101
|
Grabner A, Schramm K, Silswal N, Hendrix M, Yanucil C, Czaya B, Singh S, Wolf M, Hermann S, Stypmann J, Di Marco GS, Brand M, Wacker MJ, Faul C. FGF23/FGFR4-mediated left ventricular hypertrophy is reversible. Sci Rep 2017; 7:1993. [PMID: 28512310 PMCID: PMC5434018 DOI: 10.1038/s41598-017-02068-6] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/12/2017] [Indexed: 12/14/2022] Open
Abstract
Fibroblast growth factor (FGF) 23 is a phosphaturic hormone that directly targets cardiac myocytes via FGF receptor (FGFR) 4 thereby inducing hypertrophic myocyte growth and the development of left ventricular hypertrophy (LVH) in rodents. Serum FGF23 levels are highly elevated in patients with chronic kidney disease (CKD), and it is likely that FGF23 directly contributes to the high rates of LVH and cardiac death in CKD. It is currently unknown if the cardiac effects of FGF23 are solely pathological, or if they potentially can be reversed. Here, we report that FGF23-induced cardiac hypertrophy is reversible in vitro and in vivo upon removal of the hypertrophic stimulus. Specific blockade of FGFR4 attenuates established LVH in the 5/6 nephrectomy rat model of CKD. Since CKD mimics a form of accelerated cardiovascular aging, we also studied age-related cardiac remodeling. We show that aging mice lacking FGFR4 are protected from LVH. Finally, FGF23 increases cardiac contractility via FGFR4, while known effects of FGF23 on aortic relaxation do not require FGFR4. Taken together, our data highlight a role of FGF23/FGFR4 signaling in the regulation of cardiac remodeling and function, and indicate that pharmacological interference with cardiac FGF23/FGFR4 signaling might protect from CKD- and age-related LVH.
Collapse
MESH Headings
- Animals
- Biopsy
- Diet
- Disease Models, Animal
- Fibroblast Growth Factor-23
- Fibroblast Growth Factors/blood
- Fibroblast Growth Factors/metabolism
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Mice
- Mice, Knockout
- Myocardial Contraction/genetics
- Myocytes, Cardiac/metabolism
- Rats
- Receptor, Fibroblast Growth Factor, Type 4/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 4/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- 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
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Duke University, Durham, North Carolina, USA
| | - Karla Schramm
- 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 Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Neerupma Silswal
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Matt Hendrix
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, 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
- Department of Cell Biology and Anatomy, 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
| | - 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
- Department of Cell Biology and Anatomy, 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
| | - 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
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Myles Wolf
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Duke University, Durham, North Carolina, USA
| | - Sven Hermann
- European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Jörg Stypmann
- Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany
| | | | - Marcus Brand
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Michael J Wacker
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - 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.
- Department of Cell Biology and Anatomy, 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.
| |
Collapse
|
102
|
Afsar B, Rossignol P, van Heerebeek L, Paulus WJ, Damman K, Heymans S, van Empel V, Sag A, Maisel A, Kanbay M. Heart failure with preserved ejection fraction: a nephrologist-directed primer. Heart Fail Rev 2017; 22:765-773. [DOI: 10.1007/s10741-017-9619-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
103
|
α-Klotho expression determines nitric oxide synthesis in response to FGF-23 in human aortic endothelial cells. PLoS One 2017; 12:e0176817. [PMID: 28463984 PMCID: PMC5413063 DOI: 10.1371/journal.pone.0176817] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 04/18/2017] [Indexed: 01/03/2023] Open
Abstract
Endothelial cells (ECs) express fibroblast growth factor (FGF) receptors and are metabolically active after treatment with FGF-23. It is not known if this effect is α-Klotho independent or mediated by humoral or endogenous endothelial α-Klotho. In the present study, we aimed to characterize EC α-Klotho expression within the human vascular tree and to investigate the potential role of α-Klotho in determining FGF-23 mediated EC regulation. Human tissue and ECs from various organs were used for immunohistochemistry and Western blot. Primary cultures of human aortic endothelial cells (HAECs) and human brain microvascular endothelial cells (HBMECs) were used to generate in vitro cell models. We found endogenous α-Klotho expression in ECs from various organs except in microvascular ECs from human brain. Furthermore, FGF-23 stimulated endothelial nitric oxide synthase (eNOS) expression, nitric oxide (NO) production, and cell proliferation in HAECs. Interestingly, these effects were not observed in our HBMEC model in vitro. High phosphate treatment and endothelial α-Klotho knockdown mitigated FGF-23 mediated eNOS induction, NO production, and cell proliferation in HAECs. Rescue treatment with soluble α-Klotho did not reverse endothelial FGF-23 resistance caused by reduced or absent α-Klotho expression in HAECs. These novel observations provide evidence for differential α-Klotho functional expression in the human endothelium and its presence may play a role in determining the response to FGF-23 in the vascular tree. α-Klotho was not detected in cerebral microvascular ECs and its absence may render these cells nonresponsive to FGF-23.
Collapse
|
104
|
Abstract
Chronic kidney disease (CKD) is associated with an increased risk of cardiovascular mortality, infections, and impaired cognitive function. It is characterized by excessively increased levels of the phosphaturic hormone fibroblast growth factor 23 (FGF23) and a deficiency of its co-receptor Klotho. Despite the important physiological effect of FGF23 in maintaining phosphate homeostasis, there is increasing evidence that higher FGF23 levels are a risk factor for mortality and cardiovascular disease. FGF23 directly induces left ventricular hypertrophy via activation of the FGF receptor 4/calcineurin/nuclear factor of activated T cells signaling pathway. By contrast, the impact of FGF23 on endothelial function and the development of atherosclerosis are poorly understood. The results of recent experimental studies indicate that FGF23 directly impacts on hippocampal neurons and may thereby impair learning and memory function in CKD patients. Finally, it has been shown that FGF23 interferes with the immune system by directly acting on polymorphonuclear leukocytes and macrophages. In this review, we discuss recent data from clinical and experimental studies on the extrarenal effects of FGF23 with respect to the cardiovascular, central nervous, and immune systems.
Collapse
|
105
|
Mencke R, Hillebrands JL. The role of the anti-ageing protein Klotho in vascular physiology and pathophysiology. Ageing Res Rev 2017; 35:124-146. [PMID: 27693241 DOI: 10.1016/j.arr.2016.09.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/23/2016] [Indexed: 02/08/2023]
Abstract
Klotho is an anti-ageing protein that functions in many pathways that govern ageing, like regulation of phosphate homeostasis, insulin signaling, and Wnt signaling. Klotho expression levels and levels in blood decline during ageing. The vascular phenotype of Klotho deficiency features medial calcification, intima hyperplasia, endothelial dysfunction, arterial stiffening, hypertension, and impaired angiogenesis and vasculogenesis, with characteristics similar to aged human arteries. Klotho-deficient phenotypes can be prevented and rescued by Klotho gene expression or protein supplementation. High phosphate levels are likely to be directly pathogenic and are a prerequisite for medial calcification, but more important determinants are pathways that regulate cellular senescence, suggesting that deficiency of Klotho renders cells susceptible to phosphate toxicity. Overexpression of Klotho is shown to ameliorate medial calcification, endothelial dysfunction, and hypertension. Endogenous vascular Klotho expression is a controversial subject and, currently, no compelling evidence exists that supports the existence of vascular membrane-bound Klotho expression, as expressed in kidney. In vitro, Klotho has been shown to decrease oxidative stress and apoptosis in both SMCs and ECs, to reduce SMC calcification, to maintain the contractile SMC phenotype, and to prevent μ-calpain overactivation in ECs. Klotho has many protective effects with regard to the vasculature and constitutes a very promising therapeutic target. The purpose of this review is to explore the etiology of the vascular phenotype of Klotho deficiency and the therapeutic potential of Klotho in vascular disease.
Collapse
|
106
|
Zoccali C, Vanholder R, Massy ZA, Ortiz A, Sarafidis P, Dekker FW, Fliser D, Fouque D, Heine GH, Jager KJ, Kanbay M, Mallamaci F, Parati G, Rossignol P, Wiecek A, London G. The systemic nature of CKD. Nat Rev Nephrol 2017; 13:344-358. [PMID: 28435157 DOI: 10.1038/nrneph.2017.52] [Citation(s) in RCA: 282] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The accurate definition and staging of chronic kidney disease (CKD) is one of the major achievements of modern nephrology. Intensive research is now being undertaken to unravel the risk factors and pathophysiologic underpinnings of this disease. In particular, the relationships between the kidney and other organs have been comprehensively investigated in experimental and clinical studies in the last two decades. Owing to technological and analytical limitations, these links have been studied with a reductionist approach focusing on two organs at a time, such as the heart and the kidney or the bone and the kidney. Here, we discuss studies that highlight the complex and systemic nature of CKD. Energy balance, innate immunity and neuroendocrine signalling are highly integrated biological phenomena. The diseased kidney disrupts such integration and generates a high-risk phenotype with a clinical profile encompassing inflammation, protein-energy wasting, altered function of the autonomic and central nervous systems and cardiopulmonary, vascular and bone diseases. A systems biology approach to CKD using omics techniques will hopefully enable in-depth study of the pathophysiology of this systemic disease, and has the potential to unravel critical pathways that can be targeted for CKD prevention and therapy.
