1
|
Chen G, Chen L, Li X, Mohammadi M. FGF-based drug discovery: advances and challenges. Nat Rev Drug Discov 2025:10.1038/s41573-024-01125-w. [PMID: 39875570 DOI: 10.1038/s41573-024-01125-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2024] [Indexed: 01/30/2025]
Abstract
The fibroblast growth factor (FGF) family comprises 15 paracrine-acting and 3 endocrine-acting polypeptides, which govern a multitude of processes in human development, metabolism and tissue homeostasis. Therapeutic endocrine FGFs have recently advanced in clinical trials, with FGF19 and FGF21-based therapies on the cusp of approval for the treatment of primary sclerosing cholangitis and metabolic syndrome-associated steatohepatitis, respectively. By contrast, while paracrine FGFs were once thought to be promising drug candidates for wound healing, burns, tissue repair and ischaemic ailments based on their potent mitogenic and angiogenic properties, repeated failures in clinical trials have led to the widespread perception that the development of paracrine FGF-based drugs is not feasible. However, the observation that paracrine FGFs can exert FGF hormone-like metabolic activities has restored interest in these FGFs. The recent structural elucidation of the FGF cell surface signalling machinery and the formulation of a new threshold model for FGF signalling specificity have paved the way for therapeutically harnessing paracrine FGFs for the treatment of a range of metabolic diseases.
Collapse
Affiliation(s)
- Gaozhi Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lingfeng Chen
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Moosa Mohammadi
- Institute of Cell Growth Factor, Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health, Wenzhou, Zhejiang, China.
| |
Collapse
|
2
|
Rivoira MA, Peralta López ME, Areco V, Díaz de Barboza G, Dionisi MP, Tolosa de Talamoni N. Emerging concepts on the FGF23 regulation and activity. Mol Cell Biochem 2025; 480:75-89. [PMID: 38581553 DOI: 10.1007/s11010-024-04982-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/28/2024] [Indexed: 04/08/2024]
Abstract
Fibroblast growth factor 23 (FGF23) discovery has provided new insights into the regulation of Pi and Ca homeostasis. It is secreted by osteoblasts and osteocytes, and acts mainly in the kidney, parathyroid, heart, and bone. The aim of this review is to highlight the current knowledge on the factors modulating the synthesis of FGF23, the canonical and non-canonical signaling pathways of the hormone, the role of FGF23 in different pathophysiological conditions, and the anti-FGF23 therapy. This is a narrative review based on the search of PubMed database in the range of years 2000-2023 using the keywords local and systemic regulators of FGF23 synthesis, FGF23 receptors, canonical and non-canonical pathways, pathophysiological conditions and FGF23, and anti-FGF23 therapy, focusing the data on the molecular mechanisms. The regulation of FGF23 synthesis is complex and multifactorial. It is regulated by local factors and systemic regulators mainly involved in bone mineralization. The excessive FGF23 production is associated with different congenital diseases and with diseases occurring with a secondary high FGF23 production such as in chronic disease kidney and tumor-induced osteomalacia (TIO). The anti-FGF23 therapy appears to be useful to treat chromosome X-linked hypophosphatemia and TIO, but there are doubts about the handle of excessive FGF23 production in CKD. FGF23 biochemistry and pathophysiology are generating a plethora of knowledge to reduce FGF23 bioactivity at many levels that might be useful for future therapeutics of diseases associated with high-serum FGF23 levels.
Collapse
Affiliation(s)
- María Angélica Rivoira
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do, Piso, Ciudad Universitaria, 5000, Córdoba, Argentina
| | - María Elena Peralta López
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do, Piso, Ciudad Universitaria, 5000, Córdoba, Argentina
| | - Vanessa Areco
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do, Piso, Ciudad Universitaria, 5000, Córdoba, Argentina
- Instituto Multidisciplinario de Investigación y Transferencia Agroalimentaria y Biotecnológica (IMITAB, CONICET-UNVM), Córdoba, Argentina
| | - Gabriela Díaz de Barboza
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do, Piso, Ciudad Universitaria, 5000, Córdoba, Argentina
| | - María Paula Dionisi
- Cátedra de Clínica Médica II - UHMI Nº 2, Hospital San Roque, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nori Tolosa de Talamoni
- Laboratorio "Dr. Fernando Cañas", Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Pabellón Argentina, 2do, Piso, Ciudad Universitaria, 5000, Córdoba, Argentina.
| |
Collapse
|
3
|
Nakagawa Y, Komaba H. Roles of Parathyroid Hormone and Fibroblast Growth Factor 23 in Advanced Chronic Kidney Disease. Endocrinol Metab (Seoul) 2024; 39:407-415. [PMID: 38752265 PMCID: PMC11220210 DOI: 10.3803/enm.2024.1978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 03/26/2024] [Accepted: 03/29/2024] [Indexed: 06/29/2024] Open
Abstract
Parathyroid hormone (PTH) and fibroblast growth factor 23 (FGF23) each play a central role in the pathogenesis of chronic kidney disease (CKD)-mineral and bone disorder. Levels of both hormones increase progressively in advanced CKD and can lead to damage in multiple organs. Secondary hyperparathyroidism (SHPT), characterized by parathyroid hyperplasia with increased PTH secretion, is associated with fractures and mortality. Emerging evidence suggests that these associations may be partially explained by PTH-induced browning of adipose tissue and increased energy expenditure. Observational studies suggest a survival benefit of PTHlowering therapy, and a recent study comparing parathyroidectomy and calcimimetics further suggests the importance of intensive PTH control. The mechanisms underlying the regulation of FGF23 secretion by osteocytes in response to phosphate load have been unclear, but recent experimental studies have identified glycerol-3-phosphate, a byproduct of glycolysis released by the kidney, as a key regulator of FGF23 production. Elevated FGF23 levels have been shown to be associated with mortality, and experimental data suggest off-target adverse effects of FGF23. However, the causal role of FGF23 in adverse outcomes in CKD patients remains to be established. Further studies are needed to determine whether intensive SHPT control improves clinical outcomes and whether treatment targeting FGF23 can improve patient outcomes.
Collapse
Affiliation(s)
- Yosuke Nakagawa
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan
| | - Hirotaka Komaba
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan
- Interactive Translational Research Center for Kidney Diseases, Tokai University School of Medicine, Isehara, Japan
- The Institute of Medical Sciences, Tokai University, Isehara, Japan
| |
Collapse
|
4
|
Martínez-Heredia L, Canelo-Moreno JM, García-Fontana B, Muñoz-Torres M. Non-Classical Effects of FGF23: Molecular and Clinical Features. Int J Mol Sci 2024; 25:4875. [PMID: 38732094 PMCID: PMC11084844 DOI: 10.3390/ijms25094875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/21/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
This article reviews the role of fibroblast growth factor 23 (FGF23) protein in phosphate metabolism, highlighting its regulation of vitamin D, parathyroid hormone, and bone metabolism. Although it was traditionally thought that phosphate-calcium homeostasis was controlled exclusively by parathyroid hormone (PTH) and calcitriol, pathophysiological studies revealed the influence of FGF23. This protein, expressed mainly in bone, inhibits the renal reabsorption of phosphate and calcitriol formation, mediated by the α-klotho co-receptor. In addition to its role in phosphate metabolism, FGF23 exhibits pleiotropic effects in non-renal systems such as the cardiovascular, immune, and metabolic systems, including the regulation of gene expression and cardiac fibrosis. Although it has been proposed as a biomarker and therapeutic target, the inhibition of FGF23 poses challenges due to its potential side effects. However, the approval of drugs such as burosumab represents a milestone in the treatment of FGF23-related diseases.
Collapse
Affiliation(s)
- Luis Martínez-Heredia
- Instituto de Investigación Biosanitaria de Granada, 18014 Granada, Spain;
- Biomedical Research Network in Fragility and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | | | - Beatriz García-Fontana
- Instituto de Investigación Biosanitaria de Granada, 18014 Granada, Spain;
- Biomedical Research Network in Fragility and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- Department of Cell Biology, University of Granada, 18016 Granada, Spain
| | - Manuel Muñoz-Torres
- Instituto de Investigación Biosanitaria de Granada, 18014 Granada, Spain;
- Biomedical Research Network in Fragility and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- Department of Medicine, University of Granada, 18016 Granada, Spain
| |
Collapse
|
5
|
Ogata H, Sugawara H, Yamamoto M, Ito H. Phosphate and Coronary Artery Disease in Patients with Chronic Kidney Disease. J Atheroscler Thromb 2024; 31:1-14. [PMID: 37766573 PMCID: PMC10776333 DOI: 10.5551/jat.rv22012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/07/2023] [Indexed: 09/29/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in patients with chronic kidney disease (CKD). Both traditional and CKD-related factors are associated with CVD in CKD patients. Traditional factors that play an important role in the atherosclerotic process directly contribute to a higher risk of coronary artery disease in patients with early-stage CKD. Among CKD-related factors, CKD-mineral and bone disorder plays a critical role in the pathomechanism of nonatherosclerotic diseases, which increases the risk of cardiovascular morbidity and mortality in patients with advanced CKD. Higher serum phosphate levels were significantly associated with cardiovascular events and all-cause mortality in patients with or without CKD. An increased phosphate load, directly and indirectly, promotes arterial medial calcification and left ventricular hypertrophy, both of which predispose patients to coronary artery disease. Calciprotein particles that form in a hyperphosphatemic state promote the transformation of vascular smooth muscle cells (VSMCs) into osteoblastic cells, thereby providing a scaffold for medial calcification in the artery. Increases in fibroblast growth factor-23 and disturbed vitamin D metabolism induced by an excessive phosphate load play a significant role in the development of cardiomyocyte hypertrophy and cardiac fibrosis. Recently, hyperphosphatemia was reported to promote de novo cholesterol synthesis in VSMCs and macrophages, which is likely to contribute to statin resistance in patients with end-stage kidney disease. This review outlines the association between increased phosphate load and coronary artery disease in patients with CKD.
Collapse
Affiliation(s)
- Hiroaki Ogata
- Division of Nephrology, Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
- Department of Medical Education, Showa University School of Medicine, Tokyo, Japan
| | - Hirohito Sugawara
- Division of Nephrology, Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
| | - Masahiro Yamamoto
- Division of Nephrology, Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
| | - Hidetoshi Ito
- Division of Nephrology, Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
| |
Collapse
|
6
|
Watanabe K, Fujii H, Okamoto K, Kono K, Goto S, Nishi S. Exploring the implications of blocking renin-angiotensin-aldosterone system and fibroblast growth factor 23 in early left ventricular hypertrophy without chronic kidney disease. Front Endocrinol (Lausanne) 2023; 14:1276664. [PMID: 38174329 PMCID: PMC10762797 DOI: 10.3389/fendo.2023.1276664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Background Whether fibroblast growth factor 23 (FGF23) directly induces left ventricular hypertrophy (LVH) remains controversial. Recent studies showed an association between FGF23 and the renin-angiotensin-aldosterone system (RAAS). The aim of this study was to investigate changes in FGF23 levels and RAAS parameters and their influences on LVH. Methods In the first experiment, male C57BL/6J mice were divided into sham and transverse aortic constriction (TAC) groups. The TAC group underwent TAC at 8 weeks of age. At 1, 2, 3, and 4 weeks after TAC, the mice were sacrificed, and blood and urine samples were obtained. Cardiac expressions of FGF23 and RAAS-related factors were evaluated, and cardiac histological analyses were performed. In the second experiment, the sham and TAC groups were treated with vehicle, angiotensin-converting enzyme (ACE) inhibitor, or FGF receptor 4 (FGFR4) inhibitor and then evaluated in the same way as in the first experiment. Results In the early stage of LVH without chronic kidney disease, serum FGF23 levels did not change but cardiac FGF23 expression significantly increased along with LVH progression. Moreover, serum aldosterone and cardiac ACE levels were significantly elevated, and cardiac ACE2 levels were significantly decreased. ACE inhibitor did not change serum FGF23 levels but significantly decreased cardiac FGF23 levels with improvements in LVH and RAAS-related factors, while FGFR4 inhibitor did not change the values. Conclusions Not serum FGF23 but cardiac FGF23 levels and RAAS parameters significantly changed in the early stage of LVH without chronic kidney disease. RAAS blockade might be more crucial than FGF23 blockade for preventing LVH progression in this condition.
