W.A.)
| |
Collapse
|
15
|
Kon V, Yang HC, Smith LE, Vickers KC, Linton MF. High-Density Lipoproteins in Kidney Disease. Int J Mol Sci 2021; 22:ijms22158201. [PMID: 34360965 PMCID: PMC8348850 DOI: 10.3390/ijms22158201] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Decades of epidemiological studies have established the strong inverse relationship between high-density lipoprotein (HDL)-cholesterol concentration and cardiovascular disease. Recent evidence suggests that HDL particle functions, including anti-inflammatory and antioxidant functions, and cholesterol efflux capacity may be more strongly associated with cardiovascular disease protection than HDL cholesterol concentration. These HDL functions are also relevant in non-cardiovascular diseases, including acute and chronic kidney disease. This review examines our current understanding of the kidneys’ role in HDL metabolism and homeostasis, and the effect of kidney disease on HDL composition and functionality. Additionally, the roles of HDL particles, proteins, and small RNA cargo on kidney cell function and on the development and progression of both acute and chronic kidney disease are examined. The effect of HDL protein modification by reactive dicarbonyls, including malondialdehyde and isolevuglandin, which form adducts with apolipoprotein A-I and impair proper HDL function in kidney disease, is also explored. Finally, the potential to develop targeted therapies that increase HDL concentration or functionality to improve acute or chronic kidney disease outcomes is discussed.
Collapse
Affiliation(s)
- Valentina Kon
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (V.K.); (H.-C.Y.)
| | - Hai-Chun Yang
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (V.K.); (H.-C.Y.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Loren E. Smith
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Kasey C. Vickers
- Atherosclerosis Research Unit, Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - MacRae F. Linton
- Atherosclerosis Research Unit, Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
- Correspondence:
| |
Collapse
|
16
|
The Framingham Risk Score Is Associated with Chronic Graft Failure in Renal Transplant Recipients. J Clin Med 2021; 10:jcm10153287. [PMID: 34362071 PMCID: PMC8348129 DOI: 10.3390/jcm10153287] [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: 06/17/2021] [Revised: 07/07/2021] [Accepted: 07/23/2021] [Indexed: 12/01/2022] Open
Abstract
Predicting chronic graft failure in renal transplant recipients (RTR) is an unmet clinical need. Chronic graft failure is often accompanied by transplant vasculopathy, the formation of de novo atherosclerosis in the transplanted kidney. Therefore, we determined whether the 10-year Framingham risk score (FRS), an established atherosclerotic cardiovascular disease prediction module, is associated with chronic graft failure in RTR. In this prospective longitudinal study, 600 well-characterised RTR were followed for 10 years. The association with death-censored chronic graft failure (n = 81, 13.5%) was computed. An extended Cox model showed that each one percent increase of the FRS significantly increased the risk of chronic graft failure by 4% (HR: 1.04, p < 0.001). This association remained significant after adjustment for potential confounders, including eGFR (HR: 1.03, p = 0.014). Adding the FRS to eGFR resulted in a higher AUC in a receiver operating curve (AUC = 0.79, p < 0.001) than eGFR alone (AUC = 0.75, p < 0.001), and an improvement in the model likelihood ratio statistic (67.60 to 88.39, p < 0.001). These results suggest that a combination of the FRS and eGFR improves risk prediction. The easy to determine and widely available FRS has clinical potential to predict chronic graft failure in RTR.
Collapse
|
17
|
Speer T, Ridker PM, von Eckardstein A, Schunk SJ, Fliser D. Lipoproteins in chronic kidney disease: from bench to bedside. Eur Heart J 2021; 42:2170-2185. [PMID: 33393990 DOI: 10.1093/eurheartj/ehaa1050] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/16/2020] [Accepted: 12/08/2020] [Indexed: 12/24/2022] Open
Abstract
Chronic kidney disease (CKD) is associated with high cardiovascular risk. CKD patients exhibit a specific lipoprotein pattern termed 'uraemic dyslipidaemia', which is characterized by rather normal low-density lipoprotein cholesterol, low high-density lipoprotein cholesterol, and high triglyceride plasma levels. All three lipoprotein classes are involved in the pathogenesis of CKD-associated cardiovascular diseases (CVDs). Uraemia leads to several modifications of the structure of lipoproteins such as changes of the proteome and the lipidome, post-translational protein modifications (e.g. carbamylation) and accumulation of small-molecular substances within the lipoprotein moieties, which affect their functionality. Lipoproteins from CKD patients interfere with lipid transport and promote inflammation, oxidative stress, endothelial dysfunction as well as other features of atherogenesis, thus contributing to the development of CKD-associated CVD. While, lipid-modifying therapies play an important role in the management of CKD patients, their efficacy is modulated by kidney function. Novel therapeutic agents to prevent the adverse remodelling of lipoproteins in CKD and to improve their functional properties are highly desirable and partially under development.
Collapse
Affiliation(s)
- Thimoteus Speer
- Translational Cardio-Renal Medicine, Saarland University, Kirrberger Strasse, Building 41, D-66421 Homburg/Saar, Germany.,Department of Internal Medicine IV, Saarland University Hospital, Nephrology and Hypertension, Kirrberger Strasse, Building 41, D-66421 Homburg/Saar, Germany
| | - Paul M Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, 900 Commonwealth Avenue, Boston, MA 02215, USA
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University Hospital Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland
| | - Stefan J Schunk
- Translational Cardio-Renal Medicine, Saarland University, Kirrberger Strasse, Building 41, D-66421 Homburg/Saar, Germany
| | - Danilo Fliser
- Translational Cardio-Renal Medicine, Saarland University, Kirrberger Strasse, Building 41, D-66421 Homburg/Saar, Germany
| |
Collapse
|
18
|
Sotomayor CG, te Velde-Keyzer CA, Diepstra A, van Londen M, Pol RA, Post A, Gans RO, Nolte IM, Slart RH, de Borst MH, Berger SP, Rodrigo R, Navis GJ, de Boer RA, Bakker SJ. Galectin-3 and Risk of Late Graft Failure in Kidney Transplant Recipients: A 10-year Prospective Cohort Study. Transplantation 2021; 105:1106-1115. [PMID: 32639409 PMCID: PMC8078111 DOI: 10.1097/tp.0000000000003359] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 05/04/2020] [Accepted: 06/01/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Galectin-3 may play a causal role in kidney inflammation and fibrosis, which may also be involved in the development of kidney graft failure. With novel galectin-3-targeted pharmacological therapies increasingly coming available, we aimed to investigate whether galectin-3 is associated with risk of late graft failure in kidney transplant recipients (KTR). METHODS We studied adult KTR who participated in TransplantLines Insulin Resistance and Inflammation Biobank and Cohort Study, recruited in a university setting (2001-2003). Follow-up was performed for a median of 9.5 (interquartile range, 6.2-10.2) years. Overall and stratified (Pinteraction < 0.05) multivariable-adjusted Cox proportional-hazards regression analyses were performed to study the association of galectin-3 with risk of graft failure (restart of dialysis or retransplantation). RESULTS Among 561 KTR (age 52 ± 12 y; 54% males), baseline median galectin-3 was 21.1 (interquartile range, 17.0-27.2) ng/mL. During follow-up, 72 KTR developed graft failure (13, 18, and 44 events over increasing tertiles of galectin-3). Independent of adjustment for donor, recipient, and transplant characteristics, galectin-3-associated with increased risk of graft failure (hazard ratios [HR] per 1 SD change, 2.12; 95% confidence interval [CI], 1.63-2.75; P < 0.001), particularly among KTR with systolic blood pressure ≥140 mmHg (HR, 2.29; 95% CI, 1.80-2.92; P < 0.001; Pinteraction = 0.01) or smoking history (HR, 2.56; 95% CI, 1.95-3.37; P < 0.001; Pinteraction = 0.03). Similarly, patients in the highest tertile of galectin-3 were consistently at increased risk of graft failure. CONCLUSIONS Serum galectin-3 levels are elevated in KTR, and independently associated with increased risk of late graft failure. Whether galectin-3-targeted therapies may represent novel opportunities to decrease the long-standing high burden of late graft failure in stable KTR warrants further studies.
Collapse
Affiliation(s)
- Camilo G. Sotomayor
- Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Radiology, Clinical Hospital of the University of Chile, University of Chile, Santiago, Chile
| | - Charlotte A. te Velde-Keyzer
- Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Arjan Diepstra
- Division of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marco van Londen
- Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Robert A. Pol
- Division of Transplantation Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Adrian Post
- Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Rijk O.B. Gans
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ilja M. Nolte
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Riemer H.J.A. Slart
- Department of Nuclear and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Martin H. de Borst
- Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stefan P. Berger
- Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ramón Rodrigo
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Gerjan J. Navis
- Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Rudolf A. de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stephan J.L. Bakker
- Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
19
|
Steffen HLM, Anderson JLC, Poot ML, Lei Y, Connelly MA, Bakker SJL, Öörni K, Tietge UJF. Proteoglycan binding as proatherogenic function metric of apoB-containing lipoproteins and chronic kidney graft failure. J Lipid Res 2021; 62:100083. [PMID: 33939983 PMCID: PMC8173310 DOI: 10.1016/j.jlr.2021.100083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 04/14/2021] [Indexed: 12/31/2022] Open
Abstract
Lipoprotein-proteoglycan binding is an early key event in atherosclerotic lesion formation and thus conceivably could play a major role in vasculopathy-driven chronic graft failure and cardiovascular mortality in renal transplant recipients. The present study investigated whether lipoprotein-proteoglycan binding susceptibility (LPBS) of apoB-containing lipoproteins and levels of the classical atherosclerosis biomarker LDL-C were associated with cardiovascular mortality (n = 130) and graft failure (n = 73) in 589 renal transplant recipients who were followed up from at least 1 year after transplantation for 9.5 years. At baseline, LPBS was significantly higher in patients who subsequently developed graft failure than in those with a surviving graft (1.68 ± 0.93 vs. 1.46 ± 0.49 nmol/mmol, P = 0.001). Cox regression analysis showed an association between LPBS and chronic graft failure in an age- and sex-adjusted model (hazard ratio: 1.45; 95% CI, 1.14-1.85; P = 0.002), but no association was observed with cardiovascular mortality. LDL-C levels were not associated with graft failure or cardiovascular mortality. This study shows that measurement of cholesterol retention outperformed the traditionally used quantitative parameter of LDL-C levels in predicting graft failure, suggesting a higher relevance of proatherogenic function than the quantity of apoB-containing lipoproteins in chronic kidney graft failure.
