1
|
Yottasan P, Chu T, Chhetri PD, Cil O. Repurposing calcium-sensing receptor activator drug cinacalcet for ADPKD treatment. Transl Res 2024; 265:17-25. [PMID: 37990828 PMCID: PMC10922239 DOI: 10.1016/j.trsl.2023.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/17/2023] [Accepted: 10/27/2023] [Indexed: 11/23/2023]
Abstract
ADPKD is characterized by progressive cyst formation and enlargement leading to kidney failure. Tolvaptan is currently the only FDA-approved treatment for ADPKD; however, it can cause serious adverse effects including hepatotoxicity. There remains an unmet clinical need for effective and safe treatments for ADPKD. The extracellular Ca2+-sensing receptor (CaSR) is a regulator of epithelial ion transport. FDA-approved CaSR activator cinacalcet can reduce cAMP-induced Cl- and fluid secretion in various epithelial cells by activating phosphodiesterases (PDE) that hydrolyze cAMP. Since elevated cAMP is a key mechanism of ADPKD progression by promoting cell proliferation, cyst formation and enlargement (via Cl- and fluid secretion), here we tested efficacy of cinacalcet in cell and animal models of ADPKD. Cinacalcet treatment reduced cAMP-induced Cl- secretion and CFTR activity in MDCK cells as suggested by ∼70 % lower short-circuit current (Isc) changes in response to forskolin and CFTRinh-172, respectively. Cinacalcet treatment inhibited forskolin-induced cAMP elevation by 60 % in MDCK cells, and its effect was completely reversed by IBMX (PDE inhibitor). In MDCK cells treated with forskolin, cinacalcet treatment concentration-dependently reduced cell proliferation, cyst formation and cyst enlargement by up to 50 % without affecting cell viability. Cinacalcet treatment (20 mg/kg/day for 7 days, subcutaneous) reduced renal cyst index in a mouse model of ADPKD (Pkd1flox/flox;Ksp-Cre) by 20 %. Lastly, cinacalcet treatment reduced cyst enlargement and cell proliferation in human ADPKD cells by 60 %. Considering its efficacy as shown here, and favorable safety profile including extensive post-approval data, cinacalcet can be repurposed as a novel ADPKD treatment.
Collapse
Affiliation(s)
- Pattareeya Yottasan
- Department of Pediatrics, University of California, San Francisco, 513 Parnassus Avenue, HSE 1244, San Francisco, CA, 94143, United States
| | - Tifany Chu
- Department of Pediatrics, University of California, San Francisco, 513 Parnassus Avenue, HSE 1244, San Francisco, CA, 94143, United States
| | - Parth D Chhetri
- Department of Pediatrics, University of California, San Francisco, 513 Parnassus Avenue, HSE 1244, San Francisco, CA, 94143, United States
| | - Onur Cil
- Department of Pediatrics, University of California, San Francisco, 513 Parnassus Avenue, HSE 1244, San Francisco, CA, 94143, United States.
| |
Collapse
|
2
|
Chu T, Yottasan P, Goncalves LDS, Oak AA, Lin R, Tse M, Donowitz M, Cil O. Calcium-sensing receptor activator cinacalcet for treatment of cyclic nucleotide-mediated secretory diarrheas. Transl Res 2024; 263:45-52. [PMID: 37678755 PMCID: PMC11071643 DOI: 10.1016/j.trsl.2023.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 07/24/2023] [Accepted: 09/01/2023] [Indexed: 09/09/2023]
Abstract
Cyclic nucleotide elevation in intestinal epithelial cells is the key pathology causing intestinal fluid loss in secretory diarrheas such as cholera. Current secretory diarrhea treatment is primarily supportive, and oral rehydration solution is the mainstay of cholera treatment. There is an unmet need for safe, simple and effective diarrhea treatments. By promoting cAMP hydrolysis, extracellular calcium-sensing receptor (CaSR) is a regulator of intestinal fluid transport. We studied the antidiarrheal mechanisms of FDA-approved CaSR activator cinacalcet and tested its efficacy in clinically relevant human cell, mouse and intestinal organoid models of secretory diarrhea. By using selective inhibitors, we found that cAMP agonists-induced secretory short-circuit currents (Isc) in human intestinal T84 cells are mediated by collective actions of apical membrane cystic fibrosis transmembrane conductance regulator (CFTR) and Clc-2 Cl- channels, and basolateral membrane K+ channels. 30 μM cinacalcet pretreatment inhibited all 3 components of forskolin and cholera toxin-induced secretory Isc by ∼75%. In mouse jejunal mucosa, cinacalcet inhibited forskolin-induced secretory Isc by ∼60% in wild type mice, with no antisecretory effect in intestinal epithelia-specific Casr knockout mice (Casr-flox; Vil1-cre). In suckling mouse model of cholera induced by oral cholera toxin, single dose (30 mg/kg) oral cinacalcet treatment reduced intestinal fluid accumulation by ∼55% at 20 hours. Lastly, cinacalcet inhibited forskolin-induced secretory Isc by ∼75% in human colonic and ileal organoids. Our findings suggest that CaSR activator cinacalcet has antidiarrheal efficacy in distinct human cell, organoid and mouse models of secretory diarrhea. Considering its excellent clinical safety profile, cinacalcet can be repurposed as a treatment for cyclic nucleotide-mediated secretory diarrheas including cholera.