Collapse
Affiliation(s)
| | - Hideki Fujii
- Division of Nephrology and Kidney Center, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | | | | | | |
Collapse
|
7
|
Michon-Colin A, Metzger M, Bankir L, Gauci C, Brunel M, Baron S, Prot-Bertoye C, Stengel B, Thervet E, Haymann JP, Boffa JJ, Vrtovsnik F, Flamant M, Houillier P, Prie D, Courbebaisse M. Fibroblast growth factor 23 but not copeptin is independently associated with kidney failure and mortality in patients with chronic kidney disease. Clin Kidney J 2023; 16:2472-2481. [PMID: 38046034 PMCID: PMC10689138 DOI: 10.1093/ckj/sfad149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Indexed: 12/05/2023] Open
Abstract
Background Copeptin and intact fibroblast growth factor 23 (iFGF23) increase early during chronic kidney disease (CKD) and may be predictive of unfavourable outcomes. The aim of this study was to evaluate their respective associations with renal and vital outcomes in CKD patients. Methods We included CKD patients from the NephroTest cohort with concomitant measurements of plasma copeptin and iFGF23 concentrations and isotopic glomerular filtration rate measurement (mGFR). The primary endpoint was a composite outcome including kidney failure (KF) (dialysis initiation, pre-emptive transplantation or a 57% decrease of mGFR, corresponding to doubling of serum creatinine) or death before KF. Hazard ratios (HRs) of the primary endpoint associated with log-transformed copeptin and iFGF23 concentrations were estimated by Cox models. The slope of mGFR over time was analysed using a linear mixed model. Results A total of 329 CKD patients (243 men, mean age 60.3 ± 14.6 years) were included. Among them, 301 with an mGFR >15 ml/min/1.73 m2 were included in survival and mGFR slope analyses. During a median follow-up of 4.61 years (quartile 1-quartile 3: 3.72-6.07), 61 KFs and 32 deaths occurred. Baseline iFGF23 concentrations were associated with the composite outcome after multiple adjustments {HR 2.72 [95% confidence interval (CI) 1.85-3.99]}, whereas copeptin concentrations were not [HR 1.01 (95% CI 0.74-1.39)]. Neither copeptin nor iFGF23 were associated with mGFR slope over time. Conclusion Our study shows for the first time in population of CKD patients an independent association between iFGF23 and unfavourable renal and vital outcomes and shows no such association regarding copeptin, encouraging the integration of iFGF23 measurement into the follow-up of CKD.
Collapse
Affiliation(s)
- Arthur Michon-Colin
- Université Paris Cité, Paris, France
- Explorations Fonctionnelles Rénales – Physiologie, Hôpital Européen Georges-Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
- Centre de Référence des Maladies Rénales Héréditaires de l’Enfant et de l’Adulte, Paris, France
- Centre de Référence des Maladies Rares du Calcium et du Phosphate, Paris, France
| | - Marie Metzger
- INSERM UMRS 1018, Equipe d'Epidémiologie Clinique, CESP, Université Paris-Saclay, Villejuif, France
| | - Lise Bankir
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Paris, France
- CNRS, ERL 8228, Laboratory of Kidney Physiology and Tubulopathies, Paris, France
| | - Cédric Gauci
- Explorations Fonctionnelles Rénales – Physiologie, Hôpital Européen Georges-Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
- INSERM UMRS 1018, Equipe d'Epidémiologie Clinique, CESP, Université Paris-Saclay, Villejuif, France
| | - Mélanie Brunel
- Université Paris Cité, Paris, France
- Explorations Fonctionnelles Rénales – Physiologie, Hôpital Européen Georges-Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
- Centre de Référence des Maladies Rénales Héréditaires de l’Enfant et de l’Adulte, Paris, France
- Centre de Référence des Maladies Rares du Calcium et du Phosphate, Paris, France
| | - Stéphanie Baron
- Université Paris Cité, Paris, France
- Explorations Fonctionnelles Rénales – Physiologie, Hôpital Européen Georges-Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Paris, France
- Centre de Référence des Maladies Rénales Héréditaires de l’Enfant et de l’Adulte, Paris, France
- Centre de Référence des Maladies Rares du Calcium et du Phosphate, Paris, France
| | - Caroline Prot-Bertoye
- Explorations Fonctionnelles Rénales – Physiologie, Hôpital Européen Georges-Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Paris, France
- CNRS, ERL 8228, Laboratory of Kidney Physiology and Tubulopathies, Paris, France
- Centre de Référence des Maladies Rénales Héréditaires de l’Enfant et de l’Adulte, Paris, France
- Centre de Référence des Maladies Rares du Calcium et du Phosphate, Paris, France
| | - Bénédicte Stengel
- INSERM UMRS 1018, Equipe d'Epidémiologie Clinique, CESP, Université Paris-Saclay, Villejuif, France
| | - Eric Thervet
- Université Paris Cité, Paris, France
- Néphrologie et Hémodialyse, Hôpital Européen Georges-Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Jean-Philippe Haymann
- Explorations Fonctionnelles Multidisciplinaires, Sorbonne Université Paris, France
- Explorations Fonctionnelles Multidisciplinaires, Hôpital Tenon, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Jean-Jacques Boffa
- Explorations Fonctionnelles Multidisciplinaires, Sorbonne Université Paris, France
- Néphrologie et Dialyse, Hôpital Tenon, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - François Vrtovsnik
- Université Paris Cité, Paris, France
- Néphrologie, Hôpital Bichat, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Martin Flamant
- Université Paris Cité, Paris, France
- Explorations Fonctionnelles Multidisciplinaires, Hôpital Bichat, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Pascal Houillier
- Université Paris Cité, Paris, France
- Explorations Fonctionnelles Rénales – Physiologie, Hôpital Européen Georges-Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Paris, France
- Centre de Référence des Maladies Rénales Héréditaires de l’Enfant et de l’Adulte, Paris, France
- Centre de Référence des Maladies Rares du Calcium et du Phosphate, Paris, France
| | - Dominique Prie
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Département de Physiologie, Hôpital Necker, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Marie Courbebaisse
- Université Paris Cité, Paris, France
- Explorations Fonctionnelles Rénales – Physiologie, Hôpital Européen Georges-Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Centre de Référence des Maladies Rénales Héréditaires de l’Enfant et de l’Adulte, Paris, France
- Centre de Référence des Maladies Rares du Calcium et du Phosphate, Paris, France
| |
Collapse
|
8
|
Komaba H. Roles of PTH and FGF23 in kidney failure: a focus on nonclassical effects. Clin Exp Nephrol 2023; 27:395-401. [PMID: 36977891 PMCID: PMC10104924 DOI: 10.1007/s10157-023-02336-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/27/2023] [Indexed: 03/30/2023]
Abstract
Parathyroid hormone (PTH) and fibroblast growth factor 23 (FGF23) each play a central role in the pathogenesis of chronic kidney disease-mineral and bone disorder (CKD-MBD). Both hormones increase as kidney function declines, presumably as a response to maintain normal phosphate balance, but when patients reach kidney failure, PTH and FGF23 fail to exert their phosphaturic effects, leading to hyperphosphatemia and further elevations in PTH and FGF23. In patients with kidney failure, the major target organ for PTH is the bone, but elevated PTH is also associated with mortality presumably through skeletal and nonskeletal mechanisms. Indeed, accumulated evidence suggests improved survival with PTH-lowering therapies, and a more recent study comparing parathyroidectomy and calcimimetic treatment further suggests a notion of "the lower, the better" for PTH control. Emerging data suggest that the link between SHPT and mortality could in part be explained by the action of PTH to induce adipose tissue browning and wasting. In the absence of a functioning kidney, the classical target organ for FGF23 is the parathyroid gland, but FGF23 loses its hormonal effect to suppress PTH secretion owing to the depressed expression of parathyroid Klotho. In this setting, experimental data suggest that FGF23 exerts adverse nontarget effects, but it remains to be confirmed whether FGF23 directly contributes to multiple organ injury in patients with kidney failure and whether targeting FGF23 can improve patient outcomes. Further efforts should be made to determine whether intensive control of SHPT improves clinical outcomes and whether nephrologists should aim at controlling FGF23 levels just as with PTH levels.
Collapse
Affiliation(s)
- Hirotaka Komaba
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, 143 Shimo-Kasuya, Isehara, 259-1193, Japan.
- Interactive Translational Research Center for Kidney Diseases, Tokai University School of Medicine, Isehara, Japan.
- The Institute of Medical Sciences, Tokai University, Isehara, Japan.
| |
Collapse
|
9
|
Nakano T, Kishimoto H, Tokumoto M. Direct and indirect effects of fibroblast growth factor 23 on the heart. Front Endocrinol (Lausanne) 2023; 14:1059179. [PMID: 36909314 PMCID: PMC9999118 DOI: 10.3389/fendo.2023.1059179] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 01/30/2023] [Indexed: 03/14/2023] Open
Abstract
Fibroblast growth factor (FGF)23 is a bone-derived phosphotropic hormone that regulates phosphate and mineral homeostasis. Recent studies have provided evidence that a high plasma concentration of FGF23 is associated with cardiac disease, including left ventricular hypertrophy (LVH), heart failure, atrial fibrillation, and cardiac death. Experimental studies have shown that FGF23 activates fibroblast growth factor receptor 4 (FGFR4)/phospholipase Cγ/calcineurin/nuclear factor of activated T-cells signaling in cardiomyocytes and induces cardiac hypertrophy in rodents. Activation of FGFR4 by FGF23 normally requires the co-receptor α-klotho, and klotho-independent signaling occurs only under conditions characterized by extremely high FGF23 concentrations. Recent studies have demonstrated that FGF23 activates the renin-angiotensin-aldosterone system (RAAS) and induces LVH, at least in part as a result of lower vitamin D activation. Moreover, crosstalk between FGF23 and RAAS results in the induction of cardiac hypertrophy and fibrosis. In this review, we summarize the results of studies regarding the relationships between FGF23 and cardiac events, and describe the potential direct and indirect mechanisms whereby FGF23 induces LVH.
Collapse
Affiliation(s)
- Toshiaki Nakano
- Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- *Correspondence: Toshiaki Nakano,
| | - Hiroshi Kishimoto
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masanori Tokumoto
- Department of Nephrology, Fukuoka Red Cross Hospital, Fukuoka, Japan
| |
Collapse
|
10
|
Clemmer JS, Shafi T, Obi Y. Physiological Mechanisms of Hypertension and Cardiovascular Disease in End-Stage Kidney Disease. Curr Hypertens Rep 2022; 24:413-424. [PMID: 35708820 PMCID: PMC10041674 DOI: 10.1007/s11906-022-01203-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE OF REVIEW In this article, we summarize recent advances in understanding hypertension and cardiovascular disease in patients with end-stage kidney disease. RECENT FINDINGS Factors such as anemia, valvular and vascular calcification, vasoconstrictors, uremic toxins, hypoglycemia, carbamylated proteins, oxidative stress, and inflammation have all been associated with the progression of cardiovascular disease in end-stage kidney disease but the causality of these mechanisms has not been proven. The high risk of cardiovascular mortality has not improved as in the general population despite many advancements in cardiovascular care over the last two decades. Mechanisms that increase hypertension risk in these patients are centered on the control of extracellular fluid volume; however, over-correction of volume with dialysis can increase risks of intradialytic hypotension and death in these patients. This review presents both recent and classic work that increases our understanding of hypertension and cardiovascular disease in end-stage kidney disease.
