1
|
Van Mulders L, Locquet L, Kaandorp C, Janssens GPJ. An overview of nutritional factors in the aetiopathogenesis of myocardial fibrosis in great apes. Nutr Res Rev 2025; 38:37-52. [PMID: 38343129 DOI: 10.1017/s0954422424000076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
The main cause of mortality in great apes in zoological settings is cardiovascular disease (CVD), affecting all four taxa: chimpanzee (Pan troglodytes), bonobo (Pan paniscus), gorilla (Gorilla spp.) and orangutan (Pongo spp.). Myocardial fibrosis, the most typical histological characterisation of CVD in great apes, is non-specific, making it challenging to understand the aetiopathogenesis. A multifactorial origin of disease is assumed whereby many potential causative factors are directly or indirectly related to the diet, which in wild-living great apes mainly consists of high-fibre, low-carbohydrate and very low-sodium components. Diets of great apes housed in zoological settings are often different compared with the situation in the wild. Moreover, low circulating vitamin D levels have recently been recognised in great apes housed in more northern regions. Evaluation of current supplementation guidelines shows that, despite implementation of different dietary strategies, animals stay vitamin D insufficient. Therefore, recent hypotheses designate vitamin D deficiency as a potential underlying factor in the pathogenesis of myocardial fibrosis. The aim of this literature review is to: (i) examine important differences in nutritional factors between zoological and wild great ape populations; (ii) explain the potential detrimental effects of the highlighted dietary discrepancies on cardiovascular function in great apes; and (iii) elucidate specific nutrition-related pathophysiological mechanisms that may underlie the development of myocardial fibrosis. This information may contribute to understanding the aetiopathogenesis of myocardial fibrosis in great apes and pave the way for future clinical studies and a more preventive approach to great ape CVD management.
Collapse
Affiliation(s)
- Laurens Van Mulders
- Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Royal Zoological Society of Antwerp (KMDA), Antwerpen, Belgium
| | - Laurent Locquet
- Department of Veterinary Medicine and Sciences, University of Notingham, Nottingham, UK
- Dick White Referrals, Cambridgeshire, UK
| | - Christine Kaandorp
- Safari Park Beekse Bergen, Hilvarenbeek, The Netherlands
- Gaia zoo, Kerkrade, The Netherlands
- Zooparc Overloon, Overloon, The Netherlands
- Dierenrijk, Mierlo, The Netherlands
| | | |
Collapse
|
2
|
Hu L, Li J, Wang W, Li Y, Gu B, Tian Y, Ruan Q, Deng J. Hypertension accelerates the development of a diabetic nephropathy model with more relevant clinical features in Dahl salt-sensitive rat. J Hypertens 2025:00004872-990000000-00676. [PMID: 40394981 DOI: 10.1097/hjh.0000000000004038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/31/2025] [Indexed: 05/22/2025]
Abstract
OBJECTIVE Diabetic nephropathy is becoming an increasing health problem and the major cause of chronic kidney disease. The lack of experimental models that reproduce all structural and functional alterations of human diabetic nephropathy is one of the barriers to solve these clinical needs in basic research. We aimed to establish a simple and rapid method for diabetic nephropathy induction in Dahl salt-sensitive rats and achieved more clinical relevant features. METHODS AND RESULTS Six or seven-week-old male Dahl salt-sensitive rats were used to induce diabetes by a single injection of streptozotocin (STZ; 45 mg/kg, intraperitoneally). After 7 days, rats with diabetes (will refer as STZ-DS rats) will be used for further studies. STZ-DS rats will be grouped and were fed with water or water containing 0.3%/0.9%/2% NaCl and a regular chow throughout the study. We found that STZ-treated Dahl salt-sensitive rat fed with 0.9% NaCl in drinking water (STZ-DS + 0.9% NaCl) displayed most of the characteristics of human diabetic nephropathy and the model meets the criteria of the diabetic nephropathy standards proposed by Animal Models of Diabetic Complications Consortium (AMDCC); which includes a 10-fold increase in albuminuria, more than 50% decline in estimated glomerular filtration rate over the lifetime, hypertrophy, thickening of the glomerular basement membrane, expansion of the mesangial matrix, any degree of arteriolar hyalinosis, severe glomerulosclerosis, and tubulointerstitial fibrosis. CONCLUSION STZ-DS + 0.9% NaCl is an improved diabetes nephropathy model and more suited for the study of the signaling pathways and mechanisms in the progression of diabetes-induced renal disease. The model will facilitate the development of new therapies for diabetes nephropathy.
Collapse
Affiliation(s)
| | - Jing Li
- Sunshine Lake Pharma Co., Ltd
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Sunshine Lake Pharma Co., Ltd, Dongguan, Guangdong, China
| | | | - Yong Li
- Sunshine Lake Pharma Co., Ltd
| | | | | | | | | |
Collapse
|
3
|
Secondulfo C, Visco V, Virtuoso N, Fortunato M, Migliarino S, Rispoli A, La Mura L, Stellato A, Caliendo G, Settembre E, Galluccio F, Hamzeh S, Bilancio G. Vitamin D: A Bridge between Kidney and Heart. Life (Basel) 2024; 14:617. [PMID: 38792638 PMCID: PMC11123235 DOI: 10.3390/life14050617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/30/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Chronic kidney disease (CKD) and cardiovascular disease (CVD) are highly prevalent conditions, each significantly contributing to the global burden of morbidity and mortality. CVD and CKD share a great number of common risk factors, such as hypertension, diabetes, obesity, and smoking, among others. Their relationship extends beyond these factors, encompassing intricate interplay between the two systems. Within this complex network of pathophysiological processes, vitamin D has emerged as a potential linchpin, exerting influence over diverse physiological pathways implicated in both CKD and CVD. In recent years, scientific exploration has unveiled a close connection between these two prevalent conditions and vitamin D, a crucial hormone traditionally recognized for its role in bone health. This article aims to provide an extensive review of vitamin D's multifaceted and expanding actions concerning its involvement in CKD and CVD.
Collapse
Affiliation(s)
- Carmine Secondulfo
- Department “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| | - Valeria Visco
- Department “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| | - Nicola Virtuoso
- Cardiology Unit, Salerno University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Martino Fortunato
- Cardiology Unit, Salerno University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Serena Migliarino
- Cardiology Unit, Salerno University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Antonella Rispoli
- Cardiology Unit, Salerno University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Lucia La Mura
- Centro Medico Ascione Srl, 80059 Torre del Greco, Italy
| | - Adolfo Stellato
- Department “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| | - Giuseppe Caliendo
- Department “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| | - Emanuela Settembre
- Department “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
| | - Fabiana Galluccio
- Department of Medicine and Surgery, University of Naples “Federico II”, 80138 Napoli, Italy
| | - Sarah Hamzeh
- Department of Medicine and Surgery, University of Naples “Federico II”, 80138 Napoli, Italy
| | - Giancarlo Bilancio
- Department “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy
- Nephrology Unit, Salerno University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| |
Collapse
|
4
|
Hyeon J, Kim S, Ye BM, Kim SR, Lee DW, Lee SB, Kim IY. Association of 1,25 dihydroxyvitamin D with left ventricular hypertrophy and left ventricular diastolic dysfunction in patients with chronic kidney disease. PLoS One 2024; 19:e0302849. [PMID: 38722953 PMCID: PMC11081214 DOI: 10.1371/journal.pone.0302849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 04/11/2024] [Indexed: 05/13/2024] Open
Abstract
Left ventricular hypertrophy (LVH) and left ventricular diastolic dysfunction (LVDD) are highly prevalent predictors of cardiovascular disease in individuals with chronic kidney disease (CKD). Vitamin D, particularly 25-hydroxyvitamin D [25(OH)D], deficiency has been reported to be associated with cardiac structure and function in CKD patients. In the current study, we investigated the association between 1,25-dihydroxyvitamin D [1,25(OH)2D], the active form of 25(OH)D, and LVH/LVDD in CKD patients. We enrolled 513 non-dialysis CKD patients. The presence of LVH and LVDD was determined using transthoracic echocardiography. In multivariable analysis, serum 1,25(OH)2D levels, but not serum 25(OH)D, were independently associated with LVH [odds ratio (OR): 0.90, 95% confidential interval (CI): 0.88-0.93, P < 0.001]. Additionally, age, systolic blood pressure, and intact parathyroid hormone levels were independently associated with LVH. Similarly, multivariable analysis demonstrated that serum 1,25(OH)2D levels, but not 25(OH)D levels, were independently associated with LVDD (OR: 0.88, 95% CI: 0.86-0.91, P < 0.001) with systolic blood pressure showing independent association with LVDD. The optimal cut-off values for serum 1,25(OH)2D levels for identifying LVH and LVDD were determined as ≤ 12.7 pg/dl and ≤ 18.1 pg/dl, respectively. Our findings suggest that serum 1,25(OH)2D levels have independent association with LVH and LVDD in CKD patients, underscoring their potential as biomarkers for these conditions in this patient population.
Collapse
Affiliation(s)
- Jemin Hyeon
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Suji Kim
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Byung Min Ye
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Seo Rin Kim
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Dong Won Lee
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Soo Bong Lee
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Il Young Kim
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| |
Collapse
|
5
|
Padoan F, Guarnaroli M, Brugnara M, Piacentini G, Pietrobelli A, Pecoraro L. Role of Nutrients in Pediatric Non-Dialysis Chronic Kidney Disease: From Pathogenesis to Correct Supplementation. Biomedicines 2024; 12:911. [PMID: 38672265 PMCID: PMC11048674 DOI: 10.3390/biomedicines12040911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/05/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Nutrition management is fundamental for children with chronic kidney disease (CKD). Fluid balance and low-protein and low-sodium diets are the more stressed fields from a nutritional point of view. At the same time, the role of micronutrients is often underestimated. Starting from the causes that could lead to potential micronutrient deficiencies in these patients, this review considers all micronutrients that could be administered in CKD to improve the prognosis of this disease.
Collapse
Affiliation(s)
| | | | - Milena Brugnara
- Pediatric Unit, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37126 Verona, Italy (A.P.)
| | | | | | | |
Collapse
|
6
|
Kallmeyer A, Pello A, Cánovas E, Aceña Á, González‐Casaus ML, Tarín N, Cristóbal C, Gutiérrez‐Landaluce C, Huelmos A, Rodríguez‐Valer A, González‐Lorenzo Ó, Alonso J, López‐Bescós L, Egido J, Mahillo I, Lorenzo Ó, Tuñón J. Fibroblast growth factor 23 independently predicts adverse outcomes after an acute coronary syndrome. ESC Heart Fail 2024; 11:240-250. [PMID: 37950429 PMCID: PMC10804179 DOI: 10.1002/ehf2.14568] [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: 07/20/2023] [Revised: 09/10/2023] [Accepted: 10/08/2023] [Indexed: 11/12/2023] Open
Abstract
AIMS Abnormalities of mineral metabolism (MM) have been related to cardiovascular disorders. There are no reports on the prognostic role of MM after an acute coronary syndrome (ACS). We aim to assess the prognostic role of MM after an ACS. METHODS AND RESULTS Plasma levels of components of MM [fibroblast growth factor 23 (FGF23), calcidiol, parathormone, klotho, and phosphate], high-sensitivity C-reactive protein, and N-terminal-pro-brain natriuretic peptide were measured in 1190 patients at discharge from an ACS. The primary outcome was a combination of acute ischaemic events, heart failure (HF) and death. Secondary outcomes were the separate components of the primary outcome. Age was 61.7 ± 12.2 years, and 77.1% were men. Median follow-up was 5.44 (3.03-7.46) years. Two hundred and ninety-four patients developed the primary outcome. At multivariable analysis FGF23 (hazard ratio, HR 1.18 [1.08-1.29], P < 0.001), calcidiol (HR 0.86 [0.74-1.00], P = 0.046), previous coronary or cerebrovascular disease, and hypertension were independent predictors of the primary outcome. The predictive power of FGF23 was homogeneous across different subgroups of population. FGF23 (HR 1.45 [1.28-1.65], P < 0.001) and parathormone (HR 1.06 1.01-1.12]; P = 0.032) resulted as independent predictors of HF. FGF23 (HR 1.21 [1.07-1.37], P = 0.002) and calcidiol (HR 0.72 [0.54-0.97), P = 0.028) were independent predictors of death. No biomarker predicted acute ischaemic events. FGF23 predicted independently the primary outcome in patients with estimated glomerular filtration rate > 60 mL/min/1.73 m2 . CONCLUSIONS FGF23 and other components of MM are independent predictors of HF and death after an ACS. This effect is homogeneous across different subgroups of population, and it is not limited to patients with chronic kidney disease.