Collapse
Affiliation(s)
- Hannah L M Steffen
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Josephine L C Anderson
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Margot L Poot
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Yu Lei
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Margery A Connelly
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, NC, USA
| | - Stephan J L Bakker
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Katariina Öörni
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland; Molecular and Integrative Bioscience Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Uwe J F Tietge
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
20
|
Anderson JLC, Bakker SJL, Tietge UJF. Triglyceride/HDL cholesterol ratio and premature all-cause mortality in renal transplant recipients. Nephrol Dial Transplant 2021; 36:936-938. [PMID: 33313807 DOI: 10.1093/ndt/gfaa321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/12/2020] [Indexed: 11/14/2022] Open
Affiliation(s)
- Josephine L C Anderson
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Uwe J F Tietge
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Laboratory Medicine, Division of Clinical Chemistry, Karolinska Institutet, Stockholm, Sweden.,Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
21
|
Sotomayor CG, Bustos NI, Yepes-Calderon M, Arauna D, de Borst MH, Berger SP, Rodrigo R, Dullaart RPF, Navis GJ, Bakker SJL. Plasma Vitamin C and Risk of Late Graft Failure in Kidney Transplant Recipients: Results of the TransplantLines Biobank and Cohort Study. Antioxidants (Basel) 2021; 10:631. [PMID: 33919075 PMCID: PMC8143099 DOI: 10.3390/antiox10050631] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 11/16/2022] Open
Abstract
Recent studies have shown that depletion of vitamin C is frequent in outpatient kidney transplant recipients (KTR) and that vitamin C is inversely associated with risk of death. Whether plasma vitamin C is associated with death-censored kidney graft failure remains unknown. We investigated KTR who participated in the TransplantLines Insulin Resistance and Inflammation Biobank and Cohort Study. The primary outcome was graft failure (restart of dialysis or re-transplantation). Overall and stratified (pinteraction < 0.1) multivariable-adjusted Cox regression analyses are presented here. Among 598 KTR (age 51 ± 12 years-old; 55% males), baseline median (IQR) plasma vitamin C was 44.0 (31.0-55.3) µmol/L. Through a median follow-up of 9.5 (IQR, 6.3‒10.2) years, 75 KTR developed graft failure (34, 26, and 15 events over increasing tertiles of vitamin C, log-rank p < 0.001). Plasma vitamin C was inversely associated with risk of graft failure (HR per 1-SD increment, 0.69; 95% CI 0.54-0.89; p = 0.004), particularly among KTR with triglycerides ≥1.9 mmol/L (HR 0.46; 95% CI 0.30-0.70; p < 0.001; pinteraction = 0.01) and among KTR with HDL cholesterol ≥0.91 mmol/L (HR 0.56; 95% CI 0.38-0.84; p = 0.01; pinteraction = 0.04). These findings remained materially unchanged in multivariable-adjusted analyses (donor, recipient, and transplant characteristics, including estimated glomerular filtration rate and proteinuria), were consistent in categorical analyses according to tertiles of plasma vitamin C, and robust after exclusion of outliers. Plasma vitamin C in outpatient KTR is inversely associated with risk of late graft failure. Whether plasma vitamin C‒targeted therapeutic strategies represent novel opportunities to ease important burden of graft failure necessitates further studies.
Collapse
Affiliation(s)
- Camilo G. Sotomayor
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (M.Y.-C.); (M.H.d.B.); (S.P.B.); (G.J.N.); (S.J.L.B.)
- Faculty of Medicine, University of Chile, 8330033 Santiago, Chile; (N.I.B.); (R.R.)
| | - Nicolas I. Bustos
- Faculty of Medicine, University of Chile, 8330033 Santiago, Chile; (N.I.B.); (R.R.)
| | - Manuela Yepes-Calderon
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (M.Y.-C.); (M.H.d.B.); (S.P.B.); (G.J.N.); (S.J.L.B.)
| | - Diego Arauna
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, 3460000 Talca, Chile;
| | - Martin H. de Borst
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (M.Y.-C.); (M.H.d.B.); (S.P.B.); (G.J.N.); (S.J.L.B.)
| | - Stefan P. Berger
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (M.Y.-C.); (M.H.d.B.); (S.P.B.); (G.J.N.); (S.J.L.B.)
| | - Ramón Rodrigo
- Faculty of Medicine, University of Chile, 8330033 Santiago, Chile; (N.I.B.); (R.R.)
| | - Robin P. F. Dullaart
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
| | - Gerjan J. Navis
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (M.Y.-C.); (M.H.d.B.); (S.P.B.); (G.J.N.); (S.J.L.B.)
| | - Stephan J. L. Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (M.Y.-C.); (M.H.d.B.); (S.P.B.); (G.J.N.); (S.J.L.B.)
| |
Collapse
|
22
|
Yubero-Serrano EM, Alcalá-Diaz JF, Gutierrez-Mariscal FM, Arenas-de Larriva AP, Peña-Orihuela PJ, Blanco-Rojo R, Martinez-Botas J, Torres-Peña JD, Perez-Martinez P, Ordovas JM, Delgado-Lista J, Gómez-Coronado D, Lopez-Miranda J. Association between cholesterol efflux capacity and peripheral artery disease in coronary heart disease patients with and without type 2 diabetes: from the CORDIOPREV study. Cardiovasc Diabetol 2021; 20:72. [PMID: 33766036 PMCID: PMC7993540 DOI: 10.1186/s12933-021-01260-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 03/13/2021] [Indexed: 01/09/2023] Open
Abstract
Background Peripheral artery disease (PAD) is recognized as a significant predictor of mortality and adverse cardiovascular outcomes in patients with coronary heart disease (CHD). In fact, coexisting PAD and CHD is strongly associated with a greater coronary event recurrence compared with either one of them alone. High-density lipoprotein (HDL)-mediated cholesterol efflux capacity (CEC) is found to be inversely associated with an increased risk of incident CHD. However, this association is not established in patients with PAD in the context of secondary prevention. In this sense, our main aim was to evaluate the association between CEC and PAD in patients with CHD and whether the concurrent presence of PAD and T2DM influences this association. Methods CHD patients (n = 1002) from the CORDIOPREV study were classified according to the presence or absence of PAD (ankle-brachial index, ABI ≤ 0.9 and ABI > 0.9 and < 1.4, respectively) and T2DM status. CEC was quantified by incubation of cholesterol-loaded THP-1 cells with the participants' apoB-depleted plasma was performed. Results The presence of PAD determined low CEC in non-T2DM and newly-diagnosed T2DM patients. Coexisting PAD and newly-diagnosed T2DM provided and additive effect providing an impaired CEC compared to non-T2DM patients with PAD. In established T2DM patients, the presence of PAD did not determine differences in CEC, compared to those without PAD, which may be restored by glucose-lowering treatment. Conclusions Our findings suggest an inverse relationship between CEC and PAD in CHD patients. These results support the importance of identifying underlying mechanisms of PAD, in the context of secondary prevention, that provide potential therapeutic targets, that is the case of CEC, and establishing strategies to prevent or reduce the high risk of cardiovascular events of these patients. Trial registrationhttps://clinicaltrials.gov/ct2/show/NCT00924937. Unique Identifier: NCT00924937![]()
Collapse
Affiliation(s)
- Elena M Yubero-Serrano
- Lipids and Atherosclerosis Unit. Servicio de Medicina Interna, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research in Córdoba, University of Córdoba, Córdoba, Spain. .,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain.
| | - Juan F Alcalá-Diaz
- Lipids and Atherosclerosis Unit. Servicio de Medicina Interna, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research in Córdoba, University of Córdoba, Córdoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| | - Francisco M Gutierrez-Mariscal
- Lipids and Atherosclerosis Unit. Servicio de Medicina Interna, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research in Córdoba, University of Córdoba, Córdoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| | - Antonio P Arenas-de Larriva
- Lipids and Atherosclerosis Unit. Servicio de Medicina Interna, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research in Córdoba, University of Córdoba, Córdoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| | - Patricia J Peña-Orihuela
- Lipids and Atherosclerosis Unit. Servicio de Medicina Interna, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research in Córdoba, University of Córdoba, Córdoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| | - Ruth Blanco-Rojo
- Research and Development Department, Biosearch Life, Granada, Spain
| | - Javier Martinez-Botas
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain.,Department of Biochemistry-Research, Hospital Universitario Ramón Y Cajal, Instituto Ramón Y Cajal de Investigacion Sanitaria (IRyCIS), Madrid, Spain
| | - Jose D Torres-Peña
- Lipids and Atherosclerosis Unit. Servicio de Medicina Interna, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research in Córdoba, University of Córdoba, Córdoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| | - Pablo Perez-Martinez
- Lipids and Atherosclerosis Unit. Servicio de Medicina Interna, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research in Córdoba, University of Córdoba, Córdoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| | - Jose M Ordovas
- Jean Mayer US Department of Agriculture Human Nutrition Research Center On Aging, Tufts University School of Medicine, Boston, MA, USA.,IMDEA-Food Institute, CEI UAM + CSIC, Madrid, Spain
| | - Javier Delgado-Lista
- Lipids and Atherosclerosis Unit. Servicio de Medicina Interna, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research in Córdoba, University of Córdoba, Córdoba, Spain.,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| | - Diego Gómez-Coronado
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain.,Department of Biochemistry-Research, Hospital Universitario Ramón Y Cajal, Instituto Ramón Y Cajal de Investigacion Sanitaria (IRyCIS), Madrid, Spain
| | - Jose Lopez-Miranda
- Lipids and Atherosclerosis Unit. Servicio de Medicina Interna, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research in Córdoba, University of Córdoba, Córdoba, Spain. .,CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
23
|
Kawahira Y, Shiga Y, Inoue H, Suematsu Y, Tashiro K, Kato Y, Fujimi K, Takamiya Y, Kuwano T, Sugihara M, Miura SI. Association between high-density lipoprotein cholesterol levels and major adverse cardiovascular events in patients who underwent coronary computed tomography angiography: FU-CCTA registry. Heart Vessels 2021; 36:1457-1465. [PMID: 33744994 DOI: 10.1007/s00380-021-01831-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 03/05/2021] [Indexed: 11/29/2022]
Abstract
It is unclear whether higher levels of serum high-density lipoprotein cholesterol (HDL-C) prevent major adverse cardiovascular events (MACE). We prospectively evaluated 501 patients who had undergone coronary computed tomography angiography at Fukuoka University Hospital and either were clinically suspected of having coronary artery disease (CAD) or had at least one cardiovascular risk factor with a follow-up of up to 5 years. The primary endpoint was MACE (cardiovascular death, ischemic stroke, acute myocardial infarction and coronary revascularization). The patients were divided into tertiles according to the HDL-C level: 47 mg/dl ≥ HDL-C level [n = 167, lower HDL-C level (L-HDL)], 58 mg/dl ≥ HDL-C level ≥ 48 mg/dl [n = 167, middle HDL-C level (M-HDL)] and HDL-C level ≥ 59 mg/dl [n = 167, higher HDL-C level (H-HDL)] groups. There were significant differences in %CAD among the L-HDL, M-HDL and H-HDL groups. Unexpectedly, there was no difference in %MACE between M-HDL and H-HDL, although %MACE in M-HDL was significantly lower than that in L-HDL (p < 0.05). By a multivariate logistic regression analysis, MACE in H-HDL-C was independently associated with diabetes mellitus (DM) (p = 0.03). A Kaplan-Meier curve according to the HDL subgroup indicated that M-HDL, not H-HDL, enjoyed the greatest freedom from MACE among the 3 groups (log-rank test p = 0.047). Finally, the results of a Cox regression model indicated that L-HDL and H-HDL had significantly higher risk of MACE than M-HDL. In conclusions, patients with middle HDL-C levels, not higher HDL-C levels, showed the greatest freedom from MACE. Patients with higher HDL-C levels need to be strictly managed for DM to prevent MACE.