Collapse
Affiliation(s)
- John S Clemmer
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Tariq Shafi
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA.,Division of Nephrology, University of Mississippi Medical Center, 2500 North State Street, Suite L-504, Jackson, MS, 39216, USA
| | - Yoshitsugu Obi
- Division of Nephrology, University of Mississippi Medical Center, 2500 North State Street, Suite L-504, Jackson, MS, 39216, USA.
| |
Collapse
|
11
|
Verbueken D, Moe OW. Strategies to lower fibroblast growth factor 23 bioactivity. Nephrol Dial Transplant 2022; 37:1800-1807. [PMID: 33502502 PMCID: PMC9494132 DOI: 10.1093/ndt/gfab012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Indexed: 12/26/2022] Open
Abstract
Fibroblast growth factor 23 (FGF23) is a circulating hormone derived from the bone whose release is controlled by many factors and exerts a multitude of systemic actions. There are congenital and acquired disorders of increased and decreased FGF23 levels. In chronic kidney disease (CKD), elevations of FGF23 levels can be 1000-fold above the upper physiological limit. It is still debated whether this high FGF23 in CKD is a biomarker or causally related to morbidity and mortality. Data from human association studies support pathogenicity, while experimental data are less robust. Knowledge of the biology and pathobiology of FGF23 has generated a plethora of means to reduce FGF23 bioactivity at many levels that will be useful for therapeutic translations. This article summarizes these approaches and addresses several critical questions that still need to be answered.
Collapse
Affiliation(s)
- Devin Verbueken
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | | |
Collapse
|
12
|
Association between Anti-Erythropoietin Receptor Antibodies and Cardiac Function in Patients on Hemodialysis: A Multicenter Cross-Sectional Study. Biomedicines 2022; 10:biomedicines10092092. [PMID: 36140193 PMCID: PMC9495431 DOI: 10.3390/biomedicines10092092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiac dysfunction is an important prognostic predictor of cardiovascular mortality in patients on hemodialysis (HD). Erythropoietin (EPO) has been reported to improve cardiac function by binding to the EPO receptor (EPOR) on cardiomyocytes. This study investigated whether anti-EPOR antibodies were associated with left ventricular cardiac function in patients undergoing HD. This multicenter, cross-sectional observational study included 377 patients (median age, 70 years; 267 (70.8%) males) with chronic kidney disease (CKD) undergoing stable maintenance HD. Serum levels of anti-EPOR antibodies were measured, and echocardiography was used to assess the left ventricular mass index (LVMI) and left ventricular ejection fraction (LVEF). Anti-EPOR antibodies were found in 17 patients (4.5%). LVMI was greater (median of 135 g/m2 vs. 115 g/m2, p = 0.042), and the prevalence of LVEF < 50% was higher (35.3% vs. 15.6%, p = 0.032) in patients with anti-EPOR antibodies than in those without. Multivariable linear regression and logistic regression analysis (after adjusting for known risk factors of heart failure) revealed that anti-EPOR antibodies were independently associated with LVMI (coefficient 16.2%; 95% confidence interval (CI) 1.0−35.0%, p = 0.043) and LVEF <50% (odds ratio 3.20; 95% CI 1.05−9.73, p = 0.041). Thus, anti-EPOR antibody positivity was associated with left ventricular dysfunction in patients undergoing HD.
Collapse
|
13
|
Ishigami J, Honda Y, Karger AB, Coresh J, Selvin E, Lutsey PL, Matsushita K. Changes in Serum Intact Fibroblast Growth Factor 23 Concentrations From Midlife to Late Life and Their Predictors in the Community: The ARIC Study. Mayo Clin Proc Innov Qual Outcomes 2022; 6:209-217. [PMID: 35517245 PMCID: PMC9062741 DOI: 10.1016/j.mayocpiqo.2022.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Objective To investigate longitudinal changes in the blood concentration of fibroblast growth factor 23 (FGF23) from midlife to late life and their major predictors in the general population. Patients and Methods In 14,444 participants of the Atherosclerosis Risk in Communities Study, we analyzed the association of 31,095 measurements of serum intact FGF23 with age using data from 3 visits (visit 2 [N=13,460; mean age, 57 years]; visit 3 [N=12,323; mean age, 60 years]; and visit 5 [N=6122; mean age, 76 years]) and a linear mixed-effects model. Among 5804 participants who had FGF23 measurements at both visits 3 and 5, we explored predictors of FGF23 change from midlife to late life using linear regression models. Prespecified risk factors were estimated glomerular filtration rate, body mass index, ever smoking, ever drinker, diabetes, hypertension, history of cardiovascular disease, total cholesterol, and high-density lipoprotein cholesterol. Results Median FGF23 concentrations were 41.9 pg/mL (interquartile interval [IQI], 33.9 to 51.8 pg/mL) at visit 2, 38.3 pg/mL (IQI, 30.6 to 48.3 pg/mL) at visit 3, and 55.0 pg/mL (IQI, 44.4 to 70.3 pg/mL) at visit 5. A linear mixed-effects model showed that the association of FGF23 with age was nonlinear, with a slight decline or no change in age 45-60 years and a monotonic increase in age greater than or equal to 65 years (FGF23, +10 to 15 pg/mL per 10 years of age). In a multivariable linear regression model, significantly greater increases in FGF23 were noted, with midlife estimated glomerular filtration rate less than 60 mL/min per 1.73 m2 vs more than or equal to 60 mL/min per 1.73 m2 (ΔFGF23, +4.4 pg/mL [95% CI, 0.9 to 8.0]), diabetes vs no diabetes (ΔFGF23, +6.2 pg/mL [95% CI, 4.1 to 8.3]), and hypertension vs no hypertension (ΔFGF23, +4.1 pg/mL [95% CI, 2.7 to 5.4]). Conclusion FGF23 did not show any major changes in midlife but increased linearly in late life. Reduced kidney function, diabetes, and hypertension were robustly associated with a greater increase in FGF23. Further investigations are needed to understand the potential mechanisms linking these conditions to an increase in FGF23 concentrations.
Collapse
Affiliation(s)
- Junichi Ishigami
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Yasuyuki Honda
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Amy B. Karger
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Pamela L. Lutsey
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
14
|
Vervloet MG. Shedding Light on the Complex Regulation of FGF23. Metabolites 2022; 12:metabo12050401. [PMID: 35629904 PMCID: PMC9147863 DOI: 10.3390/metabo12050401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/23/2022] [Accepted: 04/26/2022] [Indexed: 12/10/2022] Open
Abstract
Early research has suggested a rather straightforward relation between phosphate exposure, increased serum FGF23 (Fibroblast Growth Factor 23) concentrations and clinical endpoints. Unsurprisingly, however, subsequent studies have revealed a much more complex interplay between autocrine and paracrine factors locally in bone like PHEX and DMP1, concentrations of minerals in particular calcium and phosphate, calciprotein particles, and endocrine systems like parathyroid hormone PTH and the vitamin D system. In addition to these physiological regulators, an expanding list of disease states are shown to influence FGF23 levels, usually increasing it, and as such increase the burden of disease. While some of these physiological or pathological factors, like inflammatory cytokines, may partially confound the association of FGF23 and clinical endpoints, others are in the same causal path, are targetable and hence hold the promise of future treatment options to alleviate FGF23-driven toxicity, for instance in chronic kidney disease, the FGF23-associated disease with the highest prevalence by far. These factors will be reviewed here and their relative importance described, thereby possibly opening potential means for future therapeutic strategies.
Collapse
Affiliation(s)
- Marc G. Vervloet
- Amsterdam UMC, Location Vrije Universiteit Amsterdam, Nephrology, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; ; Tel.: +31-20-4442671
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
15
|
Charoenphandhu N, Sooksawanwit S, Aeimlapa R, Thonapan N, Upanan P, Adulyaritthikul P, Krungchanuchat S, Panupinthu N, Teerapornpuntakit J, Rojviriya C, Lertsuwan K, Svasti S, Wongdee K. Mild-intensity physical activity prevents cardiac and osseous iron deposition without affecting bone mechanical property or porosity in thalassemic mice. Sci Rep 2022; 12:5959. [PMID: 35396390 PMCID: PMC8993875 DOI: 10.1038/s41598-022-09997-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 03/30/2022] [Indexed: 02/07/2023] Open
Abstract
Thalassemia causes anemia, ineffective erythropoiesis, bone loss and iron accumulation in several tissues, e.g., liver, bone and heart, the last of which leads to lethal cardiomyopathy and arrhythmia. Although exercise reportedly improves bone density in thalassemic mice, exercise performance is compromised and might pose risk of cardiovascular accident in thalassemic patients. Therefore, we sought to explore whether mild-intensity physical activity (MPA) with 30–50% of maximal oxygen consumption was sufficient to benefit the heart and bone. Herein, male hemizygous β-globin knockout (BKO) mice and wild-type littermates were subjected to voluntary wheel running 1 h/day, 5 days/week for 3 months (MPA group) or kept sedentary (SDN; control). As determined by atomic absorption spectroscopy, BKO-MPA mice had less iron accumulation in heart and bone tissues compared with BKO-SDN mice. Meanwhile, the circulating level of fibroblast growth factor-23—a factor known to reduce serum iron and intestinal calcium absorption—was increased early in young BKO-MPA mice. Nevertheless, MPA did not affect duodenal calcium transport or body calcium retention. Although MPA restored the aberrant bone calcium-phosphorus ratio to normal range, it did not change vertebral calcium content or femoral mechanical properties. Microstructural porosity in tibia of BKO-MPA mice remained unaltered as determined by synchrotron radiation X-ray tomographic microscopy. In conclusion, MPA prevents cardiac and bone iron accumulation, which is beneficial to thalassemic patients with limited physical fitness or deteriorated cardiac performance. However, in contrast to moderate-intensity exercise, MPA does not improve bone mechanical properties or reduce bone porosity.
Collapse
Affiliation(s)
- Narattaphol Charoenphandhu
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.,Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.,Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, 73170, Thailand.,The Academy of Science, The Royal Society of Thailand, Bangkok, 10300, Thailand
| | - Supagarn Sooksawanwit
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.,Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Ratchaneevan Aeimlapa
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.,Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Natchayaporn Thonapan
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.,Molecular Medicine Graduate Program, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Pornpailin Upanan
- Faculty of Allied Health Sciences, Burapha University, Long-Hard Bangsaen Road, Chonburi, 20131, Thailand
| | - Punyanuch Adulyaritthikul
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.,Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Saowalak Krungchanuchat
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.,Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Nattapon Panupinthu
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.,Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Jarinthorn Teerapornpuntakit
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.,Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand
| | - Catleya Rojviriya
- Synchrotron Light Research Institute (Public Organization), Nakhon Ratchasima, 30000, Thailand
| | - Kornkamon Lertsuwan
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.,Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Saovaros Svasti
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Kannikar Wongdee
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand. .,Faculty of Allied Health Sciences, Burapha University, Long-Hard Bangsaen Road, Chonburi, 20131, Thailand.
| |
Collapse
|
16
|
Okamoto K, Fujii H, Watanabe K, Goto S, Kono K, Nishi S. Changes of FGF23 and the Renin-Angiotensin-System in Male Mouse Models of Chronic Kidney Disease and Cardiac Hypertrophy. J Endocr Soc 2022; 6:bvab187. [PMID: 35047715 PMCID: PMC8758403 DOI: 10.1210/jendso/bvab187] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Indexed: 11/19/2022] Open
Abstract
Serum fibroblast growth factor 23 (FGF23) levels and the renin-angiotensin-aldosterone system (RAAS) are elevated in chronic kidney disease (CKD) patients, and their association with left ventricular hypertrophy (LVH) has been reported. However, whether the FGF23 elevation is the cause or result of LVH remains unclear. At 10 weeks, male C57BL/6J mice were divided into 4 groups: sham, CKD (5/6 nephrectomy), LVH (transaortic constriction), and CKD/LVH group. At 16 weeks, the mice were euthanized, and blood and urine, cardiac expressions of FGF23 and RAAS-related factors, and cardiac histological analyses were performed. Heart weight, serum FGF23 levels, and cardiac expression of FGF23 and RAAS-related factors, except for angiotensin-converting enzyme 2, were more increased in the CKD/LVH group compared to the other groups. A significant correlation between LVH and cardiac expressions of FGF23 and RAAS-related factors was observed. Furthermore, there was a significantly close correlation of the cardiac expression of FGF23 with LVH and RAAS-related factors. The coexisting CKD and LVH increased serum and cardiac FGF23 and RAAS-related factors, and there was a significant correlation between them. A close correlation of cardiac, but not serum FGF23, with LVH and RAAS suggests that local FGF23 levels may be associated with LVH and RAAS activation.