Collapse
Affiliation(s)
- Carmine Zoccali
- CNR-IFC Clinical Epidemiology and Pathophysiology of Renal Diseases and Hypertension Unit, Ospedali Riuniti 89124 Reggio Calabria, Italy
| | - Raymond Vanholder
- Ghent University Hospital, Department of Nephrology, Department of Internal Medicine, University Hospital Gent, De Pintelaan 185, B9000 Ghent, Belgium
| | - Ziad A Massy
- Division of Nephrology, Ambroise Paré Hospital, Assistance Publique Hôpitaux de Paris, 9 Avenue Charles de Gaulle, 92100 Boulogne-Billancourt, Paris.,University of Paris Ouest-Versailles-Saint-Quentin-en-Yvelines (UVSQ), 55 Avenue de Paris, 78000 Versailles, France.,Inserm U-1018, Centre de recherche en épidémiologie et santé des populations (CESP), Equipe 5, Hôpital Paul-Brousse, 16 avenue Paul Vaillant-Couturier, 94807 Villejuif Cedex, France.,Paris-Sud University (PSU), 15 Rue Georges Clemenceau, 91400 Orsay, France.,French-Clinical Research Infrastructure Network (F-CRIN), Pavillon Leriche 2è étage CHU de Toulouse, Place Dr Baylac TSA40031, 31059 TOULOUSE Cedex 3, France
| | - Alberto Ortiz
- Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Fundación Renal Iñigo Alvarez de Toledo, Madrid, Av. Reyes Católicos, 2, 28040 Madrid, Spain
| | - Pantelis Sarafidis
- Department of Nephrology, Hippokration Hospital, Thessaloniki, Konstantinoupoleos 49, Thessaloniki 546 42, Greece
| | - Friedo W Dekker
- Department of Clinical Epidemiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Danilo Fliser
- Department Internal Medicine IV-Renal and Hypertensive Disease-Saarland University Medical Centre Kirrberger Straß 66421 Homburg, Saar, Germany
| | - Denis Fouque
- Université de Lyon, UCBL, Carmen, Department of Nephrology, Centre Hospitalier Lyon-Sud, F-69495 Pierre Bénite, France
| | - Gunnar H Heine
- Department Internal Medicine IV-Renal and Hypertensive Disease-Saarland University Medical Centre Kirrberger Straß 66421 Homburg, Saar, Germany
| | - Kitty J Jager
- European Renal Association-European Dialysis and Transplant Association (ERA-EDTA) Registry, Department of Medical Informatics, Meibergdreef 9, 1105 AZ Amsterdam-Zuidoost, The Netherlands
| | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine,Koç University, Rumelifeneri Yolu 34450 Sarıyer Istanbul, Turkey
| | - Francesca Mallamaci
- CNR-IFC Clinical Epidemiology and Pathophysiology of Renal Diseases and Hypertension Unit, Ospedali Riuniti 89124 Reggio Calabria, Italy.,Nephrology, Dialysis and Transplantation Unit Ospedali Riuniti, 89124 Reggio Calabria Italy
| | - Gianfranco Parati
- Department of Cardiovascular, Neural and Metabolic Sciences, S. Luca Hospital, Istituto Auxologico Italiano &Department of Medicine and Surgery, University of Milan-Bicocca, Piazzale Brescia 20, Milan 20149, Italy
| | - Patrick Rossignol
- French-Clinical Research Infrastructure Network (F-CRIN), Pavillon Leriche 2è étage CHU de Toulouse, Place Dr Baylac TSA40031, 31059 TOULOUSE Cedex 3, France.,Inserm, Centre d'Investigations Cliniques-Plurithématique 1433, Cardiovascular and Renal Clinical Trialists (INI-CRCT), Institut Lorrain du Cœur et des Vaisseaux Louis Mathieu, 4 rue Morvan, 54500 Vandoeuvre-les-Nancy, France.,Inserm U1116, Faculté de Médecine, Bâtiment D 1er étage, 9 avenue de la forêt de Haye - BP 184, 54500 Vandœuvre-lès-Nancy Cedex, France.,CHU Nancy, Département de Cardiologie, Institut Lorrain du Cœur et des Vaisseaux, 5 Rue du Morvan, 54500 Vandœuvre-lès-Nancy, France.,Université de Lorraine, 34 Cours Léopold, 54000 Nancy, France
| | - Andrzej Wiecek
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, Francuska 20/24 Street, Pl-40-027 Katowice, Poland
| | - Gerard London
- INSERM U970, Hopital Européen Georges Pompidou, 20 Rue Leblanc, 75015 Paris, France
| | | |
Collapse
|
107
|
Kunasegaran T, Mustafa MR, Achike FI, Murugan DD. Quercetin and pioglitazone synergistically reverse endothelial dysfunction in isolated aorta from fructose-streptozotocin (F-STZ)-induced diabetic rats. Eur J Pharmacol 2017; 799:160-170. [DOI: 10.1016/j.ejphar.2017.02.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 02/11/2017] [Accepted: 02/13/2017] [Indexed: 12/15/2022]
|
108
|
Sharaf El Din UA, Salem MM, Abdulazim DO. Is Fibroblast growth factor 23 the leading cause of increased mortality among chronic kidney disease patients? A narrative review. J Adv Res 2017; 8:271-278. [PMID: 28337344 PMCID: PMC5347517 DOI: 10.1016/j.jare.2017.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/20/2017] [Accepted: 02/20/2017] [Indexed: 12/16/2022] Open
Abstract
The death rate among chronic kidney disease patients is the highest compared to other chronic diseases. 60% of these fatalities are cardiovascular. Cardiovascular calcifications and chronic inflammation affect almost all chronic kidney disease patients and are associated with cardiovascular mortality. Fibroblast growth factor 23 is associated with vascular calcification. Systemic inflammation in chronic kidney disease patients is multifactorial. The role of systemic inflammation in the pathogenesis of vascular calcification was recently reappraised. Fibroblast growth factor 23 was accused as a direct stimulus of left ventricular hypertrophy, uremic inflammation, and impaired neutrophil function. This review will discuss the underlying mechanisms that underlie the link between Fibroblast growth factor 23 and increased mortality encountered among chronic kidney disease patients.
Collapse
Affiliation(s)
- Usama A.A. Sharaf El Din
- Nephrology Unit, Internal Medicine Department, School of Medicine, Cairo University, Egypt
- Corresponding author.
| | - Mona M. Salem
- Endocrinology Unit, Internal Medicine Department, School of Medicine, Cairo University, Egypt
| | - Dina O. Abdulazim
- Rheumatology and Rehabilitation Department, School of Medicine, Cairo University, Egypt
| |
Collapse
|
109
|
Hu X, Ma X, Luo Y, Xu Y, Xiong Q, Pan X, Bao Y, Jia W. Contribution of fibroblast growth factor 23 to Framingham risk score for identifying subclinical atherosclerosis in Chinese men. Nutr Metab Cardiovasc Dis 2017; 27:147-153. [PMID: 28017526 DOI: 10.1016/j.numecd.2016.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/11/2016] [Accepted: 11/14/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND AIMS Fibroblast growth factor 23 (FGF23) was demonstrated to be involved in the occurrence and development of cardiovascular disease (CVD). The goal of the present study was to investigate the relationship between serum FGF23 levels and carotid intima-media thickness (C-IMT) in men with a low-to-moderate CVD risk. METHODS AND RESULTS Subjects with normal kidney function were selected from the Shanghai Obesity Study. Serum FGF23 levels were determined by sandwich enzyme-linked immunosorbent assay. C-IMT was measured by ultrasonography. The Framingham risk score (FRS) was used to assess CVD risk. A total of 392 men with low CVD risk and 372 men with moderate CVD risk were enrolled. The recognition rate of an elevated C-IMT was 85.66% with the combination of a moderate CVD risk and high serum FGF23 levels, which was greater than that with either parameter alone (65.44% and 61.03%, respectively). Subjects with high serum FGF23 levels, and either low or moderate CVD risk, were more likely to have elevated C-IMT than those with low serum FGF23 levels and low CVD risk (P = 0.014 and 0.001, respectively). The serum FGF23 levels were independently and positively associated with C-IMT in subjects with low or moderate CVD risk (both P = 0.007). CONCLUSION In men with low-to-moderate CVD risk, serum FGF23 levels were associated independently and positively with C-IMT. As a complementary index, serum FGF23 levels strengthen the capacity of the FRS to identify subclinical atherosclerosis.
Collapse
Affiliation(s)
- X Hu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China; Shanghai Clinical Center for Diabetes, Shanghai 200233, China; Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200233, China; Shanghai Diabetes Institute, Shanghai 200233, China; Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China
| | - X Ma
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China; Shanghai Clinical Center for Diabetes, Shanghai 200233, China; Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200233, China; Shanghai Diabetes Institute, Shanghai 200233, China; Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China
| | - Y Luo
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China; Shanghai Clinical Center for Diabetes, Shanghai 200233, China; Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200233, China; Shanghai Diabetes Institute, Shanghai 200233, China; Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China
| | - Y Xu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China; Shanghai Clinical Center for Diabetes, Shanghai 200233, China; Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200233, China; Shanghai Diabetes Institute, Shanghai 200233, China; Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China
| | - Q Xiong
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China; Shanghai Clinical Center for Diabetes, Shanghai 200233, China; Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200233, China; Shanghai Diabetes Institute, Shanghai 200233, China; Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China
| | - X Pan
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China; Shanghai Clinical Center for Diabetes, Shanghai 200233, China; Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200233, China; Shanghai Diabetes Institute, Shanghai 200233, China; Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China
| | - Y Bao
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China; Shanghai Clinical Center for Diabetes, Shanghai 200233, China; Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200233, China; Shanghai Diabetes Institute, Shanghai 200233, China; Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China.
| | - W Jia
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China; Shanghai Clinical Center for Diabetes, Shanghai 200233, China; Shanghai Key Clinical Center for Metabolic Disease, Shanghai 200233, China; Shanghai Diabetes Institute, Shanghai 200233, China; Shanghai Key Laboratory of Diabetes Mellitus, Shanghai 200233, China
| |
Collapse
|
110
|
Yamada S, Tsuruya K, Tokumoto M, Yoshida H, Tatsumoto N, Ooboshi H, Kitazono T. Total Phosphate Elimination is Negatively Associated With Increased Serum Fibroblast Growth Factor 23 Levels in Patients who Undergo Peritoneal Dialysis. Ther Apher Dial 2017; 21:71-78. [PMID: 28078749 DOI: 10.1111/1744-9987.12478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 06/05/2016] [Accepted: 06/30/2016] [Indexed: 12/15/2022]
Abstract
As fibroblast growth factor 23 (FGF23) has been shown to induce cardiovascular disease directly in patients with chronic kidney disease, identification of factors and treatments that can modulate serum FGF23 (sFGF23) level is clinically important. This retrospective longitudinal study investigated factors that modulate sFGF23 in 49 patients who underwent peritoneal dialysis (PD). sFGF23 ratio (sFGF23 at 18 months/baseline sFGF23) was used as an indicator of changes in sFGF23 level. Total phosphate elimination was the sum of both renal phosphate excretion and dialysate phosphate elimination. In multivariate analysis, log sFGF23 ratio was associated negatively with total phosphate elimination and the use of cinacalcet at baseline, and positively with the use of vitamin D receptor activators at baseline, even after adjusting for potential confounding factors. Our study indicates that maintaining phosphate elimination can prevent increased sFGF23, thereby preventing cardiovascular events in patients who undergo PD.