Collapse
Affiliation(s)
- Andrea Kallmeyer
- Department of CardiologyIIS‐Fundación Jiménez DíazMadridSpain
- Faculty of MedicineAutónoma UniversityMadridSpain
| | - Ana Pello
- Department of CardiologyIIS‐Fundación Jiménez DíazMadridSpain
- Faculty of MedicineAutónoma UniversityMadridSpain
| | - Ester Cánovas
- Department of CardiologyIIS‐Fundación Jiménez DíazMadridSpain
| | - Álvaro Aceña
- Department of CardiologyIIS‐Fundación Jiménez DíazMadridSpain
- Faculty of MedicineAutónoma UniversityMadridSpain
| | | | - Nieves Tarín
- Department of CardiologyHospital Universitario de MóstolesMadridSpain
- Faculty of MedicineRey Juan Carlos UniversityMadridSpain
| | - Carmen Cristóbal
- Faculty of MedicineRey Juan Carlos UniversityMadridSpain
- Department of CardiologyHospital Universitario de FuenlabradaMadridSpain
| | | | - Ana Huelmos
- Department of CardiologyHospital Universitario Fundación AlcorcónMadridSpain
| | | | - Óscar González‐Lorenzo
- Department of CardiologyIIS‐Fundación Jiménez DíazMadridSpain
- Faculty of MedicineAutónoma UniversityMadridSpain
| | | | | | - Jesús Egido
- CIBERDEMMadridSpain
- Department of NephrologyIIS‐Fundación Jiménez DíazMadridSpain
- Laboratory of Vascular PathologyIIS‐Fundación Jiménez DíazMadridSpain
| | - Ignacio Mahillo
- Laboratory of EpidemiologyIIS‐Fundación Jiménez DíazMadridSpain
| | - Óscar Lorenzo
- Faculty of MedicineAutónoma UniversityMadridSpain
- CIBERDEMMadridSpain
- Laboratory of Vascular PathologyIIS‐Fundación Jiménez DíazMadridSpain
| | - José Tuñón
- Department of CardiologyIIS‐Fundación Jiménez DíazMadridSpain
- Faculty of MedicineAutónoma UniversityMadridSpain
- Laboratory of Vascular PathologyIIS‐Fundación Jiménez DíazMadridSpain
- CIBERCV, ISCIIIMadridSpain
| |
Collapse
|
7
|
Arroyo E, Leber CA, Burney HN, Li Y, Li X, Lu TS, Jones G, Kaufmann M, Ting SMS, Hiemstra TF, Zehnder D, Lim K. Epimeric vitamin D and cardiovascular structure and function in advanced CKD and after kidney transplantation. Nephrol Dial Transplant 2024; 39:264-276. [PMID: 37468453 PMCID: PMC10828205 DOI: 10.1093/ndt/gfad168] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND 25-hydroxyvitamin D can undergo C-3 epimerization to produce 3-epi-25(OH)D3. 3-epi-25(OH)D3 levels decline in chronic kidney disease (CKD), but its role in regulating the cardiovascular system is unknown. Herein, we examined the relationship between 3-epi-25(OH)D3, and cardiovascular functional and structural endpoints in patients with CKD. METHODS We examined n = 165 patients with advanced CKD from the Cardiopulmonary Exercise Testing in Renal Failure and After Kidney Transplantation (CAPER) study cohort, including those who underwent kidney transplant (KTR, n = 76) and waitlisted patients who did not (NTWC, n = 89). All patients underwent cardiopulmonary exercise testing and echocardiography at baseline, 2 months and 12 months. Serum 3-epi-25(OH)D3 was analyzed by liquid chromatography-tandem mass spectrometry. RESULTS Patients were stratified into quartiles of baseline 3-epi-25(OH)D3 (Q1: <0.4 ng/mL, n = 51; Q2: 0.4 ng/mL, n = 26; Q3: 0.5-0.7 ng/mL, n = 47; Q4: ≥0.8 ng/mL, n = 41). Patients in Q1 exhibited lower peak oxygen uptake [VO2Peak = 18.4 (16.2-20.8) mL/min/kg] compared with Q4 [20.8 (18.6-23.2) mL/min/kg; P = .009]. Linear mixed regression model showed that 3-epi-25(OH)D3 levels increased in KTR [from 0.47 (0.30) ng/mL to 0.90 (0.45) ng/mL] and declined in NTWC [from 0.61 (0.32) ng/mL to 0.45 (0.29) ng/mL; P < .001]. Serum 3-epi-25(OH)D3 was associated with VO2Peak longitudinally in both groups [KTR: β (standard error) = 2.53 (0.56), P < .001; NTWC: 2.73 (0.70), P < .001], but was not with left ventricular mass or arterial stiffness. Non-epimeric 25(OH)D3, 24,25(OH)2D3 and the 25(OH)D3:24,25(OH)2D3 ratio were not associated with any cardiovascular outcome (all P > .05). CONCLUSIONS Changes in 3-epi-25(OH)D3 levels may regulate cardiovascular functional capacity in patients with advanced CKD.
Collapse
Affiliation(s)
- Eliott Arroyo
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cecilia A Leber
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Heather N Burney
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yang Li
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xiaochun Li
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tzong-shi Lu
- Renal Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Glenville Jones
- Department of Biomedical and Molecular Sciences and Medicine, Queen's University, Kingston, Ontario, Canada
| | - Martin Kaufmann
- Department of Biomedical and Molecular Sciences and Medicine, Queen's University, Kingston, Ontario, Canada
| | - Stephen M S Ting
- Department of Medicine, University Hospitals Birmingham National Health Service Foundation Trust, Birmingham, UK
| | - Thomas F Hiemstra
- Cambridge Clinical Trials Unit, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, UK
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Daniel Zehnder
- Department of Nephrology
- Department of Acute Medicine, North Cumbria University Hospital National Health Service Trust, Carlisle, UK
| | - Kenneth Lim
- Division of Nephrology & Hypertension, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
8
|
Saito T, Mizobuchi M, Kato T, Ogata H, Koiwa F, Honda H. Fibroblast Growth Factor 23 Exacerbates Cardiac Fibrosis in Deoxycorticosterone Acetate-Salt Mice With Hypertension. J Transl Med 2023; 103:100003. [PMID: 36748187 DOI: 10.1016/j.labinv.2022.100003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/26/2022] [Accepted: 09/20/2022] [Indexed: 01/18/2023] Open
Abstract
Fibroblast growth factor 23 (FGF23) is associated with cardiovascular disease in patients with chronic kidney disease; however, the mechanisms underlying the effect of FGF23 on cardiac function remain to be investigated. Herein, we studied the effect of continuous intravenous (CIV) FGF23 loading in a deoxycorticosterone acetate (DOCA)-salt mouse model with mild chronic kidney disease and hypertension as well as heart failure with a preserved ejection fraction. Wild-type male mice were randomly allocated to 4 groups: normal control, vehicle-treated DOCA-salt mice, FGF23-treated DOCA-salt mice, and FGF23- and calcitriol-treated DOCA-salt mice. The DOCA-salt mice received the agents via the CIV route for 10 days using an infusion minipump. DOCA-salt mice that received FGF23 showed a marked increase in the serum FGF23 level, and echocardiography in these mice revealed heart failure with a preserved ejection fraction. These mice also showed exacerbation of myocardial fibrosis, concomitant with an inverse and significant correlation with Cyp27b1 expression. Calcitriol treatment attenuated FGF23-induced cardiac fibrosis and improved diastolic function via inhibition of transforming growth factor-β signaling. This effect was independent of the systemic and local levels of FGF23. These results suggest that CIV FGF23 loading exacerbates cardiac fibrosis and that locally abnormal vitamin D metabolism is involved in this mechanism. Calcitriol attenuates this exacerbation by mediating transforming growth factor-β signaling independently of the FGF23 levels.
Collapse
Affiliation(s)
- Tomohiro Saito
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Masahide Mizobuchi
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan.
| | - Tadashi Kato
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hiroaki Ogata
- Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
| | - Fumihiko Koiwa
- Division of Nephrology, Department of Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Hirokazu Honda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
9
|
Hypertension and cardiomyopathy associated with chronic kidney disease: epidemiology, pathogenesis and treatment considerations. J Hum Hypertens 2023; 37:1-19. [PMID: 36138105 PMCID: PMC9831930 DOI: 10.1038/s41371-022-00751-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/09/2022] [Accepted: 08/31/2022] [Indexed: 01/31/2023]
Abstract
Chronic kidney disease (CKD) is a complex condition with a prevalence of 10-15% worldwide. An inverse-graded relationship exists between cardiovascular events and mortality with kidney function which is independent of age, sex, and other risk factors. The proportion of deaths due to heart failure and sudden cardiac death increase with progression of chronic kidney disease with relatively fewer deaths from atheromatous, vasculo-occlusive processes. This phenomenon can largely be explained by the increased prevalence of CKD-associated cardiomyopathy with worsening kidney function. The key features of CKD-associated cardiomyopathy are increased left ventricular mass and left ventricular hypertrophy, diastolic and systolic left ventricular dysfunction, and profound cardiac fibrosis on histology. While these features have predominantly been described in patients with advanced kidney disease on dialysis treatment, patients with only mild to moderate renal impairment already exhibit structural and functional changes consistent with CKD-associated cardiomyopathy. In this review we discuss the key drivers of CKD-associated cardiomyopathy and the key role of hypertension in its pathogenesis. We also evaluate existing, as well as developing therapies in the treatment of CKD-associated cardiomyopathy.
Collapse
|
10
|
Chandra A, Picard MH, Huang S, Gupta DK, Agusala K, Buring JE, Lee I, Cook NR, Manson JE, Thadhani RI, Wang TJ. Impact of Vitamin D3 Versus Placebo on Cardiac Structure and Function: A Randomized Clinical Trial. J Am Heart Assoc 2022; 11:e025008. [PMID: 36285795 PMCID: PMC9673634 DOI: 10.1161/jaha.121.025008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background
Vitamin D supplementation leads to regression of left ventricular (LV) hypertrophy and improves LV function in animal models. However, limited data exist from prospective human studies. We examined whether vitamin D supplementation improved cardiac structure and function in midlife/older individuals in a large randomized trial.
Methods and Results
The VITAL (Vitamin D and OmegA‐3 Trial) was a nationwide double‐blind, placebo‐controlled randomized trial that tested the effects of vitamin D3 (2000 IU/d) and n−3 fatty acids (1 g/d) on cardiovascular and cancer risk in 25 871 individuals aged ≥50 years. We conducted a substudy of VITAL in which participants underwent echocardiography at baseline and 2 years. Images were interpreted by a blinded investigator at a central core laboratory. The primary end point was change in LV mass. Among 1054 Greater Boston–area participants attending in‐clinic visits, we enrolled 1025 into this study. Seventy‐nine percent returned for follow‐up and had analyzable echocardiograms at both visits. At baseline, the median age was 64 years (interquartile range, 60–69 years), 52% were men, and 43% had hypertension. After 2 years, the change in LV mass did not significantly differ between the vitamin D and placebo arms (median +1.4 g versus +2.6 g, respectively;
P
=0.32). Changes in systolic and diastolic LV function also did not differ significantly between arms. There were no significant changes in cardiac structure and function between the n−3 fatty acids and placebo arms.
Conclusions
Among adults aged ≥50 years, neither vitamin D3 nor n−3 fatty acids supplementation had significant effects on cardiac structure and function after 2 years.
Registration
URL:
https://clinicaltrials.gov/
; Unique identifiers: NCT01169259 (VITAL) and NCT01630213 (VITAL‐Echo)
Collapse
Affiliation(s)
- Alvin Chandra
- Department of Internal Medicine UT Southwestern Medical Center Dallas TX
| | | | - Shi Huang
- Department of Biostatistics Vanderbilt University Medical Center Nashville TN
- Vanderbilt Translational and Clinical Cardiovascular Research Center Vanderbilt University School of Medicine Nashville TN
| | - Deepak K. Gupta
- Vanderbilt Translational and Clinical Cardiovascular Research Center Vanderbilt University School of Medicine Nashville TN
| | - Kartik Agusala
- Department of Internal Medicine UT Southwestern Medical Center Dallas TX
| | - Julie E. Buring
- Division of Preventive Medicine Brigham and Women’s Hospital, Harvard Medical School Boston MA
| | - I‐Min Lee
- Division of Preventive Medicine Brigham and Women’s Hospital, Harvard Medical School Boston MA
| | - Nancy R. Cook
- Division of Preventive Medicine Brigham and Women’s Hospital, Harvard Medical School Boston MA
| | - JoAnn E. Manson
- Division of Preventive Medicine Brigham and Women’s Hospital, Harvard Medical School Boston MA
- Department of Epidemiology Harvard T. H. Chan School of Public Health Boston MA
| | | | - Thomas J. Wang
- Department of Internal Medicine UT Southwestern Medical Center Dallas TX
| |
Collapse
|
11
|
Oka T, Sakaguchi Y, Isaka Y, Ishii H, Kabata D, Shintani A, Nakatani S, Morioka T, Mori K, Inaba M, Emoto M, Shoji T. Effects of alfacalcidol on cardiovascular outcomes according to alkaline phosphatase levels in the J-DAVID trial. Sci Rep 2022; 12:15463. [PMID: 36104443 PMCID: PMC9475027 DOI: 10.1038/s41598-022-19820-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
In the Japan Dialysis Active Vitamin D (J-DAVID) trial, oral alfacalcidol numerically, but not significantly, increased the risk of cardiovascular events among patients undergoing hemodialysis. Because the cardiovascular effect of alfacalcidol could be modulated by bone turnover status, this post-hoc analysis of the J-DAVID examined how alkaline phosphatase (ALP), a more precise marker of bone turnover than parathyroid hormone (PTH), modifies the impact of alfacalcidol. The J-DAVID was a 48-month, open-label, randomized controlled trial comparing oral alfacalcidol with no vitamin D receptor activators use in terms of cardiovascular events among 976 hemodialysis patients without secondary hyperparathyroidism. This post-hoc analysis included 959 patients with available data on baseline ALP. The median [25–75th percentile] baseline ALP level was 234 [183–296] U/L. In a Cox proportional hazards model, ALP did not significantly modify the effect of alfacalcidol on the rate of cardiovascular events or all-cause death (P for effect modification = 0.54 and 0.74, respectively). The effect of alfacalcidol on time-series changes in calcium, phosphate, and intact PTH were similar across ALP subgroups. In conclusion, oral alfacalcidol did not significantly affect cardiovascular outcomes irrespective of bone turnover status.
Collapse
|
12
|
Yang S, Wang C, Ruan C, Chen M, Cao R, Sheng L, Chang N, Xu T, Zhao P, Liu X, Zhu F, Xiao Q, Gao S. Novel Insights into the Cardioprotective Effects of Calcitriol in Myocardial Infarction. Cells 2022; 11:1676. [PMID: 35626713 PMCID: PMC9139780 DOI: 10.3390/cells11101676] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/10/2022] [Accepted: 05/14/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Increasing evidence indicates that vitamin D deficiency negatively affects the cardiovascular system. Here we studied the therapeutic effects of calcitriol in myocardial infarction (MI) and investigated its underlying mechanisms. METHODS A MI model of Kun-ming mice induced by left anterior descending coronary artery ligation was utilized to study the potential therapeutic effects of calcitriol on MI. AC16 human cardiomyocyte-like cells treated with TNF-α were used for exploring the mechanisms that underlie the cardioprotective effects of calcitriol. RESULTS We observed that calcitriol reversed adverse cardiovascular function and cardiac remodeling in post-MI mice. Mechanistically, calcitriol suppressed MI-induced cardiac inflammation, ameliorated cardiomyocyte death, and promoted cardiomyocyte proliferation. Specifically, calcitriol exerted these cellular effects by upregulating Vitamin D receptor (VDR). Increased VDR directly interacted with p65 and retained p65 in cytoplasm, thereby dampening NF-κB signaling and suppressing inflammation. Moreover, up-regulated VDR was translocated into nuclei where it directly bound to IL-10 gene promoters to activate IL-10 gene transcription, further inhibiting inflammation. CONCLUSION We provide new insights into the cellular and molecular mechanisms underlying the cardioprotective effects of calcitriol, and we present comprehensive evidence to support the preventive and therapeutic effects of calcitriol on MI.