Collapse
Affiliation(s)
- Tifany Chu
- Department of Pediatrics, University of California, San Francisco, California
| | - Pattareeya Yottasan
- Department of Pediatrics, University of California, San Francisco, California
| | | | - Apurva A Oak
- Department of Pediatrics, University of California, San Francisco, California
| | - Ruxian Lin
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ming Tse
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mark Donowitz
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Onur Cil
- Department of Pediatrics, University of California, San Francisco, California.
| |
Collapse
|
3
|
ter Braake AD, Vervloet MG, de Baaij JHF, Hoenderop JGJ. Magnesium to prevent kidney disease-associated vascular calcification: crystal clear? Nephrol Dial Transplant 2022; 37:421-429. [PMID: 33374019 PMCID: PMC8875474 DOI: 10.1093/ndt/gfaa222] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular calcification is a prognostic marker for cardiovascular mortality in chronic kidney disease (CKD) patients. In these patients, magnesium balance is disturbed, mainly due to limited ultrafiltration of this mineral, changes in dietary intake and the use of diuretics. Observational studies in dialysis patients report that a higher blood magnesium concentration is associated with reduced risk to develop vascular calcification. Magnesium prevents osteogenic vascular smooth muscle cell transdifferentiation in in vitro and in vivo models. In addition, recent studies show that magnesium prevents calciprotein particle maturation, which may be the mechanism underlying the anti-calcification properties of magnesium. Magnesium is an essential protective factor in the calcification milieu, which helps to restore the mineral-buffering system that is overwhelmed by phosphate in CKD patients. The recognition that magnesium is a modifier of calciprotein particle maturation and mineralization of the extracellular matrix renders it a promising novel clinical tool to treat vascular calcification in CKD. Consequently, the optimal serum magnesium concentration for patients with CKD may be higher than in the general population.
Collapse
Affiliation(s)
- Anique D ter Braake
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marc G Vervloet
- Amsterdam Cardiovascular Sciences, Amsterdam UMC, Location VU University Medical Center, Amsterdam, The Netherlands
| | - Jeroen H F de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
4
|
Kim IY, Ye BM, Kim MJ, Kim SR, Lee DW, Kim HJ, Rhee H, Song SH, Seong EY, Lee SB. 1,25-dihydroxyvitamin D deficiency is independently associated with cardiac valve calcification in patients with chronic kidney disease. Sci Rep 2022; 12:915. [PMID: 35042976 PMCID: PMC8766529 DOI: 10.1038/s41598-022-04981-x] [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: 07/13/2021] [Accepted: 01/05/2022] [Indexed: 11/12/2022] Open
Abstract
Cardiac valve calcification is highly prevalent in patients with chronic kidney disease (CKD). Low vitamin D levels are associated with vascular calcification in CKD. However, the association between vitamin D levels and cardiac valve calcification is unknown. A total of 513 patients with pre-dialysis CKD were included in this cross-sectional study. Aortic valve calcification (AVC) and mitral valve calcification (MVC) were assessed using two-dimensional echocardiography. The associations between AVC and MVC and baseline variables were investigated using logistic regression analyses. In multivariable analysis, serum 1,25(OH)2D level was independently associated with AVC (odds ratio [OR], 0.87; P < 0.001) and MVC (OR, 0.92; P < 0.001). Additionally, age, diabetes, coronary heart disease, calcium × phosphate product, and intact parathyroid hormone levels were independently associated with AVC and MVC. Systolic blood pressure was independently associated with AVC. A receiver-operating characteristic (ROC) curve analysis showed that the best cutoff values of serum 1,25(OH)2D levels for predicting AVC and MVC were ≤ 12.5 and ≤ 11.9 pg/dl, respectively. Serum 1,25(OH)2D deficiency is independently associated with AVC and MVC in patients with CKD, suggesting that serum 1,25(OH)2D level may be a potential biomarker of AVC and MVC in these patients.