Collapse
Affiliation(s)
- Kohei Okamoto
- Division of Nephrology and Kidney Center, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Hideki Fujii
- Division of Nephrology and Kidney Center, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Kentaro Watanabe
- Division of Nephrology and Kidney Center, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Shunsuke Goto
- Division of Nephrology and Kidney Center, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Keiji Kono
- Division of Nephrology and Kidney Center, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Shinichi Nishi
- Division of Nephrology and Kidney Center, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| |
Collapse
|
17
|
Eitner F, Richter B, Schwänen S, Szaroszyk M, Vogt I, Grund A, Thum T, Heineke J, Haffner D, Leifheit-Nestler M. Comprehensive Expression Analysis of Cardiac Fibroblast Growth Factor 23 in Health and Pressure-induced Cardiac Hypertrophy. Front Cell Dev Biol 2022; 9:791479. [PMID: 35118076 PMCID: PMC8804498 DOI: 10.3389/fcell.2021.791479] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/13/2021] [Indexed: 12/18/2022] Open
Abstract
Enhanced fibroblast growth factor 23 (FGF23) is associated with left ventricular hypertrophy (LVH) in patients with chronic kidney and heart disease. Experimentally, FGF23 directly induces cardiac hypertrophy and vice versa cardiac hypertrophy stimulates FGF23. Besides the bone, FGF23 is expressed by cardiac myocytes, whereas its synthesis in other cardiac cell types and its paracrine role in the heart in health and disease is unknown. By co-immunofluorescence staining of heart tissue of wild-type mice, we show that Fgf23 is expressed by cardiac myocytes, fibroblasts and endothelial cells. Cardiac Fgf23 mRNA and protein level increases from neonatal to six months of age, whereas no age-related changes in bone Fgf23 mRNA expression were noted. Cardiac myocyte-specific disruption of Fgf23 using Cre-LoxP system (Fgf23fl/fl/cre+) caused enhanced mortality, but no differences in cardiac function or structure. Although pressure overload-induced cardiac hypertrophy induced by transverse aortic constriction (TAC) resulted in a slightly worse phenotype with a more severe reduced ejection fraction, higher end-systolic volume and more enlarged systolic LV diameter in Fgf23fl/fl/cre+ mice compared to controls, this was not translated to any worse cellular hypertrophy, fibrosis or chamber remodeling. TAC induced Fgf23 mRNA expression in whole cardiac tissue in both genotypes. Interestingly, co-immunofluorescence staining revealed enhanced Fgf23 synthesis in cardiac fibroblasts and endothelial cells but not in cardiac myocytes. RNA sequencing of isolated adult cardiac myocytes, cardiac fibroblasts and endothelial cells confirmed significantly higher Fgf23 transcription in cardiac fibroblasts and endothelial cells after TAC. Our data indicate that Fgf23 is physiologically expressed in various cardiac cell types and that cardiac fibroblasts and endothelial cells might be an important source of FGF23 in pathological conditions. In addition, investigations in Fgf23fl/fl/cre+ mice suggest that cardiac myocyte-derived FGF23 is needed to maintain cardiac function during pressure overload.
Collapse
Affiliation(s)
- Fiona Eitner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Hannover, Germany
| | - Beatrice Richter
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Hannover, Germany
| | - Saskia Schwänen
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Hannover, Germany
| | - Malgorzata Szaroszyk
- Department for Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Isabel Vogt
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Hannover, Germany
| | - Andrea Grund
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Joerg Heineke
- Department for Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Hannover, Germany
| | - Maren Leifheit-Nestler
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Hannover, Germany
- *Correspondence: Maren Leifheit-Nestler,
| |
Collapse
|
18
|
Shan SK, Yan T, Yuan LQ. Editorial: The effects of circulating non-sex hormones in cardiovascular disease. Front Endocrinol (Lausanne) 2022; 13:1065155. [PMID: 36605935 PMCID: PMC9808377 DOI: 10.3389/fendo.2022.1065155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Su-Kang Shan
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Tong Yan
- Center for Obesity and Metabolic Health, Department of General Surgery, The Third People’s Hospital of Chengdu and the Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
- Center for Diabetes Care, Pangang Group Chengdu Hospital, Chengdu, Sichuan, China
| | - Ling-Qing Yuan
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Ling-Qing Yuan,
| |
Collapse
|
19
|
Navarro-García JA, González-Lafuente L, Fernández-Velasco M, Ruilope LM, Ruiz-Hurtado G. Fibroblast Growth Factor-23-Klotho Axis in Cardiorenal Syndrome: Mediators and Potential Therapeutic Targets. Front Physiol 2021; 12:775029. [PMID: 34867481 PMCID: PMC8634640 DOI: 10.3389/fphys.2021.775029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiorenal syndrome (CRS) is a complex disorder that refers to the category of acute or chronic kidney diseases that induce cardiovascular disease, and inversely, acute or chronic heart diseases that provoke kidney dysfunction. There is a close relationship between renal and cardiovascular disease, possibly due to the presence of common risk factors for both diseases. Thus, it is well known that renal diseases are associated with increased risk of developing cardiovascular disease, suffering cardiac events and even mortality, which is aggravated in those patients with end-stage renal disease or who are undergoing dialysis. Recent works have proposed mineral bone disorders (MBD) as the possible link between kidney dysfunction and the development of cardiovascular outcomes. Traditionally, increased serum phosphate levels have been proposed as one of the main factors responsible for cardiovascular damage in kidney patients. However, recent studies have focused on other MBD components such as the elevation of fibroblast growth factor (FGF)-23, a phosphaturic bone-derived hormone, and the decreased expression of the anti-aging factor Klotho in renal patients. It has been shown that increased FGF-23 levels induce cardiac hypertrophy and dysfunction and are associated with increased cardiovascular mortality in renal patients. Decreased Klotho expression occurs as renal function declines. Despite its expression being absent in myocardial tissue, several studies have demonstrated that this antiaging factor plays a cardioprotective role, especially under elevated FGF-23 levels. The present review aims to collect the recent knowledge about the FGF-23-Klotho axis in the connection between kidney and heart, focusing on their specific role as new therapeutic targets in CRS.
Collapse
Affiliation(s)
- José Alberto Navarro-García
- Cardiorenal Translational Laboratory, Institute of Research i + 12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Laura González-Lafuente
- Cardiorenal Translational Laboratory, Institute of Research i + 12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Luis M Ruilope
- Cardiorenal Translational Laboratory, Institute of Research i + 12, Hospital Universitario 12 de Octubre, Madrid, Spain.,CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain.,School of Doctoral Studies and Research, European University of Madrid, Madrid, Spain
| | - Gema Ruiz-Hurtado
- Cardiorenal Translational Laboratory, Institute of Research i + 12, Hospital Universitario 12 de Octubre, Madrid, Spain.,CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
| |
Collapse
|
20
|
Acute cardiac overload does not induce cardiac or skeletal expression of fibroblast growth factor 23 in rats. Cardiovasc Endocrinol Metab 2021; 10:204-209. [PMID: 34765890 PMCID: PMC8575438 DOI: 10.1097/xce.0000000000000249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/05/2021] [Indexed: 12/05/2022]
Abstract
Elevated fibroblast growth factor 23 (FGF23) is associated with cardiovascular events, particularly heart failure. Although FGF23 has been reported to induce cardiac hypertrophy, recent studies demonstrated that cardiac hypertrophy and myocardial infarction induce FGF23 production by cardiomyocytes. We aimed to explore whether acute cardiac overload increases cardiac and skeletal FGF23 expression and circulating FGF23 levels.
Collapse
|
21
|
Komaba H, Fukagawa M. Jury still out on whether FGF23 is a direct contributor, a useful biomarker, or neither. Kidney Int 2021; 100:989-993. [PMID: 34688390 DOI: 10.1016/j.kint.2021.04.045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/20/2021] [Accepted: 04/30/2021] [Indexed: 11/19/2022]
Affiliation(s)
- Hirotaka Komaba
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan; Interactive Translational Research Center for Kidney Diseases, Tokai University School of Medicine, Isehara, Japan; The Institute of Medical Sciences, Tokai University, Isehara, Japan
| | - Masafumi Fukagawa
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Isehara, Japan.
| |
Collapse
|
22
|
Sellier AB, Seiler-Mußler S, Emrich IE, Böhm M, Fliser D, Zawada AM, Heine GH. FGFR4 and Klotho Polymorphisms Are Not Associated with Cardiovascular Outcomes in Chronic Kidney Disease. Am J Nephrol 2021; 52:808-816. [PMID: 34673637 DOI: 10.1159/000519274] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/17/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION High plasma fibroblast growth factor 23 (FGF-23) predicts cardiovascular events in chronic kidney disease (CKD) patients. Experimental evidence suggests FGF receptor 4 (FGFR4) activation by FGF-23, and deficiency of the soluble form of its co-receptor Klotho promotes left-ventricular hypertrophy (LVH). To evaluate the clinical relevance of these findings, a Mendelian randomization study analyzed the association of genetic variants of FGFR4 and Klotho with echocardiographic parameters and cardiac events in CKD patients. METHODS The prospective Cardiovascular and Renal Outcome in CKD 2-4 Patients-The Fourth Homburg Evaluation study recruited CKD G2-G4 patients, of whom 519 consented to SNP genotyping (FGFR4: rs351855; Klotho: rs9536314). Echocardiographic examinations at baseline and 5 years later assessed prevalence of LVH by measurement of left-ventricular mass index (LVMI). Patients were followed for 5.1 ± 2.1 years for the primary endpoints of cardiac decompensation and atherosclerotic cardiovascular disease (ASCVD). RESULTS Carriers of the different alleles did neither differ in baseline LVMI (rs351855: p = 0.861; rs9536314: p = 0.379) nor in LVMI changes between baseline and follow-up (rs351855: p = 0.181; rs9536314: p = 0.995). Hundred and four patients suffered cardiac decompensation, and 144 patients had ASCVD. Time to cardiac decompensation (rs351855: p = 0.316; rs9536314: p = 0.765) and ASCVD (p = 0.508 and p = 0.800, respectively) did not differ between carriers of different alleles. DISCUSSION/CONCLUSION rs351855 and rs9536314 were not associated with LVMI or cardiac events. These findings do not provide evidence for a relevant clinical role of either FGFR4 stimulation or soluble form of Klotho deficiency in LVH development.