Collapse
Affiliation(s)
- Simin Yang
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
| | - Chunmiao Wang
- Department of Cardiology, The First Affiliated Hospital, Anhui Medical University, Hefei 230022, China;
| | - Chengshao Ruan
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
| | - Meiling Chen
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
- Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China;
| | - Ran Cao
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
- Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China;
| | - Liang Sheng
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
- Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China;
| | - Naiying Chang
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
- Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China;
| | - Tong Xu
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
| | - Peiwen Zhao
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
| | - Xuesheng Liu
- Department of Anesthesiology, The First Affiliated Hospital, Anhui Medical University, Hefei 230022, China;
| | - Fengqin Zhu
- Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China;
| | - Qingzhong Xiao
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Shan Gao
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
| |
Collapse
|
13
|
Jafarzadeh J, Payahoo L, Yousefi M, Barzegar A. The comprehensive mechanistic insight into the effects of vitamin D on dementia – a review. NUTRITION & FOOD SCIENCE 2022; 52:698-721. [DOI: 10.1108/nfs-08-2021-0256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
PurposeThis paper aims to depict the mechanistic role of vitamin D on dementia prevention, relief of the severity and the complication of the disease. All papers indexed in scientific databases, including Scopus, Elsevier, PubMed, Embase and Google Scholar between 2000 and 2021 were extracted and discussed. To present the mechanistic role of vitamin D in declining the severity of dementia, keywords including dementia, vitamin D, oxidative stress, inflammation, amyloid beta-Peptides were used.Design/methodology/approachDementia is a prevalent cognitive disorder worldwide, especially in elderly people, which is accompanied by serious disabilities. Besides genetic, biological and lifestyle factors are involved in the incidence of dementia. An unhealthy diet along with micronutrient deficiencies are among modifiable factors. Vitamin D is one of the important micronutrients in brain health. Besides the involvement in gene expression, bone mineralization, apoptosis, inflammation, skeletal maturation, neurotropic action and hemostasis of phosphate and calcium, vitamin D also exerts neuroprotective effects via genomic and non-genomic pathways.FindingsVitamin D up-regulates the expression of various genes involved in dementia incidence via various mechanisms. Decreasing oxidative stress and the neuro-inflammatory cytokines levels, regulation of the expression of alternated Proteins including Tau and Amyloid-ß, calcium homeostasis in the central nervous system and also vascular are considered main mechanisms.Originality/valueConsidering the importance of diet in preventing dementia, adherence to a healthy diet that provides essential nutrients to brain function seems to be urgent. Controlling serum levels of vitamin D periodically and providing vitamin D by related sources or supplements, if there is a deficiency, is recommended. Future studies are needed to clarify other related mechanisms.
Collapse
|
14
|
Bai J, Zhang X, Zhang A, Zhang Y, Ren K, Ren Z, Zhao C, Wang Q, Cao N. Cardiac valve calcification is associated with mortality in hemodialysis patients: a retrospective cohort study. BMC Nephrol 2022; 23:43. [PMID: 35065601 PMCID: PMC8783521 DOI: 10.1186/s12882-022-02670-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 01/12/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Cardiac valve calcification (CVC) is common in end-stage renal disease (ESRD). We investigated the effect of CVC on all-cause and cardiovascular (CV) mortality in maintenance hemodialysis (MHD) patients.
Methods
A retrospective cohort study was conducted on 434 hemodialysis patients who underwent echocardiography for qualitative assessment of valve calcification with complete follow-up data from January 1, 2014, to April 30, 2021. The baseline data between the CVC and non-CVC groups were compared. The Kaplan–Meier method was used to analyse all-cause and cardiovascular mortality. The association of CVC with all-cause and cardiovascular mortality was evaluated using multivariate Cox regression analysis.
Results
Overall, 27.2% of patients had mitral valve calcification (MVC), and 31.8% had aortic valve calcification (AVC) on echocardiography. Patients with CVC showed significantly higher all-cause (log-rank P < 0.001) and cardiovascular (log-rank P < 0.001) mortality rates than patients without CVC. In multivariate regression analyses, MVC (HR: 1.517, P = 0.010) and AVC (HR: 1.433, P = 0.028) were significant factors associated with all-cause mortality. MVC (HR: 2.340, P < 0.001) and AVC (HR: 2.410, P < 0.001) were also significant factors associated with cardiovascular mortality.
Conclusions
MVC and AVC increased the risk of all-cause and cardiovascular mortality in MHD patients. Regular follow-up with echocardiography could be a useful method for risk stratification in MHD patients.
Collapse
|
15
|
Navarro-García JA, González-Lafuente L, Fernández-Velasco M, Ruilope LM, Ruiz-Hurtado G. Fibroblast Growth Factor-23-Klotho Axis in Cardiorenal Syndrome: Mediators and Potential Therapeutic Targets. Front Physiol 2021; 12:775029. [PMID: 34867481 PMCID: PMC8634640 DOI: 10.3389/fphys.2021.775029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiorenal syndrome (CRS) is a complex disorder that refers to the category of acute or chronic kidney diseases that induce cardiovascular disease, and inversely, acute or chronic heart diseases that provoke kidney dysfunction. There is a close relationship between renal and cardiovascular disease, possibly due to the presence of common risk factors for both diseases. Thus, it is well known that renal diseases are associated with increased risk of developing cardiovascular disease, suffering cardiac events and even mortality, which is aggravated in those patients with end-stage renal disease or who are undergoing dialysis. Recent works have proposed mineral bone disorders (MBD) as the possible link between kidney dysfunction and the development of cardiovascular outcomes. Traditionally, increased serum phosphate levels have been proposed as one of the main factors responsible for cardiovascular damage in kidney patients. However, recent studies have focused on other MBD components such as the elevation of fibroblast growth factor (FGF)-23, a phosphaturic bone-derived hormone, and the decreased expression of the anti-aging factor Klotho in renal patients. It has been shown that increased FGF-23 levels induce cardiac hypertrophy and dysfunction and are associated with increased cardiovascular mortality in renal patients. Decreased Klotho expression occurs as renal function declines. Despite its expression being absent in myocardial tissue, several studies have demonstrated that this antiaging factor plays a cardioprotective role, especially under elevated FGF-23 levels. The present review aims to collect the recent knowledge about the FGF-23-Klotho axis in the connection between kidney and heart, focusing on their specific role as new therapeutic targets in CRS.
Collapse
Affiliation(s)
- José Alberto Navarro-García
- Cardiorenal Translational Laboratory, Institute of Research i + 12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Laura González-Lafuente
- Cardiorenal Translational Laboratory, Institute of Research i + 12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Luis M Ruilope
- Cardiorenal Translational Laboratory, Institute of Research i + 12, Hospital Universitario 12 de Octubre, Madrid, Spain.,CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain.,School of Doctoral Studies and Research, European University of Madrid, Madrid, Spain
| | - Gema Ruiz-Hurtado
- Cardiorenal Translational Laboratory, Institute of Research i + 12, Hospital Universitario 12 de Octubre, Madrid, Spain.,CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
| |
Collapse
|
16
|
Koshi-Ito E, Inaguma D, Koide S, Takahashi K, Hayashi H, Tsuboi N, Hasegawa M, Maruyama S, Yuzawa Y. Relationship between selection of dosage forms of vitamin D receptor activators and short-term survival of patients on hemodialysis. Ren Fail 2021; 43:1528-1538. [PMID: 34787531 PMCID: PMC8604548 DOI: 10.1080/0886022x.2021.1995423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 10/30/2022] Open
Abstract
BACKGROUND The benefits of vitamin D receptor activators (VDRAs) for patients with chronic kidney disease are well recognized. However, the optimal criteria for patient selection, dosage forms, and duration providing the highest benefit and the least potential risk remain to be confirmed. MATERIALS AND METHODS The study population was derived from the Aichi Cohort Study of Prognosis in Patients Newly Initiated into Dialysis, a multicenter prospective cohort study of 1520 incident dialysis patients. According to the VDRA usage status in March 2015 (interim report), the 967 patients surviving after March 2015 were classified into three groups: without VDRA (NV, n = 177), oral VDRA (OV, n = 447), and intravenous VDRA (IV, n = 343). Mortality rates were compared using the log-rank test, and factors contributing to all-cause mortality were examined using both univariate and multivariate Cox proportional hazard regression analyses. RESULTS There were 104 deaths (NV, n = 27; OV, n = 53; IV, n = 24) during the follow-up period (1360 days, median), and significant differences in cumulative survival rates were observed between the three groups (p = 0.010). Moreover, lower all-cause mortality was associated with IV versus NV (hazard ratio, 0.46 [95% confidence interval 0.24-0.89]; p = 0.020). CONCLUSION This study demonstrated the impact of the VDRA dosage form on the short-term survival of incident hemodialysis patients during the introduction period. Our results suggest that relatively early initiation of intravenous VDRA in patients beginning hemodialysis may have some clinical potential.
Collapse
Affiliation(s)
- Eri Koshi-Ito
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Japan
- The Aichi Cohort Study of Prognosis in Patients Newly Initiated into Dialysis (AICOPP) Group, Aichi, Japan
| | - Daijo Inaguma
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Japan
- The Aichi Cohort Study of Prognosis in Patients Newly Initiated into Dialysis (AICOPP) Group, Aichi, Japan
- Department of Internal Medicine, Fujita Health University Bantane Hospital, Nagoya, Japan
| | - Shigehisa Koide
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Japan
- The Aichi Cohort Study of Prognosis in Patients Newly Initiated into Dialysis (AICOPP) Group, Aichi, Japan
| | - Kazuo Takahashi
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Hiroki Hayashi
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Naotake Tsuboi
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Midori Hasegawa
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- The Aichi Cohort Study of Prognosis in Patients Newly Initiated into Dialysis (AICOPP) Group, Aichi, Japan
| | - Yukio Yuzawa
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Japan
| |
Collapse
|
17
|
Abstract
Vitamin D represents a group of secosteroids involved in the calcium and phosphate metabolism. The active form of vitamin D, 1,25-dihydroxylcalciferol, exerts its biological mechanisms via the VDR (vitamin D receptor) which acts as a regulator of several target genes. Hypovitaminosis D is associated with many diseases, which are not only limited to the metabolism of the skeleton, but growing evidence links the deficit of vitamin D to cardiovascular, metabolic, immune, and neoplastic diseases. In regard to the cardiovascular system, current evidence shows the presence of VDR in endothelial cells. Moreover, both in vitro and animal experimental models demonstrated that the deficit of vitamin D can promote endothelial dysfunction and atherosclerosis development. Vitamin D can interfere with vascular functions also by affecting the production of vasodilator mediators. VDR is also expressed in left ventricle cardiomyocytes, and hypovitaminosis D can relate to cardiac hypertrophy and heart failure. Randomized clinical trials (RCT) designed to prove the therapeutic role of vitamin D supplementation have been inconclusive to date. The aim of this review is to highlight the main interactions between vitamin D metabolism and cardiovascular diseases; thus, focusing on pathogenic mechanisms and related clinical manifestations.
Collapse
|
18
|
Dai L, Liu M, Chen L. Association of Serum 25-Hydroxyvitamin D Concentrations With All-Cause and Cause-Specific Mortality Among Adult Patients With Existing Cardiovascular Disease. Front Nutr 2021; 8:740855. [PMID: 34631770 PMCID: PMC8496747 DOI: 10.3389/fnut.2021.740855] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/30/2021] [Indexed: 01/04/2023] Open
Abstract
Background: Vitamin D insufficiency and deficiency are common in patients with cardiovascular disease (CVD). We aimed to prospectively examine the associations of serum 25-hydroxyvitamin D [25(OH)D] concentrations with all-cause and cause-specific mortality among adult patients with existing CVD. Methods: We included 37,079 patients with CVD from the UK Biobank study, a prospective cohort of half a million participants aged 40–69 years. We defined patients with CVD as those who suffered coronary heart disease, atrial fibrillation, heart failure, or stroke. The associations of serum 25(OH)D concentration with all-cause and cause-specific mortality were examined by using multivariable Cox regression models and competing risk analyses. Results: Among 37,079 patients with CVD at baseline, 57.5% were subjected to vitamin D deficiency (i.e., 25[OH]D <50 nmol/L). During a median follow-up of 11.7 years, 6,319 total deaths occurred, including 2,161 deaths from CVD, 2,230 deaths from cancer, 623 deaths from respiratory disease, and 1,305 other-cause deaths. We observed non-linear inverse associations for all-cause, cancer, respiratory disease, and other-cause mortality (P-non-linearity <0.01) and approximately linear inverse associations for CVD mortality (P-non-linearity = 0.074). Among CVD patients with vitamin D deficiency, per 10 nmol/L increment in serum 25(OH)D concentrations was associated with an 12% reduced risk for all-cause mortality and 9% reduced risk for CVD mortality. Conclusion: Among patients with existing CVD, increasing levels in serum 25(OH)D were independently associated with a decreased risk of all-cause and cause-specific mortality. These findings suggest that elevated serum 25(OH)D concentration benefits CVD patients with vitamin D deficiency.
Collapse
Affiliation(s)
- Lei Dai
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Man Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liangkai Chen
- Hubei Key Laboratory of Food Nutrition and Safety, Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Banerjee D, Chitalia N, Ster IC, Appelbaum E, Thadhani R, Kaski JC, Goldsmith D. Impact of vitamin D on cardiac structure and function in chronic kidney disease patients with hypovitaminosis D: a randomized controlled trial and meta-analysis. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2021; 7:302-311. [PMID: 31830258 PMCID: PMC8302255 DOI: 10.1093/ehjcvp/pvz080] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/10/2019] [Accepted: 12/09/2019] [Indexed: 01/08/2023]
Abstract
AIMS Vitamin D deficiency is associated with cardiovascular events in chronic kidney disease (CKD) yet the impact of supplementation is controversial. Previous active vitamin D supplementation studies did not show improvement in cardiac structure or function but the effect of native vitamin D supplementation in CKD patients with low vitamin D levels is unknown. We have addressed this question via both a randomized double-blind prospective study and a meta-analysis of three randomized placebo-controlled studies. METHODS AND RESULTS We conducted a randomized double-blind, placebo-controlled trial of vitamin D supplementation in stable, non-diabetic, CKD three to four patients with circulating vitamin D <75nmol/L, who were receiving treatment with ACEi or ARB and had high-normal left ventricular (LV) mass. Patients were randomized to receive six directly observed doses of 100 000 IU cholecalciferol (n = 25) or matched placebo (n = 23). The primary endpoint was changed in LV mass index (LVMI) over 52 weeks, as assessed by cardiac magnetic resonance imaging. Secondary endpoints included changes in LV ejection fraction (LVEF); LV and right ventricular volumes and left and right atrial area. Vitamin D concentration increased with the administration of cholecalciferol. The change in LVMI with cholecalciferol [median (inter-quartile range), -0.25 g (-7.20 to 5.30)] was no different from placebo [-4.30 g (9.70 to 2.60)]. There was no difference in changes of LVEF; LV and right ventricular volumes and left and right atrial area. The meta-analysis of three 52-week, randomized placebo-controlled studies using active/native vitamin D supplementation showed no differences in LVMI measurements. CONCLUSION Vitamin D supplementation does not have beneficial effects on LV mass in CKD patients.