Collapse
Affiliation(s)
- Il Young Kim
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, South Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Byung Min Ye
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, South Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Min Jeong Kim
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, South Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Seo Rin Kim
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, South Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Dong Won Lee
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, South Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea
| | - Hyo Jin Kim
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, South Korea
- Medical Research Institute, Pusan National University Hospital, Busan, South Korea
| | - Harin Rhee
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, South Korea
- Medical Research Institute, Pusan National University Hospital, Busan, South Korea
| | - Sang Heon Song
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, South Korea
- Medical Research Institute, Pusan National University Hospital, Busan, South Korea
| | - Eun Young Seong
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, South Korea
- Medical Research Institute, Pusan National University Hospital, Busan, South Korea
| | - Soo Bong Lee
- Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, South Korea.
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, South Korea.
| |
Collapse
|
5
|
Aguilera-Méndez A, Boone-Villa D, Nieto-Aguilar R, Villafaña-Rauda S, Molina AS, Sobrevilla JV. Role of vitamins in the metabolic syndrome and cardiovascular disease. Pflugers Arch 2021; 474:117-140. [PMID: 34518916 DOI: 10.1007/s00424-021-02619-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 08/14/2021] [Accepted: 08/30/2021] [Indexed: 12/25/2022]
Abstract
The prevalence of metabolic syndrome and cardiovascular disease has increased and continues to be the leading cause of mortality worldwide. The etiology of these diseases includes a complex phenotype derived from interactions between genetic, environmental, and nutritional factors. In this regard, it is common to observe vitamin deficiencies in the general population and even more in patients with cardiometabolic diseases due to different factors. Vitamins are essential micronutrients for cellular metabolism and their deficiencies result in diseases. In addition to its role in nutritional functions, increasingly, vitamins are being recognized as modulators of genetics expression and signals transduction, when consumed at pharmacological concentrations. Numerous randomized preclinical and clinical trials have evaluated the use of vitamin supplementation in the prevention and treatment of metabolic syndrome and cardiovascular disease. However, it is controversy regarding its efficacy in the treatment and prevention of these diseases. In this review, we investigated chemical basics, physiological effect and recommended daily intake, problems with deficiency and overdose, preclinical and clinical studies, and mechanisms of action of vitamin supplementation in the treatment and prevention of metabolic syndrome and cardiovascular disease.
Collapse
Affiliation(s)
- Asdrubal Aguilera-Méndez
- Institute of Biological Chemistry Research, Universidad Michoacana de San Nicolás de Hidalgo, Av. J. Mújica, Edificio B3, Ciudad Universitaria, CP, 58030, Morelia, Michoacán, México.