Collapse
Affiliation(s)
- Yuto Kawahira
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan.,Department of Cardiology, Fukuoka University Nishijin Hospital, Fukuoka, Japan
| | - Yuhei Shiga
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Hiroko Inoue
- Department of Cardiology, Fukuoka University Nishijin Hospital, Fukuoka, Japan
| | - Yasunori Suematsu
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Kohei Tashiro
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Yuta Kato
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Kanta Fujimi
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan.,Cardiac Rehabilitation Center, Fukuoka University Hospital, Fukuoka, Japan
| | - Yosuke Takamiya
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Takashi Kuwano
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Makoto Sugihara
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Shin-Ichiro Miura
- Department of Cardiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan. .,Department of Cardiology, Fukuoka University Nishijin Hospital, Fukuoka, Japan. .,Cardiac Rehabilitation Center, Fukuoka University Hospital, Fukuoka, Japan.
| |
Collapse
|
24
|
Kuusisto S, Kostara C, Kangas AJ, Perola M, Salomaa V, Kettunen J, Ala-Korpela M. HDL-Mediated Cholesterol Efflux Associates with Incident Kidney Disease. Clin Chem 2021; 67:689-691. [PMID: 33723592 DOI: 10.1093/clinchem/hvab024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Sanna Kuusisto
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland.,Center for Life Course Health Research, University of Oulu, Oulu, Finland.,NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Christina Kostara
- Laboratory of Clinical Chemistry, School of Medicine, University of Ioannina, Ioannina, Greece
| | | | - Markus Perola
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland.,Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Veikko Salomaa
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Johannes Kettunen
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland.,Center for Life Course Health Research, University of Oulu, Oulu, Finland.,Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Mika Ala-Korpela
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland.,Center for Life Course Health Research, University of Oulu, Oulu, Finland.,NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
25
|
Adorni MP, Ronda N, Bernini F, Zimetti F. High Density Lipoprotein Cholesterol Efflux Capacity and Atherosclerosis in Cardiovascular Disease: Pathophysiological Aspects and Pharmacological Perspectives. Cells 2021; 10:cells10030574. [PMID: 33807918 PMCID: PMC8002038 DOI: 10.3390/cells10030574] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
Over the years, the relationship between high-density lipoprotein (HDL) and atherosclerosis, initially highlighted by the Framingham study, has been revealed to be extremely complex, due to the multiple HDL functions involved in atheroprotection. Among them, HDL cholesterol efflux capacity (CEC), the ability of HDL to promote cell cholesterol efflux from cells, has emerged as a better predictor of cardiovascular (CV) risk compared to merely plasma HDL-cholesterol (HDL-C) levels. HDL CEC is impaired in many genetic and pathological conditions associated to high CV risk such as dyslipidemia, chronic kidney disease, diabetes, inflammatory and autoimmune diseases, endocrine disorders, etc. The present review describes the current knowledge on HDL CEC modifications in these conditions, focusing on the most recent human studies and on genetic and pathophysiologic aspects. In addition, the most relevant strategies possibly modulating HDL CEC, including lifestyle modifications, as well as nutraceutical and pharmacological interventions, will be discussed. The objective of this review is to help understanding whether, from the current evidence, HDL CEC may be considered as a valid biomarker of CV risk and a potential pharmacological target for novel therapeutic approaches.
Collapse
Affiliation(s)
- Maria Pia Adorni
- Unit of Neurosciences, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy;
| | - Nicoletta Ronda
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (N.R.); (F.Z.)
| | - Franco Bernini
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (N.R.); (F.Z.)
- Correspondence:
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (N.R.); (F.Z.)
| |
Collapse
|
26
|
Anderson JLC, Bakker SJL, Tietge UJF. The triglyceride to HDL-cholesterol ratio and chronic graft failure in renal transplantation. J Clin Lipidol 2021; 15:301-310. [PMID: 33589404 DOI: 10.1016/j.jacl.2021.01.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 01/09/2021] [Accepted: 01/23/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND Transplant vasculopathy (TV) is a major contributing factor to chronic graft failure in renal transplant recipients (RTR). TV lesions resemble atherosclerosis in several ways, and it is plausible to believe that some risk factors influence both atherosclerotic plaque formation and formation of TV. OBJECTIVE The objective of this prospective longitudinal study was to determine if dyslipidemia reflected by the triglyceride (TG)/high-density lipoprotein cholesterol (HDL-C) ratio is prospectively associated with death censored chronic graft failure in RTR. METHOD 454 prospectively included RTR with a functioning graft for at least one year, were followed for a median of 7 years. RTR were matched based on propensity scores to avoid potential confounding and subsequently the association of the TG/HDL-C ratio with the endpoint chronic graft failure, defined as return to dialysis or re-transplantation, was investigated. RESULTS Linear regression analysis showed that concentration of insulin, male gender, BMI and number of antihypertensives predict the TG/HDL-C ratio. Cox regression showed that the TG/HDL-C ratio is associated with chronic graft failure (HR = 1.43, 95%CI = 1.12-1.84, p = 0.005) in competing risk analysis for mortality. Interaction testing indicated that the relationship of the TG/HDL-C ratio with graft failure is stronger in subjects with a higher insulin concentration. CONCLUSION Our results demonstrate that the TG/HDL-C ratio has the potential to act as a predictive clinical biomarker. Furthermore, there is a need for closer attention to lipid management in RTR in clinical practice with a focus on triglyceride metabolism.
Collapse
Affiliation(s)
- Josephine L C Anderson
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Uwe J F Tietge
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
27
|
Wallimann-Annema W. [The Current Significance of Measuring HDL-Cholesterol in Cardiovascular Risk Assessment]. PRAXIS 2021; 110:383-390. [PMID: 34019448 DOI: 10.1024/1661-8157/a003677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The Current Significance of Measuring HDL-Cholesterol in Cardiovascular Risk Assessment Abstract. In clinical practice, high-density lipoprotein cholesterol (HDL-C) levels are frequently used for cardiovascular risk prediction. HDL particles perform numerous functions that theoretically protect against atherosclerosis. Accordingly, extensive studies have clearly demonstrated that low HDL-C is an important independent risk factor for cardiovascular diseases. However, it is now considered questionable whether very high HDL-C levels are always cardioprotective. This may be explained by the structural heterogeneity of HDL particles and the loss of HDL protective functions in the context of disease, which cannot be detected by the simple measurement of HDL-C. In the future new markers of the functional capacity of HDL particles may replace HDL-C as a traditional parameter for cardiovascular risk assessment.
Collapse
|
28
|
Tashiro K, Inoue H, Shiga Y, Tsukihashi Y, Imaizumi T, Norimatsu K, Idemoto Y, Kuwano T, Sugihara M, Nishikawa H, Katsuda Y, Miura SI. Associations Between High Levels of High-Density Lipoprotein Cholesterol and the Presence and Severity of Coronary Artery Disease in Patients Who Have Undergone Coronary Computed Tomography Angiography. J Clin Med Res 2020; 12:734-739. [PMID: 33224375 PMCID: PMC7665872 DOI: 10.14740/jocmr4367] [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: 10/06/2020] [Accepted: 10/17/2020] [Indexed: 01/08/2023] Open
Abstract
Background Although a recent study in a Japanese cohort indicated that extremely high-density lipoprotein cholesterol (HDL-C, ≥ 90 mg/dL) had an adverse effect on atherosclerotic cardiovascular disease mortality, we could not conclude that high levels of HDL-C were associated with the presence or severity of coronary artery disease (CAD). Methods We enrolled 1,016 patients who were clinically suspected to have CAD and who underwent coronary computed tomography angiography (CCTA). The number of significantly stenosed coronary vessels (vessel disease (VD), ≥ 50% coronary stenosis is diagnosed as CAD) and the Gensini score were quantified using CCTA, and the lipid profile was measured. The patients were divided into four groups according to the HDL-C level: < 40 mg/dL (n = 115, low), 40 - 59 mg/dL (n = 530, normal), 60 - 89 mg/dL (n = 335, high) and ≥ 90 mg/dL (n = 36, very-high). Results The percentage (%) of CAD in the low, normal, high and very-high groups was 69%, 55%, 42% and 25%, respectively (P for trend < 0.01). The Gensini score in the low, normal, high and very-high groups was 20 ± 25, 12 ± 16, 8 ± 12 and 4 ± 6, respectively (P for trend < 0.01). The very-high group showed the lowest triglyceride (TG) levels among the four groups. There were no significant differences in the level of low-density lipoprotein cholesterol or % use of statin among the four groups. Finally, the presence of CAD was independently associated with a low level of HDL-C, in addition to age, male, high systolic blood pressure and hemoglobin A1c, but not TG, by a multivariate logistic regression analysis. Conclusions High levels of HDL-C at the time of CCTA for screening were associated with a reduced presence and severity of CAD.