Collapse
Affiliation(s)
- Debasish Banerjee
- Renal and Transplantation Unit, St George’s University Hospital NHS Foundation Trust, G 2.113, Grosvenor Wing, Blackshaw Road, Tooting, London SW17 0QT, UK
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research Centre, St George’s University of London, London, UK
| | - Nihil Chitalia
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research Centre, St George’s University of London, London, UK
- Renal Medicine, Darent Valley Hospital, Dartford, Kent, UK
| | - Irina Chis Ster
- Institute of Infection and Immunity, St George's University of London, London, UK
| | | | - Ravi Thadhani
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Juan Carlos Kaski
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research Centre, St George’s University of London, London, UK
| | - David Goldsmith
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research Centre, St George’s University of London, London, UK
| |
Collapse
|
20
|
The Complexity of FGF23 Effects on Cardiomyocytes in Normal and Uremic Milieu. Cells 2021; 10:cells10051266. [PMID: 34065339 PMCID: PMC8161087 DOI: 10.3390/cells10051266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
Fibroblast growth factor-23 (FGF23) appears to be one of the most promising biomarkers and predictors of cardiovascular risk in patients with heart disease and normal kidney function, but moreover in those with chronic kidney disease (CKD). This review summarizes the current knowledge of FGF23 mechanisms of action in the myocardium in the physiological and pathophysiological state of CKD, as well as its cross-talk to other important signaling pathways in cardiomyocytes. In this regard, current therapeutic possibilities and future perspectives are also discussed.
Collapse
|
21
|
Komaba H, Ketteler M, Cunningham J, Fukagawa M. Old and New Drugs for the Management of Bone Disorders in CKD. Calcif Tissue Int 2021; 108:486-495. [PMID: 33386480 DOI: 10.1007/s00223-020-00788-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/04/2020] [Indexed: 12/20/2022]
Abstract
Disturbances in mineral and bone metabolism are common in patients with chronic kidney disease (CKD), especially those undergoing dialysis. Renal osteodystrophy, which describes an alteration of bone morphology, is an important component of this systemic disorder and may explain the elevated risk of fracture which adversely affects morbidity and mortality. The most common form of renal osteodystrophy is high-turnover bone disease (osteitis fibrosa), which is induced by secondary hyperparathyroidism (SHPT). During the past decade, there has been considerable advances in the management of SHPT, with the introduction of the calcimimetic agents, the optimized use of nutritional and active vitamin D, and the accumulated experience with surgical parathyroidectomy. Studies supported that these advances could translate into improvement of renal bone disease and fracture prevention, as well as decreasing the risk of cardiovascular events and mortality. In this review, we summarize the available clinical evidence on the effect of old and new drugs on bone disorders in patients with CKD.
Collapse
Affiliation(s)
- Hirotaka Komaba
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, 143 Shimo-Kasuya, Isehara, 259-1193, Japan
- The Institute of Medical Sciences, Tokai University, Isehara, Japan
| | - Markus Ketteler
- Department of General Internal Medicine and Nephrology, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | | | - Masafumi Fukagawa
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, 143 Shimo-Kasuya, Isehara, 259-1193, Japan.
| |
Collapse
|
22
|
Soh V, Tan SJX, Sehgal R, Shirke MM, Ashry A, Harky A. The Relationship Between Vitamin D Status and Cardiovascular Diseases. Curr Probl Cardiol 2021; 46:100836. [PMID: 33848960 DOI: 10.1016/j.cpcardiol.2021.100836] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022]
Abstract
With cardiovascular conditions being a leading cause of mortality and morbidity globally, several studies have identified that there is an important correlation between the level of Vitamin D and cardiovascular diseases, including an increased risk of hypertension, heart failure, and coronary artery diseases. Current published studies are in the form of both in vivo and in vitro studies and they primarily showed the evidence of how Vitamin D can downregulate Renin-Angiotensin-Aldosterone system activity and therefore providing a cardioprotective role. Nevertheless, most of these studies are observational, and there yet to be large-scale randomized controlled trials which would increase the evidence of the findings.This review aims to capture the current evidence of Vitamin D as a metabolite which is critical in reducing cardiovascular conditions and the possible physiological pathways that it works via.
Collapse
Affiliation(s)
- Vernie Soh
- Department of Medicine, Queen's University Belfast, School of Medicine, Belfast, UK
| | - Shawn Jia Xiang Tan
- Department of Medicine, Queen's University Belfast, School of Medicine, Belfast, UK
| | - Rijuvani Sehgal
- Department of Medicine, Queen's University Belfast, School of Medicine, Belfast, UK
| | - Manasi Mahesh Shirke
- Department of Medicine, Queen's University Belfast, School of Medicine, Belfast, UK
| | - Amr Ashry
- Department of Paediatric Cardiac Surgery, Alder Hey Children Hospital, Liverpool, UK; Department of Cardiothoracic Surgery, Assiut University Hospital, Assiut, Egypt
| | - Amer Harky
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, UK; Department of Cardiothoracic Surgery, Liverpool Heart and Chest Hospital, Liverpool, UK.
| |
Collapse
|
23
|
Lee TL, Lee MH, Chen YC, Lee YC, Lai TC, Lin HYH, Hsu LF, Sung HC, Lee CW, Chen YL. Vitamin D Attenuates Ischemia/Reperfusion-Induced Cardiac Injury by Reducing Mitochondrial Fission and Mitophagy. Front Pharmacol 2020; 11:604700. [PMID: 33362559 PMCID: PMC7758530 DOI: 10.3389/fphar.2020.604700] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/16/2020] [Indexed: 01/23/2023] Open
Abstract
Myocardial infarction is the leading cause of morbidity and mortality worldwide. Although myocardial reperfusion after ischemia (I/R) is an effective method to save ischemic myocardium, it can cause adverse reactions, including increased oxidative stress and cardiomyocyte apoptosis. Mitochondrial fission and mitophagy are essential factors for mitochondrial quality control, but whether they play key roles in cardiac I/R injury remains unknown. New pharmacological or molecular interventions to alleviate reperfusion injury are currently considered desirable therapies. Vitamin D3 (Vit D3) regulates cardiovascular function, but its physiological role in I/R-exposed hearts, especially its effects on mitochondrial homeostasis, remains unclear. An in vitro hypoxia/reoxygenation (H/R) model was established in H9c2 cells to simulate myocardial I/R injury. H/R treatment significantly reduced H9c2 cell viability, increased apoptosis, and activated caspase 3. In addition, H/R treatment increased mitochondrial fission, as manifested by increased expression of phosphorylated dynein-related protein 1 (p-Drp1) and mitochondrial fission factor (Mff) as well as increased mitochondrial translocation of Drp1. Treatment with the mitochondrial reactive oxygen species scavenger MitoTEMPO increased cell viability and decreased mitochondrial fission. H/R conditions elicited excessive mitophagy, as indicated by increased expression of BCL2-interacting protein 3 (BNIP3) and light chain (LC3BII/I) and increased formation of autolysosomes. In contrast, Vit D3 reversed these effects. In a mouse model of I/R, apoptosis, mitochondrial fission, and mitophagy were induced. Vit D3 treatment mitigated apoptosis, mitochondrial fission, mitophagy, and myocardial ultrastructural abnormalities. The results indicate that Vit D3 exerts cardioprotective effects against I/R cardiac injury by protecting mitochondrial structural and functional integrity and reducing mitophagy.
Collapse
Affiliation(s)
- Tzu-Lin Lee
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Hsueh Lee
- Division of Neurosurgery, Department of Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan.,Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi, Taiwan
| | - Yu-Chen Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Chieh Lee
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tsai-Chun Lai
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hugo You-Hsien Lin
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Lee-Fen Hsu
- Division of Neurosurgery, Department of Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan.,Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi, Taiwan
| | - Hsin-Ching Sung
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Aesthetic Medical Center, Department of Dermatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chiang-Wen Lee
- Department of Nursing, Division of Basic Medical Sciences, and Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Chiayi, Taiwan.,Research Center for Industry of Human Ecology and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yuh-Lien Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
24
|
Maxacalcitol (22-Oxacalcitriol (OCT)) Retards Progression of Left Ventricular Hypertrophy with Renal Dysfunction Through Inhibition of Calcineurin-NFAT Activity. Cardiovasc Drugs Ther 2020; 35:381-397. [PMID: 33206298 DOI: 10.1007/s10557-020-07111-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/10/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE Left ventricular hypertrophy (LVH) is a cardiovascular complication highly prevalent in patients with chronic kidney disease (CKD). Previous studies analyzing 1α-hydroxylase or vitamin D receptor (Vdr) knockout mice revealed active vitamin D as a promising agent inhibiting LVH progression. Paricalcitol, an active vitamin D analog, failed to suppress the progression of LV mass index (LVMI) in pre-dialysis patients with CKD. As target genes of activated VDR differ depending on its agonists, we examined the effects of maxacalcitol (22-oxacalcitriol: OCT), a less calcemic active vitamin D analog, on LVH in hemodialysis patients and animal LVH models with renal insufficiency. METHODS In retrospective cohort study, patients treated with OCT who underwent hemodialysis were enrolled. Using cardiac echocardiography, LV mass was evaluated by the area-length method. In animal study, angiotensin II (Ang II)-infused Wister rats with heminephrectomy or Ang II-stimulated neonatal rat ventricular myocytes (NRVM) were treated with OCT. RESULTS OCT significantly inhibited the progression of LVMI in hemodialysis patients. In Ang II-infused heminephrectomized rats, OCT suppressed the progression of LVH in a blood pressure-independent manner. OCT also suppressed the activity of calcineurin in the left ventricle of model rats. Specifically, OCT reduced the protein levels of calcineurin A, but not the mRNA levels of Ppp3ca (calcineurin Aα). Luciferase assays showed that OCT increased the promoter activity of Fbxo32 (atrogin1), an E3 ubiquitin ligase targeting calcineurin A. Finally, OCT promoted ubiquitination and degradation of calcineurin A. CONCLUSION Our works indicated that OCT retards progression of LVH through calcineurin-NFAT pathway, which reveal a novel aspect of OCT in attenuating pathological LVH.
Collapse
|
25
|
Vitamin D and Cardiovascular Disease, with Emphasis on Hypertension, Atherosclerosis, and Heart Failure. Int J Mol Sci 2020; 21:ijms21186483. [PMID: 32899880 PMCID: PMC7555466 DOI: 10.3390/ijms21186483] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/24/2020] [Accepted: 08/31/2020] [Indexed: 12/14/2022] Open
Abstract
Vitamin D deficiency is the most common nutritional deficiency, affecting almost one billion people worldwide. Vitamin D is mostly known for its role in intestinal calcium absorption and bone mineralization. However, the observation of seasonal changes in blood pressure and the subsequent identification of vitamin D receptor (VDR) and 1α-hydroxylase in cardiomyocytes, as well as endothelial and vascular smooth muscle cells, implicated a role of vitamin D in the cardiovascular system. Animal studies provided compelling evidence that vitamin D signaling is essential for cardiovascular integrity, especially for the regulation of vascular tone and as an antifibrotic and antihypertrophic signaling pathway in the heart. In addition, observational studies reported an association between vitamin D deficiency and risk of hypertension, atherosclerosis, and heart failure. However, recent clinical intervention studies failed to prove the causal relationship between vitamin D supplementation and beneficial effects on cardiovascular health. In this review, we aim to highlight our current understanding of the role of vitamin D in the cardiovascular system and to find potential explanations for the large discrepancies between the outcome of experimental studies and clinical intervention trials.
Collapse
|
26
|
The protective effect of 1,25(OH) 2D 3 against cardiac hypertrophy is mediated by the cyclin-dependent kinase inhibitor p21. Eur J Pharmacol 2020; 888:173510. [PMID: 32861664 DOI: 10.1016/j.ejphar.2020.173510] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 08/14/2020] [Accepted: 08/24/2020] [Indexed: 12/29/2022]
Abstract
As a critical regulator of the cell cycle, cyclin-dependent kinase (CDK) inhibitor p21 or p21 is involved in the development of cardiac hypertrophy and heart failure. Calcitriol, or 1,25(OH)2D3, the bioactive form of vitamin D (VD), can activate p21 expression and attenuate cardiac hypertrophy. To simulate cardiac hypertrophy in vitro and ex vivo, respectively, mice and cardiomyocytes were treated with isoproterenol (ISO). Moreover, the p21 signaling pathway was examined in ISO + VD and ISO + VD p21 inhibitor-treated cardiomyocytes. We found that calcitriol treatment led to a significant decrease in cardiac size and the mRNA levels of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) in ISO-treated mice. Furthermore, the surface area of cardiomyocytes and the expression of ANP and BNP were decreased, and the expression of p21 was increased in the ISO + VD group compared with those in the ISO group. Furthermore, the surface area of cardiomyocytes and the expression of ANP and BNP were markedly upregulated in the ISO + VD p21 inhibitor group relative to the ISO + VD group, whereas the difference was not statistically significant compared with those of the ISO p21 inhibitor group. Therefore, our findings indicate that 1,25(OH)2D3 protects against cardiac hypertrophy in mice through upregulating p21 expression.
Collapse
|
27
|
Vitamin D Analogues and Coronary Calcification in CKD Stages 3 and 4: A Randomized Controlled Trial of Calcitriol Versus Paricalcitol. Kidney Med 2020; 2:450-458. [PMID: 32775985 PMCID: PMC7406841 DOI: 10.1016/j.xkme.2020.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Rationale & Objective Mineral and bone disorder in chronic kidney disease (CKD) is associated with progression of coronary artery calcification (CAC). Mineral and bone disorder often is treated with calcitriol and other vitamin D receptor activators, including paricalcitol, agents that may have differential effects on calcium, phosphate, and parathyroid hormone levels. Accordingly, we investigated whether these agents have differential effects on CAC progression in patients with CKD. Study Design Randomized, double-concealed, 48-week clinical trial. Setting & Participants CKD stage 3 or 4 with secondary hyperparathyroidism with CAC score > 0 and no prior treatment with activated vitamin D. Intervention Calcitriol versus paricalcitol. Outcomes The primary outcome was log-transformed CAC change. Secondary outcomes included percent change in CAC volume, valvular calcifications, and bone mineral metabolism markers. Results Among 44 individuals randomly assigned, mean age was 65 years and mean estimated glomerular filtration rate was 27 mL/min/1.73 m2. Median CAC score was 140 (IQR, 55-277) Agatston units at baseline. There was no significant difference in CAC progression between treatment arms (P = 0.06). After adjustment for baseline CAC score (log), treatment group remains nonsignificant (P = 0.08). Further adjustment for creatinine level and/or CKD stage did not change the association. In secondary analyses adjusting for dose level of activated vitamin D, treatment group was significant (P = 0.01), and when dose level was also included in the model, the coefficient for individuals in the paricalcitol group was significantly associated with CAC progression (P = 0.02). An interaction term between dosing level and CKD stage was significant at the highest dosing level (P = 0.04). Limitations Pilot single-center study. Conclusions In patients with CKD with secondary hyperparathyroidism naive to activated vitamin D therapy, there was no difference in CAC or valvular progression in participants receiving calcitriol compared with paricalcitol during a 48-week period. Funding Abbvie, Inc. Trial Registration NCT00752102.