| | - Daniel Boone-Villa
- School of Medicine, North Section, Universidad Autónoma de Coahuila, Piedras Negras, 26090, Coahuila, México
| | - Renato Nieto-Aguilar
- University Center for Postgraduate Studies and Research, School of Dentistry, Universidad Michoacana de San Nicolás de Hidalgo, 58337, Morelia, Michoacán, México
| | - Santiago Villafaña-Rauda
- Postgraduate Section, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, México
| | - Alfredo Saavedra Molina
- Institute of Biological Chemistry Research, Universidad Michoacana de San Nicolás de Hidalgo, Av. J. Mújica, Edificio B3, Ciudad Universitaria, CP, 58030, Morelia, Michoacán, México
| | - Janeth Ventura Sobrevilla
- School of Medicine, North Section, Universidad Autónoma de Coahuila, Piedras Negras, 26090, Coahuila, México
| |
Collapse
|
6
|
Donato M, Faggin E, Cinetto F, Felice C, Lupo MG, Ferri N, Rattazzi M. The Emerging Role of Nutraceuticals in Cardiovascular Calcification: Evidence from Preclinical and Clinical Studies. Nutrients 2021; 13:nu13082603. [PMID: 34444763 PMCID: PMC8401694 DOI: 10.3390/nu13082603] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 02/03/2023] Open
Abstract
Cardiovascular calcification is the ectopic deposition of calcium-phosphate crystals within the arterial wall and the aortic valve leaflets. This pathological process leads to increased vascular stiffness, reduced arterial elasticity, and aortic valve stenosis, increasing the risk of cardiovascular diseases. Although cardiovascular calcification is an increasing health care burden, to date no medical therapies have been approved for treating or preventing it. Considering the current lack of therapeutic strategies and the increasing prevalence of cardiovascular calcification, the investigation of some nutraceuticals to prevent this pathological condition has become prevalent in recent years. Recent preclinical and clinical studies evaluated the potential anti-calcific role of nutraceuticals (including magnesium, zinc, iron, vitamin K, and phytate) in the progression of vascular calcification, providing evidence for their dietary supplementation, especially in high-risk populations. The present review summarizes the current knowledge and latest advances for nutraceuticals with the most relevant preclinical and clinical data, including magnesium, zinc, iron, vitamin K, and phytate. Their supplementation might be recommended as a cost-effective strategy to avoid nutritional deficiency and to prevent or treat cardiovascular calcification. However, the optimal dose of nutraceuticals has not been identified and large interventional trials are warranted to support their protective effects on cardiovascular disease.
Collapse
Affiliation(s)
- Maristella Donato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padua, Italy; (M.D.); (M.G.L.); (N.F.)
| | - Elisabetta Faggin
- Department of Medicine—DIMED, University of Padova, 35122 Padua, Italy; (E.F.); (F.C.); (C.F.)
| | - Francesco Cinetto
- Department of Medicine—DIMED, University of Padova, 35122 Padua, Italy; (E.F.); (F.C.); (C.F.)
- Medicina Generale I^, Ca’ Foncello Hospital, 31100 Treviso, Italy
| | - Carla Felice
- Department of Medicine—DIMED, University of Padova, 35122 Padua, Italy; (E.F.); (F.C.); (C.F.)
- Medicina Generale I^, Ca’ Foncello Hospital, 31100 Treviso, Italy
| | - Maria Giovanna Lupo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padua, Italy; (M.D.); (M.G.L.); (N.F.)
| | - Nicola Ferri
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padua, Italy; (M.D.); (M.G.L.); (N.F.)
| | - Marcello Rattazzi
- Department of Medicine—DIMED, University of Padova, 35122 Padua, Italy; (E.F.); (F.C.); (C.F.)
- Medicina Generale I^, Ca’ Foncello Hospital, 31100 Treviso, Italy
- Correspondence: ; Tel.: +39-04-9821-1867 or +39-04-2232-2207
| |
Collapse
|
7
|
Inflammation: a putative link between phosphate metabolism and cardiovascular disease. Clin Sci (Lond) 2021; 135:201-227. [PMID: 33416083 PMCID: PMC7796315 DOI: 10.1042/cs20190895] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 02/06/2023]
Abstract
Dietary habits in the western world lead to increasing phosphate intake. Under physiological conditions, extraosseous precipitation of phosphate with calcium is prevented by a mineral buffering system composed of calcification inhibitors and tight control of serum phosphate levels. The coordinated hormonal regulation of serum phosphate involves fibroblast growth factor 23 (FGF23), αKlotho, parathyroid hormone (PTH) and calcitriol. A severe derangement of phosphate homeostasis is observed in patients with chronic kidney disease (CKD), a patient collective with extremely high risk of cardiovascular morbidity and mortality. Higher phosphate levels in serum have been associated with increased risk for cardiovascular disease (CVD) in CKD patients, but also in the general population. The causal connections between phosphate and CVD are currently incompletely understood. An assumed link between phosphate and cardiovascular risk is the development of medial vascular calcification, a process actively promoted and regulated by a complex mechanistic interplay involving activation of pro-inflammatory signalling. Emerging evidence indicates a link between disturbances in phosphate homeostasis and inflammation. The present review focuses on critical interactions of phosphate homeostasis, inflammation, vascular calcification and CVD. Especially, pro-inflammatory responses mediating hyperphosphatemia-related development of vascular calcification as well as FGF23 as a critical factor in the interplay between inflammation and cardiovascular alterations, beyond its phosphaturic effects, are addressed.