Collapse
Affiliation(s)
- Kohei Tashiro
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan.,These authors contributed equally to this manuscript
| | - Hiroko Inoue
- Department of Cardiology, Fukuoka University Nishijin Hospital, Fukuoka, Japan.,These authors contributed equally to this manuscript
| | - Yuhei Shiga
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan.,These authors contributed equally to this manuscript
| | - Yohei Tsukihashi
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan.,Department of Cardiology, Fukuoka University Nishijin Hospital, Fukuoka, Japan
| | - Tomoki Imaizumi
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Kenji Norimatsu
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Yoshiaki Idemoto
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Takashi Kuwano
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Makoto Sugihara
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Hiroaki Nishikawa
- Department of Cardiology, Fukuoka University Nishijin Hospital, Fukuoka, Japan
| | - Yousuke Katsuda
- Department of Cardiology, Fukuoka University Nishijin Hospital, Fukuoka, Japan
| | - Shin-Ichiro Miura
- Department of Cardiology, Fukuoka University School of Medicine, Fukuoka, Japan.,Department of Cardiology, Fukuoka University Nishijin Hospital, Fukuoka, Japan
| |
Collapse
|
29
|
Current Understanding of the Relationship of HDL Composition, Structure and Function to Their Cardioprotective Properties in Chronic Kidney Disease. Biomolecules 2020; 10:biom10091348. [PMID: 32967334 PMCID: PMC7564231 DOI: 10.3390/biom10091348] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 12/27/2022] Open
Abstract
In the general population, the ability of high-density lipoproteins (HDLs) to promote cholesterol efflux is a predictor of cardiovascular events, independently of HDL cholesterol levels. Although patients with chronic kidney disease (CKD) have a high burden of cardiovascular morbidity and mortality, neither serum levels of HDL cholesterol, nor cholesterol efflux capacity associate with cardiovascular events. Important for the following discussion on the role of HDL in CKD is the notion that traditional atherosclerotic cardiovascular risk factors only partially account for this increased incidence of cardiovascular disease in CKD. As a potential explanation, across the spectrum of cardiovascular disease, the relative contribution of atherosclerotic cardiovascular disease becomes less important with advanced CKD. Impaired renal function directly affects the metabolism, composition and functionality of HDL particles. HDLs themselves are a heterogeneous population of particles with distinct sizes and protein composition, all of them affecting the functionality of HDL. Therefore, a more specific approach investigating the functional and compositional features of HDL subclasses might be a valuable strategy to decipher the potential link between HDL, cardiovascular disease and CKD. This review summarizes the current understanding of the relationship of HDL composition, metabolism and function to their cardio-protective properties in CKD, with a focus on CKD-induced changes in the HDL proteome and reverse cholesterol transport capacity. We also will highlight the gaps in the current knowledge regarding important aspects of HDL biology.
Collapse
|
30
|
van der Boom T, Jia C, Lefrandt JD, Connelly MA, Links TP, Tietge UJF, Dullaart RPF. HDL Cholesterol Efflux Capacity is Impaired in Severe Short-Term Hypothyroidism Despite Increased HDL Cholesterol. J Clin Endocrinol Metab 2020; 105:5881625. [PMID: 32761088 PMCID: PMC7947992 DOI: 10.1210/clinem/dgaa411] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/28/2020] [Indexed: 01/01/2023]
Abstract
CONTEXT Severe hypothyroidism has profound effects on lipoprotein metabolism including high-density lipoprotein (HDL) cholesterol elevations but effects on HDL function metrics are unknown. OBJECTIVE To determine the impact of severe short-term hypothyroidism on HDL particle characteristics, HDL cholesterol efflux capacity (CEC), and HDL antioxidative capacity. DESIGN Observational study with variables measured during severe short-term hypothyroidism (median TSH 81 mU/L) and after 20 weeks of thyroid hormone supplementation (median TSH 0.03 mU/L) (Netherlands Trial Registry ID 7228). SETTING University hospital setting in The Netherlands. PATIENTS Seventeen patients who had undergone a total thyroidectomy for differentiated thyroid carcinoma. MAIN OUTCOME MEASURES HDL particle characteristics (nuclear magnetic resonance spectrometry), CEC (human THP-1-derived macrophage foam cells and apolipoprotein B-depleted plasma), and HDL anti-oxidative capacity (inhibition of low-density lipoprotein oxidation). RESULTS During hypothyroidism plasma total cholesterol, HDL cholesterol and apolipoprotein A-I were increased (P ≤ 0.001). HDL particle concentration was unchanged, but there was a shift in HDL subclasses toward larger HDL particles (P < 0.001). CEC was decreased (P = 0.035), also when corrected for HDL cholesterol (P < 0.001) or HDL particle concentration (P = 0.011). HDL antioxidative capacity did not change. CONCLUSION During severe short-term hypothyroidism CEC, an important antiatherogenic metric of HDL function, is impaired. HDL cholesterol and larger HDL particles are increased but HDL particle concentration is unchanged. Combined, these findings suggest that HDL quality and quantity are not improved, reflecting dysfunctional HDL in hypothyroidism.
Collapse
Affiliation(s)
- Trynke van der Boom
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, RB Groningen, The Netherlands
- Department of Vascular Medicine, University of Groningen, University Medical Center Groningen, RB Groningen, The Netherlands
| | - Congzhuo Jia
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, RB Groningen, The Netherlands
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Joop D Lefrandt
- Department of Vascular Medicine, University of Groningen, University Medical Center Groningen, RB Groningen, The Netherlands
| | - Margery A Connelly
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, North Carolina
| | - Thera P Links
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, RB Groningen, The Netherlands
- Correspondence and Reprint Requests: T.P. Links, MD, PhD, University Medical Center Groningen, Department of Endocrinology, PO Box 30.001, 9700 RB Groningen, The Netherlands. E-mail:
| | - Uwe J F Tietge
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, RB Groningen, The Netherlands
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Robin P F Dullaart
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, RB Groningen, The Netherlands
| |
Collapse
|
31
|
Szili-Torok T, Bakker SJL, Tietge UJF. Statin Use Is Prospectively Associated With New-Onset Diabetes After Transplantation in Renal Transplant Recipients. Diabetes Care 2020; 43:1945-1947. [PMID: 32444455 DOI: 10.2337/dc19-1212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 04/14/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE New-onset diabetes after transplantation (NODAT) is frequent and worsens graft and patient outcomes in renal transplant recipients (RTRs). In the general population, statins are diabetogenic. This study investigated whether statins also increase NODAT risk in RTRs. RESEARCH DESIGN AND METHODS From a prospective longitudinal study of 606 RTRs (functioning allograft >1 year, single academic center, follow-up: median 9.6 [range, 6.6-10.2] years), 95 patients using statins were age- and sex-matched to RTRs not on statins (all diabetes-free at inclusion). RESULTS NODAT incidence was 7.2% (73.3% of these on statins). In Kaplan-Meier (log-rank test, P = 0.017) and Cox regression analyses (HR 3.86 [95% CI 1.21-12.27]; P = 0.022), statins were prospectively associated with incident NODAT, even independent of several relevant confounders including immunosuppressive medication and biomarkers of glucose homeostasis. CONCLUSIONS This study demonstrates that statin use is prospectively associated with the development of NODAT in RTRs independent of other recognized risk factors.
Collapse
Affiliation(s)
- Tamas Szili-Torok
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Uwe J F Tietge
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands .,Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
32
|
Huang X, Jia Y, Zhu X, Zhang Y, Jiang L, Wei X, Zhao D, Zhao X, Du Y. Effects of Statins on Lipid Profile of Kidney Transplant Recipients: A Meta-Analysis of Randomized Controlled Trials. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9094543. [PMID: 32462035 PMCID: PMC7212277 DOI: 10.1155/2020/9094543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 03/21/2020] [Accepted: 04/07/2020] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To assess the benefits of statins on lipid profile in kidney transplant recipients via a meta-analysis. METHODS We systematically identified peer-reviewed clinical trials, review articles, and treatment guidelines from PubMed, Embase, the Cochrane Library, Wanfang, Chinese National Knowledge Infrastructure (CNKI), SinoMed (CBM), and Chongqing VIP databases from inception to April 2019. In the analysis, only randomized controlled clinical trials performed in human were included. RESULTS Eight articles were included in the analysis, involving 335 kidney transplant recipients who received statins and 350 kidney transplant patients as the control group. Results revealed that statins improved the lipid profile of kidney transplant recipients. Specifically, statin therapy significantly reduced total cholesterol and low-density lipoprotein cholesterol. However, it had no effects on high-density lipoprotein cholesterol and triglyceride levels. CONCLUSIONS The present study provides valuable knowledge on the potential benefits of statins in kidney transplant recipients. This meta-analysis shows that statin therapy modifies the lipid profile in this patient population.