Collapse
Affiliation(s)
- Alexander B Sellier
- Department of Internal Medicine IV-Nephrology and Hypertension, Saarland University Hospital and Saarland University Faculty of Medicine, Homburg, Germany
| | - Sarah Seiler-Mußler
- Department of Internal Medicine IV-Nephrology and Hypertension, Saarland University Hospital and Saarland University Faculty of Medicine, Homburg, Germany
- Vauban Praxis, Saarlouis, Germany
| | - Insa E Emrich
- Department of Internal Medicine III-Cardiology, Angiology and Intensive Care Medicine, Saarland University Hospital and Saarland University Faculty of Medicine, Homburg, Germany
| | - Michael Böhm
- Department of Internal Medicine III-Cardiology, Angiology and Intensive Care Medicine, Saarland University Hospital and Saarland University Faculty of Medicine, Homburg, Germany
| | - Danilo Fliser
- Department of Internal Medicine IV-Nephrology and Hypertension, Saarland University Hospital and Saarland University Faculty of Medicine, Homburg, Germany
| | - Adam M Zawada
- Department of Internal Medicine IV-Nephrology and Hypertension, Saarland University Hospital and Saarland University Faculty of Medicine, Homburg, Germany
| | - Gunnar H Heine
- Department of Internal Medicine IV-Nephrology and Hypertension, Saarland University Hospital and Saarland University Faculty of Medicine, Homburg, Germany
- Department of Internal Medicine II, Agaplesion Markus Krankenhaus, Frankfurt, Germany
| |
Collapse
|
23
|
Leifheit-Nestler M, Haffner D. How FGF23 shapes multiple organs in chronic kidney disease. Mol Cell Pediatr 2021; 8:12. [PMID: 34536161 PMCID: PMC8449753 DOI: 10.1186/s40348-021-00123-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/07/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease (CKD) is associated with distinct alterations in mineral metabolism in children and adults resulting in multiple organ dysfunctions. Children with advanced CKD often suffer from impaired bone mineralization, bone deformities and fractures, growth failure, muscle weakness, and vascular and soft tissue calcification, a complex which was recently termed CKD-mineral and bone disorder (CKD-MBD). The latter is a major contributor to the enhanced cardiovascular disease comorbidity and mortality in these patients. Elevated circulating levels of the endocrine-acting phosphaturic hormone fibroblast growth factor (FGF) 23 are the first detectable alteration of mineral metabolism and thus CKD-MBD. FGF23 is expressed and secreted from osteocytes and osteoblasts and rises, most likely due to increased phosphate load, progressively as kidney function declines in order to maintain phosphate homeostasis. Although not measured in clinical routine yet, CKD-mediated increased circulating levels of FGF23 in children are associated with pathological cardiac remodeling, vascular alterations, and increased cognitive risk. Clinical and experimental studies addressing other FGF23-mediated complications of kidney failure, such as hypertension and impaired bone mineralization, show partly conflicting results, and the causal relationships are not always entirely clear. This short review summarizes regulators of FGF23 synthesis altered in CKD and the main CKD-mediated organ dysfunctions related to high FGF23 levels.
Collapse
Affiliation(s)
- Maren Leifheit-Nestler
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School Children's Hospital, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School Children's Hospital, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| |
Collapse
|
24
|
Matsumoto A, Matsui I, Mano K, Mizuno H, Katsuma Y, Yasuda S, Shimada K, Inoue K, Oki T, Hanai T, Kojima K, Kaneko T, Isaka Y. Recurrent membranous nephropathy with a possible alteration in the etiology: a case report. BMC Nephrol 2021; 22:253. [PMID: 34229600 PMCID: PMC8258946 DOI: 10.1186/s12882-021-02457-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 06/24/2021] [Indexed: 11/13/2022] Open
Abstract
Background Phospholipase A2 receptor 1 (PLA2R1) and thrombospondin type-1 domain-containing 7A (THSD7A) are the two major pathogenic antigens for membranous nephropathy (MN). It has been reported that THSD7A-associated MN has a higher prevalence of comorbid malignancy than PLA2R1-associated MN. Here we present a case of MN whose etiology might change from idiopathic to malignancy-associated MN during the patient’s clinical course. Case presentation A 68-year-old man with nephrotic syndrome was diagnosed with MN by renal biopsy. Immunohistochemistry showed that the kidney specimen was negative for THSD7A. The first course of corticosteroid therapy achieved partial remission; however, nephrotic syndrome recurred 1 year later. Two years later, his abdominal echography revealed a urinary bladder tumor, but he did not wish to undergo additional diagnostic examinations. Because his proteinuria increased consecutively, corticosteroid therapy was resumed, but it failed to achieve remission. Another kidney biopsy was performed and revealed MN with positive staining for THSD7A. PLA2R1 staining levels were negative for both first and second biopsies. Because his bladder tumor had gradually enlarged, he agreed to undergo bladder tumor resection. Pathological examination indicated that the tumor was THDS7A-positive bladder cancer. Subsequently, his proteinuria decreased and remained in remission. Conclusions This case suggests that the etiology of MN might be altered during the therapeutic course. Intensive screening for malignancy may be preferable in patients with unexpected recurrence of proteinuria and/or change in therapy response. Supplementary Information The online version contains supplementary material available at 10.1186/s12882-021-02457-0.
Collapse
Affiliation(s)
- Ayumi Matsumoto
- Department of Nephrology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Isao Matsui
- Department of Nephrology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| | - Keiji Mano
- Department of Nephrology, Daini Osaka Police Hospital, 2-6-40, Karasuga-tsuji, Tennoji, Osaka, 543-8922, Japan
| | - Hitoshi Mizuno
- Department of Nephrology, Daini Osaka Police Hospital, 2-6-40, Karasuga-tsuji, Tennoji, Osaka, 543-8922, Japan
| | - Yusuke Katsuma
- Department of Nephrology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Seiichi Yasuda
- Department of Nephrology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Karin Shimada
- Department of Nephrology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Kazunori Inoue
- Department of Nephrology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Takashi Oki
- Department of Urology, Mimihara General Hospital, 4-465, kyowacho, Sakai-ku, Sakai, Osaka, 590-8505, Japan
| | - Tadashi Hanai
- Department of Urology, Daini Osaka Police Hospital, 2-6-40, Karasuga-tsuji, Tennoji, Osaka, 543-8922, Japan
| | - Keiko Kojima
- Department of Pathology, Daini Osaka Police Hospital, 2-6-40, Karasuga-tsuji, Tennoji, Osaka, 543-8922, Japan
| | - Tetsuya Kaneko
- Department of Nephrology, Daini Osaka Police Hospital, 2-6-40, Karasuga-tsuji, Tennoji, Osaka, 543-8922, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
25
|
Memmos E, Papagianni A. New Insights into the Role of FGF-23 and Klotho in Cardiovascular Disease in Chronic Kidney Disease Patients. Curr Vasc Pharmacol 2021; 19:55-62. [PMID: 32310050 DOI: 10.2174/1570161118666200420102100] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 12/29/2022]
Abstract
Alterations of fibroblast growth factor 23 (FGF-23) and Klotho levels are considered to be the earliest biochemical abnormality of chronic kidney disease - mineral and bone disease (CKDMBD) syndrome. Moreover, emerging data suggests that the dysregulated FGF-23 and Klotho axis has many effects on the cardiovascular (CV) system and contributes significantly to the increased CV morbidity and mortality rates of CKD patients. This review examines recent evidence on the role of FGF-23 and Klotho in the development and progression of CV complications of uremia namely cardiac hypertrophy, uremic cardiomyopathy, and atherosclerotic and arteriosclerotic vascular lesions. Moreover, the available evidence on their associations with adverse clinical outcomes are summarized. Undoubtedly, more studies are needed to further elucidate the effects of FGF-23 and Klotho on the heart and vessels and to gain insights into their prognostic value as CV risk factors. Finally, large prospective studies are required to test the hypothesis that modification of their levels would have a favourable impact on the unacceptably high mortality rates of these patient populations.
Collapse
Affiliation(s)
- Evangelos Memmos
- Department of Nephrology, Aristotle University of Thessaloniki, General Hospital "Hippokratio", Thessaloniki, Greece
| | - Aikaterini Papagianni
- Department of Nephrology, Aristotle University of Thessaloniki, General Hospital "Hippokratio", Thessaloniki, Greece
| |
Collapse
|
26
|
Grund A, Sinha MD, Haffner D, Leifheit-Nestler M. Fibroblast Growth Factor 23 and Left Ventricular Hypertrophy in Chronic Kidney Disease-A Pediatric Perspective. Front Pediatr 2021; 9:702719. [PMID: 34422725 PMCID: PMC8372151 DOI: 10.3389/fped.2021.702719] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/05/2021] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular diseases (CVD) are a hallmark in pediatric patients with chronic kidney disease (CKD) contributing to an enhanced risk of all-cause and CV morbidity and mortality in these patients. The bone-derived phosphaturic hormone fibroblast growth factor (FGF) 23 progressively rises with declining kidney function to maintain phosphate homeostasis, with up to 1,000-fold increase in patients with kidney failure requiring dialysis. FGF23 is associated with the development of left ventricular hypertrophy (LVH) and thereby accounts to be a CVD risk factor in CKD. Experimentally, FGF23 directly induces hypertrophic growth of cardiac myocytes in vitro and LVH in vivo. Further, clinical studies in adult CKD have observed cardiotoxicity associated with FGF23. Data regarding prevalence and determinants of FGF23 excess in children with CKD are limited. This review summarizes current data and discusses whether FGF23 may be a key driver of LVH in pediatric CKD.
Collapse
Affiliation(s)
- Andrea Grund
- Department of Paediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School Children's Hospital, Hanover, Germany.,Paediatric Research Centre, Hannover Medical School, Hanover, Germany
| | - Manish D Sinha
- Department of Paediatric Nephrology, King's College London, Evelina London Children's Hospital, London, United Kingdom
| | - Dieter Haffner
- Department of Paediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School Children's Hospital, Hanover, Germany.,Paediatric Research Centre, Hannover Medical School, Hanover, Germany
| | - Maren Leifheit-Nestler
- Department of Paediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School Children's Hospital, Hanover, Germany.,Paediatric Research Centre, Hannover Medical School, Hanover, Germany
| |
Collapse
|
27
|
High-phosphorus diets reduce aortic lesions and cardiomyocyte size and modify lipid metabolism in Ldl receptor knockout mice. Sci Rep 2020; 10:20748. [PMID: 33247205 PMCID: PMC7695849 DOI: 10.1038/s41598-020-77509-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022] Open
Abstract
The consumption of phosphorus in Western populations largely exceeds the recommended intake, while vitamin D supply is often insufficient. Both situations are linked to an increased cardiovascular risk. A 17-week two-factorial study with Ldl receptor-/- mice was conducted to investigate the cardiovascular impact of dietary phosphorus [adequate (0.3%; P0.3) vs. high (1.5%; P1.5)] in combination with a low (50 IU/kg; D50) or adequate vitamin D diet (1000 IU/kg; D1000). The data demonstrate that mice fed the P1.5 vs. P0.3 diets developed smaller vascular lesions (p = 0.013) and cardiac hypotrophy (p = 0.011), which were accompanied by diminished IGF1 and insulin signalling activity in their hearts. Vitamin D showed no independent effect on atherogenesis and heart morphology. Feeding P1.5 vs. P0.3 diets resulted in markedly reduced serum triacylglycerols (p < 0.0001) and cholesterol (p < 0.0001), higher faecal lipid excretion (p < 0.0001) and a reduced mRNA abundance of hepatic sterol exporters and lipoprotein receptors. Minor hypocholesterolaemic and hypotriglyceridaemic effects were also found in mice fed the D1000 vs. D50 diets (p = 0.048, p = 0.026). To conclude, a high phosphorus intake strongly affected the formation of vascular lesions, cardiac morphology, and lipid metabolism, although these changes are not indicative of an increased cardiovascular risk.