Collapse
Affiliation(s)
- Shunsuke Yamada
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, Japan.,Department of Internal Medicine, Fukuoka Dental College, Sawara-ku, Fukuoka, Japan
| | - Kazuhiko Tsuruya
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, Japan.,Department of Integrated Therapy for Chronic Kidney Disease, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, Japan
| | - Masanori Tokumoto
- Department of Internal Medicine, Fukuoka Dental College, Sawara-ku, Fukuoka, Japan
| | - Hisako Yoshida
- Department of Integrated Therapy for Chronic Kidney Disease, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, Japan
| | - Narihito Tatsumoto
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, Japan
| | - Hiroaki Ooboshi
- Department of Internal Medicine, Fukuoka Dental College, Sawara-ku, Fukuoka, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, Japan
| |
Collapse
|
111
|
Nishi T, Shuto E, Ogawa M, Ohya M, Nakanishi M, Masuda M, Katsumoto M, Yamanaka-Okumura H, Sakai T, Takeda E, Sakaue H, Taketani Y. Excessive dietary phosphorus intake impairs endothelial function in young healthy men: a time- and dose-dependent study. THE JOURNAL OF MEDICAL INVESTIGATION 2017; 62:167-72. [PMID: 26399342 DOI: 10.2152/jmi.62.167] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Excessive dietary phosphorus (P) has been speculated to be a risk factor for cardiovascular disease (CVD). Here, we performed a double-blinded crossover study to investigate the time- and dose-dependent effects of dietary P intake on endothelial function in healthy subjects. Sixteen healthy male volunteers were given meals containing 400, 800, and 1,200 mg P (P400, P800, and P1200 meals, respectively) with at least 7 days between doses. There were no differences in nutritional composition among the experimental diets except for P content. Blood biochemistry data and flow-mediated dilation (%FMD) of the brachial artery were measured while fasted, at 0 h, 1 h, 2 h, and 4 h after meal ingestion, and the next morning while fasted. The P800 and P1200 meals significantly increased serum P levels at 1-4 h after ingestion. A significant decrease in %FMD was observed between 1-4 h,while the P400 meal did not affect %FMD. We observed no differences among meals in serum P levels or %FMD the next morning. A significant negative correlation was observed between %FMD and serum P. These results indicate that excessive dietary P intake can acutely impair endothelial function in healthy people.
Collapse
Affiliation(s)
- Tamae Nishi
- Department of Clinical Nutrition and Food Management, University of Tokushima Graduate School
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Vanhoutte PM, Shimokawa H, Feletou M, Tang EHC. Endothelial dysfunction and vascular disease - a 30th anniversary update. Acta Physiol (Oxf) 2017; 219:22-96. [PMID: 26706498 DOI: 10.1111/apha.12646] [Citation(s) in RCA: 629] [Impact Index Per Article: 78.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/27/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023]
Abstract
The endothelium can evoke relaxations of the underlying vascular smooth muscle, by releasing vasodilator substances. The best-characterized endothelium-derived relaxing factor (EDRF) is nitric oxide (NO) which activates soluble guanylyl cyclase in the vascular smooth muscle cells, with the production of cyclic guanosine monophosphate (cGMP) initiating relaxation. The endothelial cells also evoke hyperpolarization of the cell membrane of vascular smooth muscle (endothelium-dependent hyperpolarizations, EDH-mediated responses). As regards the latter, hydrogen peroxide (H2 O2 ) now appears to play a dominant role. Endothelium-dependent relaxations involve both pertussis toxin-sensitive Gi (e.g. responses to α2 -adrenergic agonists, serotonin, and thrombin) and pertussis toxin-insensitive Gq (e.g. adenosine diphosphate and bradykinin) coupling proteins. New stimulators (e.g. insulin, adiponectin) of the release of EDRFs have emerged. In recent years, evidence has also accumulated, confirming that the release of NO by the endothelial cell can chronically be upregulated (e.g. by oestrogens, exercise and dietary factors) and downregulated (e.g. oxidative stress, smoking, pollution and oxidized low-density lipoproteins) and that it is reduced with ageing and in the course of vascular disease (e.g. diabetes and hypertension). Arteries covered with regenerated endothelium (e.g. following angioplasty) selectively lose the pertussis toxin-sensitive pathway for NO release which favours vasospasm, thrombosis, penetration of macrophages, cellular growth and the inflammatory reaction leading to atherosclerosis. In addition to the release of NO (and EDH, in particular those due to H2 O2 ), endothelial cells also can evoke contraction of the underlying vascular smooth muscle cells by releasing endothelium-derived contracting factors. Recent evidence confirms that most endothelium-dependent acute increases in contractile force are due to the formation of vasoconstrictor prostanoids (endoperoxides and prostacyclin) which activate TP receptors of the vascular smooth muscle cells and that prostacyclin plays a key role in such responses. Endothelium-dependent contractions are exacerbated when the production of nitric oxide is impaired (e.g. by oxidative stress, ageing, spontaneous hypertension and diabetes). They contribute to the blunting of endothelium-dependent vasodilatations in aged subjects and essential hypertensive and diabetic patients. In addition, recent data confirm that the release of endothelin-1 can contribute to endothelial dysfunction and that the peptide appears to be an important contributor to vascular dysfunction. Finally, it has become clear that nitric oxide itself, under certain conditions (e.g. hypoxia), can cause biased activation of soluble guanylyl cyclase leading to the production of cyclic inosine monophosphate (cIMP) rather than cGMP and hence causes contraction rather than relaxation of the underlying vascular smooth muscle.
Collapse
Affiliation(s)
- P. M. Vanhoutte
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| | - H. Shimokawa
- Department of Cardiovascular Medicine; Tohoku University; Sendai Japan
| | - M. Feletou
- Department of Cardiovascular Research; Institut de Recherches Servier; Suresnes France
| | - E. H. C. Tang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
- School of Biomedical Sciences; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| |
Collapse
|
113
|
Lu X, Hu MC. Klotho/FGF23 Axis in Chronic Kidney Disease and Cardiovascular Disease. KIDNEY DISEASES 2016; 3:15-23. [PMID: 28785560 DOI: 10.1159/000452880] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 10/26/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Membrane αKlotho (hereinafter called Klotho) is highly expressed in the kidney and functions as a coreceptor of FGF receptors (FGFRs) to activate specific fibroblast growth factor 23 (FGF23) signal pathway. FGF23 is produced in bones and participates in the maintenance of mineral homeostasis. The extracellular domain of transmembrane Klotho can be cleaved by secretases and released into the circulation as soluble Klotho. Soluble Klotho does not only weakly activate FGFRs to transduce the FGF23 signaling pathway, but also functions as an enzyme and hormonal substance to play a variety of biological functions. FGF23 exerts its biological effects through activation of FGFRs in a Klotho-dependent manner. However, extremely high FGF23 can exert its pathological action in a Klotho-independent manner. SUMMARY The decline in serum and urinary Klotho followed by a rise in serum FGF23 at an early stage of chronic kidney disease (CKD) functions as an early biomarker for kidney dysfunction and can also serve as a predictor for risk of cardiovascular disease (CVD) and mortality in both CKD patients and the general population. Moreover, Klotho deficiency is a pathogenic factor for CKD progression and CVD. FGF23 may also contribute to CVD. Prevention of Klotho decline, reactivation of endogenous Klotho production, or supplementation of exogenous Klotho attenuate renal fibrosis, retard CKD progression, improve mineral metabolism, ameliorate cardiomyopathy, and alleviate vascular calcification in CKD. However, the poor CVD outcome after depletion of FGF23 with FGF23 antibody stimulates the generation of a more specific inhibitor of FGF23 for CKD treatment. KEY MESSAGE Klotho/FGF23 may not only be diagnostic and/or prognostic biomarkers for CKD and CVD, but are also pathogenic contributors to CKD progression and CVD development. The Klotho/FGF23 axis should be a novel target for renal clinics.
Collapse
Affiliation(s)
- Xiang Lu
- Department of Geriatrics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, PR China
| | - Ming Chang Hu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
114
|
Elevation in fibroblast growth factor 23 and its value for identifying subclinical atherosclerosis in first-degree relatives of patients with diabetes. Sci Rep 2016; 6:34696. [PMID: 27698482 PMCID: PMC5048154 DOI: 10.1038/srep34696] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 09/19/2016] [Indexed: 11/08/2022] Open
Abstract
Accumulating evidence supported an association between diabetes and fibroblast growth factor 23 (FGF23). The goal of the present study was to explore alteration in serum FGF23 levels and to assess its value for identifying subclinical atherosclerosis in normoglycemic individuals with a first-degree family history of diabetes (FHD). The study enrolled 312 subjects with a first-degree FHD and 1407 subjects without an FHD. Serum FGF23 levels were detected by a sandwich enzyme-linked immunosorbent assay. Serum FGF23 levels were much higher in subjects with a first-degree FHD than in those without an FHD (P = 0.006). A first-degree FHD was positively associated with serum FGF23 levels, independent of C-IMT and cardiovascular factors (both P < 0.05). In subjects with a first-degree FHD, only those with serum FGF23 levels in the upper quartile were more likely to have an increased C-IMT (odds ratio = 2.263, P < 0.05). As conclusions, a first-degree FHD contributes to the increased serum FGF23 levels independently. Subjects with a first-degree FHD need higher serum FGF23 levels to indicate subclinical atherosclerosis. The influence of a first-degree FHD on serum FGF23 levels should be considered to avoid overestimating the risk of cardiovascular disease in normoglycemic individuals with a first-degree FHD.
Collapse
|
115
|
Abstract
Fibroblast growth factor-23 (FGF23) is a bone-derived hormone known to suppress phosphate reabsorption and vitamin D hormone production in the kidney. Klotho was originally discovered as an anti-aging factor, but the functional role of Klotho is still a controversial issue. Three major functions have been proposed, a hormonal function of soluble Klotho, an enzymatic function as glycosidase, and the function as an obligatory co-receptor for FGF23 signaling. The purpose of this review is to highlight the recent advances in the area of FGF23 and Klotho signaling in the kidney, in the parathyroid gland, in the cardiovascular system, in bone, and in the central nervous system. During recent years, major new functions of FGF23 and Klotho have been discovered in these organ systems. Based on these novel findings, FGF23 has emerged as a pleiotropic endocrine and auto-/paracrine factor influencing not only mineral metabolism but also cardiovascular function.
Collapse
|
116
|
Yamada H, Kuro-o M, Hara K, Ueda Y, Kusaka I, Kakei M, Ishikawa SE. The Urinary Phosphate to Serum Fibroblast Growth Factor 23 Ratio Is a Useful Marker of Atherosclerosis in Early-Stage Chronic Kidney Disease. PLoS One 2016; 11:e0160782. [PMID: 27504998 PMCID: PMC4978402 DOI: 10.1371/journal.pone.0160782] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 07/25/2016] [Indexed: 01/15/2023] Open
Abstract
Background Fibroblast growth factor 23 (FGF23) regulates mineral homeostasis. In developed renal dysfunction, FGF23 levels increase to maintain the phosphate excretion capacity. However, in diabetic patients with early-stage renal impairment, the FGF23 elevation is not very sensitive. We hypothesized that urinary phosphate (U-P)/serum FGF23 ratio would theoretically be an index that reflects the number of nephrons (nephron index). In this study, we determined whether the nephron index would be associated with renal function and vascular diseases in diabetic patients. Methods In total, 142 patients with diabetes mellitus were enrolled. The nephron index was calculated using the following formula: U-P (mg/day)/ serum FGF23 (pg/ml). Results The mean age was 63 ± 11 years and eGFR levels were 79.5 ± 25.4 ml/min/1.73 m2, respectively. Thirty patients had a medical history of macroangiopathy. The Nephron index was significantly decreased in subjects with macroangiopathy compared with those without macroangiopathy. A multivariate analysis of risk factors for macroangiopathy revealed that duration of diabetes, eGFR, and nephron index were significantly associated with a higher frequency of arteriosclerotic disease. Conclusion These findings suggest that a decrease in nephron index reflects early-stage renal impairment and is an independent risk factor of macroangiopathy in diabetic patients.