Collapse
Affiliation(s)
- Xiu Huang
- Department of Nephrology, The First Hospital of Jilin University, Changchun 130000, China
| | - Yong Jia
- School of Nursing, Jilin University, Changchun 130000, China
| | - Xiaoyu Zhu
- Department of Nephrology, The First Hospital of Jilin University, Changchun 130000, China
| | - Yangyang Zhang
- Department of Nephrology, The First Hospital of Jilin University, Changchun 130000, China
| | - Lili Jiang
- Department of Nephrology, The First Hospital of Jilin University, Changchun 130000, China
| | - Xuejiao Wei
- Department of Nephrology, The First Hospital of Jilin University, Changchun 130000, China
| | - Dan Zhao
- Department of Nephrology, The First Hospital of Jilin University, Changchun 130000, China
| | - Xiaoxia Zhao
- Department of Nephrology, The First Hospital of Jilin University, Changchun 130000, China
| | - Yujun Du
- Department of Nephrology, The First Hospital of Jilin University, Changchun 130000, China
| |
Collapse
|
33
|
Soria-Florido MT, Schröder H, Grau M, Fitó M, Lassale C. High density lipoprotein functionality and cardiovascular events and mortality: A systematic review and meta-analysis. Atherosclerosis 2020; 302:36-42. [PMID: 32438197 DOI: 10.1016/j.atherosclerosis.2020.04.015] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 03/11/2020] [Accepted: 04/22/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND AIMS The aim of this systematic review and meta-analysis is to synthesize studies assessing the associations between high-density lipoprotein functionality and risk of cardiovascular disease and mortality. METHODS We searched Medline and Embase for the identification of observational studies meeting the inclusion criteria. This meta-analysis was conducted following the PRISMA statement and was registered in PROSPERO (CRD42017065857). We pooled risk estimates with a random-effect model separately for cardiovascular disease (fatal and non-fatal) and all-cause mortality. RESULTS Out of 29 manuscripts, 20 articles investigated cholesterol efflux capacity (13 prospective and 7 cross-sectional), 10 antioxidant capacity (7 prospective and 3 cross-sectional) and two anti-inflammatory capacity of high-density lipoprotein (1 prospective and 1 cross-sectional). A greater cholesterol efflux capacity was associated with lower risk of cardiovascular disease in 8 studies (RR for 1SD increase: 0.86; 95% CI: 0.76-0.98) and of mortality in 5 studies (RR for 1SD increase: 0,77; 0.60-1.00). Better antioxidant capacity was non-significantly associated with lower cardiovascular disease risk in 2 studies (RR for 1SD increase 0.70; 0.32-1.53) and significantly with mortality in 3 studies (RR for 1SD increase 0.48; 0.28-0.81). High-density lipoprotein anti-inflammatory ability was associated with a lower cardiovascular disease risk in the only prospective study. CONCLUSIONS Greater high-density lipoprotein cholesterol efflux capacity and antioxidant/anti-inflammatory capacities were associated with lower risk of cardiovascular disease. However, the heterogeneity between studies and evidence of publication bias warrants caution and highlights the need for larger prospective studies with standardized assays and specific outcomes.
Collapse
Affiliation(s)
- Maria T Soria-Florido
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; PhD Program in Food Sciences and Nutrition, Department of Nutrition, Food Science and Gastronomy, School of Pharmacy and Food Science, Campus de l'Alimentació Torribera, University of Barcelona, Barcelona, Spain; Department of Behavioural Science and Health, University College London, London, United Kingdom
| | - Helmut Schröder
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; CIBER of Epidemiology and Public Health (CIBERESP), Institute of Health Carlos III, Madrid, Spain
| | - María Grau
- Registre Gironí del COR. Group, Cardiovascular, Epidemiology and Genetics Research Group, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; CIBER of Cardiovascular Diseases (CIBERCV), Institute of Health Carlos III, Madrid, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Montserrat Fitó
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| | - Camille Lassale
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain; Department of Behavioural Science and Health, University College London, London, United Kingdom; CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
34
|
Hafiane A, Favari E, Daskalopoulou SS, Vuilleumier N, Frias MA. High-density lipoprotein cholesterol efflux capacity and cardiovascular risk in autoimmune and non-autoimmune diseases. Metabolism 2020; 104:154141. [PMID: 31923386 DOI: 10.1016/j.metabol.2020.154141] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/23/2019] [Accepted: 01/05/2020] [Indexed: 12/22/2022]
Abstract
Functional assessment of cholesterol efflux capacity (CEC) to high-density lipoprotein (HDL) is an emerging tool for evaluating morbidity and mortality associated with cardiovascular disease (CVD). By promoting macrophage reverse cholesterol transport (RCT), HDL-mediated CEC is believed to play an important role in atherosclerotic lesion progression in the vessel wall. Furthermore, recent evidence indicates that the typical inverse associations between various forms of CEC and CV events may be strongly modulated by environmental systemic factors and traditional CV risk factors, in addition to autoimmune diseases. These factors influence the complex and dynamic composition of HDL particles, which in turn positively or negatively affect HDL-CEC. Herein, we review recent findings connecting HDL-CEC to traditional CV risk factors and cardiometabolic conditions (non-autoimmune diseases) as well as autoimmune diseases, with a specific focus on how these factors may influence the associations between HDL-CEC and CVD risk.
Collapse
Affiliation(s)
- Anouar Hafiane
- Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, 1001 Decarie Blvd, Bloc E01. 3370H, Montréal, Qc H4A 3J1, Canada.
| | - Elda Favari
- Department of Food and Drug, University of Parma, Parco Area delle Scienze, 27/A, 43124 Parma, Italy.
| | - Stella S Daskalopoulou
- Department of Medicine, Division of Internal Medicine, McGill University, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, EM1.2230, Montreal, Quebec H4A 3J1, Canada.
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, 1211 Geneva, Switzerland; Division of Laboratory Medicine, Department of Medical Specialties, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland.
| | - Miguel A Frias
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, 1211 Geneva, Switzerland; Division of Laboratory Medicine, Department of Medical Specialties, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland.
| |
Collapse
|
35
|
Sotomayor CG, Gomes-Neto AW, Eisenga MF, Nolte IM, Anderson JLC, de Borst MH, Osté MCJ, Rodrigo R, Gans ROB, Berger SP, Navis GJ, Bakker SJL. Consumption of fruits and vegetables and cardiovascular mortality in renal transplant recipients: a prospective cohort study. Nephrol Dial Transplant 2020; 35:357-365. [PMID: 30165500 DOI: 10.1093/ndt/gfy248] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 06/27/2018] [Indexed: 08/18/2024] Open
Abstract
BACKGROUND It currently remains understudied whether low consumption of fruits and vegetables after kidney transplantation may be a modifiable cardiovascular risk factor. We aimed to investigate the associations between consumption of fruits and vegetables and cardiovascular mortality in renal transplant recipients (RTRs). METHODS Consumption of fruits and vegetables was assessed in an extensively phenotyping cohort of RTRs. Multivariable-adjusted Cox proportional hazards regression analyses were performed to assess the risk of cardiovascular mortality. RESULTS We included 400 RTRs (age 52 ± 12 years, 54% males). At a median follow-up of 7.2 years, 23% of RTRs died (53% were due to cardiovascular causes). Overall, fruit consumption was not associated with cardiovascular mortality {hazard ratio [HR] 0.82 [95% confidence interval (CI) 0.60-1.14]; P = 0.24}, whereas vegetable consumption was inversely associated with cardiovascular mortality [HR 0.49 (95% CI 0.34-0.71); P < 0.001]. This association remained independent of adjustment for several potential confounders. The association of fruit consumption with cardiovascular mortality was significantly modified by estimated glomerular filtration rate (eGFR; Pinteraction = 0.01) and proteinuria (Pinteraction = 0.01), with significant inverse associations in patients with eGFR > 45 mL/min/1.73 m2 [HR 0.56 (95% CI 0.35-0.92); P = 0.02] or the absence of proteinuria [HR 0.62 (95% CI 0.41-0.92); P = 0.02]. CONCLUSIONS In RTRs, a relatively higher vegetable consumption is independently and strongly associated with lower cardiovascular mortality. A relatively higher fruit consumption is also associated with lower cardiovascular mortality, although particularly in RTRs with eGFR > 45 mL/min/1.73 m2 or an absence of proteinuria. Further studies seem warranted to investigate whether increasing consumption of fruits and vegetables may open opportunities for potential interventional pathways to decrease the burden of cardiovascular mortality in RTRs.
Collapse
Affiliation(s)
- Camilo G Sotomayor
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - António W Gomes-Neto
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Michele F Eisenga
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Josephine L C Anderson
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Martin H de Borst
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maryse C J Osté
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ramón Rodrigo
- Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Rijk O B Gans
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Stefan P Berger
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gerjan J Navis
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
36
|
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
|
37
|
Gacitúa TA, Sotomayor CG, Groothof D, Eisenga MF, Pol RA, de Borst MH, Gans RO, Berger SP, Rodrigo R, Navis GJ, Bakker SJ. Plasma Vitamin C and Cancer Mortality in Kidney Transplant Recipients. J Clin Med 2019; 8:E2064. [PMID: 31771233 PMCID: PMC6947225 DOI: 10.3390/jcm8122064] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/21/2019] [Accepted: 11/21/2019] [Indexed: 02/07/2023] Open
Abstract
There is a changing trend in mortality causes in kidney transplant recipients (KTR), with a decline in deaths due to cardiovascular causes along with a relative increase in cancer mortality rates. Vitamin C, a well-known antioxidant with anti-inflammatory and immune system enhancement properties, could offer protection against cancer. We aimed to investigate the association of plasma vitamin C with long-term cancer mortality in a cohort of stable outpatient KTR without history of malignancies other than cured skin cancer. Primary and secondary endpoints were cancer and cardiovascular mortality, respectively. We included 598 KTR (mean age 51 ± 12 years old, 55% male). Mean (SD) plasma vitamin C was 44 ± 20 μmol/L. At a median follow-up of 7.0 (IQR, 6.2-7.5) years, 131 patients died, of which 24% deaths were due to cancer. In Cox proportional hazards regression analyses, vitamin C was inversely associated with cancer mortality (HR 0.50; 95%CI 0.34-0.74; P < 0.001), independent of potential confounders, including age, smoking status and immunosuppressive therapy. In secondary analyses, vitamin C was not associated with cardiovascular mortality (HR 1.16; 95%CI 0.83-1.62; P = 0.40). In conclusion, plasma vitamin C is inversely associated with cancer mortality risk in KTR. These findings underscore that relatively low circulating plasma vitamin C may be a meaningful as yet overlooked modifiable risk factor of cancer mortality in KTR.