Collapse
|
8
|
Singh A, Tandon S, Tandon C. An update on vascular calcification and potential therapeutics. Mol Biol Rep 2021; 48:887-896. [PMID: 33394226 DOI: 10.1007/s11033-020-06086-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 12/12/2020] [Indexed: 02/07/2023]
Abstract
Pathological calcification is a major cause of cardiovascular morbidities primarily in population with chronic kidney disease (CKD), end stage renal diseases (ERSD) and metabolic disorders. Investigators have accepted the fact that vascular calcification is not a passive process but a highly complex, cell mediated, active process in patients with cardiovascular disease (CVD) resulting from, metabolic insults of bone fragility, diabetes, hypertension, dyslipidemia and atherosclerosis. Over the years, studies have revealed various mechanisms of vascular calcification like induction of bone formation, apoptosis, alteration in Ca-P balance and loss of inhibition. Novel clinical studies targeting cellular mechanisms of calcification provide promising and potential avenues for drug development. The interventions include phosphate binders, sodium thiosulphate, vitamin K, calcimimetics, vitamin D, bisphosphonates, Myoinositol hexaphosphate (IP6), Denosumab and TNAP inhibitors. Concurrently investigators are also working towards reversing or curing pathological calcification. This review focuses on the relationship of vascular calcification to clinical diseases, regulators and factors causing calcification including genetics which have been identified. At present, there is lack of any significant preventive measures for calcifications and hence this review explores further possibilities for drug development and treatment modalities.
Collapse
Affiliation(s)
- Anubha Singh
- Amity Institute of Biotechnology (AIB), Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Simran Tandon
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Chanderdeep Tandon
- Amity Institute of Biotechnology (AIB), Amity University Uttar Pradesh, Noida, Uttar Pradesh, India.
| |
Collapse
|
9
|
Lim K, Molostvov G, Lubczanska M, Fletcher S, Bland R, Hiemstra TF, Zehnder D. Impaired arterial vitamin D signaling occurs in the development of vascular calcification. PLoS One 2020; 15:e0241976. [PMID: 33211721 PMCID: PMC7676703 DOI: 10.1371/journal.pone.0241976] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 10/24/2020] [Indexed: 01/10/2023] Open
Abstract
Conflicting data exists as to whether vitamin D receptor agonists (VDRa) are protective of arterial calcification. Confounding this, is the inherent physiological differences between human and animal experimental models and our current fragmented understanding of arterial vitamin D metabolism, their alterations in disease states and responses to VDRa's. Herein, the study aims to address these problems by leveraging frontiers in human arterial organ culture models. Human arteries were collected from a total of 24 patients (healthy controls, n = 12; end-stage CKD, n = 12). Cross-sectional and interventional studies were performed using arterial organ cultures treated with normal and calcifying (containing 5mmol/L CaCl2 and 5mmol/L β-glycerophosphate) medium, ex vivo. To assess the role of VDRa therapy, arteries were treated with either calcitriol or paricalcitol. We found that human arteries express a functionally active vitamin D system, including the VDR, 1α-hydroxylase and 24-hydroxylase (24-OHase) components and these were dysregulated in CKD arteries. VDRa therapy increased VDR expression in healthy arteries (p<0.01) but not in CKD arteries. Arterial 1α-OHase (p<0.05) and 24-OHase mRNA and protein expression were modulated differentially in healthy and CKD arteries by VDRa therapy. VDRa exposure suppressed Runx2 and MMP-9 expression in CKD arteries, however only paricalcitol suppressed MMP-2. VDRa exposure did not modulate arterial calcification in all organ culture models. However, VDRa reduced expression of senescence associated β-galactosidase (SAβG) staining in human aortic-smooth muscle cells under calcifying conditions, in vitro. In conclusion, maladaptation of arterial vitamin D signaling components occurs in CKD. VDRa exposure can exert vasculo-protective effects and seems critical for the regulation of arterial health in CKD.