Collapse
|
28
|
Maxacalcitol (22-Oxacalcitriol (OCT)) Retards Progression of Left Ventricular Hypertrophy with Renal Dysfunction Through Inhibition of Calcineurin-NFAT Activity. Cardiovasc Drugs Ther 2020; 35:381-397. [PMID: 33206298 DOI: 10.1007/s10557-020-07111-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/10/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE Left ventricular hypertrophy (LVH) is a cardiovascular complication highly prevalent in patients with chronic kidney disease (CKD). Previous studies analyzing 1α-hydroxylase or vitamin D receptor (Vdr) knockout mice revealed active vitamin D as a promising agent inhibiting LVH progression. Paricalcitol, an active vitamin D analog, failed to suppress the progression of LV mass index (LVMI) in pre-dialysis patients with CKD. As target genes of activated VDR differ depending on its agonists, we examined the effects of maxacalcitol (22-oxacalcitriol: OCT), a less calcemic active vitamin D analog, on LVH in hemodialysis patients and animal LVH models with renal insufficiency. METHODS In retrospective cohort study, patients treated with OCT who underwent hemodialysis were enrolled. Using cardiac echocardiography, LV mass was evaluated by the area-length method. In animal study, angiotensin II (Ang II)-infused Wister rats with heminephrectomy or Ang II-stimulated neonatal rat ventricular myocytes (NRVM) were treated with OCT. RESULTS OCT significantly inhibited the progression of LVMI in hemodialysis patients. In Ang II-infused heminephrectomized rats, OCT suppressed the progression of LVH in a blood pressure-independent manner. OCT also suppressed the activity of calcineurin in the left ventricle of model rats. Specifically, OCT reduced the protein levels of calcineurin A, but not the mRNA levels of Ppp3ca (calcineurin Aα). Luciferase assays showed that OCT increased the promoter activity of Fbxo32 (atrogin1), an E3 ubiquitin ligase targeting calcineurin A. Finally, OCT promoted ubiquitination and degradation of calcineurin A. CONCLUSION Our works indicated that OCT retards progression of LVH through calcineurin-NFAT pathway, which reveal a novel aspect of OCT in attenuating pathological LVH.
Collapse
|
29
|
Ghuman J, Cai X, Patel RB, Khan SS, Hecktman J, Redfield MM, Lewis G, Shah SJ, Wolf M, Isakova T, Mehta R. Fibroblast Growth Factor 23 and Exercise Capacity in Heart Failure with Preserved Ejection Fraction. J Card Fail 2020; 27:309-317. [PMID: 33035687 DOI: 10.1016/j.cardfail.2020.09.477] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/24/2020] [Accepted: 09/25/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is characterized by left ventricular hypertrophy and decreased exercise capacity. Fibroblast growth factor 23 (FGF23), a hormone involved in phosphate, vitamin D, and iron homeostasis, is linked to left ventricular hypertrophy and HF. We measured c-terminal FGF23 (cFGF23) and intact FGF23 (iFGF23) levels and examined their associations with exercise capacity in patients with HFpEF. METHODS AND RESULTS Using multivariable linear regression and linear mixed models, we studied the associations of cFGF23 and iFGF23 with baseline and mean weekly change over 24 weeks in peak oxygen consumption and 6-minute walk distance in individuals enrolled in the Phosphodiesterase-5 Inhibition to Improve Clinical Status and Exercise Capacity in HFpEF trial. Our study population included 172 individuals with available plasma for cFGF23 and iFGF23 measurements. Median (25th-75th percentile) baseline cFGF23 and iFGF23 levels were 208.7 RU/mL (132.1-379.5 RU/mL) and 90.3 pg/mL (68.6-128.5 pg/mL), respectively. After adjustment for cardiovascular disease and hematologic and kidney parameters, higher cFGF23 was independently associated with a lower peak oxygen consumption at baseline. Higher iFGF23 was independently associated with shorter 6-minute walk distance at baseline. No significant associations were appreciated with the longitudinal outcomes. CONCLUSIONS In patients with HFpEF, higher FGF23 levels are independently associated with decreased exercise capacity at baseline.
Collapse
Affiliation(s)
- Jasleen Ghuman
- McGaw Medical Center of Northwestern University Feinberg School of Medicine, Department of Medicine, Chicago, Illinois
| | - Xuan Cai
- Center for Translational Metabolism and Health, Institute of Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ravi B Patel
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Sadiya S Khan
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jonathan Hecktman
- Center for Translational Metabolism and Health, Institute of Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - Gregory Lewis
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sanjiv J Shah
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Myles Wolf
- Division of Nephrology, Department of Medicine, Duke University, Durham, North Carolina
| | - Tamara Isakova
- Center for Translational Metabolism and Health, Institute of Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Division of Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Rupal Mehta
- Center for Translational Metabolism and Health, Institute of Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Division of Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Division of Nephrology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois.
| |
Collapse
|
30
|
Bao JF, Hu PP, She QY, Li A. A Land of Controversy: Fibroblast Growth Factor-23 and Uremic Cardiac Hypertrophy. J Am Soc Nephrol 2020; 31:1423-1434. [PMID: 32527977 PMCID: PMC7351013 DOI: 10.1681/asn.2020010081] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiac hypertrophy is a common feature in patients with CKD. Recent studies revealed that two phosphate regulators, fibroblast growth factor-23 and α-Klotho, are highly involved in the pathophysiologic process of CKD-induced cardiac hypertrophy. With decreasing renal function, elevated fibroblast growth factor-23 and decreased α-Klotho may contribute to cardiac hypertrophy by targeting the heart directly or by inducing systemic changes, such as vascular injury, hemodynamic disorders, and inflammation. However, several studies have demonstrated that disturbances in the fibroblast growth factor-23/α-Klotho axis do not lead to cardiac hypertrophy. In this review, we describe the cardiac effects of the fibroblast growth factor-23/α-Klotho axis and summarize recent progress in this field. In addition, we present not only the main controversies in this field but also provide possible directions to resolve these disputes.
Collapse
Affiliation(s)
- Jing-Fu Bao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pan-Pan Hu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qin-Ying She
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Aiqing Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
31
|
Kondo Y, Komaba H, Fukagawa M. Endocrine fibroblast growth factors as potential biomarkers for chronic kidney disease. Expert Rev Mol Diagn 2020; 20:715-724. [PMID: 32513031 DOI: 10.1080/14737159.2020.1780918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Among the family of fibroblast growth factors (FGFs), FGF19, FGF21, and FGF23 act as circulating hormones and are called endocrine FGFs. FGF19 and FGF21 regulate bile acid and energy homeostasis, respectively, whereas FGF23 regulates vitamin D and phosphate homeostasis. Accumulating evidence suggests that FGF23 plays a critical role in disturbed mineral metabolisms, left ventricular hypertrophy, immunosuppression, inflammation, among others in patients with chronic kidney disease (CKD), highlighting the potential both as a biomarker and a therapeutic target. Several studies have also examined the potential role of FGF19 and FGF21 in CKD patients. AREAS COVERED In this review, we present a brief overview of the biology of FGF19, FGF21, and FGF23, and summarize recent clinical and experimental studies on the pathophysiological roles of endocrine FGFs, mainly FGF23, in CKD patients. EXPERT OPINION Among the endocrine FGFs, FGF23 represents the most promising biomarker in CKD patients. If future studies confirm that FGF23 is directly toxic in CKD patients, FGF23 could be regarded as a therapeutic target and its measurement would be valuable if applied in clinical practice. Despite their potentially important roles, the clinical relevance of FGF19 and FGF21 in CKD patients is unclear, and much more studies are required.
Collapse
Affiliation(s)
- Yuichiro Kondo
- Interactive Translational Research Center for Kidney Diseases, Tokai University School of Medicine , Isehara, Japan
| | - Hirotaka Komaba
- Interactive Translational Research Center for Kidney Diseases, Tokai University School of Medicine , Isehara, Japan.,Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine , Isehara, Japan.,The Institute of Medical Sciences, Tokai University , Isehara, Japan
| | - Masafumi Fukagawa
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine , Isehara, Japan
| |
Collapse
|
32
|
Vervloet MG. FGF23 measurement in chronic kidney disease: What is it really reflecting? Clin Chim Acta 2020; 505:160-166. [PMID: 32156608 DOI: 10.1016/j.cca.2020.03.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 12/20/2022]
Abstract
Fibroblast growth factor can be measured in clinical practice using ELISA, with acceptable validity. Different from many metabolites and minerals, its value can differ by a thousand-fold between individuals, largely because of differences in kidney function and dietary habits. This wide range complicates the proper interpretation of the concentration of FGF23, both in terms of the appropriateness of a given value for a given estimated GFR, and in terms of estimating the magnitude of risk for clinical events, with which FGF23 is clearly associated. In this narrative review, the impact of kidney function, exposure to phosphate from diet, and novel emerging factors that influence FGF23 concentrations are discussed. These and yet to define determinants of FGF23 question the causality of the association of FGF23 with hard (cardiovascular) endpoints, as observed in several epidemiological studies.
Collapse
Affiliation(s)
- Marc G Vervloet
- Amsterdam University Medical Center, Department of Nephrology, and Amsterdam Cardiovascular Sciences (ACS), Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| |
Collapse
|
33
|
Bouma-de Krijger A, Vervloet MG. Fibroblast growth factor 23: are we ready to use it in clinical practice? J Nephrol 2020; 33:509-527. [PMID: 32130720 PMCID: PMC7220896 DOI: 10.1007/s40620-020-00715-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 02/15/2020] [Indexed: 12/15/2022]
Abstract
Patients with chronic kidney disease (CKD) have a greatly enhanced risk of cardiovascular morbidity and mortality. Over the past decade it has come clear that a disturbed calcium-phosphate metabolism, with Fibroblast Growth Factor-23 as a key hormone, is partly accountable for this enhanced risk. Numerous studies have been performed unravelling FGF23s actions and its association with clinical conditions. As FGF23 is strongly associated with adverse outcome it may be a promising biomarker for risk prediction or, even more important, targeting FGF23 may be a strategy to improve patient outcome. This review elaborates on the clinical usefulness of FGF23 measurement. Firstly it discusses the reliability of the FGF23 measurement. Secondly, it evaluates whether FGF23 measurement may lead to improved patient risk classification. Finally, and possibly most importantly, this review evaluates if lowering of FGF23 should be a target for therapy. For this, the review discusses the current evidence indicating that FGF23 may be in the causal pathway to cardiovascular pathology, provides an overview of strategies to lower FGF23 levels and discusses the current evidence concerning the benefit of lowering FGF23.
Collapse
Affiliation(s)
- Annet Bouma-de Krijger
- Department of Nephrology, Amsterdam Cardiovascular Science, Amsterdam University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Marc G. Vervloet
- Department of Nephrology, Amsterdam Cardiovascular Science, Amsterdam University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
34
|
Rodríguez M. FGF23: Is It Another Biomarker for Phosphate-Calcium Metabolism? Adv Ther 2020; 37:73-79. [PMID: 32236868 DOI: 10.1007/s12325-019-01181-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Indexed: 12/13/2022]
Abstract
Fibroblast growth factor 23 (FGF23) is a protein produced by mature osteoblasts involved in mineral homeostasis by binding to its receptor complex FGFR/Klotho located mainly in the kidneys. Although this protein participates in numerous biological processes, increase in the levels of FGF23 is responsible for many pathologies, such as X-linked hypophosphataemia (XLH), chronic kidney disease, cardiovascular disease or even mortality. For this reason, both FGF23 and its receptors have become elements of interest for the development of treatments. However, FGF23 can be altered for many other reasons, such as inflammatory processes, iron, hypoxia, heart failure or erythropoietin, that negatively affect mortality. This article will review the role of FGF23 in phosphate homeostasis, its relationship to mortality, fractures and chronic renal failure, and how the levels of this factor can be reduced.
Collapse
|
35
|
Abstract
FGF23 is a protein secreted in the plasma by bone cells. In the kidney, FGF23 can activate an FGF receptor in the presence of its co-receptor αKlotho. FGF23 controls the renal phosphate reabsorption and calcitriol metabolism. When renal function declines, plasma FGF23 concentration raises and FGF23 can stimulate FGFRs in the absence of αKlotho. This induces cardiac hypertrophy, modifies cardiomyocyte contractility and increases the risk of arrhythmic events in cardiac cells. There is still no evidence that decreasing FGF23 concentration in patients on dialysis could improve their survival. In different cardiac disorders cardiomyocyte can produce FGF23, which can reveal a way of adaptation to the stress.