Collapse
Affiliation(s)
- Tomás A. Gacitúa
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (T.A.G.); (D.G.); (M.F.E.); (M.H.d.B.); (S.P.B.); (G.J.N.)
| | - Camilo G. Sotomayor
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (T.A.G.); (D.G.); (M.F.E.); (M.H.d.B.); (S.P.B.); (G.J.N.)
| | - Dion Groothof
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (T.A.G.); (D.G.); (M.F.E.); (M.H.d.B.); (S.P.B.); (G.J.N.)
| | - Michele F. Eisenga
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (T.A.G.); (D.G.); (M.F.E.); (M.H.d.B.); (S.P.B.); (G.J.N.)
| | - Robert A. Pol
- Department of Surgery, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
| | - Martin H. de Borst
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (T.A.G.); (D.G.); (M.F.E.); (M.H.d.B.); (S.P.B.); (G.J.N.)
| | - Rijk O.B. Gans
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (T.A.G.); (D.G.); (M.F.E.); (M.H.d.B.); (S.P.B.); (G.J.N.)
| | - Stefan P. Berger
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (T.A.G.); (D.G.); (M.F.E.); (M.H.d.B.); (S.P.B.); (G.J.N.)
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, CP 8380453 Santiago, Chile;
| | - Gerjan J. Navis
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (T.A.G.); (D.G.); (M.F.E.); (M.H.d.B.); (S.P.B.); (G.J.N.)
| | - Stephan J.L. Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (T.A.G.); (D.G.); (M.F.E.); (M.H.d.B.); (S.P.B.); (G.J.N.)
| |
Collapse
|
38
|
Sotomayor CG, Rodrigo R, Gomes-Neto AW, Gormaz JG, Pol RA, Minović I, Eggersdorfer ML, Vos M, Riphagen IJ, de Borst MH, Nolte IM, Berger SP, Navis GJ, Bakker SJL. Plasma versus Erythrocyte Vitamin E in Renal Transplant Recipients, and Duality of Tocopherol Species. Nutrients 2019; 11:E2821. [PMID: 31752254 PMCID: PMC6893692 DOI: 10.3390/nu11112821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 12/22/2022] Open
Abstract
Redox imbalance is an adverse on-going phenomenon in renal transplant recipients (RTR). Vitamin E has important antioxidant properties that counterbalance its deleterious effects. However, plasma vitamin E affinity with lipids challenges interpretation of its levels. To test the hypothesis that erythrocyte membranes represent a lipids-independent specimen to estimate vitamin E status, we performed a cross-sectional study in a cohort of adult RTR (n = 113) recruited in a university setting (2015-2018). We compared crude and total lipids-standardized linear regression-derived coefficients of plasma and erythrocyte tocopherol species in relation to clinical and laboratory parameters. Strongly positive associations of fasting lipids with plasma tocopherol became inverse, rather than absent, in total lipids-standardized analyses, indicating potential overadjustment. Whilst, no variables from the lipids domain were associated with the tocopherol species measured from erythrocyte specimens. In relation to inflammatory status and clinical parameters with antioxidant activity, we found associations in directions that are consistent with either beneficial or adverse effects concerning α- or γ-tocopherol, respectively. In conclusion, erythrocytes offer a lipids-independent alternative to estimate vitamin E status and investigate its relationship with parameters over other biological domains. In RTR, α- and γ-tocopherol may serve as biomarkers of relatively lower or higher vulnerability to oxidative stress and inflammation, noticeably in opposite directions.
Collapse
Affiliation(s)
- Camilo G. Sotomayor
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (A.W.G.-N.); (M.H.d.B.); (S.P.B.); (G.J.N.); (S.J.L.B.)
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, CP 8380453 Santiago, Chile;
| | - António W. Gomes-Neto
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (A.W.G.-N.); (M.H.d.B.); (S.P.B.); (G.J.N.); (S.J.L.B.)
| | | | - Robert A. Pol
- Division of Transplantation Surgery, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
| | - Isidor Minović
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (I.M.); (M.L.E.); (M.V.); (I.J.R.)
| | - Manfred L. Eggersdorfer
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (I.M.); (M.L.E.); (M.V.); (I.J.R.)
| | - Michel Vos
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (I.M.); (M.L.E.); (M.V.); (I.J.R.)
| | - Ineke J. Riphagen
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (I.M.); (M.L.E.); (M.V.); (I.J.R.)
| | - Martin H. de Borst
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (A.W.G.-N.); (M.H.d.B.); (S.P.B.); (G.J.N.); (S.J.L.B.)
| | - Ilja M. Nolte
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
| | - Stefan P. Berger
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (A.W.G.-N.); (M.H.d.B.); (S.P.B.); (G.J.N.); (S.J.L.B.)
| | - Gerjan J. Navis
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (A.W.G.-N.); (M.H.d.B.); (S.P.B.); (G.J.N.); (S.J.L.B.)
| | - Stephan J. L. Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (A.W.G.-N.); (M.H.d.B.); (S.P.B.); (G.J.N.); (S.J.L.B.)
| |
Collapse
|
39
|
Szili-Torok T, Annema W, Anderson JLC, Bakker SJL, Tietge UJF. HDL Cholesterol Efflux Predicts Incident New-Onset Diabetes After Transplantation (NODAT) in Renal Transplant Recipients Independent of HDL Cholesterol Levels. Diabetes 2019; 68:1915-1923. [PMID: 31375510 DOI: 10.2337/db18-1267] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 07/29/2019] [Indexed: 11/13/2022]
Abstract
In renal transplant recipients (RTRs), new-onset diabetes after transplantation (NODAT) is a frequent and serious complication limiting survival of graft and patient. However, the underlying pathophysiology remains incompletely understood. In vitro and in preclinical models, HDL can preserve β-cell function, largely by mediating cholesterol efflux, but this concept has not been evaluated in humans. This study investigated whether baseline cholesterol efflux capacity (CEC) in RTRs is associated with incident NODAT during follow-up. This prospective longitudinal study included 405 diabetes-free RTRs with a functioning graft for >1 year. During a median (interquartile range) follow-up of 9.6 (6.6-10.2) years, 57 patients (14.1%) developed NODAT. HDL CEC was quantified using incubation of human macrophage foam cells with apolipoprotein B-depleted plasma. Baseline CEC was significantly lower in patients developing NODAT during follow-up (median 6.84% [interquartile range 5.84-7.50%]) compared with the NODAT-free group (7.44% [6.46-8.60%]; P = 0.001). Kaplan-Meier analysis showed a lower risk for incident NODAT with increasing sex-stratified tertiles of HDL efflux capacity (P = 0.004). Linear regression analysis indicated that CEC is independently associated with incident NODAT (P = 0.04). In Cox regression analyses, CEC was significantly associated with NODAT (hazard ratio 0.53 [95% CI 0.38-0.76]; P < 0.001), independent of HDL cholesterol levels (P = 0.015), adiposity (P = 0.018), immunosuppressive medication (P = 0.001), and kidney function (P = 0.01). Addition of CEC significantly improved the predictive power of the Framingham Diabetes Risk Score (P = 0.004). This study establishes HDL CEC as a strong predictor of NODAT in RTRs, independent of several other recognized risk factors.
Collapse
Affiliation(s)
- Tamas Szili-Torok
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Wijtske Annema
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Josephine L C Anderson
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Uwe J F Tietge
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
40
|
Sotomayor CG, Gomes-Neto AW, van Londen M, Gans ROB, Nolte IM, Berger SP, Navis GJ, Rodrigo R, Leuvenink HGD, Schalkwijk CG, Bakker SJL. Circulating Advanced Glycation Endproducts and Long-Term Risk of Cardiovascular Mortality in Kidney Transplant Recipients. Clin J Am Soc Nephrol 2019; 14:1512-1520. [PMID: 31530552 PMCID: PMC6777589 DOI: 10.2215/cjn.00540119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 07/10/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND OBJECTIVES In kidney transplant recipients, elevated circulating advanced glycation endproducts (AGEs) are the result of increased formation and decreased kidney clearance. AGEs trigger several intracellular mechanisms that ultimately yield excess cardiovascular disease. We hypothesized that, in stable kidney transplant recipients, circulating AGEs are associated with long-term risk of cardiovascular mortality, and that such a relationship is mediated by inflammatory, oxidative stress, and endothelial dysfunction biomarkers. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Prospective cohort study of stable kidney transplant recipients recruited between 2001 and 2003 in a university setting. We performed multivariable-adjusted Cox regression analyses to assess the association of AGEs (i.e., Nε -[Carboxymethyl]lysine (CML) and Nε -[Carboxyethyl]lysine (CEL), measured by tandem mass spectrometry) with cardiovascular mortality. Mediation analyses were performed according to Preacher and Hayes's procedure. RESULTS We included 555 kidney transplant recipients (age 51±12 years, 56% men). During a median follow-up of 6.9 years, 122 kidney transplant recipients died (52% deaths were due to cardiovascular causes). CML and CEL concentrations were directly associated with cardiovascular mortality (respectively, hazard ratio, 1.55; 95% confidence interval, 1.24 to 1.95; P<0.001; and hazard ratio, 1.53; 95% confidence interval 1.18 to 1.98; P=0.002), independent of age, diabetes, smoking status, body mass index, eGFR and proteinuria. Further adjustments, including cardiovascular history, did not materially change these findings. In mediation analyses, free thiol groups and soluble vascular cell adhesion molecule-1 consistently explained approximately 35% of the association of CML and CEL with cardiovascular mortality. CONCLUSIONS In stable kidney transplant recipients, circulating levels of AGEs are independently associated with long-term risk of cardiovascular mortality. PODCAST This article contains a podcast at https://www.asn-online.org/media/podcast/CJASN/2019_09_17_CJN00540119.mp3.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ramón Rodrigo
- Faculty of Medicine, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; and
| | - Henri G D Leuvenink
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine and.,Cardiovascular Research Institute Maastricht School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | | |
Collapse
|
41
|
Ebtehaj S, Gruppen EG, Bakker SJ, Dullaart RP, Tietge UJ. HDL (High-Density Lipoprotein) Cholesterol Efflux Capacity Is Associated With Incident Cardiovascular Disease in the General Population. Arterioscler Thromb Vasc Biol 2019; 39:1874-1883. [DOI: 10.1161/atvbaha.119.312645] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Objective:
Focus is shifting from HDL-C (high-density lipoprotein cholesterol) as predictive biomarker for cardiovascular disease (CVD) towards antiatherogenic HDL functionalities. Still, limited data exist on the prospective association of HDL function metrics with CVD events. The current work aimed to determine, if baseline HDL-C efflux capacity (CEC) is associated with future CVD events in the general population.