Collapse
|
28
|
Tamayo M, Martín-Nunes L, Val-Blasco A, G M-Piedras MJ, Navarro-García JA, Lage E, Prieto P, Ruiz-Hurtado G, Fernández-Velasco M, Delgado C. Beneficial effects of paricalcitol on cardiac dysfunction and remodelling in a model of established heart failure. Br J Pharmacol 2020; 177:3273-3290. [PMID: 32154912 DOI: 10.1111/bph.15048] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND AND PURPOSE The synthetic vitamin D3 analogue paricalcitol acts as a selective activator of the vitamin D receptor (VDR). While there is evidence for cardioprotective effects of paricalcitol associated with the VDR pathway, less information is available about the structural and functional cardiac effects of paricalcitol on established heart failure (HF) and particularly its effects on associated electrophysiological or Ca2+ handling remodelling. EXPERIMENTAL APPROACH We used a murine model of transverse aortic constriction (TAC) to study the effect of paricalcitol on established HF. Treatment was initiated 4 weeks after surgery over five consecutive weeks, and mice were sacrificed 9 weeks after surgery. Cardiac MRI (CMRI) was performed 4 and 9 weeks after surgery. Hearts were used for biochemical and histological studies and to isolate ventricular myocytes for electrophysiological and calcium imaging studies. KEY RESULTS CMRI analysis revealed that, compared with vehicle, paricalcitol treatment prevented the progression of ventricular dilation and hypertrophy after TAC and halted the corresponding decline in ejection fraction. These beneficial effects were related to the attenuation of intracellular Ca2+ mishandling remodelling, antifibrotic and antihypertrophic effects and potentially antiarrhythmic effects by preventing the reduction of K+ current density and the long QT, JT and TpTe intervals observed in HF animals. CONCLUSION AND IMPLICATIONS The results suggest that paricalcitol treatment in established HF hampers disease progression and improves adverse electrophysiological and Ca2+ handling remodelling, attenuating the vulnerability to HF-associated ventricular arrhythmias. Paricalcitol may emerge as a potential therapeutic option in the treatment of HF.
Collapse
Affiliation(s)
- María Tamayo
- Department of Metabolism and Cell Signalling, Biomedical Research Institute "Alberto Sols" CSIC-UAM/CIBER-CV, Madrid, Spain
| | - Laura Martín-Nunes
- Department of Metabolism and Cell Signalling, Biomedical Research Institute "Alberto Sols" CSIC-UAM/CIBER-CV, Madrid, Spain
| | - Almudena Val-Blasco
- Innate Immune Response Group, IdiPAZ/CIBER-CV, La Paz University Hospital, Madrid, Spain
| | - Maria José G M-Piedras
- Department of Anatomy, Faculty of Medicine, Universidad Francisco de Vitoria, Madrid, Spain
| | - José Alberto Navarro-García
- Cardiorenal Translational Laboratory, Institute of Research i+12/CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Eduardo Lage
- Department of Electronics and Communications Technology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Patricia Prieto
- Department of Metabolism and Cell Signalling, Biomedical Research Institute "Alberto Sols" CSIC-UAM/CIBER-CV, Madrid, Spain
| | - Gema Ruiz-Hurtado
- Cardiorenal Translational Laboratory, Institute of Research i+12/CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Carmen Delgado
- Department of Metabolism and Cell Signalling, Biomedical Research Institute "Alberto Sols" CSIC-UAM/CIBER-CV, Madrid, Spain
| |
Collapse
|
29
|
Grundmann SM, Schutkowski A, Schreier B, Rabe S, König B, Gekle M, Stangl GI. Vitamin D Receptor Deficiency Does Not Affect Blood Pressure and Heart Function. Front Physiol 2019; 10:1118. [PMID: 31555149 PMCID: PMC6727788 DOI: 10.3389/fphys.2019.01118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/13/2019] [Indexed: 01/07/2023] Open
Abstract
Vitamin D is thought to play a role in blood pressure regulation, which in turn can influence cardiovascular risk. Several meta-analyses of cohort studies found low serum levels of 25-hydroxyvitamin D to be associated with increased blood pressure or increased cardiovascular morbidity and mortality in the general population. Active vitamin D mediates its function via the vitamin D receptor (Vdr), which is a ligand-activated transcription factor. A suitable model to examine the causal role of vitamin D in blood pressure regulation and heart function is the Vdr knockout (Vdr–/–) mouse. To elucidate the role of vitamin D on blood pressure, heart function, and cardiac myocyte size, we conducted a long-term study using Vdr–/– mice and well-defined diets. Group 1 comprised Vdr–/– mice that received a high-calcium, high-phosphorus rescue diet to prevent hypocalcemia and a rickets phenotype. Groups 2 and 3 included Vdr+/+ mice that were fed either the rescue diet or a control diet containing normal amounts of these minerals. As Vdr is a nuclear factor that regulates transcription, we analyzed the renal mRNA expression and serum concentration of renin and found that the Vdr–/– group had an almost 50% higher renin mRNA expression in the kidney compared to both groups of Vdr+/+ mice. Additionally, serum concentration of renin in Vdr–/– mice was significantly higher than that of Vdr+/+ mice that received the rescue or control diet (+ 17%,+ 32%; P < 0.05). In contrast, renin activity was lower in Vdr–/– mice than in both groups of Vdr+/+ mice (P < 0.05). However, blood pressure, heart rate, cardiac myocyte sizes, and the expression of renal renin receptor, hepatic angiotensinogen and angiotensin II receptor, type 1, in kidney, liver and heart, did not differ between the three groups of mice. Additionally, data from transthoracic echocardiography did not indicate the role of Vdr on heart function, as the left ventricular ejection fraction, fractional shortening, and velocity of blood flow were comparable between the three groups. To conclude, the roles of Vdr and therefore most probably of vitamin D, in blood pressure regulation and heart function, were not confirmed by our findings.
Collapse
Affiliation(s)
- Sarah M Grundmann
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.,Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Leipzig, Germany
| | - Alexandra Schutkowski
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Barbara Schreier
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Leipzig, Germany.,Julius Bernstein Institute of Physiology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Sindy Rabe
- Julius Bernstein Institute of Physiology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Bettina König
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.,Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Leipzig, Germany
| | - Michael Gekle
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Leipzig, Germany.,Julius Bernstein Institute of Physiology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Gabriele I Stangl
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.,Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Leipzig, Germany
| |
Collapse
|
30
|
Renin Activity in Heart Failure with Reduced Systolic Function-New Insights. Int J Mol Sci 2019; 20:ijms20133182. [PMID: 31261774 PMCID: PMC6651297 DOI: 10.3390/ijms20133182] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 12/27/2022] Open
Abstract
Regardless of the cause, symptomatic heart failure (HF) with reduced ejection fraction (rEF) is characterized by pathological activation of the renin–angiotensin–aldosterone system (RAAS) with sodium retention and extracellular fluid expansion (edema). Here, we review the role of active renin, a crucial, upstream enzymatic regulator of the RAAS, as a prognostic and diagnostic plasma biomarker of heart failure with reduced ejection fraction (HFrEF) progression; we also discuss its potential as a pharmacological bio-target in HF therapy. Clinical and experimental studies indicate that plasma renin activity is elevated with symptomatic HFrEF with edema in patients, as well as in companion animals and experimental models of HF. Plasma renin activity levels are also reported to be elevated in patients and animals with rEF before the development of symptomatic HF. Modulation of renin activity in experimental HF significantly reduces edema formation and the progression of systolic dysfunction and improves survival. Thus, specific assessment and targeting of elevated renin activity may enhance diagnostic and therapeutic precision to improve outcomes in appropriate patients with HFrEF.
Collapse
|
31
|
Jahanifar F, Astani A, Shekarforoosh S, Jamhiri M, Safari F, Zarei F, Safari F. 1.25 Dihydroxyvitamin D3 Attenuates Hypertrophy Markers in Cardiomyoblast H9c2 Cells: Evaluation of Sirtuin3 mRNA and Protein Level. INT J VITAM NUTR RES 2019; 89:144-151. [PMID: 30856082 DOI: 10.1024/0300-9831/a000469] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Background: The cellular and molecular mechanisms of cardioprotective effects of Vitamin D are poorly understood. Given the essential role of sirtuin-3 (SIRT3) as an endogenous negative regulator of cardiac hypertrophy, this study was designed to investigate the effect of 1, 25-dihydroxyvitamin D3 (calcitriol) on hypertrophy markers and SIRT3 mRNA and protein levels following angiotensin II induced - hypertrophy in cardiomyoblast H9c2 cells. Methods: Rat cardiomyoblast H9c2 cells were treated for 48 hr with angiotensin II alone (Ang group) or in combination with 1, 10 and 100 nM of calcitriol (C + Ang groups). Intact cells served as control (Ctl). The cell area was measured using methylene blue staining. Atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP) and SIRT3 transcription levels were measured by real time RT-PCR. SIRT3 protein expression was evaluated using western blot technique. Results: The results showed that in Ang group cell size was increase by 128.4 ± 15% (P < 0.001 vs. Ctl) whereas in C100 + Ang group it was increased by 21.3 ± 6% (P < 0.001 vs. Ang group). Calcitriol pretreatment decreased ANP mRNA level significantly (P < 0.05) in comparison with Ang group (Ang: 75.5 ± 15%, C100 + Ang: 19.2 ± 9%). There were no significant differences between Ang group and cells pretreated with 1 and 10 nM of calcitriol. SIRT3 at mRNA and protein levels did not change significantly among the experimental groups. Conclusions: In conclusion, pretreatment with calcitriol (100 nM) prevents Ang II-induced hypertrophy in cardiomyoblast H9c2 cells. This probably occurs through other pathways except SIRT3 upregulation.
Collapse
Affiliation(s)
- Fatemeh Jahanifar
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Akram Astani
- Department of Microbiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Mohabbat Jamhiri
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Farideh Zarei
- Premature Neonates Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Safari
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
32
|
Kleisarchaki AN, Papadopoulou-Legbelou K, Kotanidou EP, Kotanidis CP, Eboriadou-Petikopoulou M, Galli-Tsinopoulou A. Could vitamin D deficiency influence left heart ventricular geometry in youngsters with type 1 diabetes mellitus? Hippokratia 2019; 23:9-14. [PMID: 32256032 PMCID: PMC7124876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
BACKGROUND Experimental and clinical studies have shown the cardio-protective, anti-inflammatory, and anti-atherosclerotic actions of vitamin D. MATERIAL AND METHODS We aimed to investigate a possible correlation between vitamin D status and heart geometry using echocardiographic parameters of the left ventricle in youngsters with type 1 diabetes mellitus (T1D). Seventy-eight pediatric patients (aged 13.47 ± 2.86 years) with T1D of more than two years duration and 74 healthy controls (aged 12.04 ± 2.79 years) were enrolled in this case-control study. Anthropometric parameters were recorded, vitamin D and parathormone serum levels were measured, and trans-thoracic echocardiographic study was performed. RESULTS Vitamin D deficiency was found in 74 % T1D patients and in 72 % of the controls, while parathormone levels were normal in both groups. T1D patients presented significantly higher values of interventricular septal thickness at diastole (IVSD) compared to controls (0.76 ± 0.16 cm vs 0.71 ± 0.14 cm, p =0.043). All other echocardiographic parameters did not exhibit significant differences between patients and controls. The diastolic function of the left ventricle (LV) was normal in both groups. After sub-grouping, the participants according to the deficiency or not of vitamin D, only patients with T1D and low vitamin D levels had increased values of IVSD compared to controls (0.78 ± 0.17 vs 0.71 ± 0.14, p =0.008). Patients with T1D and normal vitamin D levels presented similar values of IVSD compared to controls (0.71 ± 0.12 vs 0.73 ± 0.15, p =NS). CONCLUSIONS Children and adolescents with T1D and normal vitamin D levels do not exhibit changes in LV dimensions or diastolic function, except for increased IVSD, compared to controls. Larger and longitudinal studies are required to confirm and consolidate this finding. HIPPOKRATIA 2019, 23(1): 9-14.
Collapse
Affiliation(s)
- A N Kleisarchaki
- 4 Department of Paediatrics, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Thessaloniki, Greece
| | - K Papadopoulou-Legbelou
- 4 Department of Paediatrics, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Thessaloniki, Greece
| | - E P Kotanidou
- 4 Department of Paediatrics, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Thessaloniki, Greece
| | - C P Kotanidis
- 4 Department of Paediatrics, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Thessaloniki, Greece
| | - M Eboriadou-Petikopoulou
- 4 Department of Paediatrics, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Thessaloniki, Greece
| | - A Galli-Tsinopoulou
- 4 Department of Paediatrics, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Thessaloniki, Greece
| |
Collapse
|
33
|
Czaya B, Seeherunvong W, Singh S, Yanucil C, Ruiz P, Quiroz Y, Grabner A, Katsoufis C, Swaminathan S, Abitbol C, Rodriguez-Iturbe B, Faul C, Freundlich M. Cardioprotective Effects of Paricalcitol Alone and in Combination With FGF23 Receptor Inhibition in Chronic Renal Failure: Experimental and Clinical Studies. Am J Hypertens 2019; 32:34-44. [PMID: 30329020 DOI: 10.1093/ajh/hpy154] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/13/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND In uremic animals, vitamin D receptor (VDR) agonists like paricalcitol (Pc) attenuate cardiac hypertrophy, but this effect has not been replicated consistently in humans with chronic kidney disease. Elevated fibroblast growth factor 23 (FGF23) levels cause cardiac hypertrophy with activation of the myocardial calcineurin/nuclear factor of activated T cell (NFAT) axis and may antagonize the cardioprotective effects of VDR agonist therapy. We hypothesized that the effectiveness of Pc may depend on the prevailing circulating levels of FGF23 and could be potentiated by the combined administration of a pan-FGF23 receptor (FGFR) blocker agent (PD173074). METHODS In rats with 5/6 nephrectomy treated with Pc or PD173074 or both agents concurrently, myocardial mRNA expression of renin-angiotensin system, VDR, FGFR4, and calcineurin/NFAT target genes was determined. In adolescents on hemodialysis, we analyzed sequential echocardiograms, blood pressures and serial FGF23 measurements, and their relations to the cumulative administered dose of parenteral Pc. RESULTS The ratio of Pc dose/plasma levels of FGF23 correlated inversely (P < 0.005) with the cardiac mass in uremic rats and in hemodialysis patients, independently of hypertension. Despite persistently elevated FGF23 levels and myocardial FGFR4 activation, Pc suppressed upregulated myocardial calcineurin/NFAT target genes, and the effects were amplified by coadministration of PD173074. CONCLUSIONS The beneficial effects of Pc on uremic cardiac hypertrophy are counterbalanced by the increased FGF23 levels. Blockade of FGF23-mediated signaling increased the Pc-induced suppression of the myocardial calcineurin/NFAT system. Higher doses of Pc should be considered in the treatment of patients with uremic cardiomyopathy.