Collapse
Affiliation(s)
- Dominique Prié
- Faculté de santé de l'université de Paris, hôpital Necker Enfants Malades GHU Centre-Université de Paris AP-HP, INEM INSERM U1151, 149, rue de Sèvres 75015 Paris, France.
| |
Collapse
|
36
|
Rroji M, Figurek A, Spasovski G. Should We Consider the Cardiovascular System While Evaluating CKD-MBD? Toxins (Basel) 2020; 12:toxins12030140. [PMID: 32106499 PMCID: PMC7150959 DOI: 10.3390/toxins12030140] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/12/2020] [Accepted: 02/20/2020] [Indexed: 12/25/2022] Open
Abstract
Cardiovascular (CV) disease is highly prevalent in the population with chronic kidney disease (CKD), where the risk of CV death in early stages far exceeds the risk of progression to dialysis. The presence of chronic kidney disease-mineral and bone disorder (CKD-MBD) has shown a strong correlation with CV events and mortality. As a non-atheromatous process, it could be partially explained why standard CV disease-modifying drugs do not provide such an impact on CV mortality in CKD as observed in the general population. We summarize the potential association of CV comorbidities with the older (parathyroid hormone, phosphate) and newer (FGF23, Klotho, sclerostin) CKD-MBD biomarkers.
Collapse
Affiliation(s)
- Merita Rroji
- University Department of Nephrology, Faculty of Medicine, University of Medicine Tirana, Tirana 1001, Albania
- Correspondence:
| | - Andreja Figurek
- Institute of Anatomy, University of Zurich, Zurich 8057, Switzerland;
| | - Goce Spasovski
- University Department of Nephrology, Medical Faculty, University of Skopje, Skopje 1000, North Macedonia;
| |
Collapse
|
37
|
Stöhr R, Brandenburg VM, Heine GH, Maeder MT, Leibundgut G, Schuh A, Jeker U, Pfisterer M, Sanders-van Wijk S, Brunner-la Rocca HP. Limited role for fibroblast growth factor 23 in assessing prognosis in heart failure patients: data from the TIME-CHF trial. Eur J Heart Fail 2020; 22:701-709. [PMID: 32020782 DOI: 10.1002/ejhf.1749] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/30/2019] [Accepted: 12/27/2019] [Indexed: 12/16/2022] Open
Abstract
AIM Fibroblast growth factor 23 (FGF23) is an intensively studied biomarker at the crossroads of cardiovascular disease, heart failure (HF) and chronic kidney disease. Independent associations between increasing FGF23 levels and cardiovascular events were found in many, but not all studies. By analysing data from the TIME-CHF cohort, we sought to investigate the prognostic value of FGF23 in an elderly, multimorbid HF patient cohort. We determined differences between intact (iFGF23) and C-terminal FGF23 (cFGF23) regarding their prognostic value and their levels over time in different HF subgroups according to left ventricular ejection fraction (LVEF). METHODS AND RESULTS In this multicentre trial of 622 patients with symptomatic HF aged ≥60 years, we determined iFGF23 and cFGF23 at baseline, 3, 6 and 12-month follow-up. In unadjusted analyses, cFGF23 significantly predicted all HF-related outcomes at all time points. The predictive value of iFGF23 was less and not statistically significant at baseline. After multivariable adjustments, the association between both cFGF23 and iFGF23 and outcome lost statistical significance apart from cFGF23 at month 3. Overall, patients with preserved and mid-range LVEF had higher levels of iFGF23 and cFGF23 than those with reduced LVEF. Levels decreased significantly during the first 3 months in mid-range and reduced LVEF patients, but did not significantly change over time in those with preserved LVEF. CONCLUSIONS Fibroblast growth factor 23 is of limited value regarding risk prediction in this elderly HF population. Potentially heterogeneous roles of FGF23 in different LVEF groups deserve further investigation.
Collapse
Affiliation(s)
- Robert Stöhr
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Germany
| | | | - Gunnar H Heine
- AGAPLESION MARKUS KRANKENHAUS, Frankfurt am Main, Germany
| | - Micha T Maeder
- Department of Cardiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Gregor Leibundgut
- Department of Internal Medicine, Cardiology, Kantonsspital Baselland, Liestal, Switzerland
| | - Alexander Schuh
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Germany
| | - Urs Jeker
- Heart Center, Luzerner Kantonsspital, Lucerne, Switzerland
| | | | | | - Hans-Peter Brunner-la Rocca
- Department of Cardiology, University Hospital Basel, Basel, Switzerland.,Department of Cardiology, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
38
|
Fukumoto S, Takashi Y, Tsoumpra MK, Sawatsubashi S, Matsumoto T. How do we sense phosphate to regulate serum phosphate level? J Bone Miner Metab 2020; 38:1-6. [PMID: 31797064 DOI: 10.1007/s00774-019-01066-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 11/05/2019] [Indexed: 12/25/2022]
Abstract
Abnormal phosphate levels result in several pathological conditions such as rickets/osteomalacia and ectopic calcification indicating that there must be a system that regulates phosphate level within a narrow range. FGF23 has been shown to be an essential hormone regulating serum phosphate level. FGF23 binds to Klotho-FGF receptor complex to reduce serum phosphate level. Several reports suggested that FGF receptor is involved in the regulation of FGF23 production. It has been also shown that high extracellular phosphate can activate several intracellular signaling pathways. However, it has been unclear whether and how phosphate regulates FGF23 production in vivo. Our recent results indicate that high extracellular phosphate directly activates FGF receptor 1 and the downstream intracellular signaling enhances FGF23 production. Thus, there is a negative feedback system for the regulation of serum phosphate level involving FGF receptor and FGF23. We propose that FGF receptor works at least as one of phosphate sensors in the maintenance of serum phosphate level.
Collapse
Affiliation(s)
- Seiji Fukumoto
- Department of Molecular Endocrinology, Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima, Tokushima, 770-8503, Japan.
| | - Yuichi Takashi
- Department of Molecular Endocrinology, Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima, Tokushima, 770-8503, Japan
- Department of Endocrinology and Diabetes Mellitus, Fukuoka University Chikushi Hospital, 1-1-1 Zokumyoin, Chikushino, 818-8502, Japan
| | - Maria K Tsoumpra
- Department of Molecular Endocrinology, Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima, Tokushima, 770-8503, Japan
- Department of Molecular Therapy, National Institute of Neuroscience, National Center for Neurology and Psychiatry, 4-1-1 Ogawahigashi-cho, Kodaira, Tokyo, 187-8502, Japan
| | - Shun Sawatsubashi
- Department of Molecular Endocrinology, Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima, Tokushima, 770-8503, Japan
| | - Toshio Matsumoto
- Department of Molecular Endocrinology, Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima, Tokushima, 770-8503, Japan
| |
Collapse
|
39
|
Abstract
Fibroblast growth factor 23 (FGF23) is a hormone with a central role in the regulation of phosphate homeostasis. This regulation is accomplished by the coordinated modulation of renal phosphate handling, vitamin D metabolism and parathyroid hormone secretion. Patients with kidney disease have increased circulating levels of FGF23 and in other patient populations and in healthy individuals, FGF23 levels also rise following an increase in dietary phosphate intake. Maladaptive increases in FGF23 have a detrimental effect on several organs and tissues and, importantly, these pathological changes most likely contribute to increased morbidity and mortality. For example, in the context of heart disease, FGF23 is involved in the development of pathological hypertrophy that can lead to congestive heart failure. Increased FGF23 concentrations can also lead to microcirculatory changes, in particular reduced vasodilatory capacity, and collectively these cardiovascular changes can compromise tissue perfusion. In addition, FGF23 is associated with inflammation and an increased risk of infection; other potentially detrimental effects of FGF23 are likely to emerge in the future. Most importantly, recent insights demonstrate that FGF23 can be therapeutically targeted, which holds promise for the treatment of many patients in a variety of clinical settings.
Collapse
|
40
|
Wagner CA, Rubio-Aliaga I, Egli-Spichtig D. Fibroblast growth factor 23 in chronic kidney disease: what is its role in cardiovascular disease? Nephrol Dial Transplant 2019; 34:1986-1990. [PMID: 30903187 DOI: 10.1093/ndt/gfz044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 02/10/2019] [Indexed: 11/14/2022] Open
Affiliation(s)
- Carsten A Wagner
- National Center of Competence in Research Kidney, CH, Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Isabel Rubio-Aliaga
- National Center of Competence in Research Kidney, CH, Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Daniela Egli-Spichtig
- National Center of Competence in Research Kidney, CH, Institute of Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
41
|
Smith ER, Holt SG, Hewitson TD. αKlotho-FGF23 interactions and their role in kidney disease: a molecular insight. Cell Mol Life Sci 2019; 76:4705-4724. [PMID: 31350618 PMCID: PMC11105488 DOI: 10.1007/s00018-019-03241-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 07/09/2019] [Accepted: 07/22/2019] [Indexed: 12/16/2022]
Abstract
Following the serendipitous discovery of the ageing suppressor, αKlotho (αKl), several decades ago, a growing body of evidence has defined a pivotal role for its various forms in multiple aspects of vertebrate physiology and pathology. The transmembrane form of αKl serves as a co-receptor for the osteocyte-derived mineral regulator, fibroblast growth factor (FGF)23, principally in the renal tubules. However, compelling data also suggest that circulating soluble forms of αKl, derived from the same source, may have independent homeostatic functions either as a hormone, glycan-cleaving enzyme or lectin. Chronic kidney disease (CKD) is of particular interest as disruption of the FGF23-αKl axis is an early and common feature of disease manifesting in markedly deficient αKl expression, but FGF23 excess. Here we critically discuss recent findings in αKl biology that conflict with the view that soluble αKl has substantive functions independent of FGF23 signalling. Although the issue of whether soluble αKl can act without FGF23 has yet to be resolved, we explore the potential significance of these contrary findings in the context of CKD and highlight how this endocrine pathway represents a promising target for novel anti-ageing therapeutics.
Collapse
Affiliation(s)
- Edward R Smith
- Department of Nephrology, The Royal Melbourne Hospital, Melbourne, Australia.
- Department of Medicine, University of Melbourne, Grattan Street, Parkville, VIC, 3050, Australia.
| | - Stephen G Holt
- Department of Nephrology, The Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, University of Melbourne, Grattan Street, Parkville, VIC, 3050, Australia
| | - Tim D Hewitson
- Department of Nephrology, The Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, University of Melbourne, Grattan Street, Parkville, VIC, 3050, Australia
| |
Collapse
|
42
|
Ding J, Tang Q, Luo B, Zhang L, Lin L, Han L, Hao M, Li M, Yu L, Li M. Klotho inhibits angiotensin II-induced cardiac hypertrophy, fibrosis, and dysfunction in mice through suppression of transforming growth factor-β1 signaling pathway. Eur J Pharmacol 2019; 859:172549. [PMID: 31325434 DOI: 10.1016/j.ejphar.2019.172549] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 07/13/2019] [Accepted: 07/15/2019] [Indexed: 12/14/2022]
Abstract
Recent studies have revealed critical roles of transforming growth factor-β1 (TGF-β1) and microRNA-132 (miR-132), a downstream mediator of TGF-β1, in the pathogenesis of cardiac remodeling. In this study, we tested whether the antiaging protein klotho modifies angiotensin II (Ang II)-induced cardiac remodeling through regulating TGF-β1-miR-132 axis. We found that both klotho and the TGF-β1 inhibitor LY364947 significantly inhibited cardiac hypertrophy, fibrosis, and dysfunction in Ang II-infused mice, as evidenced by the ratios of heart weight to body weight (HW/BW), heart weight to tibial length (HW/TL), cardiomyocyte cross-sectional area, fibrotic area, and expression of prohypertrophic genes (ANP, β-MHC) and fibrotic marker genes (α-SMA, collagen I), echocardiographic parameters. Meanwhile, klotho also significantly inhibited Ang II-induced protein expression of TGF-β1 and phosphorylated Smad2/3 in the heart tissues and cultured cardiomyocytes and cardiac fibroblasts. In vitro experiments demonstrated that Ang II-induced cardiomyocyte hypertrophy and proliferation and activation of cardiac fibroblasts were markedly inhibited by klotho, LY364947 or the miR-132 inhibitor anti-miR-132. Both klotho and the TGF-β1 inhibitor LY364947 downregulated the miR-132 expression. Additionally, klotho decreased Ang II-induced protein expressions of cardiac fibroblast growth factor (FGF)23 in vivo and in vitro. The decreased protein levels of klotho in serum and renal tissues of Ang II-infused mice were elevated by klotho. Klotho downregulated the protein levels of TGF-β1 in renal tissues of Ang II-infused mice. In conclusion, our results suggest that klotho prevents Ang II-induced cardiac remodeling and dysfunction through modifying the TGF-β1-miR-132 axis, providing an experimental basis for clinical treatment on cardiac remodeling.