Collapse
Affiliation(s)
- Hodaka Yamada
- Department of Medicine, Jichi Medical University Saitama Medical Center, 1-847 Amanuma-cho, Omiya-ku, Saitama, 330-8503, Japan
- * E-mail:
| | - Makoto Kuro-o
- Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji Shimotsuke, Tochigi, 329-0498, Japan
| | - Kazuo Hara
- Department of Medicine, Jichi Medical University Saitama Medical Center, 1-847 Amanuma-cho, Omiya-ku, Saitama, 330-8503, Japan
| | - Yuichiro Ueda
- Department of Medicine, Jichi Medical University Saitama Medical Center, 1-847 Amanuma-cho, Omiya-ku, Saitama, 330-8503, Japan
| | - Ikuyo Kusaka
- Department of Medicine, Jichi Medical University Saitama Medical Center, 1-847 Amanuma-cho, Omiya-ku, Saitama, 330-8503, Japan
| | - Masafumi Kakei
- Department of Medicine, Jichi Medical University Saitama Medical Center, 1-847 Amanuma-cho, Omiya-ku, Saitama, 330-8503, Japan
| | - San-e Ishikawa
- Division of Endocrinology and Metabolism, International University of Health and Welfare Hospital, 537-3 Iguchi, Nasushiobara, Tochigi, 329-2763, Japan
| |
Collapse
|
117
|
Klotho modulates FGF23-mediated NO synthesis and oxidative stress in human coronary artery endothelial cells. Pflugers Arch 2016; 468:1621-35. [PMID: 27448998 DOI: 10.1007/s00424-016-1858-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/28/2016] [Accepted: 07/12/2016] [Indexed: 12/27/2022]
Abstract
Chronic kidney disease (CKD) is a state of Klotho deficiency and excess of the phosphaturic hormone fibroblast growth factor 23 (FGF23). Both dysregulations were shown to be associated with endothelial dysfunction in humans, but direct vascular effects of FGF23 remain largely elusive. In vitro experiments were performed to assess the effects of FGF23 (10 ng/mL) in relation to its co-receptor Klotho on nitric oxide (NO) synthesis and reactive oxygen species (ROS) formation and detoxification in human coronary artery endothelial cells (HCAEC). Membrane-bound Klotho is expressed in HCAEC, and FGF23 increases the expression of the Klotho shedding protease ADAM17, and consequently the secretion of soluble Klotho. FGF23 activates FGF receptor 1 and stimulates NO release via Akt-dependent activation of endothelial NO synthase (eNOS). Both FGF receptor (FGFR)-dependent ROS formation via activation of NADPH oxidase 2 (Nox2) as well as ROS degradation via superoxide dismutase 2 (SOD2) and catalase (CAT) is stimulated by FGF23. Pre-incubation with a Klotho inhibitor blunts the FGF23-stimulated Akt-eNOS activation and NO synthesis, and decreases ROS degradation by blocking SOD2 and CAT enzymes, whereas FGF23-stimulated ROS synthesis via Nox2 is unaffected, resulting in low NO bioavailability and increased oxidative stress. Our data indicate that in the presence of Klotho, FGF23 induces NO release in HCAEC and its stimulating effects on ROS production are counterbalanced by increased ROS degradation. In states of Klotho deficiency, e.g., CKD, FGF23-mediated NO synthesis is blunted and ROS formation overrules ROS degradation. Thus, FGF23 excess may primarily promote oxidative stress and thus endothelial dysfunction.
Collapse
|
118
|
Dipeptidyl peptidase-4 inhibition with linagliptin prevents western diet-induced vascular abnormalities in female mice. Cardiovasc Diabetol 2016; 15:94. [PMID: 27391040 PMCID: PMC4938903 DOI: 10.1186/s12933-016-0414-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/23/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Vascular stiffening, a risk factor for cardiovascular disease, is accelerated, particularly in women with obesity and type 2 diabetes. Preclinical evidence suggests that dipeptidylpeptidase-4 (DPP-4) inhibitors may have cardiovascular benefits independent of glycemic lowering effects. Recent studies show that consumption of a western diet (WD) high in fat and simple sugars induces aortic stiffening in female C57BL/6J mice in advance of increasing blood pressure. The aims of this study were to determine whether administration of the DPP-4 inhibitor, linagliptin (LGT), prevents the development of aortic and endothelial stiffness induced by a WD in female mice. METHODS C56Bl6/J female mice were fed a WD for 4 months. Aortic stiffness and ex vivo endothelial stiffness were evaluated by Doppler pulse wave velocity (PWV) and atomic force microscopy (AFM), respectively. In addition, we examined aortic vasomotor responses and remodeling markers via immunohistochemistry. Results were analyzed via 2-way ANOVA, p < 0.05 was considered as statistically significant. RESULTS Compared to mice fed a control diet (CD), WD-fed mice exhibited a 24 % increase in aortic PWV, a five-fold increase in aortic endothelial stiffness, and impaired endothelium-dependent vasodilation. In aorta, these findings were accompanied by medial wall thickening, adventitial fibrosis, increased fibroblast growth factor 23 (FGF-23), decreased Klotho, enhanced oxidative stress, and endothelial cell ultrastructural changes, all of which were prevented with administration of LGT. CONCLUSIONS The present findings support the notion that DPP-4 plays a role in development of WD-induced aortic stiffening, vascular oxidative stress, endothelial dysfunction, and vascular remodeling. Whether, DPP-4 inhibition could be a therapeutic tool used to prevent the development of aortic stiffening and the associated cardiovascular complications in obese and diabetic females remains to be elucidated.
Collapse
|
119
|
Zoccali C, Torino C, Curatola G, Panuccio V, Tripepi R, Pizzini P, Versace M, Bolignano D, Cutrupi S, Ghiadoni L, Thadhani R, Tripepi G, Mallamaci F. Serum phosphate modifies the vascular response to vitamin D receptor activation in chronic kidney disease (CKD) patients. Nutr Metab Cardiovasc Dis 2016; 26:581-589. [PMID: 27113290 DOI: 10.1016/j.numecd.2016.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/01/2016] [Accepted: 03/14/2016] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND AIMS Vitamin D receptor activation (VDRA) ameliorates endothelial dysfunction in CKD patients but also increases phosphate and FGF-23, which may attenuate the beneficial effect of VDRA on endothelial function. METHODS AND RESULTS This is a pre-specified secondary analysis of the PENNY trial (NCT01680198) testing the effect of phosphate and FGF-23 on the flow mediated vasodilatory (FMD) response to paricalcitol (PCT, 2 μg/day) and placebo over a 12-weeks treatment period. Eighty-eight stage G3-4 CKD patients were randomized to PCT (n = 44) and Placebo (n = 44). Endothelial function was assessed by measuring endothelium dependent forearm blood flow (FBF) response to ischemia. The FMD response was by the 61% higher in PCT treated patients than in those on placebo (P = 0.01). Phosphate (+11%, P = 0.039), calcium (+3%, P = 0.01) and, particularly so, FGF23 (+164%, P < 0.001) increased in PCT treated patients. Changes in FMD by PCT associated inversely with phosphate (r = -0.37, P = 0.01) but were independent of FGF-23, calcium and PTH changes. The response to PCT was maximal in patients with no changes in phosphate (1st tertile), attenuated in those with mild-to-moderate rise in phosphate (2nd tertile) and abolished in those with the most pronounced phosphate increase (3rd tertile) (effect modification P = 0.009). No effect modification by FGF-23 and other variables was observed. CONCLUSIONS The beneficial effect of PCT on endothelial function in CKD is maximal in patients with no or minimal changes in phosphate and it is abolished in patients with a pronounced phosphate rise. These findings generate the hypothesis that the endothelium protective effect by VDRA may be potentiated by phosphate lowering interventions.
Collapse
Affiliation(s)
- C Zoccali
- CNR-IBIM/IFC Clinical Epidemiology and Pathophysiology of Renal Diseases and Hypertension, Ospedali Riuniti, Reggio Calabria, Italy.