Collapse
Affiliation(s)
- Brian Czaya
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Wacharee Seeherunvong
- Division of Pediatric Nephrology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Saurav Singh
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Christopher Yanucil
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Phillip Ruiz
- Department of Surgery and Immunopathology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Yasmir Quiroz
- Hospital Universitario, Servicio de Nefrologia y Laboratorio de Inmunobiologia, Instituto Venezolano de Investigaciones Científicas (IVIC)-Zulia, Maracaibo, Venezuela
| | - Alexander Grabner
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Chryso Katsoufis
- Division of Pediatric Nephrology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sethuraman Swaminathan
- Division of Pediatric Cardiology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Carolyn Abitbol
- Division of Pediatric Nephrology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Bernardo Rodriguez-Iturbe
- Hospital Universitario, Servicio de Nefrologia y Laboratorio de Inmunobiologia, Instituto Venezolano de Investigaciones Científicas (IVIC)-Zulia, Maracaibo, Venezuela
| | - Christian Faul
- Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Michael Freundlich
- Division of Pediatric Nephrology, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
34
|
Shoji T, Inaba M, Fukagawa M, Ando R, Emoto M, Fujii H, Fujimori A, Fukui M, Hase H, Hashimoto T, Hirakata H, Honda H, Hosoya T, Ikari Y, Inaguma D, Inoue T, Isaka Y, Iseki K, Ishimura E, Itami N, Ito C, Kakuta T, Kawai T, Kawanishi H, Kobayashi S, Kumagai J, Maekawa K, Masakane I, Minakuchi J, Mitsuiki K, Mizuguchi T, Morimoto S, Murohara T, Nakatani T, Negi S, Nishi S, Nishikawa M, Ogawa T, Ohta K, Ohtake T, Okamura M, Okuno S, Shigematsu T, Sugimoto T, Suzuki M, Tahara H, Takemoto Y, Tanaka K, Tominaga Y, Tsubakihara Y, Tsujimoto Y, Tsuruya K, Ueda S, Watanabe Y, Yamagata K, Yamakawa T, Yano S, Yokoyama K, Yorioka N, Yoshiyama M, Nishizawa Y. Effect of Oral Alfacalcidol on Clinical Outcomes in Patients Without Secondary Hyperparathyroidism Receiving Maintenance Hemodialysis: The J-DAVID Randomized Clinical Trial. JAMA 2018; 320:2325-2334. [PMID: 30535217 PMCID: PMC6583075 DOI: 10.1001/jama.2018.17749] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 10/18/2018] [Indexed: 12/28/2022]
Abstract
IMPORTANCE Patients with chronic kidney disease have impaired vitamin D activation and elevated cardiovascular risk. Observational studies in patients treated with hemodialysis showed that the use of active vitamin D sterols was associated with lower risk of all-cause mortality, regardless of parathyroid hormone levels. OBJECTIVE To determine whether vitamin D receptor activators reduce cardiovascular events and mortality in patients without secondary hyperparathyroidism undergoing hemodialysis. DESIGN, SETTING, AND PARTICIPANTS Randomized, open-label, blinded end point multicenter study of 1289 patients in 207 dialysis centers in Japan. The study included 976 patients receiving maintenance hemodialysis with serum intact parathyroid hormone levels less than or equal to 180 pg/mL. The first and last participants were enrolled on August 18, 2008, and January 26, 2011, respectively. The final date of follow-up was April 4, 2015. INTERVENTIONS Treatment with 0.5 μg of oral alfacalcidol per day (intervention group; n = 495) vs treatment without vitamin D receptor activators (control group; n = 481). MAIN OUTCOMES AND MEASURES The primary outcome was a composite measure of fatal and nonfatal cardiovascular events, including myocardial infarctions, hospitalizations for congestive heart failure, stroke, aortic dissection/rupture, amputation of lower limb due to ischemia, and cardiac sudden death; coronary revascularization; and leg artery revascularization during 48 months of follow-up. The secondary outcome was all-cause death. RESULTS Among 976 patients who were randomized from 108 dialysis centers, 964 patients were included in the intention-to-treat analysis (median age, 65 years; 386 women [40.0%]), and 944 (97.9%) completed the trial. During follow-up (median, 4.0 years), the primary composite outcome of cardiovascular events occurred in 103 of 488 patients (21.1%) in the intervention group and 85 of 476 patients (17.9%) in the control group (absolute difference, 3.25% [95% CI, -1.75% to 8.24%]; hazard ratio, 1.25 [95% CI, 0.94-1.67]; P = .13). There was no significant difference in the secondary outcome of all-cause mortality between the groups (18.2% vs 16.8%, respectively; hazard ratio, 1.12 [95% CI, 0.83-1.52]; P = .46). Of the 488 participants in the intervention group, 199 (40.8%) experienced serious adverse events that were classified as cardiovascular, 64 (13.1%) experienced adverse events classified as infection, and 22 (4.5%) experienced malignancy-related serious adverse events. Of 476 participants in the control group, 191 (40.1%) experienced cardiovascular-related serious adverse events, 63 (13.2%) experienced infection-related serious adverse events, and 21 (4.4%) experienced malignancy-related adverse events. CONCLUSIONS AND RELEVANCE Among patients without secondary hyperparathyroidism undergoing maintenance hemodialysis, oral alfacalcidol compared with usual care did not reduce the risk of a composite measure of select cardiovascular events. These findings do not support the use of vitamin D receptor activators for patients such as these. TRIAL REGISTRATION UMIN-CTR Identifier: UMIN000001194.
Collapse
Affiliation(s)
- Tetsuo Shoji
- Department of Vascular Medicine, Osaka City University Graduate School of Medicine, Japan
- Vascular Science Center for Translational Research, Osaka City University Graduate School of Medicine, Japan
| | - Masaaki Inaba
- Vascular Science Center for Translational Research, Osaka City University Graduate School of Medicine, Japan
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Japan
| | - Masafumi Fukagawa
- Division of Nephrology, Endocrinology, and Metabolism, Tokai University School of Medicine, Kanagawa, Japan
| | - Ryoichi Ando
- Department of Nephrology, Musashino Red Cross Hospital, Tokyo, Japan
| | - Masanori Emoto
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Japan
| | - Hisako Fujii
- Department of Drug and Food Evaluation, Osaka City University Graduate School of Medicine, Japan
| | - Akira Fujimori
- Blood Purification and Kidney Center, Konan Hospital, Hyogo, Japan
| | - Mitsuru Fukui
- Laboratory of Statistics, Osaka City University Graduate School of Medicine, Japan
| | - Hiroki Hase
- Department of Nephrology, Toho University School of Medicine, Tokyo, Japan
| | | | - Hideki Hirakata
- Division of Nephrology, Fukuoka Renal Clinic, Fukuoka, Japan
| | - Hirokazu Honda
- Division of Nephrology, Department of Medicine, Showa University Koto Toyosu Hospital, Tokyo, Japan
| | - Tatsuo Hosoya
- Department of Pathophysiology and Therapy in Chronic Kidney Disease, The Jikei University School of Medicine, Tokyo, Japan
| | - Yuji Ikari
- Department of Cardiology, Tokai University School of Medicine, Kanagawa, Japan
| | - Daijo Inaguma
- Department of Nephrology, Fujita Health University School of Medicine, Aichi, Japan
| | | | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Japan
| | - Kunitoshi Iseki
- Clinical Research Support Center, Tomishiro Central Hospital, Japan
| | - Eiji Ishimura
- Department of Nephrology, Osaka City University Graduate School of Medicine, Japan
| | - Noritomo Itami
- Department of Nephrology, Itami Kidney Clinic, Hokkaido, Japan
| | - Chiharu Ito
- Department of Internal Medicine, Haga Red Cross Hospital, Tochigi, Japan
| | - Toshitaka Kakuta
- Division of Nephrology, Endocrinology, and Metabolism, Tokai University Hachioji Hospital, Tokyo, Japan
| | - Toru Kawai
- Medical Corporation Chuou Naika Clinic, Hiroshima, Japan
| | - Hideki Kawanishi
- Department of Artificial Organs, Tsuchiya General Hospital, Hiroshima, Japan
| | - Shuzo Kobayashi
- Department of Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kanagawa, Japan
| | - Junko Kumagai
- Akane Foundation Omachi Tsuchiya Clinic, Hiroshima, Japan
| | | | | | - Jun Minakuchi
- Department of Kidney Disease, Kawashima Hospital, Tokushima, Japan
| | - Koji Mitsuiki
- Nephrology and Dialysis Center, Japanese Red Cross Fukuoka Hospital, Japan
| | - Takashi Mizuguchi
- Department of Hematology, Dialysis, and Diabetes Mellitus, Kochi-Takasu Hospital, Kochi, Japan
| | - Satoshi Morimoto
- Department of Medicine, Endocrinology, and Hypertension, Tokyo Women's Medical University, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Tatsuya Nakatani
- Department of Urology, Osaka City University Graduate School of Medicine, Japan
| | - Shigeo Negi
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Shinichi Nishi
- Division of Nephrology and Kidney Center, Kobe University Graduate School of Medicine, Hyogo, Japan
| | | | - Tetsuya Ogawa
- Department of Medicine, Tokyo Women's Medical University Medical Center East, Tokyo, Japan
| | - Kazumichi Ohta
- Department of Urology, Kochi Takasu Hospital, Kochi, Japan
| | - Takayasu Ohtake
- Department of Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kanagawa, Japan
| | - Mikio Okamura
- Department of Internal Medicine, Kayashima Ikuno Hospital, Osaka, Japan
| | - Senji Okuno
- Department of Internal Medicine, Kidney Center, Shirasagi Hospital, Osaka, Japan
| | | | - Toshitsugu Sugimoto
- Department of Internal Medicine, Shimane University Faculty of Medicine, Shimane, Japan
| | - Masashi Suzuki
- Department of Nephrology, Shinraku-En Hospital, Niigata, Japan
| | | | - Yoshiaki Takemoto
- Department of Urology, Osaka City University Graduate School of Medicine, Japan
| | - Kenji Tanaka
- Department of Internal Medicine, Suiyukai Clinic, Nara, Japan
| | - Yoshihiro Tominaga
- Department of Transplant and Endocrine Surgery, Nagoya 2nd Red Cross Hospital Japan
| | - Yoshiharu Tsubakihara
- Department of Safety Management in Health Care Sciences, Graduate School of Health Care Sciences, Jikei Institute, Osaka, Japan
| | | | | | - Shinichiro Ueda
- Department of Clinical Pharmacology and Therapeutics, University of the Ryukyus Graduate School of Medicine, Okinawa, Japan
| | | | - Kunihiro Yamagata
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | | | - Shozo Yano
- Department of Laboratory Medicine, Shimane University Faculty of Medicine, Japan
| | - Keitaro Yokoyama
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | | | - Minoru Yoshiyama
- Department of Cardiovascular Medicine, Osaka City University Graduate School of Medicine, Japan
| | - Yoshiki Nishizawa
- Hemodialysis Center, Inoue Hospital, Soryu Medical Corporation, Osaka, Japan
| |
Collapse
|
35
|
Vila Cuenca M, Ferrantelli E, Meinster E, Pouw SM, Kovačević I, de Menezes RX, Niessen HW, Beelen RH, Hordijk PL, Vervloet MG. Vitamin D Attenuates Endothelial Dysfunction in Uremic Rats and Maintains Human Endothelial Stability. J Am Heart Assoc 2018; 7:e008776. [PMID: 30371149 PMCID: PMC6201442 DOI: 10.1161/jaha.118.008776] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 07/09/2018] [Indexed: 02/07/2023]
Abstract
Background Dysfunctional endothelium may contribute to the development of cardiovascular complications in chronic kidney disease ( CKD ). Supplementation with active vitamin D has been proposed to have vasoprotective potential in CKD , not only by direct effects on the endothelium but also by an increment of α-Klotho. Here, we explored the capacity of the active vitamin D analogue paricalcitol to protect against uremia-induced endothelial damage and the extent to which this was dependent on increased α-Klotho concentrations. Methods and Results In a combined rat model of CKD with vitamin D deficiency, renal failure induced vascular permeability and endothelial-gap formation in thoracic aorta irrespective of baseline vitamin D, and this was attenuated by paricalcitol. Downregulation of renal and serum α-Klotho was found in the CKD model, which was not restored by paricalcitol. By measuring the real-time changes of the human endothelial barrier function, we found that paricalcitol effectively improved the recovery of endothelial integrity following the addition of the pro-permeability factor thrombin and the induction of a wound. Furthermore, immunofluorescence staining revealed that paricalcitol promoted vascular endothelial-cadherin-based cell-cell junctions and diminished F-actin stress fiber organization, preventing the formation of endothelial intracellular gaps. Conclusions Our results demonstrate that paricalcitol attenuates the CKD -induced endothelial damage in the thoracic aorta and directly mediates endothelial stability in vitro by enforcing cell-cell interactions.