Approach and Results:
We performed a prospective study among participants of the PREVEND (Prevention of Renal and Vascular End-stage Disease) cohort (follow-up, 12 years). From the overall n=8592 subjects 325 with previous CVD events were excluded; of the remaining 8267 eligible participants all subjects with new CVD events during follow-up were selected and individually matched to controls for age, sex, smoking status, and HDL-C levels. CEC at baseline was quantified using human THP-1-derived macrophage foam cells and apolipoprotein B-depleted plasma. Despite identical HDL-C and apoA (apolipoprotein)-I levels between cases (n=351) and controls (n=354) CEC was significantly lower in cases (0.93±0.29 versus 1.01±0.24 arbitrary units;
P
<0.001). In all subjects combined, CEC correlated positively with HDL-C and apoA-I and negatively with body mass index, hsCRP (high-sensitivity C-reactive protein), and urinary albumin excretion. CEC was inversely associated with incident CVD events, both expressed per quartile and per 1 SD change (odds ratio, 0.73; 95% CI, 0.62–0.86;
P
<0.001); this association remained significant after adjustments for HDL-C, hsCRP, kidney function, and several other clinical covariates.
Conclusions:
Combined these data demonstrate that in the general population baseline CEC is significantly associated with the future development of CVD events independent of HDL-C and apoA-I plasma levels.
Collapse
Affiliation(s)
- Sanam Ebtehaj
- From the Department of Pediatrics (S.E., U.J.F.T.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Eke G. Gruppen
- Department of Endocrinology (E.G.G., R.P.F.D.), University of Groningen, University Medical Center Groningen, the Netherlands
- Department of Nephrology (E.G.G., S.J.L.B.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Stephan J.L. Bakker
- Department of Nephrology (E.G.G., S.J.L.B.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Robin P.F. Dullaart
- Department of Endocrinology (E.G.G., R.P.F.D.), University of Groningen, University Medical Center Groningen, the Netherlands
| | - Uwe J.F. Tietge
- From the Department of Pediatrics (S.E., U.J.F.T.), University of Groningen, University Medical Center Groningen, the Netherlands
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (U.J.F.T.)
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Sweden (U.J.F.T.)
| |
Collapse
|
42
|
Kim JE, Yu MY, Kim YC, Min SI, Ha J, Lee JP, Kim DK, Oh KH, Joo KW, Ahn C, Kim YS, Lee H. Ratio of triglyceride to high-density lipoprotein cholesterol and risk of major cardiovascular events in kidney transplant recipients. Clin Exp Nephrol 2019; 23:1407-1417. [PMID: 31468232 PMCID: PMC6848440 DOI: 10.1007/s10157-019-01776-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 08/12/2019] [Indexed: 01/23/2023]
Abstract
BACKGROUND Dyslipidemia is common in kidney transplant (KT) recipients. We analyzed the ratio of triglyceride to high-density lipoprotein cholesterol (TG/HDL-C) in KT recipients to identify risk factors for major cardiovascular events (MACE). METHODS We retrospectively included KT recipients with a lipid profile performed 1 year after transplantation. We classified patients according to the TG/HDL-C divided into quintiles. Subsequently, we analyzed the association between TG/HDL-C and MACE, defined as heart failure, coronary artery disease, and cerebrovascular disease confirmed by imaging studies. RESULTS A total of 1301 KT recipients were enrolled. The median follow-up duration was 7.4 years (interquartile range 4.4-11.1 years). During the follow-up period, 80 (6.2%) patients developed MACE, which included 38 of unstable anginas, 9 of MIs, 19 of heart failures, 18 of cerebral infarcts, and 4 of cerebral hemorrhages. The fourth and fifth quintiles of TG/HDL-C showed a significantly increased risk of MACE [fourth quintile: adjusted hazard ratio (aHR), 3.38; 95% confidence interval (CI) 1.44-7.95; p = 0.005, fifth quintile: aHR, 2.67; 95% CI 1.13-6.30; p = 0.02]) compared to the second quintile of TG/HDL-C. This association is particularly evident in subgroups of non-DM, HTN, no history of CVD, and statin users. CONCLUSIONS Higher TG/HDL-C levels may be associated with MACE risk in KT recipients.
Collapse
Affiliation(s)
- Ji Eun Kim
- Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehakro, Jongno-gu, Seoul, 03080, South Korea
| | - Mi-Yeon Yu
- Department of Internal Medicine, Hanyang University Guri Hospital, Guri, South Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehakro, Jongno-gu, Seoul, 03080, South Korea
| | - Sang-Il Min
- Department of Surgery, Seoul National University Transplantation Research Laboratory, Seoul National University Hospital, Seoul, South Korea
| | - Jongwon Ha
- Department of Surgery, Seoul National University Transplantation Research Laboratory, Seoul National University Hospital, Seoul, South Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehakro, Jongno-gu, Seoul, 03080, South Korea.,Kidney Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehakro, Jongno-gu, Seoul, 03080, South Korea
| | - Kwon-Wook Joo
- Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehakro, Jongno-gu, Seoul, 03080, South Korea.,Kidney Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Curie Ahn
- Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehakro, Jongno-gu, Seoul, 03080, South Korea.,Kidney Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehakro, Jongno-gu, Seoul, 03080, South Korea.,Kidney Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Hajeong Lee
- Department of Internal Medicine, Seoul National University College of Medicine, 103 Daehakro, Jongno-gu, Seoul, 03080, South Korea. .,Kidney Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
43
|
Suematsu Y, Kawachi E, Idemoto Y, Matsuo Y, Kuwano T, Kitajima K, Imaizumi S, Kawamura A, Saku K, Uehara Y, Miura SI. Anti-atherosclerotic effects of an improved apolipoprotein A-I mimetic peptide. Int J Cardiol 2019; 297:111-117. [PMID: 31519377 DOI: 10.1016/j.ijcard.2019.08.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/01/2019] [Accepted: 08/20/2019] [Indexed: 01/23/2023]
Abstract
BACKGROUND Apolipoprotein (Apo)A-I is a major protein component of high-density lipoprotein (HDL) that causes cholesterol efflux from peripheral cells through ATP-binding cassette transporter A1 (ABCA1) and the generation of HDL. Furthermore, it has a possible protective function against atherosclerotic cardiovascular disease (ASCVD). We previously developed a novel ApoA-I mimetic peptide without phospholipids (Fukuoka University ApoA-I Mimetic Peptide, FAMP). According to our previous studies, FAMP had an anti-arteriosclerotic effect. Since the required dose and reaction time of conventional FAMP were relatively large and short, respectively, we newly developed an improved FAMP (i-FAMP). METHODS AND RESULTS We synthesized four candidate i-FAMPs, i-FAMP-D1, -D2, -D3 and -D4, and examined which i-FAMP has greater cholesterol efflux capacity than FAMP in A172 human glioblastoma cells transiently transfected with human ABCA1 cDNA. Only i-FAMP-D1 showed significantly greater cholesterol efflux capacity than conventional FAMP. i-FAMP-D1 formed stronger α-helical conformations than FAMP as assessed by circular dichroism spectra. Thus, we selected i-FAMP-D1 for further experiments. i-FAMP-D1 had a greater atheroprotective effect than FAMP in ApoE knockout mice. In addition, i-FAMP-D1 activated cholesterol efflux from macrophage to HDL more strongly than FAMP and increased cholesterol excretion from liver to feces. CONCLUSION These results suggest that i-FAMP-D1 has a stronger anti-atherosclerotic effect than conventional FAMP.
Collapse
Affiliation(s)
- Yasunori Suematsu
- Department of Cardiology, Fukuoka University School of Medicine, Japan
| | - Emi Kawachi
- Clinical Research and Ethics Center, Fukuoka University School of Medicine, Japan
| | - Yoshiaki Idemoto
- Department of Cardiology, Fukuoka University School of Medicine, Japan
| | - Yoshino Matsuo
- Department of Cardiology, Fukuoka University School of Medicine, Japan
| | - Takashi Kuwano
- Department of Cardiology, Fukuoka University School of Medicine, Japan
| | - Ken Kitajima
- Department of Cardiology, Fukuoka University School of Medicine, Japan
| | - Satoshi Imaizumi
- Clinical Research and Ethics Center, Fukuoka University School of Medicine, Japan
| | - Akira Kawamura
- Center for Graduate Clinical Practice, Fukuoka University Hospital, Fukuoka, Japan
| | - Keijiro Saku
- General Medical Research Center, Fukuoka University School of Medicine, Japan
| | - Yoshinari Uehara
- Graduate School of Sports and Health Sciences, Fukuoka University, Fukuoka, Japan.
| | - Shin-Ichiro Miura
- Department of Cardiology, Fukuoka University School of Medicine, Japan.
| |
Collapse
|
44
|
Lipoprotein modulation of proteinuric renal injury. J Transl Med 2019; 99:1107-1116. [PMID: 31019291 PMCID: PMC6658349 DOI: 10.1038/s41374-019-0253-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/08/2019] [Accepted: 03/04/2019] [Indexed: 12/14/2022] Open
Abstract
High-density lipoprotein (HDL) and its main protein, apolipoprotein AI (apoAI), have established benefits in various cells, but whether these cytoprotective effects of HDL pertain to renal cells is unclear. We investigated the in vitro consequences of exposing damaged podocytes to normal apoAI, HDL, and apoAI mimetic (L-4F), and the in vivo effects of L-4F on kidney and atherosclerotic injury in a podocyte-specific injury model of proteinuria. In vitro, primary mouse podocytes were injured by puromycin aminonucleoside (PAN). Cellular viability, migration, production of reactive oxygen species (ROS), apoptosis, and the underlying signaling pathway were assessed. In vivo, we used a proteinuric model, Nphs1-hCD25 transgenic (NEP25+) mice, which express human CD25 on podocytes. Podocyte injury was induced by using immunotoxin (LMB2) and generated a proteinuric atherosclerosis model, NEP25+:apoE-/- mice, was generated by mating apoE-deficient (apoE-/-) mice with NEP25+ mice. Animals received L-4F or control vehicle. Renal function, podocyte injury, and atherosclerosis were assessed. PAN reduced podocyte viability, migration, and increased ROS production, all significantly lessened by apoAI, HDL, and L-4F. L-4F attenuated podocyte apoptosis and diminished PAN-induced inactivation of Janus family protein kinase-2/signal transducers and activators of transcription 3. In NEP25+ mice, L-4F significantly lessened overall proteinuria, and preserved podocyte expression of synaptopodin and cell density. Proteinuric NEP25+:apoE-/- mice had more atherosclerosis than non-proteinuric apoE-/- mice, and these lesions were significantly decreased by L-4F. Normal human apoAI, HDL, and apoAI mimetic protect against podocyte damage. ApoAI mimetic provides in vivo beneficial effects on podocytes that culminate in reduced albuminuria and atherosclerosis. The results suggest supplemental apoAI/apoAI mimetic may be a novel candidate to lessen podocyte damage and its complications.