| | - C Torino
- CNR-IBIM/IFC Clinical Epidemiology and Pathophysiology of Renal Diseases and Hypertension, Ospedali Riuniti, Reggio Calabria, Italy
| | - G Curatola
- CNR-IBIM/IFC Clinical Epidemiology and Pathophysiology of Renal Diseases and Hypertension, Ospedali Riuniti, Reggio Calabria, Italy
| | - V Panuccio
- Nephrology, Hypertension and Renal Transplantation Unit, Ospedali Riuniti, Reggio Calabria, Italy
| | - R Tripepi
- CNR-IBIM/IFC Clinical Epidemiology and Pathophysiology of Renal Diseases and Hypertension, Ospedali Riuniti, Reggio Calabria, Italy
| | - P Pizzini
- CNR-IBIM/IFC Clinical Epidemiology and Pathophysiology of Renal Diseases and Hypertension, Ospedali Riuniti, Reggio Calabria, Italy
| | - M Versace
- CNR-IBIM/IFC Clinical Epidemiology and Pathophysiology of Renal Diseases and Hypertension, Ospedali Riuniti, Reggio Calabria, Italy
| | - D Bolignano
- CNR-IBIM/IFC Clinical Epidemiology and Pathophysiology of Renal Diseases and Hypertension, Ospedali Riuniti, Reggio Calabria, Italy
| | - S Cutrupi
- CNR-IBIM/IFC Clinical Epidemiology and Pathophysiology of Renal Diseases and Hypertension, Ospedali Riuniti, Reggio Calabria, Italy
| | - L Ghiadoni
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - R Thadhani
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
| | - G Tripepi
- CNR-IBIM/IFC Clinical Epidemiology and Pathophysiology of Renal Diseases and Hypertension, Ospedali Riuniti, Reggio Calabria, Italy
| | - F Mallamaci
- CNR-IBIM/IFC Clinical Epidemiology and Pathophysiology of Renal Diseases and Hypertension, Ospedali Riuniti, Reggio Calabria, Italy; Nephrology, Hypertension and Renal Transplantation Unit, Ospedali Riuniti, Reggio Calabria, Italy
| |
Collapse
|
120
|
Katoh M. FGFR inhibitors: Effects on cancer cells, tumor microenvironment and whole-body homeostasis (Review). Int J Mol Med 2016; 38:3-15. [PMID: 27245147 PMCID: PMC4899036 DOI: 10.3892/ijmm.2016.2620] [Citation(s) in RCA: 311] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/23/2016] [Indexed: 12/13/2022] Open
Abstract
Fibroblast growth factor (FGF)2, FGF4, FGF7 and FGF20 are representative paracrine FGFs binding to heparan-sulfate proteoglycan and fibroblast growth factor receptors (FGFRs), whereas FGF19, FGF21 and FGF23 are endocrine FGFs binding to Klotho and FGFRs. FGFR1 is relatively frequently amplified and overexpressed in breast and lung cancer, and FGFR2 in gastric cancer. BCR-FGFR1, CNTRL-FGFR1, CUX1-FGFR1, FGFR1OP-FGFR1, MYO18A-FGFR1 and ZMYM2-FGFR1 fusions in myeloproliferative neoplasms are non-receptor-type FGFR kinases, whereas FGFR1-TACC1, FGFR2-AFF3, FGFR2-BICC1, FGFR2-PPHLN1, FGFR3-BAIAP2L1 and FGFR3-TACC3 fusions in solid tumors are transmembrane-type FGFRs with C-terminal alterations. AZD4547, BGJ398 (infigratinib), Debio-1347 and dovitinib are FGFR1/2/3 inhibitors; BLU9931 is a selective FGFR4 inhibitor; FIIN-2, JNJ-42756493, LY2874455 and ponatinib are pan-FGFR inhibitors. AZD4547, dovitinib and ponatinib are multi-kinase inhibitors targeting FGFRs, colony stimulating factor 1 receptor (CSF1R), vascular endothelial growth factor (VEGF)R2, and others. The tumor microenvironment consists of cancer cells and stromal/immune cells, such as cancer-associated fibroblasts (CAFs), endothelial cells, M2-type tumor-associating macrophages (M2-TAMs), myeloid-derived suppressor cells (MDSCs) and regulatory T cells. FGFR inhibitors elicit antitumor effects directly on cancer cells, as well as indirectly through the blockade of paracrine signaling. The dual inhibition of FGF and CSF1 or VEGF signaling is expected to enhance the antitumor effects through the targeting of immune evasion and angiogenesis in the tumor microenvironment. Combination therapy using tyrosine kinase inhibitors (FGFR or CSF1R inhibitors) and immune checkpoint blockers (anti-PD-1 or anti-CTLA-4 monoclonal antibodies) may be a promising choice for cancer patients. The inhibition of FGF19-FGFR4 signaling is associated with a risk of liver toxicity, whereas the activation of FGF23-FGFR4 signaling is associated with a risk of heart toxicity. Endocrine FGF signaling affects the pathophysiology of cancer patients who are prescribed FGFR inhibitors. Whole-genome sequencing is necessary for the detection of promoter/enhancer alterations of FGFR genes and rare alterations of other genes causing FGFR overexpression. To sustain the health care system in an aging society, a benefit-cost analysis should be performed with a focus on disease-free survival and the total medical cost before implementing genome-based precision medicine for cancer patients.
Collapse
Affiliation(s)
- Masaru Katoh
- Department of Omics Network, National Cancer Center, Tokyo 104-0045, Japan
| |
Collapse
|
121
|
Wacker MJ, Touchberry CD, Silswal N, Brotto L, Elmore CJ, Bonewald LF, Andresen J, Brotto M. Skeletal Muscle, but not Cardiovascular Function, Is Altered in a Mouse Model of Autosomal Recessive Hypophosphatemic Rickets. Front Physiol 2016; 7:173. [PMID: 27242547 PMCID: PMC4866514 DOI: 10.3389/fphys.2016.00173] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 04/28/2016] [Indexed: 01/29/2023] Open
Abstract
Autosomal recessive hypophosphatemic rickets (ARHR) is a heritable disorder characterized by hypophosphatemia, osteomalacia, and poor bone development. ARHR results from inactivating mutations in the DMP1 gene with the human phenotype being recapitulated in the Dmp1 null mouse model which displays elevated plasma fibroblast growth factor 23. While the bone phenotype has been well-characterized, it is not known what effects ARHR may also have on skeletal, cardiac, or vascular smooth muscle function, which is critical to understand in order to treat patients suffering from this condition. In this study, the extensor digitorum longus (EDL-fast-twitch muscle), soleus (SOL–slow-twitch muscle), heart, and aorta were removed from Dmp1 null mice and ex-vivo functional tests were simultaneously performed in collaboration by three different laboratories. Dmp1 null EDL and SOL muscles produced less force than wildtype muscles after normalization for physiological cross sectional area of the muscles. Both EDL and SOL muscles from Dmp1 null mice also produced less force after the addition of caffeine (which releases calcium from the sarcoplasmic reticulum) which may indicate problems in excitation contraction coupling in these mice. While the body weights of the Dmp1 null were smaller than wildtype, the heart weight to body weight ratio was higher. However, there were no differences in pathological hypertrophic gene expression compared to wildtype and maximal force of contraction was not different indicating that there may not be cardiac pathology under the tested conditions. We did observe a decrease in the rate of force development generated by cardiac muscle in the Dmp1 null which may be related to some of the deficits observed in skeletal muscle. There were no differences observed in aortic contractions induced by PGF2α or 5-HT or in endothelium-mediated acetylcholine-induced relaxations or endothelium-independent sodium nitroprusside-induced relaxations. In summary, these results indicate that there are deficiencies in both fast twitch and slow twitch muscle fiber type contractions in this model of ARHR, while there was less of a phenotype observed in cardiac muscle, and no differences observed in aortic function. These results may help explain skeletal muscle weakness reported by some patients with osteomalacia and need to be further investigated.
Collapse
Affiliation(s)
- Michael J Wacker
- Muscle Biology Research Group, School of Medicine, University of Missouri-Kansas City Kansas City, MO, USA
| | | | - Neerupma Silswal
- Muscle Biology Research Group, School of Medicine, University of Missouri-Kansas City Kansas City, MO, USA
| | - Leticia Brotto
- Bone-Muscle Collaborative Science, College of Nursing and Health Innovation, University of Texas at Arlington Arlington, TX, USA
| | - Chris J Elmore
- Muscle Biology Research Group, School of Medicine, University of Missouri-Kansas City Kansas City, MO, USA
| | - Lynda F Bonewald
- Bone Biology Research Group, School of Dentistry, University of Missouri-Kansas City Kansas City, MO, USA
| | - Jon Andresen
- Muscle Biology Research Group, School of Medicine, University of Missouri-Kansas City Kansas City, MO, USA
| | - Marco Brotto
- Bone-Muscle Collaborative Science, College of Nursing and Health Innovation, University of Texas at Arlington Arlington, TX, USA
| |
Collapse
|
122
|
Cao C, Hu JX, Dong YF, Zhan R, Li P, Su H, Peng Q, Wu T, Huang X, Sun WH, Wu QH, Cheng XS. Association of Endothelial and Mild Renal Dysfunction With the Severity of Left Ventricular Hypertrophy in Hypertensive Patients. Am J Hypertens 2016; 29:501-8. [PMID: 26232035 DOI: 10.1093/ajh/hpv128] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/13/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The association between impaired renal function and increase left ventricular mass was shown to be related to increase in arterial stiffness, which indicates that vascular homeostasis and remodeling may impact the left ventricular hypertrophy (LVH) in patients with renal dysfunction. METHODS We measured the peripheral arterial reactive hyperemia index (RHI) and estimated glomerular filtration rate (eGFR) in 317 hypertensive patients comprising 115 normal RHI (RHI > 1.67) and normal eGFR (eGFR ≥ 90ml/min per 1.73 m(2)), 136 low RHI (RHI ≤ 1.67), 27 low eGFR (60 ≤ eGFR < 90ml/min per 1.73 m(2)) and 39 low RHI combined with low eGFR. RESULTS Multivariate logistic regression analysis identified lg RHI (odds ratio (OR): 0.001, 95% confidence interval (CI): 10(-6) to 0.426, P = 0.024) and lg eGFR (OR: 0.009, 95% CI: 10(-4) to 0.414, P = 0.016) as independent factors correlated with LVH respectively in hypertensive patients. Compared with normal RHI and eGFR patients, the extent of LVH in patients with either low RHI (OR: 1.224 95% CI: 0.451 to 3.327, P = 0.691) or low eGFR (OR: 0.593 95% CI: 0.070 to 5.037, P = 0.632) did not significantly increase, while it increased significantly in patients with low RHI combined with low eGFR (OR: 4.629 95% CI: 1.592 to 13.458, P = 0.005). CONCLUSIONS The concurrence of endothelial and mild renal dysfunction was significantly associated with the severity of LVH in hypertensive patients.
Collapse
Affiliation(s)
- Cheng Cao
- Department of Cardiovascular Medicine, Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Jian-Xin Hu
- Department of Cardiovascular Medicine, Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Yi-Fei Dong
- Department of Cardiovascular Medicine, Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, China; Key Laboratory of Molecular Biology in Jiangxi Province, Nanchang City, Jiangxi Province, China.
| | - Rui Zhan
- Department of Cardiovascular Medicine, Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Ping Li
- Department of Cardiovascular Medicine, Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Hai Su
- Department of Cardiovascular Medicine, Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Qiang Peng
- Department of Cardiovascular Medicine, Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Tao Wu
- Department of Cardiovascular Medicine, Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Xiao Huang
- Department of Cardiovascular Medicine, Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Wen-Hua Sun
- Department of Cardiovascular Medicine, Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Qing-Hua Wu
- Department of Cardiovascular Medicine, Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, China
| | - Xiao-Shu Cheng
- Department of Cardiovascular Medicine, Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province, China
| |
Collapse
|
123
|
Abstract
Alpha-Klotho (αKlotho) protein is encoded by the gene, Klotho, and functions as a coreceptor for endocrine fibroblast growth factor-23. The extracellular domain of αKlotho is cleaved by secretases and released into the circulation where it is called soluble αKlotho. Soluble αKlotho in the circulation starts to decline in chronic kidney disease (CKD) stage 2 and urinary αKlotho in even earlier CKD stage 1. Therefore soluble αKlotho is an early and sensitive marker of decline in kidney function. Preclinical data from numerous animal experiments support αKlotho deficiency as a pathogenic factor for CKD progression and extrarenal CKD complications including cardiac and vascular disease, hyperparathyroidism, and disturbed mineral metabolism. αKlotho deficiency induces cell senescence and renders cells susceptible to apoptosis induced by a variety of cellular insults including oxidative stress. αKlotho deficiency also leads to defective autophagy and angiogenesis and promotes fibrosis in the kidney and heart. Most importantly, prevention of αKlotho decline, upregulation of endogenous αKlotho production, or direct supplementation of soluble αKlotho are all associated with attenuation of renal fibrosis, retardation of CKD progression, improvement of mineral metabolism, amelioration of cardiac function and morphometry, and alleviation of vascular calcification in CKD. Therefore in rodents, αKlotho is not only a diagnostic and prognostic marker for CKD but the enhancement of endogenous or supplement of exogenous αKlotho are promising therapeutic strategies to prevent, retard, and decrease the comorbidity burden of CKD.