Collapse
Affiliation(s)
- Marc Vila Cuenca
- Department of NephrologyVU University Medical CenterAmsterdamThe Netherlands
- Amsterdam Cardiovascular SciencesAmsterdamThe Netherlands
| | - Evelina Ferrantelli
- Department of Molecular Cell Biology and ImmunologyVU University Medical CenterAmsterdamThe Netherlands
| | - Elisa Meinster
- Department of Pathology and Cardiac SurgeryVU University Medical CenterAmsterdamThe Netherlands
| | - Stephan M. Pouw
- Department of Molecular Cell Biology and ImmunologyVU University Medical CenterAmsterdamThe Netherlands
| | - Igor Kovačević
- Amsterdam Cardiovascular SciencesAmsterdamThe Netherlands
- Department of PhysiologyVU University Medical CenterAmsterdamThe Netherlands
| | - Renné X. de Menezes
- Department of Epidemiology and BiostatisticsVU University Medical CenterAmsterdamThe Netherlands
| | - Hans W. Niessen
- Amsterdam Cardiovascular SciencesAmsterdamThe Netherlands
- Department of Pathology and Cardiac SurgeryVU University Medical CenterAmsterdamThe Netherlands
| | - Robert H.J. Beelen
- Department of Molecular Cell Biology and ImmunologyVU University Medical CenterAmsterdamThe Netherlands
| | - Peter L. Hordijk
- Amsterdam Cardiovascular SciencesAmsterdamThe Netherlands
- Department of PhysiologyVU University Medical CenterAmsterdamThe Netherlands
| | - Marc G. Vervloet
- Department of NephrologyVU University Medical CenterAmsterdamThe Netherlands
- Amsterdam Cardiovascular SciencesAmsterdamThe Netherlands
| |
Collapse
|
36
|
Edmonston D, Morris JD, Middleton JP. Working Toward an Improved Understanding of Chronic Cardiorenal Syndrome Type 4. Adv Chronic Kidney Dis 2018; 25:454-467. [PMID: 30309463 DOI: 10.1053/j.ackd.2018.08.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/09/2018] [Accepted: 08/15/2018] [Indexed: 12/17/2022]
Abstract
Chronic diseases of the heart and of the kidneys commonly coexist in individuals. Certainly combined and persistent heart and kidney failure can arise from a common pathologic insult, for example, as a consequence of poorly controlled hypertension or of severe diffuse arterial disease. However, strong evidence is emerging to suggest that cross talk exists between the heart and the kidney. Independent processes are set in motion when kidney function is chronically diminished, and these processes can have distinct adverse effects on the heart. The complex chronic heart condition that results from chronic kidney disease (CKD) has been termed cardiorenal syndrome type 4. This review will include an updated description of the cardiac morphology in patients who have CKD, an overview of the most likely CKD-sourced culprits for these cardiac changes, and the potential therapeutic strategies to limit cardiac complications in patients who have CKD.
Collapse
|
37
|
Lekawanvijit S. Cardiotoxicity of Uremic Toxins: A Driver of Cardiorenal Syndrome. Toxins (Basel) 2018; 10:toxins10090352. [PMID: 30200452 PMCID: PMC6162485 DOI: 10.3390/toxins10090352] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 08/19/2018] [Accepted: 08/30/2018] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD) is highly prevalent in the setting of chronic kidney disease (CKD). Such coexistence of CVD and CKD—the so-called “cardiorenal or renocardiac syndrome”—contributes to exponentially increased risk of cardiovascular (CV) mortality. Uremic cardiomyopathy is a characteristic cardiac pathology commonly found in CKD. CKD patients are also predisposed to heart rhythm disorders especially atrial fibrillation. Traditional CV risk factors as well as known CKD-associated CV risk factors such as anemia are insufficient to explain CV complications in the CKD population. Accumulation of uremic retention solutes is a hallmark of impaired renal excretory function. Many of them have been considered inert solutes until their biological toxicity is unraveled and they become accepted as “uremic toxins”. Direct cardiotoxicity of uremic toxins has been increasingly demonstrated in recent years. This review offers a mechanistic insight into the pathological cardiac remodeling and dysfunction contributed by uremic toxins with a main focus on fibroblastic growth factor-23, an emerging toxin playing a central role in the chronic kidney disease–mineral bone disorder, and the two most investigated non-dialyzable protein-bound uremic toxins, indoxyl sulfate and p-cresyl sulfate. Potential therapeutic strategies that could address these toxins and their relevant mediated pathways since pre-dialysis stages are also discussed.
Collapse
Affiliation(s)
- Suree Lekawanvijit
- Department of Pathology, Faculty of Medicine, Chiang Mai University, 110 Intawaroros Rd, Sribhoom, Chiang Mai 50200, Thailand.
| |
Collapse
|
38
|
Tian XY, Ma S, Tse G, Wong WT, Huang Y. Uncoupling Protein 2 in Cardiovascular Health and Disease. Front Physiol 2018; 9:1060. [PMID: 30116205 PMCID: PMC6082951 DOI: 10.3389/fphys.2018.01060] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 07/16/2018] [Indexed: 12/22/2022] Open
Abstract
Uncoupling protein 2 (UCP2) belongs to the family of mitochondrial anion carrier proteins. It uncouples oxygen consumption from ATP synthesis. UCP2 is ubiquitously expressed in most cell types to reduce oxidative stress. It is tightly regulated at the transcriptional, translational, and post-translational levels. UCP2 in the cardiovascular system is being increasingly recognized as an important molecule to defend against various stress signals such as oxidative stress in the pathology of vascular dysfunction, atherosclerosis, hypertension, and cardiac injuries. UCP2 protects against cellular dysfunction through reducing mitochondrial oxidative stress and modulation of mitochondrial function. In view of the different functions of UCP2 in various cell types that contribute to whole body homeostasis, cell type-specific modification of UCP2 expression may offer a better approach to help understanding how UCP2 governs mitochondrial function, reactive oxygen species production and transmembrane proton leak and how dysfunction of UCP2 participates in the development of cardiovascular diseases. This review article provided an update on the physiological regulation of UCP2 in the cardiovascular system, and also discussed the involvement of UCP2 deficiency and associated oxidative stress in the pathogenesis of several common cardiovascular diseases. Drugs targeting UCP2 expression and activity might serve another effective strategy to ameliorate cardiovascular dysfunction. However, more detailed mechanistic study will be needed to dissect the role of UCP2, the regulation of UCP2 expression, and the cellular responses to the changes of UCP2 expression in normal and stressed situations at different stages of cardiovascular diseases.
Collapse
Affiliation(s)
- Xiao Yu Tian
- School of Biomedical Sciences, Institute of Vascular Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Shuangtao Ma
- Division of Nanomedicine and Molecular Intervention, Department of Medicine, Michigan State University, East Lansing, MI, United States
| | - Gary Tse
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Wing Tak Wong
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu Huang
- School of Biomedical Sciences, Institute of Vascular Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
39
|
Le TY, Ogawa M, Kizana E, Gunton JE, Chong JJ. Vitamin D Improves Cardiac Function After Myocardial Infarction Through Modulation of Resident Cardiac Progenitor Cells. Heart Lung Circ 2018; 27:967-975. [DOI: 10.1016/j.hlc.2018.01.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/04/2017] [Accepted: 01/04/2018] [Indexed: 01/02/2023]
|
40
|
Moretti R, Morelli ME, Caruso P. Vitamin D in Neurological Diseases: A Rationale for a Pathogenic Impact. Int J Mol Sci 2018; 19:2245. [PMID: 30065237 PMCID: PMC6121649 DOI: 10.3390/ijms19082245] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/25/2018] [Accepted: 07/26/2018] [Indexed: 12/22/2022] Open
Abstract
It is widely known that vitamin D receptors have been found in neurons and glial cells, and their highest expression is in the hippocampus, hypothalamus, thalamus and subcortical grey nuclei, and substantia nigra. Vitamin D helps the regulation of neurotrophin, neural differentiation, and maturation, through the control operation of growing factors synthesis (i.e., neural growth factor [NGF] and glial cell line-derived growth factor (GDNF), the trafficking of the septohippocampal pathway, and the control of the synthesis process of different neuromodulators (such as acetylcholine [Ach], dopamine [DA], and gamma-aminobutyric [GABA]). Based on these assumptions, we have written this review to summarize the potential role of vitamin D in neurological pathologies. This work could be titanic and the results might have been very fuzzy and even incoherent had we not conjectured to taper our first intentions and devoted our interests towards three mainstreams, demyelinating pathologies, vascular syndromes, and neurodegeneration. As a result of the lack of useful therapeutic options, apart from the disease-modifying strategies, the role of different risk factors should be investigated in neurology, as their correction may lead to the improvement of the cerebral conditions. We have explored the relationships between the gene-environmental influence and long-term vitamin D deficiency, as a risk factor for the development of different types of neurological disorders, along with the role and the rationale of therapeutic trials with vitamin D implementation.
Collapse
Affiliation(s)
- Rita Moretti
- Neurology Clinic, Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy.
| | - Maria Elisa Morelli
- Neurology Clinic, Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy.
| | - Paola Caruso
- Neurology Clinic, Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy.
| |
Collapse
|
41
|
Gluba-Brzózka A, Franczyk B, Ciałkowska-Rysz A, Olszewski R, Rysz J. Impact of Vitamin D on the Cardiovascular System in Advanced Chronic Kidney Disease (CKD) and Dialysis Patients. Nutrients 2018; 10:E709. [PMID: 29865146 PMCID: PMC6024710 DOI: 10.3390/nu10060709] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/24/2018] [Accepted: 05/29/2018] [Indexed: 01/07/2023] Open
Abstract
In patients suffering from chronic kidney disease (CKD), the prevalence of cardiovascular disease is much more common than in the general population. The role of vitamin D deficiency had been underestimated until a significant association was found between vitamin D therapy and survival benefit in haemodialysis patients. Vitamin D deficiency is present even in the early stages of chronic kidney disease. The results of experimental studies have revealed the relationship between vitamin D deficiency and impairment of cardiac contractile function, higher cardiac mass and increased myocardial collagen content. Experimental models propose that intermediate end points for the relationship between vitamin D deficiency and higher risk of cardiovascular disease comprise diminished left ventricular hypertrophy (LVH), enhanced left ventricular diastolic function, and decreased frequency of heart failure. Multiple observational studies have demonstrated an association between the use of active vitamin D therapy in patients on dialysis and with CKD and improved survival. However, there are also many studies indicating important adverse effects of such treatment. Therefore, large randomized trials are required to analyze whether supplementation of vitamin D may affect outcomes and whether it is safe to be used in CKD patients.
Collapse
Affiliation(s)
- Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, WAM Teaching Hospital, 90-549 Lodz, Poland.
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland.
| | | | - Robert Olszewski
- Department of Ultrasound, Institute of Fundamental Technological Research, Polish Academy of Sciences (IPPT PAN), 02-106 Warsaw, Poland.
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland.
| |
Collapse
|
42
|
Leifheit-Nestler M, Grabner A, Hermann L, Richter B, Schmitz K, Fischer DC, Yanucil C, Faul C, Haffner D. Vitamin D treatment attenuates cardiac FGF23/FGFR4 signaling and hypertrophy in uremic rats. Nephrol Dial Transplant 2018; 32:1493-1503. [PMID: 28339837 DOI: 10.1093/ndt/gfw454] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 12/06/2016] [Indexed: 11/13/2022] Open
Abstract
Background Vitamin D deficiency and excess of circulating fibroblast growth factor 23 (FGF23) contribute to cardiovascular mortality in patients with chronic kidney disease (CKD). FGF23 activates FGF receptor 4 and (FGFR4) calcineurin/nuclear factor of activated T cells (NFAT) signaling in cardiac myocytes, thereby causing left ventricular hypertrophy (LVH). Here, we determined if 1,25-dihydroxyvitamin D (calcitriol) inhibits FGF23-induced cardiac signaling and LVH. Methods 5/6 nephrectomized (5/6 Nx) rats were treated with different doses of calcitriol for 4 or 10 weeks and cardiac expression of FGF23/FGFR4 and activation of calcineurin/NFAT as well as LVH were analyzed. FGFR4 activation and hypertrophic cell growth were studied in cultured cardiac myocytes that were co-treated with FGF23 and calcitriol. Results In 5/6Nx rats with LVH, we detected elevated FGF23 expression in bone and myocardium, increased cardiac expression of FGFR4 and elevated cardiac activation of calcineurin/NFAT signaling. Cardiac expression levels of FGF23 and FGFR4 significantly correlated with the presence of LVH in uremic rats. Treatment with calcitriol reduced LVH as well as cardiac FGFR4 expression and calcineurin/NFAT activation. Bone and cardiac FGF23 expression were further stimulated by calcitriol in a dose-dependent manner, but levels of intact cardiac FGF23 protein were suppressed by high-dose calcitriol. In cultured cardiac myocytes, co-treatment with calcitriol blocked FGF23-induced activation of FGFR4 and hypertrophic cell growth. Conclusions Our data suggest that in CKD, cardioprotective effects of calcitriol stem from its inhibitory actions on the cardiac FGF23/FGFR4 system, and based on their counterbalancing effects on cardiac myocytes, high FGF23 and low calcitriol synergistically contribute to cardiac hypertrophy.
Collapse
Affiliation(s)
- Maren Leifheit-Nestler
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Alexander Grabner
- Department of Medicine, Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Laura Hermann
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Beatrice Richter
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Karin Schmitz
- Department of Pediatrics, University Hospital Rostock, Rostock, Germany
| | | | - Christopher Yanucil
- Department of Medicine, Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Cell Biology and Anatomy, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Christian Faul
- Department of Medicine, Katz Family Drug Discovery Center and Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Cell Biology and Anatomy, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| |
Collapse
|
43
|
Ito E, Inaguma D, Koide S, Takahashi K, Hayashi H, Hasegawa M, Yuzawa Y. Effect of combined vitamin D receptor activator and lanthanum carbonate on serum fibroblast growth factor 23 level in predialysis patients (CVD-LAF study): design and method. Clin Exp Nephrol 2018; 22:1309-1314. [PMID: 29748907 DOI: 10.1007/s10157-018-1584-0] [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: 01/29/2018] [Accepted: 04/19/2018] [Indexed: 11/26/2022]
Abstract
BACKGROUND Whether vitamin D receptor activator (VDRA) use is beneficial in chronic kidney disease (CKD) is unclear, because it is possible that VDRA increases serum fibroblast growth factor 23 (FGF23) levels. We will conduct a randomized controlled trial in predialysis patients to determine the effect of VDRA alone or in combination with lanthanum carbonate (LC) on serum FGF23 levels. METHODS This is a single-center, open-label, randomized controlled trial. Enrollment will commence February 1, 2018, using the following inclusion criteria: (1) age ≥ 20 years, (2) CKD with an estimated glomerular filtration rate of 10-45 mL/min/1.73 m2, (3) serum adjusted calcium level < 9.5 mg/dL, (4) serum phosphate level 4.0-6.0 mg/dL, and (5) serum intact parathyroid hormone (PTH) level ≥ 60 pg/mL. Study patients will be randomized 1:1 to receive alfacalcidol alone or in combination with LC. The initial dose of alfacalcidol will be 0.25-0.5 µg once a day according to serum adjusted calcium level. The initial dose of LC will be 250 mg once a day. We will measure serum intact and C-terminal FGF23 at 0, 4, 8, 12, 24, and 52 weeks. The primary outcome will be serum FGF23 level at 24 weeks compared with baseline. DISCUSSION This study aims to determine whether low-dose oral VDRA increases serum FGF23 level and whether the combination of VDRA and LC inhibits this increase. The results will be useful in the management of CKD-mineral and bone disorder in predialysis patients. TRIAL REGISTRATION UMIN000030503. Registered 20 January 2018.