Collapse
Affiliation(s)
- Jieqiong Ding
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, 437100, PR China; Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, 437100, PR China
| | - Qiong Tang
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, 437100, PR China; Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, 437100, PR China
| | - Binhua Luo
- School of Pharmacy, Hubei University of Science and Technology, Xianning, 437100, PR China
| | - Lijun Zhang
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, 437100, PR China
| | - Li Lin
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, 437100, PR China
| | - Lu Han
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, 437100, PR China
| | - Miaomiao Hao
- School of Pharmacy, Hubei University of Science and Technology, Xianning, 437100, PR China
| | - Mingyue Li
- School of Pharmacy, Hubei University of Science and Technology, Xianning, 437100, PR China
| | - Liangzhu Yu
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, 437100, PR China; Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, 437100, PR China.
| | - Mincai Li
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, 437100, PR China; Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, 437100, PR China.
| |
Collapse
|
43
|
Bär L, Stournaras C, Lang F, Föller M. Regulation of fibroblast growth factor 23 (FGF23) in health and disease. FEBS Lett 2019; 593:1879-1900. [PMID: 31199502 DOI: 10.1002/1873-3468.13494] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 12/19/2022]
Abstract
Fibroblast growth factor 23 (FGF23) is mainly produced in the bone and, upon secretion, forms a complex with a FGF receptor and coreceptor αKlotho. FGF23 can exert several endocrine functions, such as inhibiting renal phosphate reabsorption and 1,25-dihydroxyvitamin D3 production. Moreover, it has paracrine activities on several cell types, including neutrophils and hepatocytes. Klotho and Fgf23 deficiencies result in pathologies otherwise encountered in age-associated diseases, mainly as a result of hyperphosphataemia-dependent calcification. FGF23 levels are also perturbed in the plasma of patients with several disorders, including kidney or cardiovascular diseases. Here, we review mechanisms controlling FGF23 production and discuss how FGF23 regulation is perturbed in disease.
Collapse
Affiliation(s)
- Ludmilla Bär
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Christos Stournaras
- Institute of Biochemistry, University of Crete Medical School, Heraklion, Greece
| | - Florian Lang
- Institute of Physiology, University of Tübingen, Germany
| | - Michael Föller
- Institute of Physiology, University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
44
|
Risk of cardiovascular involvement in pediatric patients with X-linked hypophosphatemia. Pediatr Nephrol 2019; 34:1077-1086. [PMID: 30607568 DOI: 10.1007/s00467-018-4180-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/29/2018] [Accepted: 12/13/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To find out if cardiovascular alterations are present in pediatric patients with X-linked hypophosphatemia (XLH). STUDY DESIGN Multicentre prospective clinical study on pediatric patients included in the RenalTube database ( www.renaltube.com ) with genetically confirmed diagnosis of XLH by mutations in the PHEX gene. The study's protocol consisted of biochemical work-up, 24-h ambulatory blood pressure monitoring (ABPM), carotid ultrasonography, and echocardiogram. All patients were on chronic treatment with phosphate supplements and 1-hydroxy vitamin D metabolites. RESULTS Twenty-four patients (17 females, from 1 to 17 years of age) were studied. Serum concentrations (X ± SD) of phosphate and intact parathyroid hormone were 2.66 ± 0.60 mg/dl and 58.3 ± 26.8 pg/ml, respectively. Serum fibroblast growth factor 23 (FGF23) concentration was 278.18 ± 294.45 pg/ml (normal < 60 pg/ml). Abnormally high carotid intima media thickness was found in one patient, who was obese and hypertensive as revealed by ABPM, which disclosed arterial hypertension in two other patients. Z scores for echocardiographic interventricular septum end diastole and left ventricular posterior wall end diastole were + 0.77 ± 0.77 and + 0.94 ± 0.86, respectively. Left ventricular mass index (LVMI) was 44.93 ± 19.18 g/m2.7, and four patients, in addition to the obese one, had values greater than 51 g/m2.7, indicative of left ventricular hypertrophy. There was no correlation between these echocardiographic parameters and serum FGF23 concentrations. CONCLUSIONS XLH pediatric patients receiving conventional treatment have echocardiographic measurements of ventricular mass within normal reference values, but above the mean, and 18% have LVMI suggestive of left ventricular hypertrophy without correlation with serum FGF23 concentrations. This might indicate an increased risk of cardiovascular involvement in XLH.
Collapse
|
45
|
Faul C. FGF23 effects on the heart-levels, time, source, and context matter. Kidney Int 2019; 94:7-11. [PMID: 29933856 DOI: 10.1016/j.kint.2018.03.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 03/20/2018] [Accepted: 03/22/2018] [Indexed: 12/21/2022]
Abstract
Fibroblast growth factor 23 (FGF23) has appeared as a hormone that is massively elevated in patients with chronic kidney disease. Whether FGF23 is a risk factor that associates with cardiac pathologies and cardiovascular mortality, as suggested by a variety of clinical studies, or additionally acts as a causative factor that induces cardiac injury, as more recently indicated by cell culture and animal studies, is under debate and the center of many ongoing experimental studies.
Collapse
Affiliation(s)
- Christian Faul
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
46
|
Bär L, Hase P, Föller M. PKC regulates the production of fibroblast growth factor 23 (FGF23). PLoS One 2019; 14:e0211309. [PMID: 30921339 PMCID: PMC6438472 DOI: 10.1371/journal.pone.0211309] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/14/2019] [Indexed: 12/20/2022] Open
Abstract
Serine/threonine protein kinase C (PKC) is activated by diacylglycerol that is released from membrane lipids by phospholipase C in response to activation of G protein-coupled receptors or receptor tyrosine kinases. PKC isoforms are particularly relevant for proliferation and differentiation of cells including osteoblasts. Osteoblasts/osteocytes produce fibroblast growth factor 23 (FGF23), a hormone regulating renal phosphate and vitamin D handling. PKC activates NFκB, a transcription factor complex controlling FGF23 expression. Here, we analyzed the impact of PKC on FGF23 synthesis. Fgf23 expression was analyzed by qRT-PCR in UMR106 osteoblast-like cells and in IDG-SW3 osteocytes, and FGF23 protein was measured by ELISA. Phorbol ester 12-O-tetradecanoylphorbol-13-acetate (PMA), a PKC activator, up-regulated FGF23 production. In contrast, PKC inhibitors calphostin C, Gö6976, sotrastaurin and ruboxistaurin suppressed FGF23 formation. NFκB inhibitor withaferin A abolished the stimulatory effect of PMA on Fgf23. PKC is a powerful regulator of FGF23 synthesis, an effect which is at least partly mediated by NFκB.
Collapse
Affiliation(s)
- Ludmilla Bär
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Philipp Hase
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Michael Föller
- Institute of Physiology, University of Hohenheim, Stuttgart, Germany
- * E-mail:
| |
Collapse
|
47
|
Negrea L. Active Vitamin D in Chronic Kidney Disease: Getting Right Back Where We Started from? KIDNEY DISEASES (BASEL, SWITZERLAND) 2019; 5:59-68. [PMID: 31019920 PMCID: PMC6465695 DOI: 10.1159/000495138] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 11/01/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND The vitamin D system is essential for optimal health in humans. Circulating calcitriol, a key metabolite in maintaining calcium and phosphorus homeostasis, is produced in the kidney. In kidney failure, calcitriol levels progressively decrease, contributing to the development of renal secondary hyperparathyroidism (SHPT). SUMMARY For years, SHPT had a central role in the disturbed mineral metabolism of renal patients. As calcitriol deficiency contributes to SHPT development, treatment with calcitriol or other compounds able to activate the vitamin D receptor (VDR) was one of the mainstays of therapy for renal patients in the last 40 years. In this review, we discuss how the treatment with VDR activators (VDRA) evolved during this time in the United States, as well as the main factors responsible for these changes. KEY MESSAGES Management of SHPT with VDRA in renal patients has undergone a few paradigm shifts over the last 40 years. When treating SHPT, the newly developed therapies as well as VDRA need to be carefully considered and used appropriately. Nephrologists need to use an integrated approach that avoids excessive use of VDRA, ensures replenishment of vitamin D stores, and avoids hypercalcemia and hyperphosphatemia.
Collapse
Affiliation(s)
- Lavinia Negrea
- Renal Division, Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
48
|
Changes in serum and intracardiac fibroblast growth factor 23 during the progression of left ventricular hypertrophy in hypertensive model rats. Clin Exp Nephrol 2018; 23:589-596. [PMID: 30539338 DOI: 10.1007/s10157-018-1680-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 12/04/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Recent clinical studies have demonstrated that serum fibroblast growth factor 23 (FGF23) levels have a significant association with left ventricular hypertrophy (LVH). Although LVH is commonly seen in hypertensive patients, the association between FGF23, hypertension, and LVH remains unclear. We aimed to examine the changes in serum and intracardiac FGF23 during the progression of hypertension using spontaneously hypertensive rats (SHR). METHODS Male SHR comprised the experimental group (HT group) and Wistar Kyoto rats served as controls. At 10 weeks, urinary and blood biochemical analyses and blood pressure measurements were performed for both the groups. At 18 weeks, the rats were sacrificed: urinary and blood biochemical analyses and real-time PCR were performed. RESULTS At 18 weeks, the relative heart weight and serum N-terminal pro-brain natriuretic peptide and aldosterone levels were significantly greater in the HT group. Serum calcium and phosphate levels were significantly lower, while serum FGF23 levels were significantly higher in the HT group compared to the control group. Further analyses showed that the mRNA expression of FGF23 in the heart was significantly increased in the HT group compared to the control group. Both serum FGF23 levels and intracardiac mRNA expression of FGF23 showed significant correlation with the relative heart weight. CONCLUSIONS During LVH progression, serum and intracardiac FGF23 increased in hypertension. Although it is unclear whether the change in FGF23 is the cause or result of LVH, the interaction between FGF23 and aldosterone may be associated with the development of LVH in hypertension.
Collapse
|
49
|
Fibroblast growth factor 23 and cardiovascular disease in patients with chronic kidney disease. RENAL REPLACEMENT THERAPY 2018. [DOI: 10.1186/s41100-018-0172-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
50
|
Zhou C, Mei C, Dai B, Xue C. Fibroblast Growth Factor-23 May Follow Cardiovascular Disease Rather than Causing It in Chronic Kidney Disease. J Am Soc Nephrol 2018; 29:2602. [PMID: 30093457 DOI: 10.1681/asn.2018050517] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Chenchen Zhou
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China; and.,Division of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Changlin Mei
- Division of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Bing Dai
- Division of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Cheng Xue
- Division of Nephrology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|