Collapse
Affiliation(s)
- Kenneth Lim
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Guerman Molostvov
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Maria Lubczanska
- Divisions of Biomedical Sciences, University of Warwick, Coventry, United Kingdom
| | - Simon Fletcher
- Department of Nephrology, University Hospital Coventry and Warwickshire NHS Trust, Coventry, United Kingdom
| | - Rosemary Bland
- Divisions of Biomedical Sciences, University of Warwick, Coventry, United Kingdom
| | - Thomas F. Hiemstra
- Cambridge Clinical Trials Unit and School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Daniel Zehnder
- Department of Nephrology and Department of Acute Medicine, North Cumbria Integrated Care University Hospital NHS Trust, Carlisle, United Kingdom
| |
Collapse
|
10
|
Bushinsky DA, Chertow GM, Cheng S, Deng H, Kopyt N, Martin KJ, Rastogi A, Ureña-Torres P, Vervloet M, Block GA. One-year safety and efficacy of intravenous etelcalcetide in patients on hemodialysis with secondary hyperparathyroidism. Nephrol Dial Transplant 2020; 35:1769-1778. [PMID: 30859218 PMCID: PMC7538239 DOI: 10.1093/ndt/gfz039] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 01/21/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Secondary hyperparathyroidism (sHPT), a common complication of chronic kidney disease, is characterized by elevated serum parathyroid hormone (PTH). Etelcalcetide is an intravenous calcimimetic that increases sensitivity of the calcium-sensing receptor to calcium and decreases PTH secretion. This open-label extension (OLE) trial evaluated the long-term effects of etelcalcetide for sHPT treatment in patients receiving hemodialysis. METHODS This 52-week, multicenter, single-arm OLE enrolled patients from three parent trials: two randomized, double-blind, placebo-controlled trials and one open-label, single-arm, 'switch' study from cinacalcet to etelcalcetide. The primary endpoint was to investigate the nature, frequency, severity and relation to treatment of all adverse events (AEs) reported throughout the trial. Secondary endpoints included the proportion of patients with >30% reduction from baseline in PTH and the percentage change from baseline in PTH, albumin-corrected calcium (Ca), phosphate (P) and the calcium-phosphate product (Ca × P).ClinicalTrials.gov identifier: NCT01785875; Amgen study: 20120231. RESULTS Overall, 89.8% of the patients experienced one or more treatment-emergent AE. The most common were decreased blood Ca (43.3%), diarrhea (10.8%), vomiting (10.4%) and nausea (9.6%); symptomatic hypocalcemia occurred in 3.7% of the patients. Approximately 68% of patients achieved >30% reduction in PTH, and ∼56% achieved PTH ≤300 pg/mL. Mean percent changes from baseline ranged from -25.4% to -26.1% for PTH, -8.3% to -9.1% for Ca, -3.6% to -4.1% for P and -12.0% to -12.6% for Ca × P. CONCLUSIONS Etelcalcetide effectively lowered PTH and its effect was sustained, while no new safety concerns emerged over a 1-year treatment period.