Collapse
Affiliation(s)
- J A Neyra
- University of Texas Southwestern Medical Center, Dallas, TX, United States; Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - M C Hu
- University of Texas Southwestern Medical Center, Dallas, TX, United States; Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
124
|
Kunasegaran T, Mustafa MR, Murugan DD, Achike FI. The bioflavonoid quercetin synergises with PPAR-γ agonist pioglitazone in reducing angiotensin-II contractile effect in fructose-streptozotocin induced diabetic rats. Biochimie 2016; 125:131-9. [PMID: 27012965 DOI: 10.1016/j.biochi.2016.03.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 03/21/2016] [Indexed: 01/13/2023]
Abstract
This study investigated the effects of combined minimal concentrations of quercetin and pioglitazone on angiotensin II-induced contraction of the aorta from fructose-streptozotocin (F-STZ)-induced type 2 diabetic rats and the possible role of superoxide anions (O2(-)) and nitric oxide (NO) in their potential therapeutic interaction. Contractile responses to Ang II of aortic rings from Sprague-Dawley (SD) and F-STZ rats were tested following pre-incubation of the tissues in the vehicle (DMSO; 0.05%), quercetin (Q, 0.1 μM), pioglitazone (P, 0.1 μM) or their combination (P + Q; 0.1 μM each). The amount of superoxide anion was evaluated by lucigenin-enhanced chemiluminescence and dihydroethidium fluorescence, and NO by assay of total nitrate/nitrite, and 4-Amino-5-Methylamino-2',7'-Difluorofluorescein (DAF-FM) diacetate. The synergistic reduction of Ang II-induced contraction of diabetic but not normal aorta with minimally effective concentrations of P + Q occurs through inhibiting O2(-) and increasing NO bioavailability. This finding opens the possibility of maximal vascular protective/antidiabetic effects with low dose pioglitazone combined with quercetin, thus minimizing the risk of adverse effects.
Collapse
Affiliation(s)
- Thubasni Kunasegaran
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mohd Rais Mustafa
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Dharmani Devi Murugan
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| | | |
Collapse
|
125
|
Silva AP, Mendes F, Fragoso A, Jeronimo T, Pimentel A, Gundlach K, Büchel J, Santos N, Neves PL. Altered serum levels of FGF-23 and magnesium are independent risk factors for an increased albumin-to-creatinine ratio in type 2 diabetics with chronic kidney disease. J Diabetes Complications 2016; 30:275-80. [PMID: 26750742 DOI: 10.1016/j.jdiacomp.2015.11.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 12/12/2022]
Abstract
AIMS To investigate the role of FGF-23 and magnesium in relation to the albumin-to-creatinine ratio in type 2 diabetics with chronic kidney disease (CKD) stages 2-4. METHODS In a cross-sectional study we included all eligible type 2 diabetic patients with CKD stages 2-4, followed in our outpatient Diabetic Kidney clinic. We used descriptive statistics, the Student'st-test, ANOVA and the chi-square tests. Our population was divided according to the UACR (G1 30-300 mg/g and G2≥300 mg/g), and compared these groups regarding several biological and laboratorial parameters. We employed a multiple regression model to identify risk factors of increased UACR. RESULTS The patients in G2 displayed a lower eGFR (p=0.0001) and, had lower levels of magnesium (p=0.004) as well as higher levels of FGF-23 (p=0.043) compared to patients in G1. FGF-23 (β=0.562, P=0.0001) and the magnesium (β=- 8.916, p=0.0001) were associated with increased UACR. CONCLUSIONS A dysregulation of mineral metabolism, reflected by altered levels of magnesium and FGF-23, correlates with an increased UACR in type 2 diabetic patients with CKD stages 2-4.
Collapse
Affiliation(s)
- Ana Paula Silva
- Nephrology, Centro Hospitalar do Algarve, Faro, Portugal; Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal.
| | - Filipa Mendes
- Nephrology, Centro Hospitalar do Algarve, Faro, Portugal
| | - André Fragoso
- Nephrology, Centro Hospitalar do Algarve, Faro, Portugal
| | | | - Ana Pimentel
- Nephrology, Centro Hospitalar do Algarve, Faro, Portugal
| | - Kristina Gundlach
- Fresenius Medical Care Deutschland GmbH, Bad Homburg vor der Höhe, Germany
| | - Janine Büchel
- Fresenius Medical Care Deutschland GmbH, Bad Homburg vor der Höhe, Germany
| | - Nélio Santos
- Pathology Clinic, Centro Hospitalar do Algarve, Faro, Portugal
| | - Pedro Leão Neves
- Nephrology, Centro Hospitalar do Algarve, Faro, Portugal; Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal
| |
Collapse
|
126
|
Dounousi E, Torino C, Pizzini P, Cutrupi S, Panuccio V, D'Arrigo G, Abd ElHafeez S, Tripepi G, Mallamaci F, Zoccali C. Intact FGF23 and α-Klotho during acute inflammation/sepsis in CKD patients. Eur J Clin Invest 2016; 46:234-41. [PMID: 26728476 DOI: 10.1111/eci.12588] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 12/30/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND High FGF23 and low α-Klotho levels associate with systemic inflammation and reduced nitric oxide (NO) bioavailability, but the dynamics of this relationship in patients with CKD has not been investigated. METHODS We sequentially measured serum intact FGF23 and carboxyl-terminal (iFGF23, cFGF23), the iFGF23/cFGF23 ratio, αKlotho, biomarkers of inflammation (hs-CRP, IL-6 and TNF-α) and sepsis (procalcitonin), nitrotyrosine (reflecting NO synthesis and oxidative stress), serum iron and ferritin and CKD-MBD biomarkers, PTH, 25(OH)VD, 1,25(OH)2 VD at peak of intercurrent sepsis and after complete resolution in a series of 17 patients with CKD. RESULTS At peak infection, biomarkers of inflammation/sepsis, ferritin and nitrotyrosine were all very high (all P < 0·01) and declined towards the normal range thereafter (P < 0·01). iFGF23 was 191 ± 10 pg/ml (geometric mean, SD) and doubled to 371 ± 8 pg/ml (P = 0·003) after the resolution of infection, while cFGF23 did not change (246 ± 5 pg/mL vs. 248 ± 5 pg/mL, P = 0·50). As a consequence, the iFGF23/cFGF23 ratio, an indicator of the proteolytic cleavage of the FGF23 molecule, was 0·78 ± 3·87 at peak infection and increased to 1·49 ± 3·00 after resolution of infection (P < 0·001). In contrast, serum α-Klotho levels were upregulated at peak infection (peak infection: 526 ± 4 pg/ml, postinfection: 447 ± 4 pg/ml, P = 0·001). The eGFR, PTH and vitamin D did not change significantly throughout. CONCLUSIONS Acute inflammation/sepsis suppresses the active form of FGF23 and activates α-Klotho, the latter effect being likely attributable to enhance proteolysis of FGF23 molecule. iFGF23 downregulation and α-Klotho upregulation during acute sepsis may participate into the counter-regulatory response to severe inflammation in CKD patients with sepsis.
Collapse
Affiliation(s)
- Evangelia Dounousi
- Department of Nephrology, Medical School, University of Ioannina, Ioannina, Greece
| | - Claudia Torino
- Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, CNR-IFC, Reggio Calabria, Italy
| | - Patrizia Pizzini
- Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, CNR-IFC, Reggio Calabria, Italy
| | - Sebastiano Cutrupi
- Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, CNR-IFC, Reggio Calabria, Italy
| | - Vincenzo Panuccio
- Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, CNR-IFC, Reggio Calabria, Italy.,Renal Unit, Reggio Calabria, Italy
| | - Graziella D'Arrigo
- Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, CNR-IFC, Reggio Calabria, Italy
| | | | - Giovanni Tripepi
- Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, CNR-IFC, Reggio Calabria, Italy
| | - Francesca Mallamaci
- Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, CNR-IFC, Reggio Calabria, Italy.,Renal Unit, Reggio Calabria, Italy
| | - Carmine Zoccali
- Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, CNR-IFC, Reggio Calabria, Italy
| |
Collapse
|
127
|
Ter Maaten JM, Damman K, Verhaar MC, Paulus WJ, Duncker DJ, Cheng C, van Heerebeek L, Hillege HL, Lam CSP, Navis G, Voors AA. Connecting heart failure with preserved ejection fraction and renal dysfunction: the role of endothelial dysfunction and inflammation. Eur J Heart Fail 2016; 18:588-98. [PMID: 26861140 DOI: 10.1002/ejhf.497] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 01/09/2016] [Accepted: 01/12/2015] [Indexed: 12/17/2022] Open
Abstract
Renal dysfunction in heart failure with preserved ejection fraction (HFpEF) is common and is associated with increased mortality. Impaired renal function is also a risk factor for developing HFpEF. A new paradigm for HFpEF, proposing a sequence of events leading to myocardial remodelling and dysfunction in HFpEF, was recently introduced, involving inflammatory, microvascular, and cardiac components. The kidney might play a key role in this systemic process. Renal impairment causes metabolic and systemic derangements in circulating factors, causing an activated systemic inflammatory state and endothelial dysfunction, which may lead to cardiomyocyte stiffening, hypertrophy, and interstitial fibrosis via cross-talk between the endothelium and cardiomyocyte compartments. Here, we review the role of endothelial dysfunction and inflammation to explain the link between renal dysfunction and HFpEF, which allows for identification of new early risk markers, prognostic factors, and unique targets for intervention.