Collapse
Affiliation(s)
- Eri Ito
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Daijo Inaguma
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan.
| | - Shigehisa Koide
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Kazuo Takahashi
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Hiroki Hayashi
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Midori Hasegawa
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Yukio Yuzawa
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| |
Collapse
|
44
|
Zhang L, Yan X, Zhang YL, Bai J, Hidru TH, Wang QS, Li HH. Vitamin D attenuates pressure overload-induced cardiac remodeling and dysfunction in mice. J Steroid Biochem Mol Biol 2018; 178:293-302. [PMID: 29337094 DOI: 10.1016/j.jsbmb.2018.01.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 12/28/2022]
Abstract
Vitamin D (VD) and its analogues play critical roles in metabolic and cardiovascular diseases. Recent studies have demonstrated that VD exerts a protective role in cardiovascular diseases. However, the beneficial effect of VD on pressure overload-induced cardiac remodeling and dysfunction and its underlying mechanisms are not fully elucidated. In this study, cardiac dysfunction and hypertrophic remodeling in mice were induced by pressure overload. Cardiac function was evaluated by echocardiography, and myocardial histology was detected by H&E and Masson's trichrome staining. Cardiomyocyte size was detected by wheat germ agglutinin staining. The protein levels of signaling mediators were examined by western blotting while mRNA expression of hypertrophic and fibrotic markers was examined by qPCR analysis. Oxidative stress was detected by dihydroethidine staining. Our results showed that administration of VD3 significantly ameliorates pressure overload-induced contractile dysfunction, cardiac hypertrophy, fibrosis and inflammation in mice. In addition, VD3 treatment also markedly inhibited cardiac oxidative stress and apoptosis. Moreover, protein levels of calcineurin A, ERK1/2, AKT, TGF-β, GRP78, cATF6, and CHOP were significantly reduced whereas SERCA2 level was upregulated in the VD3-treated hearts compared with control. These results suggest that VD3 attenuates cardiac remodeling and dysfunction induced by pressure overload, and this protective effect is associated with inhibition of multiple signaling pathways.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China; Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiao Yan
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China; Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yun-Long Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China; Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Jie Bai
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China; Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | | | - Qing-Shan Wang
- School of Public Health, Dalian Medical University, Dalian 116044, China.
| | - Hui-Hua Li
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China; Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
45
|
Navarro-García JA, Fernández-Velasco M, Delgado C, Delgado JF, Kuro-O M, Ruilope LM, Ruiz-Hurtado G. PTH, vitamin D, and the FGF-23-klotho axis and heart: Going beyond the confines of nephrology. Eur J Clin Invest 2018; 48. [PMID: 29394451 DOI: 10.1111/eci.12902] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 01/28/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Profound disturbances in mineral metabolism are closely linked to the progression of chronic kidney disease. However, increasing clinical and experimental evidence indicates that alterations in phosphate homoeostasis could have an even stronger impact on the heart. AIM The aim of this review is to provide the reader with an update of how alterations in mineral metabolism are related to direct and indirect cardiotoxic effects beyond the nephrology setting. RESULTS Evidence exists that alterations in mineral metabolism that are related to changes in parathyroid hormone (PTH), vitamin D, and the FGF-23-klotho axis have direct pathological consequences for the heart. Alterations in plasma PTH levels are associated with cardiac dysfunction and detrimental cardiac remodelling. Several clinical studies have associated vitamin D deficiency with the prevalence of cardiovascular disease (CV) and its risk factors. Recent evidences support deleterious direct and nonphosphaturic effects of FGF-23 on the heart as hypertrophy development. In contrast, reduced systemic klotho levels are related to CV damage, at least when advanced age is present. In addition, we discuss how these mineral metabolism molecules can counteract each other in some situations, in the context of failed clinical trials on cardiac protection as is the case of vitamin D supplementation. CONCLUSIONS Among all mineral components, an increase in systemic FGF-23 levels is considered to have the greatest CV impact and risk. However, it is quite possible that many intracellular mechanisms mediated by FGF-23, especially those related to cardiomyocyte function, remain to be discovered.
Collapse
Affiliation(s)
- José Alberto Navarro-García
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario de Octubre/CIBERCV, Madrid, Spain
| | | | - Carmen Delgado
- Biomedical Research Institute Alberto Sols/CIBERCV, Madrid, Spain
| | - Juan F Delgado
- Cardiology Service, Hospital Universitario 12 de Octubre/CIBERCV, Madrid, Spain
| | - Makoto Kuro-O
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Luis M Ruilope
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario de Octubre/CIBERCV, Madrid, Spain.,Department of Preventive Medicine and Public Health, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain.,European University of Madrid, Madrid, Spain
| | - Gema Ruiz-Hurtado
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario de Octubre/CIBERCV, Madrid, Spain
| |
Collapse
|
46
|
Ureña-Torres PA, Cozzolino M, Bover J. [Utilization of alfacalcidol and active vitamin D analogs in chronic kidney disease]. Nephrol Ther 2018; 14:189-200. [PMID: 29545131 DOI: 10.1016/j.nephro.2017.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/01/2017] [Accepted: 03/16/2017] [Indexed: 01/22/2023]
Abstract
Secondary hyperparathyroidism (SHPT) is one of the most frequent and deleterious complication of chronic kidney disease (CKD). SHPT is also one of the principal components of the now called CKD-mineral and bone disorders (MBD) syndrome. It is usually prevented and treated by vitamin D derivatives. However, the rationale for the prescription of vitamin D sterols in those patients is still a matter of hotly debates, mainly because of unsatisfactory results from numerous observational and not well-controlled studies. Scanty clinical data on head-to-head comparisons between the multiple vitamin D sterols are currently available. Moreover, there is crescent expectations on nutritional vitamin D, as well as vitamin D receptor activators (VDRA), regarding their putative pleiotropic effects even in CKD patients, and the promising effects of VDRA against proteinuria and myocardial hypertrophy in diabetic CKD cohorts. Nevertheless, additional randomized controlled trials (RCT) are needed to answer to many open questions and incertitude considering the effect of nutritional vitamin D and VDRA on hard end points including the risk of skeletal fractures and of mortality in CKD patients. RCT comparing VDRA to calcimimetics in the control of SHPT are also needed in dialysis patients. The present review will visit these open questions that nephrologists should ask before starting a treatment by nutritional vitamin D or VDRA.
Collapse
Affiliation(s)
- Pablo Antonio Ureña-Torres
- Ramsay générale de santé, service de néphrologie et dialyse, clinique du Landy, 23, rue du Landy, 93400 Saint-Ouen, France; Department of renal physiology, Necker hospital, university of Paris Descartes, 75015 Paris, France.
| | - Mario Cozzolino
- Renal division, San Paolo hospital, Depart of health sciences, University of Milan, Milan, Italie
| | - Jordi Bover
- Fundació Puigvert, Department of nephrology, C./Cartagena 340-350, 08025 Barcelona, Espagne
| |
Collapse
|
47
|
Ludvigsen S, Mancusi C, Kildal S, de Simone G, Gerdts E, Ytrehus K. Cardiac adaptation to hypertension in adult female Dahl salt-sensitive rats is dependent on ovarian function, but loss of ovarian function does not predict early maladaptation. Physiol Rep 2018; 6:e13593. [PMID: 29417743 PMCID: PMC5803524 DOI: 10.14814/phy2.13593] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 12/19/2017] [Accepted: 12/20/2017] [Indexed: 01/25/2023] Open
Abstract
Aim of study was to examine experimentally the adult female hypertensive heart in order to determine the role of ovary function in the response of the heart to salt-dependent hypertension. Dahl salt-sensitive rats, age 12 weeks, with/without ovariectomy were fed a standard (0.3% NaCl) or high-salt diet (8%) for 16 weeks. Mean arterial blood pressure monitored noninvasively in conscious state increased significantly by high salt. Echocardiography was performed at baseline and endpoint. Heart function and molecular changes were evaluated at endpoint by left ventricle catheterization, by sirius red staining for collagen and by gene expression using quantitative RT-PCR for selected genes. At endpoint, significant concentric hypertrophy was present with high salt. Increase in relative wall thickening with high salt compared to normal diet was more pronounced with intact ovaries (0.33 ± 0.02 and 0.57 ± 0.04 vs. 0.29 ± 0.00 and 0.46 ± 0.03) as was the reduction in midwall fractional shortening (20 ± 0.6 and 14 ± 2 vs. 19 ± 0.9 and 18 ± 1). Ovariectomy increased stroke volume and decreased the ratio of mitral peak velocity of early filling (E) to early diastolic mitral annular velocity (E') (E/E' ratio) when compared to hearts from intact rats. High salt increased expression of collagen I and III genes and perivascular collagen in the heart slightly, but % interstitial collagen by sirius red staining remained unchanged in intact rats and decreased significantly by ovariectomy. Added volume load but not deterioration of function or structure characterized the nonfailing hypertensive heart of salt-sensitive females ovariectomized at mature age when compared to corresponding intact females.
Collapse
Affiliation(s)
- Stian Ludvigsen
- Cardiovascular Research GroupDepartment of Medical BiologyUiT – The Arctic University of NorwayTromsøNorway
| | - Costantino Mancusi
- Hypertension Research CenterFederico II University of NaplesNaplesItaly
- Department of Clinical ScienceUniversity of BergenBergenNorway
| | - Simon Kildal
- Cardiovascular Research GroupDepartment of Medical BiologyUiT – The Arctic University of NorwayTromsøNorway
| | | | - Eva Gerdts
- Department of Clinical ScienceUniversity of BergenBergenNorway
| | - Kirsti Ytrehus
- Cardiovascular Research GroupDepartment of Medical BiologyUiT – The Arctic University of NorwayTromsøNorway
| |
Collapse
|
48
|
Abstract
PURPOSE Vitamin D is principally known for its role in calcium homeostasis, but preclinical studies implicate multiple pathways through which vitamin D may affect cardiovascular function and influence risk for heart failure. Many adults with cardiovascular disease have low vitamin D status, making it a potential therapeutic target. We review the rationale and potential role of vitamin D supplementation in the prevention and treatment of chronic heart failure. RECENT FINDINGS Substantial observational evidence has associated low vitamin D status with the risk of heart failure, ventricular remodeling, and clinical outcomes in heart failure, including mortality. However, trials assessing the influence of vitamin D supplementation on surrogate markers and clinical outcomes in heart failure have generally been small and inconclusive. There are insufficient data to recommend routine assessment or supplementation of vitamin D for the prevention or treatment of chronic heart failure. Prospective trials powered for clinical outcomes are warranted.
Collapse
|
49
|
Wimalawansa SJ. Vitamin D and cardiovascular diseases: Causality. J Steroid Biochem Mol Biol 2018; 175:29-43. [PMID: 28027913 DOI: 10.1016/j.jsbmb.2016.12.016] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/01/2016] [Accepted: 12/23/2016] [Indexed: 12/21/2022]
Abstract
Vitamin D regulates blood pressure, cardiac functions, and endothelial and smooth muscle cell functions, thus, playing an important role in cardiovascular health. Observational studies report associations between vitamin D deficiency with hypertension and cardiovascular-related deaths. Peer-reviewed papers were examined in several research databases as per the guidelines of the Preferred Reporting Items for Systematic Reviews, using key words that address the relationship between vitamin D and cardiovascular disease. Correlations and interpretations were made considering the risks-benefits, broader evidence, and implications. This review analyzed current knowledge regarding the effects of vitamin D on the cardiovascular system. 1,25(OH)2D and related epigenetic modifications subdue cellular inflammation, improve overall endothelial functions, reduce age-related systolic hypertension and vascular rigidity, and attenuate the actions of the renin-angiotensin-aldosterone system. Most observational and ecological studies support 25(OH)vitamin D having protective effects on the cardiovascular system. However, the association of vitamin D deficiency with cardiovascular diseases is based primarily on observational and ecological studies and thus, is a matter of controversy. Adequately powered, randomized controlled clinical trial data are not available to confirm these associations. Thus, to test the hypothesis that correction of vitamin D deficiency protects the cardiovascular system, well-designed, statistically powered, longer-term clinical trials are needed in persons with vitamin D deficiency. Nevertheless, the available data support that adequate vitamin D supplementation and/or sensible sunlight exposure to achieve optimal vitamin D status are important in the prevention of cardiovascular disease and other chronic diseases.
Collapse
Affiliation(s)
- Sunil J Wimalawansa
- Professor of Medicine, Endocrinology & Nutrition, Cardio Metabolic Institute, NJ, USA.
| |
Collapse
|
50
|
Jamali N, Sorenson CM, Sheibani N. Vitamin D and regulation of vascular cell function. Am J Physiol Heart Circ Physiol 2017; 314:H753-H765. [PMID: 29351464 DOI: 10.1152/ajpheart.00319.2017] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Vitamin D deficiency is linked to pathogenesis of many diseases including cardiovascular, cancer, and various eye diseases. In recent years, important roles for vitamin D in regulation of immune function, inflammation, angiogenesis, and aging have been demonstrated. Thus, vitamin D and its analogs have been evaluated for the treatment of various types of cancer and chronic diseases. We have previously shown that the active form of vitamin D [1,25(OH)2D3] is a potent inhibitor of angiogenesis. This activity is consistent with the important role proposed for vitamin D and its analogs in the mitigation of tumor growth through inhibition of angiogenesis. Here, we review the important nutritional value of vitamin D and the abnormalities linked to its deficiency. We will explore its potential role as a regulator of angiogenesis and vascular cell function and the role vitamin D receptor (VDR) expression plays in these activities during vascular development and neovascularization. Our studies have established an important role for 1,25(OH)2D3 and VDR in the regulation of perivascular supporting cell function. In addition, the interaction of 1,25(OH)2D3 and VDR is essential for these activities and inhibition of neovascularization. Delineating the signaling pathways involved and identification of genes that are the target of 1,25(OH)2D3 regulation in vascular cells will allow us to identify novel pathways that are targets for regulation of vascular function and angiogenesis.
Collapse
Affiliation(s)
- Nasim Jamali
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health , Madison, Wisconsin.,McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health , Madison, Wisconsin
| | - Christine M Sorenson
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health , Madison, Wisconsin.,Department of Pediatrics, University of Wisconsin School of Medicine and Public Health , Madison, Wisconsin
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health , Madison, Wisconsin.,McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health , Madison, Wisconsin.,Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health , Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin School of Medicine and Public Health , Madison, Wisconsin
| |
Collapse
|