Collapse
Affiliation(s)
- David A Bushinsky
- Department of Medicine, Division of Nephrology, University of Rochester School of Medicine, Rochester, NY, USA
| | - Glenn M Chertow
- Department of Medicine, Division of Nephrology, Stanford University, Stanford, CA, USA
| | - Sunfa Cheng
- Clinical Development, Amgen Inc., Thousand Oaks, CA, USA
| | - Hongjie Deng
- Biostatistics, Amgen Inc., Thousand Oaks, CA, USA
| | - Nelson Kopyt
- Department of Medicine, Division of Nephrology, Lehigh Valley Hospital, Allentown, PA, USA
| | - Kevin J Martin
- Department of Internal Medicine, Division of Nephrology, Saint Louis University School of Medicine, St Louis, MO, USA
| | - Anjay Rastogi
- Department of Medicine, Division of Nephrology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Pablo Ureña-Torres
- Department of Nephrology and Dialysis, Ramsay-Générale de Santé, Clinique du Landy, Saint Ouen, France and Necker Hospital, University of Paris Descartes, Paris, France
| | - Marc Vervloet
- Department of Nephrology and Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | | |
Collapse
|
11
|
Lim K, McGregor G, Coggan AR, Lewis GD, Moe SM. Cardiovascular Functional Changes in Chronic Kidney Disease: Integrative Physiology, Pathophysiology and Applications of Cardiopulmonary Exercise Testing. Front Physiol 2020; 11:572355. [PMID: 33041870 PMCID: PMC7522507 DOI: 10.3389/fphys.2020.572355] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 08/21/2020] [Indexed: 12/24/2022] Open
Abstract
The development of cardiovascular disease during renal impairment involves striking multi-tiered, multi-dimensional complex alterations encompassing the entire oxygen transport system. Complex interactions between target organ systems involving alterations of the heart, vascular, musculoskeletal and respiratory systems occur in Chronic Kidney Disease (CKD) and collectively contribute to impairment of cardiovascular function. These systemic changes have challenged our diagnostic and therapeutic efforts, particularly given that imaging cardiac structure at rest, rather than ascertainment under the stress of exercise, may not accurately reflect the risk of premature death in CKD. The multi-systemic nature of cardiovascular disease in CKD patients provides strong rationale for an integrated approach to the assessment of cardiovascular alterations in this population. State-of-the-art cardiopulmonary exercise testing (CPET) is a powerful, dynamic technology that enables the global assessment of cardiovascular functional alterations and reflects the integrative exercise response and complex machinery that form the oxygen transport system. CPET provides a wealth of data from a single assessment with mechanistic, physiological and prognostic utility. It is an underutilized technology in the care of patients with kidney disease with the potential to help advance the field of cardio-nephrology. This article reviews the integrative physiology and pathophysiology of cardio-renal impairment, critical new insights derived from CPET technology, and contemporary evidence for potential applications of CPET technology in patients with kidney disease.
Collapse
Affiliation(s)
- Kenneth Lim
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Gordon McGregor
- Coventry University Hospital, Coventry and Warwickshire NHS Trust, Coventry, United Kingdom.,Warwick Clinical Trials Unit, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Andrew R Coggan
- Department of Kinesiology, Indiana University - Purdue University, Indianapolis, IN, United States
| | - Gregory D Lewis
- Division of Cardiology, The Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Sharon M Moe
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
12
|
Lim K, Halim A, Lu TS, Ashworth A, Chong I. Klotho: A Major Shareholder in Vascular Aging Enterprises. Int J Mol Sci 2019; 20:E4637. [PMID: 31546756 PMCID: PMC6770519 DOI: 10.3390/ijms20184637] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/11/2019] [Accepted: 09/13/2019] [Indexed: 01/08/2023] Open
Abstract
Accelerated vascular aging is a condition that occurs as a complication of several highly prevalent inflammatory conditions such as chronic kidney disease, cancer, HIV infection and diabetes. Age-associated vascular alterations underlie a continuum of expression toward clinically overt cardiovascular disease. This has contributed to the striking epidemiologic transition whereby such noncommunicable diseases have taken center stage as modern-day global epidemics and public health problems. The identification of α-Klotho, a remarkable protein that confers powerful anti-aging properties has stimulated significant interest. In fact, emerging data have provided fundamental rationale for Klotho-based therapeutic intervention for vascular diseases and multiple other potential indications. However, the application of such discoveries in Klotho research remains fragmented due to significant gaps in our molecular understanding of Klotho biology, as well as hurdles in clinical research and experimental barriers that must first be overcome. These advances will be critical to establish the scientific platform from which future Klotho-based interventional trials and therapeutic enterprises can be successfully launched.
Collapse
Affiliation(s)
- Kenneth Lim
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- MGH Renal Associates, 165 Cambridge Street, Suite 302, Boston, MA 02114, USA
| | - Arvin Halim
- Renal Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02114, USA; (A.H.)
| | - Tzong-shi Lu
- Renal Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02114, USA; (A.H.)