Collapse
Affiliation(s)
- Jozine M Ter Maaten
- University of Groningen, Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Kevin Damman
- University of Groningen, Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Marianne C Verhaar
- University Medical Center Utrecht, Department of Nephrology and Hypertension, Utrecht, The Netherlands
| | - Walter J Paulus
- Department of Physiology, Cardiology, Pathology, and Surgery, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Dirk J Duncker
- Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Caroline Cheng
- University Medical Center Utrecht, Department of Nephrology and Hypertension, Utrecht, The Netherlands.,Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Loek van Heerebeek
- Department of Physiology, Cardiology, Pathology, and Surgery, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Hans L Hillege
- University of Groningen, Department of Epidemiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Carolyn S P Lam
- National Heart Centre Singapore and, Duke-National University of Singapore Graduate School Medicine, Singapore
| | - Gerjan Navis
- University of Groningen, Department of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| | - Adriaan A Voors
- University of Groningen, Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
128
|
Affiliation(s)
- Marcello Tonelli
- From Department of Medicine, University of Calgary, AB, Canada (M.T.); Department of Medicine and Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, and Howard Hughes Medical Institute, Chevy Chase, MD (S.A.K..); and Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston (R.T.)
| | - S. Ananth Karumanchi
- From Department of Medicine, University of Calgary, AB, Canada (M.T.); Department of Medicine and Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, and Howard Hughes Medical Institute, Chevy Chase, MD (S.A.K..); and Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston (R.T.)
| | - Ravi Thadhani
- From Department of Medicine, University of Calgary, AB, Canada (M.T.); Department of Medicine and Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, and Howard Hughes Medical Institute, Chevy Chase, MD (S.A.K..); and Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston (R.T.)
| |
Collapse
|
129
|
Dounousi E, Torino C, Pizzini P, Cutrupi S, Panuccio V, D'Arrigo G, Abd ElHafeez S, Tripepi G, Mallamaci F, Zoccali C. Effect of inflammation by acute sepsis on intact fibroblast growth factor 23 (iFGF23) and asymmetric dimethyl arginine (ADMA) in CKD patients. Nutr Metab Cardiovasc Dis 2016; 26:80-83. [PMID: 26712271 DOI: 10.1016/j.numecd.2015.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 10/07/2015] [Accepted: 10/12/2015] [Indexed: 12/13/2022]
Affiliation(s)
- E Dounousi
- Department of Nephrology, Medical School, University of Ioannina, Ioannina, Greece
| | - C Torino
- CNR-IFC, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Reggio Calabria, Italy
| | - P Pizzini
- CNR-IFC, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Reggio Calabria, Italy
| | - S Cutrupi
- CNR-IFC, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Reggio Calabria, Italy
| | - V Panuccio
- Renal Unit, Reggio Calabria, Italy; CNR-IFC, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Reggio Calabria, Italy
| | - G D'Arrigo
- CNR-IFC, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Reggio Calabria, Italy
| | - S Abd ElHafeez
- Epidemiology Department, High Institute of Public Health, Alexandria University, Alexandria, Egypt
| | - G Tripepi
- CNR-IFC, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Reggio Calabria, Italy
| | - F Mallamaci
- Renal Unit, Reggio Calabria, Italy; CNR-IFC, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Reggio Calabria, Italy
| | - C Zoccali
- CNR-IFC, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Reggio Calabria, Italy.
| |
Collapse
|
130
|
Nilsson IL, Norenstedt S, Granath F, Zedenius J, Pernow Y, Larsson TE. FGF23, metabolic risk factors, and blood pressure in patients with primary hyperparathyroidism undergoing parathyroid adenomectomy. Surgery 2016; 159:211-7. [DOI: 10.1016/j.surg.2015.06.057] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 04/26/2015] [Accepted: 06/03/2015] [Indexed: 01/01/2023]
|
131
|
Freedman BI, Divers J, Russell GB, Palmer ND, Bowden DW, Carr JJ, Wagenknecht LE, Hightower RC, Xu J, Smith SC, Langefeld CD, Hruska KA, Register TC. Plasma FGF23 and Calcified Atherosclerotic Plaque in African Americans with Type 2 Diabetes Mellitus. Am J Nephrol 2015; 42:391-401. [PMID: 26693712 PMCID: PMC4732898 DOI: 10.1159/000443241] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 12/06/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND Fibroblast growth factor 23 (FGF23) is a phosphaturic hormone implicated in disorders of serum phosphorus concentration and vitamin D. The role of FGF23 in vascular calcification remains controversial. METHODS Relationships between FGF23 and coronary artery calcified atherosclerotic plaque (CAC), aortoiliac calcified plaque (CP), carotid artery CP, volumetric bone mineral density (vBMD), albuminuria, and estimated glomerular filtration rate (eGFR) were determined in 545 African Americans with type 2 diabetes (T2D) and preserved kidney function in African American-Diabetes Heart Study participants. Generalized linear models were fitted to test associations between FGF23 and cardiovascular, bone, and renal phenotypes, and change in measurements over time, adjusting for age, gender, African ancestry proportion, body mass index, diabetes duration, hemoglobin A1c, blood pressure, renin-angiotensin-system inhibitors, statins, calcium supplements, serum calcium, and serum phosphate. RESULTS The sample was 56.7% female with a mean (SD) age of 55.6 (9.6) years, diabetes duration of 10.3 (8.2) years, eGFR 90.9 (22.1) ml/min/1.73 m2, urine albumin:creatinine ratio (UACR) 151 (588) (median 13) mg/g, plasma FGF23 161 (157) RU/ml, and CAC 637 (1,179) mg. In fully adjusted models, FGF23 was negatively associated with eGFR (p < 0.0001) and positively associated with UACR (p < 0.0001) and CAC (p = 0.0006), but not with carotid CP or aortic CP. Baseline FGF23 concentration did not associate with changes in vBMD or CAC after a mean of 5.1 years follow-up. CONCLUSIONS Plasma FGF23 concentrations were independently associated with subclinical coronary artery disease, albuminuria, and kidney function in the understudied African American population with T2D. Findings support relationships between FGF23 and vascular calcification, but not between FGF23 and bone mineral density, in African Americans lacking advanced nephropathy.
Collapse
Affiliation(s)
- Barry I. Freedman
- Department of Internal Medicine, Section on Nephrology, Winston-Salem, North Carolina, USA
| | - Jasmin Divers
- Division of Public Health Sciences, Department of Biostatistical Sciences, Center for Public Health Genomics, Winston-Salem, North Carolina, USA
| | - Gregory B. Russell
- Division of Public Health Sciences, Department of Biostatistical Sciences, Center for Public Health Genomics, Winston-Salem, North Carolina, USA
| | - Nicholette D. Palmer
- Department of Biochemistry, Center for Genomics and Personalized Medicine Research, Center for Diabetes Research, Winston-Salem, North Carolina, USA
| | - Donald W. Bowden
- Department of Biochemistry, Center for Genomics and Personalized Medicine Research, Center for Diabetes Research, Winston-Salem, North Carolina, USA
| | - J. Jeffrey Carr
- Department of Radiology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Lynne E. Wagenknecht
- Division of Public Health Sciences, Department of Biostatistical Sciences, Center for Public Health Genomics, Winston-Salem, North Carolina, USA
| | | | - Jianzhao Xu
- Department of Biochemistry, Center for Genomics and Personalized Medicine Research, Center for Diabetes Research, Winston-Salem, North Carolina, USA
| | - S. Carrie Smith
- Department of Internal Medicine, Section on Nephrology, Winston-Salem, North Carolina, USA
| | - Carl D. Langefeld
- Division of Public Health Sciences, Department of Biostatistical Sciences, Center for Public Health Genomics, Winston-Salem, North Carolina, USA
| | - Keith A. Hruska
- Department of Pediatrics, Nephrology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Thomas C. Register
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
132
|
Restoration of Endothelial Function in Pparα (-/-) Mice by Tempol. PPAR Res 2015; 2015:728494. [PMID: 26649033 PMCID: PMC4663011 DOI: 10.1155/2015/728494] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 10/16/2015] [Accepted: 10/20/2015] [Indexed: 12/18/2022] Open
Abstract
Peroxisome proliferator activated receptor alpha (PPARα) is one of the PPAR isoforms belonging to the nuclear hormone receptor superfamily that regulates genes involved in lipid and lipoprotein metabolism. PPARα is present in the vascular wall and is thought to be involved in protection against vascular disease. To determine if PPARα contributes to endothelial function, conduit and cerebral resistance arteries were studied in Pparα−/− mice using isometric and isobaric tension myography, respectively. Aortic contractions to PGF2α and constriction of middle cerebral arteries to phenylephrine were not different between wild type (WT) and Pparα−/−; however, relaxation/dilation to acetylcholine (ACh) was impaired. There was no difference in relaxation between WT and Pparα−/− aorta to treatment with a nitric oxide (NO) surrogate indicating impairment in endothelial function. Endothelial NO levels as well as NO synthase expression were reduced in Pparα−/− aortas, while superoxide levels were elevated. Two-week feeding with the reactive oxygen species (ROS) scavenger, tempol, normalized ROS levels and rescued the impaired endothelium-mediated relaxation in Pparα−/− mice. These results suggest that Pparα−/− mice have impaired endothelial function caused by decreased NO bioavailability. Therefore, activation of PPARα receptors may be a therapeutic target for maintaining endothelial function and protection against cardiovascular disease.
Collapse
|
133
|
Lee G, Krishnasamy R, Hawley CM, Johnson DW. The impact of fibroblast growth factor-23 on the cardiovascular system in chronic kidney disease. Expert Rev Endocrinol Metab 2015; 10:565-568. [PMID: 30289031 DOI: 10.1586/17446651.2015.1079124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Chronic kidney disease is associated with an accelerated risk of cardiovascular (CV) mortality. Seminal work over the last decade has identified abnormal bone metabolism as an important modulator of the increased CV burden in this cohort. In particular, FGF23, a phosphaturic hormone with serum levels found to be markedly elevated in chronic kidney disease, is independently associated with increased risks of all-cause mortality and CV events. This editorial will discuss the proposed mechanisms linking FGF23 to CV disease in chronic kidney disease, namely, direct cardiac myocyte toxicity, endothelial dysfunction and vascular calcification.
Collapse
Affiliation(s)
- Gavin Lee
- a 1 Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
| | - Rathika Krishnasamy
- a 1 Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
- b 2 School of Medicine, The University of Queensland, Brisbane, Australia
| | - Carmel M Hawley
- a 1 Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
- b 2 School of Medicine, The University of Queensland, Brisbane, Australia
- c 3 Translational Research Institute, Brisbane, Australia
| | - David W Johnson
- a 1 Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
- b 2 School of Medicine, The University of Queensland, Brisbane, Australia
- c 3 Translational Research Institute, Brisbane, Australia
| |
Collapse
|
134
|
Neyra JA, Moe OW, Hu MC. Fibroblast growth factor 23 and acute kidney injury. Pediatr Nephrol 2015; 30:1909-18. [PMID: 25480729 PMCID: PMC4458234 DOI: 10.1007/s00467-014-3006-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 11/03/2014] [Accepted: 11/04/2014] [Indexed: 02/07/2023]
Abstract
Fibroblast growth factor 23 (FGF23), which is produced in bone, participates in the maintenance of phosphate metabolism and can serve as a biomarker for adverse cardiovascular outcomes in patients with chronic kidney disease and end-stage renal disease. Circulating FGF23 rapidly increases after acute kidney injury (AKI), preceding other known markers such as neutrophil gelatinase-associated lipocalin and serum creatinine. The increase in FGF23 in AKI appears to be independent of parathyroid hormone, vitamin D signaling pathways, and dietary phosphate. The potential mechanisms include: (1) increased production of FGF23 in the bone by yet-to-be-identified factors; (2) ectopic production of FGF23 by injured renal tubules; and (3) decreased renal clearance of circulating FGF23. Circulating FGF23 determined by intact FGF23 enzyme-linked immunosorbent assay (ELISA) is a more reliable biomarker of AKI than FGF23 C-terminal ELISA (a mixed readout of C-terminal fragment and intact FGF23). Given that FGF23 can be ectopically expressed in differentiated renal tubules and iron modulates FGF23 metabolism, an effect of iron on FGF23 expression in renal tubules is conceivable but remains to be confirmed. More clinical and experimental studies are required to validate the use of circulating FGF23 as a biomarker for the early identification of AKI and prediction of short- and long-term adverse outcomes post-AKI. More importantly, the biologic effect of increased FGF23 in AKI needs to be defined.