Collapse
|
45
|
Anderson JLC, Pagano S, Virzi J, Dullaart RPF, Annema W, Kuipers F, Bakker SJL, Vuilleumier N, Tietge UJF. Autoantibodies to Apolipoprotein A-1 as Independent Predictors of Cardiovascular Mortality in Renal Transplant Recipients. J Clin Med 2019; 8:jcm8070948. [PMID: 31261925 PMCID: PMC6679113 DOI: 10.3390/jcm8070948] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/24/2019] [Accepted: 06/27/2019] [Indexed: 12/15/2022] Open
Abstract
Renal transplant recipients (RTRs) are known to have a high cardio-vascular disease (CVD) burden only partly explained by traditional CVD risk factors. The aim of this paper was therefore to determine: i) the prognostic value of autoantibodies against apoA-1 (anti-apoA-1 IgG) for incidence of CVD mortality, all-cause mortality and graft failure in RTR. Four hundred and sixty two (462) prospectively included RTRs were followed for 7.0 years. Baseline anti-apoA-1 IgG were determined and associations with incidence of CVD mortality (n = 48), all-cause mortality (n = 92) and graft failure (n = 39) were tested. Kaplan-Meier analyses demonstrated significant associations between tertiles of anti-apoA-1 IgG and CVD mortality (log rank test: p = 0.048). Adjusted Cox regression analysis showed a 54% increase in risk for CVD mortality for each anti-apoA-1 IgG levels standard deviation increase (hazard ratio [HR]: 1.54, 95% Confidence Interval [95%CI]: 1.14-2.05, p = 0.005), and a 33% increase for all-cause mortality (HR: 1.33; 95%CI: 1.06-1.67, p = 0.01), independent of CVD risk factors, renal function and HDL function. The association with all-cause mortality disappeared after excluding cases of CVD specific mortality. The sensitivity, specificity, positive predictive value, and negative predictive value of anti-apoA-1 positivity for CVD mortality were 18.0%, 89.3%, 17.0%, and 90.0%, respectively. HDL functionality was not associated with anti-apoA-1 IgG levels. This prospective study demonstrates that in RTR, anti-apoA-1 IgG are independent predictors of CVD mortality and are not associated with HDL functionality.
Collapse
Affiliation(s)
- Josephine L C Anderson
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 1205 Groningen, The Netherlands
| | - Sabrina Pagano
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospital, 1205 Geneva, Switzerland
- Department of Medical Specialties, Faculty of Medicine, Geneva University, 1205 Geneva, Switzerland
| | - Julien Virzi
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospital, 1205 Geneva, Switzerland
- Department of Medical Specialties, Faculty of Medicine, Geneva University, 1205 Geneva, Switzerland
| | - Robin P F Dullaart
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Wijtske Annema
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 1205 Groningen, The Netherlands
- Institute of Clinical Chemistry, University Hospital of Zurich and University of Zurich, 8006 Zurich, Switzerland
| | - Folkert Kuipers
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 1205 Groningen, The Netherlands
- Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Stephan J L Bakker
- Department of Nephrology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospital, 1205 Geneva, Switzerland
| | - Uwe J F Tietge
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, 1205 Groningen, The Netherlands.
- Division of Clinical Chemistry, Department of Laboratory Medicine H5, Karolinska Institutet, 14183 Stockholm, Sweden.
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, SE-141 86 Stockholm, Sweden.
| |
Collapse
|
46
|
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
|
47
|
van den Berg EH, Gruppen EG, Ebtehaj S, Bakker SJL, Tietge UJF, Dullaart RPF. Cholesterol efflux capacity is impaired in subjects with an elevated Fatty Liver Index, a proxy of non-alcoholic fatty liver disease. Atherosclerosis 2018; 277:21-27. [PMID: 30170220 DOI: 10.1016/j.atherosclerosis.2018.07.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/06/2018] [Accepted: 07/19/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Non-alcoholic fatty liver disease (NAFLD) parallels the obesity epidemic and associates with components of the metabolic syndrome (MetS). Cholesterol efflux capacity (CEC) represents a key metric of high density lipoprotein (HDL) function which may predict atherosclerotic cardiovascular disease (CVD). Here we assessed the relationship of CEC with NAFLD. METHODS CEC was determined from THP-1 macrophage foam cells towards apolipoprotein B-depleted plasma among 639 subjects (454 men; 36 subjects with type 2 diabetes mellitus (T2D); 226 with MetS), participating in the Prevention of Renal and Vascular End-Stage Disease (PREVEND) study. A Fatty Liver Index (FLI) ≥ 60 was used as a proxy of NAFLD. RESULTS 372 participants had a FLI ≥60, which coincided with an increased prevalence of T2D and MetS (p = 0.009 and p < 0.001), as well as with central obesity, higher systolic blood pressure, glucose, total cholesterol, triglycerides and high sensitivity C-reactive protein (hsCRP), and decreased HDL cholesterol (p < 0.001 for each). In multivariable linear regression analyses, CEC was inversely associated with an elevated FLI, when taking account of clinical covariates (fully adjusted model: β = -0.091, p = 0.043), and alternatively when taking account of systolic blood pressure, waist/hip ratio, glucose, HDL cholesterol, triglycerides and hsCRP (fully adjusted model: β = -0.103, p = 0.034). CONCLUSIONS Impaired CEC is associated with NAFLD, as inferred from a FLI≥60, even when taking account of lower HDL cholesterol and enhanced low-grade chronic inflammation. Reduced CEC could contribute to accelerated CVD in NAFLD patients.
Collapse
Affiliation(s)
- Eline H van den Berg
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, The Netherlands; Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Eke G Gruppen
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, The Netherlands; Department of Nephrology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Sanam Ebtehaj
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Stephan J L Bakker
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Uwe J F Tietge
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - Robin P F Dullaart
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, The Netherlands.
| |
Collapse
|
48
|
Moradi H, Streja E, Vaziri ND. ESRD-induced dyslipidemia-Should management of lipid disorders differ in dialysis patients? Semin Dial 2018; 31:398-405. [PMID: 29707830 DOI: 10.1111/sdi.12706] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease (CVD) is a major cause of morbidity and mortality worldwide. Although numerous modifiable risk factors in the pathogenesis of CVD and its associated mortality have been identified, dyslipidemia remains to be a key focus for therapy. In this regard, significant progress has been made in reducing cardiovascular mortality via the use of lipid-lowering agents such as HMG CoA reductase inhibitors (statins). Yet, despite the disproportionate risk of CVD and mortality in patients with advanced chronic and end stage renal disease (ESRD), treatment of dyslipidemia in this patient population has not been associated with a notable improvement in outcomes. Furthermore, observational studies have not consistently found an association between dyslipidemia and poor outcomes in patients with ESRD. However, it is imperative that examination of dyslipidemia and its association with outcomes take place in the context of the many factors that are unique to kidney disease and may contribute to the abnormalities in lipid metabolism in patients with ESRD. Understanding these intricacies and distinct features will be vital not only to the interpretation of the available clinical data in regards to outcomes, but also to the individualization of lipid therapy in ESRD. In this review, we will examine the nature and underlying mechanisms responsible for dyslipidemia, the association of serum lipids and lipoprotein concentrations with outcomes and the results of major trials targeting cholesterol (mainly statins) in patients with ESRD.
Collapse
Affiliation(s)
- Hamid Moradi
- Division of Nephrology and Hypertension, Department of Medicine, University of California, Irvine, CA, USA.,Department of Medicine, Long Beach VA Healthcare System, Long Beach, CA, USA
| | - Elani Streja
- Division of Nephrology and Hypertension, Department of Medicine, University of California, Irvine, CA, USA.,Department of Medicine, Long Beach VA Healthcare System, Long Beach, CA, USA
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, Department of Medicine, University of California, Irvine, CA, USA
| |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW Cholesterol metabolism is increasingly recognized in inflammatory diseases including transplantation. This review discusses the mechanistic underpinnings that tie macrophage cholesterol efflux capacity (CEC) of high-density lipoprotein (HDL) to chronic rejection in transplanted patients. RECENT FINDINGS Animal studies suggest that administration of apolipoprotein A-I, the main protein constituent of HDL, can prevent transplant arteriosclerosis. apoA-I administration increases CEC of HDL. In patients with cardiac allograft vasculopathy (CAV), decreased CEC has been associated with poorer survival. In addition, reduced CEC in recipients, pretransplant, has been associated with the development of CAV and renal allograft survival. SUMMARY These recent findings raise the hypothesis that increasing cholesterol efflux may prevent chronic rejection and improve allograft survival after transplant. Reconstituted HDL significantly increases CEC and is currently in clinical development for traditional atherosclerosis. Clinical trials of reconstituted HDL administration in transplantation should be performed.
Collapse
|
50
|
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
|