| | - Alan Ashworth
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco (UCSF), 1450 3rd St, San Francisco, CA 94158, USA;
| | - Irene Chong
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Chester Beatty Laboratories, 237 Fulham Road, London SW3 6JB, UK;
| |
Collapse
|
13
|
Hiemstra T, Lim K, Thadhani R, Manson JE. Vitamin D and Atherosclerotic Cardiovascular Disease. J Clin Endocrinol Metab 2019; 104:4033-4050. [PMID: 30946457 PMCID: PMC7112191 DOI: 10.1210/jc.2019-00194] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 03/29/2019] [Indexed: 02/07/2023]
Abstract
CONTEXT A large body of experimental and observational data has implicated vitamin D deficiency in the development of cardiovascular disease. However, evidence to support routine vitamin D supplementation to prevent or treat cardiovascular disease is lacking. DESIGN AND RESULTS A comprehensive literature review was performed using PubMed and other literature search engines. Mounting epidemiological evidence and data from Mendelian randomization studies support a link between vitamin D deficiency and adverse cardiovascular health outcomes, but randomized trial evidence to support vitamin D supplementation is sparse. Current public health guidelines restrict vitamin D intake recommendations to the maintenance of bone health and prevention of fractures. Two recently published large trials (VITAL and ViDA) that assessed the role of moderate- to high-dose vitamin D supplementation as primary prevention for cardiovascular outcomes in the general population had null results, and previous randomized trials have also been generally negative. These findings from general population cohorts that are largely replete in vitamin D may not be applicable to chronic kidney disease (CKD) populations, in which the use of active (1α-hydroxylated) vitamin D compounds is prevalent, or to other high-risk populations. Additionally, recent trials in the CKD population, as well as trials using vitamin D analogs, have been limited. CONCLUSIONS Current randomized trials of vitamin D supplementation do not support benefits for cardiovascular health, but the evidence remains inconclusive. Additional randomized trials assessing larger numbers of participants with low baseline vitamin D levels, having longer follow-up periods, and testing higher vitamin D dosages are needed to guide clinical practice.
Collapse
Affiliation(s)
- Thomas Hiemstra
- Cambridge Clinical Trials Unit, Addenbrookes Hospital, Cambridge, UK
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Kenneth Lim
- Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Ravi Thadhani
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - JoAnn E. Manson
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School and Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
14
|
Tangvoraphonkchai K, Davenport A. Aortic pulse wave velocity is greater in peritoneal dialysis patients with lower dual energy X-ray absorptiometry (DXA) femoral neck bone mineral density. J Nephrol 2018; 32:471-476. [PMID: 30535631 DOI: 10.1007/s40620-018-0551-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 11/03/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND Increased vascular stiffness is associated with low bone mineral density (BMD) in the general population, and both are risk factors for mortality. We wished to determine whether vascular stiffness is associated with BMD in peritoneal dialysis (PD) patients. METHODS We measured vascular stiffness by aortic pulse wave velocity (aPWV), BMD by dual electron absorptiometry (DXA) scanning, and body composition using bioimpedance. RESULTS We reviewed DXA scans in 125 PD patients, 56.8% male, mean age 64.4 ± 15.3 years, mean aPWV, 10.2 ± 2.6 m/s. We divided patients by aPWV (< 10 and > 10 m/s), and there were no statistical differences in patient demographics, body composition, PD adequacy, peritoneal and urinary calcium losses. On univariate analysis aPWV was negatively associated with total body T score (r = - 0.20, p = 0.037). On multivariable logistic regression patients with higher aPWV were prescribed fewer non-calcium containing phosphate binders, odds ratio (OR) 0.83, 95% confidence interval (CI) 0.70-0.99, p = 0.039, more had lower 25 hydroxy-vitamin D3 concentrations < 50 ng/L (OR 0.34, CI 0.12-0.93, p = 0.035, and lower femoral BMD OR 0.03 (CI 0-0.3.4), p = 0.029, but there was no association with total or lumbar spine BMD. CONCLUSION Our study reinforces the hypothesis of a link between bone disease and vascular disease in dialysis patients. As patients with higher aPWV were prescribed fewer non-calcium containing phosphate binders and fewer had higher 25 hydroxy-vitamin D3 concentrations, then this raises the possibility that differences in clinical practice and drug prescribing may help to reduce vascular stiffness, which will require testing in future trials.
Collapse
Affiliation(s)
| | - Andrew Davenport
- UCL Department of Nephrology, Royal Free hospital, University College London, Rowland Hill Street, London, NW3 2PF, UK
| |
Collapse
|