Collapse
Affiliation(s)
- Javier A Neyra
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-885, USA
| | - Orson W Moe
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-885, USA
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-885, USA
| | - Ming Chang Hu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-885, USA.
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-885, USA.
| |
Collapse
|
135
|
Zelenchuk LV, Hedge AM, Rowe PSN. Age dependent regulation of bone-mass and renal function by the MEPE ASARM-motif. Bone 2015; 79:131-42. [PMID: 26051469 PMCID: PMC4501877 DOI: 10.1016/j.bone.2015.05.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 05/07/2015] [Accepted: 05/22/2015] [Indexed: 11/28/2022]
Abstract
CONTEXT Mice with null mutations in matrix extracellular phosphoglycoprotein (MEPE) have increased bone mass, increased trabecular density and abnormal cancellous bone (MN-mice). These defects worsen with age and MEPE overexpression induces opposite effects. Also, genome wide association studies show that MEPE plays a major role in bone mass. We hypothesized that the conserved C-terminal MEPE ASARM-motif is chiefly responsible for regulating bone mass and trabecular structure. DESIGN To test our theory we overexpressed C-terminal ASARM-peptide in MN-mice using the Col1α1 promoter (MNAt-mice). We then compared the bone and renal phenotypes of the MNAt-mouse with the MN-mouse and the X-linked hypophosphatemic rickets mouse (HYP). The HYP mouse overexpresses ASARM-peptides and is defective for the PHEX gene. RESULTS The MN-mouse developed increased bone mass, bone strength and trabecular abnormalities that worsened markedly with age. Defects in bone formation were chiefly responsible with suppressed sclerostin and increased active β-catenin. Increased uric acid levels also suggested that abnormalities in purine-metabolism and a reduced fractional excretion of uric acid signaled additional renal transport changes. The MN mouse developed a worsening hyperphosphatemia and reduced FGF23 with age. An increase in the fractional excretion of phosphate (FEP) despite the hyperphosphatemia confirms an imbalance in kidney-intestinal phosphate regulation. Also, the MN mice showed an increased creatinine clearance suggesting hyperfiltration. A reversal of the MN bone-renal phenotype changes occurred with the MNAt mice including the apparent hyperfiltration. The MNAt mice also developed localized hypomineralization, hypophosphatemia and increased FGF23. CONCLUSIONS The C-terminal ASARM-motif plays a major role in regulating bone-mass and cancellous structure as mice age. In healthy mice, the processing and release of free ASARM-peptide are chiefly responsible for preserving normal bone and renal function. Free ASARM-peptide also affects renal mineral phosphate handling by influencing FGF23 expression. These findings have implications for understanding age-dependent osteoporosis, unraveling drug-targets and developing treatments.
Collapse
Affiliation(s)
- Lesya V Zelenchuk
- The Kidney Institute, Kansas University Medical Center, Kansas City, KS, USA
| | - Anne-Marie Hedge
- The Kidney Institute, Kansas University Medical Center, Kansas City, KS, USA
| | - Peter S N Rowe
- The Kidney Institute, Kansas University Medical Center, Kansas City, KS, USA.
| |
Collapse
|
136
|
Georgianos PI, Sarafidis PA, Liakopoulos V. Arterial Stiffness: A Novel Risk Factor for Kidney Injury Progression? Am J Hypertens 2015; 28:958-65. [PMID: 25687879 DOI: 10.1093/ajh/hpv004] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 01/05/2015] [Indexed: 02/06/2023] Open
Abstract
Arterial stiffness is typical feature of vascular remodeling in chronic kidney disease (CKD). Increased arterial stiffness raises flow and pressure pulsatility and is considered the principle pathogenic mechanism of isolated systolic hypertension, left ventricular hypertrophy, and congestive heart failure. Apart from the impact of arterial stiffness on left ventricular afterload, downstream transmission of pressure pulsatility to the level of microcirculation is suggested to promote injury of other susceptible organs. This may be of particular importance for kidney injury progression, since passive renal perfusion along with low resistance and input impedance in renal microvessels make kidneys particularly vulnerable to the damaging effect of systemic pulsatile pressure. Recent studies have provided evidence that arterial stiffness culminates in elevated pulsatility and resistance in renal microvasculature, promoting structural damage of small intra-renal arterioles. Further, prospective observational studies have shown that reduced aortic compliance is closely associated with the annual rate of renal function decline and represents independent predictor of kidney injury progression to end-stage renal disease among patients with CKD. This article provides insights into the cross-talk between macrocirculation and renal microcirculation and summarizes the currently available clinical evidence linking increased arterial stiffness with kidney disease progression.
Collapse
Affiliation(s)
- Panagiotis I Georgianos
- Section of Nephrology and Hypertension, 1st Department of Medicine, AHEPA Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece;
| | - Pantelis A Sarafidis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Vassilios Liakopoulos
- Section of Nephrology and Hypertension, 1st Department of Medicine, AHEPA Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
137
|
Miyamura M, Fujita SI, Morita H, Sakane K, Okamoto Y, Sohmiya K, Hoshiga M, Ishizaka N. Circulating Fibroblast Growth Factor 23 Has a U-Shaped Association With Atrial Fibrillation Prevalence. Circ J 2015; 79:1742-8. [PMID: 26016926 DOI: 10.1253/circj.cj-15-0413] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) occurs more frequently among patients with renal dysfunction. We investigated the possible association between prevalence of AF and serum fibroblast growth factor 23 (FGF23), which has been shown to be increased in subjects with renal dysfunction. METHODS AND RESULTS Among the total enrollment of 851 cardiac patients, 188 patients had AF (paroxysmal AF, 95; non-paroxysmal AF, 93). Prevalence of AF for FGF23 octile had a U-shaped relationship with the lowest prevalence at the fifth octile. On logistic regression analysis, when the third FGF23 quartile was used as the reference, the first and fourth FGF23 quartiles were associated with prevalence of AF with an odds ratio (OR) of 3.34 (95% confidence interval [CI]: 1.89-5.88) and 2.58 (95% CI: 1.45-4.58), respectively, after adjusting for confounding factors including estimated glomerular filtration rate (eGFR). Among the subgroup of 416 patients for whom serum parathyroid hormone and 25-hydroxy vitamin D data were available, OR of the first and the fourth FGF23 quartile were calculated to be 3.52 and 2.97, respectively, when further adjusted for these two variables in the statistical model. CONCLUSIONS Serum FGF23 had a U-shaped relationship with prevalence of AF among Japanese cardiac patients, which was independent of other calcium/phosphate metabolism-related parameters and eGFR. Pathophysiology underlying the observed link, if at all, awaits further investigation.
Collapse
|
138
|
Mehta R, Ying GS, Houston S, Isakova T, Nessel L, Ojo A, Go A, Lash J, Kusek J, Grunwald J, Wolf M. Phosphate, fibroblast growth factor 23 and retinopathy in chronic kidney disease: the Chronic Renal Insufficiency Cohort Study. Nephrol Dial Transplant 2015; 30:1534-41. [PMID: 25910495 DOI: 10.1093/ndt/gfv123] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/30/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Elevated circulating concentrations of phosphate and fibroblast growth factor 23 (FGF23) contribute to the pathogenesis of cardiovascular disease in chronic kidney disease (CKD). Retinopathy is a common manifestation of microvascular disease in CKD, but its associations with phosphate and FGF23 have not been studied. We tested the hypothesis that higher serum phosphate is associated with more severe retinopathy in individuals with CKD, independent of FGF23 and known risk factors for retinopathy. METHODS We tested the associations of serum phosphate and plasma FGF23 with retinopathy in a cross-sectional analysis of 1800 participants in the Chronic Renal Insufficiency Cohort Study who underwent fundus photography. Retinopathy severity was graded according to the Early Treatment of Diabetic Retinopathy Severity score, and retinal venous and arterial diameters were measured. RESULTS Mean estimated glomerular filtration rate (eGFR) was 46.5 ± 15.4 mL/min/1.73 m(2), mean serum phosphate was 3.7 ± 0.6 mg/dl and median plasma C-terminal FGF23 was 133 RU/mL (interquartile range 87.2, 217.8 RU/mL). In multivariable ordinal logistic regression models, higher serum phosphate was associated with greater retinopathy severity independent of hypertension, diabetes, CKD severity and FGF23 [adjusted odds ratio of being in one higher category of retinopathy severity: 1.19 per 1 standard deviation increase; 95% confidence interval (CI) 1.05, 1.36; P = 0.007]. Presence of diabetes or hypertension did not modify the results. Higher serum phosphate was also independently associated with greater retinal venous diameter (multivariable-adjusted 1.70 µm increase per 1 standard deviation increase in phosphate; 95% CI 0.46, 2.93; P = 0.007). FGF23 levels were not independently associated with retinopathy severity or retinal venous diameter, and neither FGF23 nor phosphate was associated with retinal arterial diameter. CONCLUSIONS Among individuals with moderate-to-severe CKD, higher serum phosphate but not FGF23 was independently associated with more severe retinopathy and microvascular retinal venous dilatation.
Collapse
Affiliation(s)
- Rupal Mehta
- Division of Nephrology and Hypertension, Department of Medicine and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | | | - Tamara Isakova
- Division of Nephrology and Hypertension, Department of Medicine and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lisa Nessel
- University of Pennsylvania, Philadelphia, PA, USA
| | | | - Alan Go
- Kaiser Permanente, Oakland, CA, USA
| | - Jim Lash
- University of Illinois at Chicago, Chicago, IL, USA
| | - John Kusek
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | | | - Myles Wolf
- Division of Nephrology and Hypertension, Department of Medicine and Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | |
Collapse
|