1
|
Javier AJS, Kennedy FM, Yi X, Wehling-Henricks M, Tidball JG, White KE, Witczak CA, Kuro-O M, Welc SS. Klotho Is Cardioprotective in the mdx Mouse Model of Duchenne Muscular Dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:923-940. [PMID: 39889824 PMCID: PMC12016860 DOI: 10.1016/j.ajpath.2024.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/18/2024] [Accepted: 12/30/2024] [Indexed: 02/03/2025]
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, progressive skeletal and cardiac myopathy. Cardiomyopathy is the leading cause of death in patients with DMD, but the molecular basis for heart failure is incompletely understood. As with humans, in the mdx mouse model of DMD, cardiac function is impaired after the onset of skeletal muscle pathology. Dysregulation of Klotho gene regulation in dystrophic skeletal muscles occurs at disease onset, affecting pathogenesis. Whether Klotho is protective against dystrophin-deficient cardiomyopathy is unknown. This study found that expression of a Klotho transgene prevented deficits in left ventricular ejection fraction and fractional shortening in mdx mice. Improvements in cardiac performance were associated with reductions in adverse cardiac remodeling, cardiac myocyte hypertrophy, and fibrosis. In addition, mdx mice expressed high concentrations of plasma fibroblast growth factor 23 (FGF23), and expression was increased locally in hearts. The cardioprotective effects of Klotho were not associated with differences in renal function or serum biochemistries, but transgene expression prevented increased expression of plasma FGF23 and cardiac Fgf23 mRNA expression. Cardiac reactive oxygen species, oxidative damage, mitochondrial damage, and apoptosis were reduced in transgenic hearts. FGF23 stimulated hypertrophic growth in dystrophic neonatal mouse ventricular myocytes in vitro, which was inhibited by co-stimulation with soluble Klotho. Taken together, these results show that Klotho prevented dystrophic cardiac remodeling and improved function.
Collapse
MESH Headings
- Animals
- Klotho Proteins
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/physiopathology
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/complications
- Muscular Dystrophy, Duchenne/genetics
- Glucuronidase/metabolism
- Glucuronidase/genetics
- Fibroblast Growth Factor-23
- Fibroblast Growth Factors/metabolism
- Fibroblast Growth Factors/blood
- Fibroblast Growth Factors/genetics
- Mice, Inbred mdx
- Disease Models, Animal
- Mice
- Cardiomyopathies/pathology
- Cardiomyopathies/metabolism
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Mice, Transgenic
- Mice, Inbred C57BL
- Male
- Apoptosis
- Reactive Oxygen Species/metabolism
- Fibrosis
- Humans
- Myocardium/pathology
- Myocardium/metabolism
Collapse
Affiliation(s)
- Areli Jannes S Javier
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana
| | - Felicia M Kennedy
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Xin Yi
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - James G Tidball
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, California
| | - Kenneth E White
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Carol A Witczak
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana; Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Makoto Kuro-O
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Steven S Welc
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana; Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
2
|
Vogt J, Daferner K, Föller M. Suppression of Fibroblast Growth Factor 23 in UMR106 Osteoblast-Like Cells and MC3T3-E1 Cells by Adipokine Chemerin. Cell Biochem Funct 2025; 43:e70051. [PMID: 39887469 PMCID: PMC11782926 DOI: 10.1002/cbf.70051] [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: 06/05/2024] [Revised: 12/09/2024] [Accepted: 01/14/2025] [Indexed: 02/01/2025]
Abstract
Endocrine fibroblast growth factor 23 (FGF23) derived from bone governs phosphate and vitamin D metabolism. Paracrine FGF23 has additional functions in different organs. Moreover, plasma FGF23 is correlated with outcomes in chronic kidney disease. FGF23 regulation is complex depending on a plethora of different factors and conditions including AMP-dependent kinase (AMPK), inflammation, and adipokines leptin and adiponectin. Chemerin is an adipokine implicated in proinflammatory processes in adipose tissue and other organs and an activator of AMPK. Here, we investigated whether chemerin is a regulator of FGF23. UMR106 osteoblast-like cells and MC3T3-E1 osteoblasts were studied. Gene expression was assessed by qRT-PCR, FGF23 protein by ELISA, and AMPK activity by western blotting. Both cell lines expressed Cmklr1 encoding chemerin chemokine-like receptor 1. Chemerin slightly but significantly reduced Fgf23 expression. Chemerin reduced FGF23 protein abundance in the cell culture supernatant, and RNAi-mediated Cmklr1 silencing upregulated Fgf23 expression in UMR106 cells. In the presence of AMPK inhibitor compound C, chemerin failed to suppress Fgf23 in UMR106 cells. In conclusion, chemerin-dependent Cmklr1 signaling downregulates FGF23 in bone cell lines. This effect requires, at least partly, AMPK.
Collapse
Affiliation(s)
- Julia Vogt
- Department of PhysiologyUniversity of HohenheimStuttgartGermany
| | - Kim Daferner
- Department of PhysiologyUniversity of HohenheimStuttgartGermany
| | - Michael Föller
- Department of PhysiologyUniversity of HohenheimStuttgartGermany
| |
Collapse
|
3
|
Płonka J, Olejnik A, Klus A, Gawrylak-Dryja E, Wężyk N, Rzepiela L, Dąbrowska K, Nalewajko K, Porażko T, Bil-Lula I, Gierlotka M. Use of FGF-23 and sαKlotho for Risk Stratification in Patients with Acute Heart Failure. J Clin Med 2025; 14:860. [PMID: 39941530 PMCID: PMC11818865 DOI: 10.3390/jcm14030860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Soluble αKlotho (sαKlotho) and fibroblast growth factor 23 (FGF-23) are increased in acute heart failure (AHF). This study aimed to assess changes in serum sαKlotho and FGF-23 concentrations during an episode of AHF as well as the usefulness of both biomarkers for predicting long-term prognosis. Methods: The study included 104 consecutive patients hospitalized in t he intensive cardiac care unit due to AHF (mean age, 65.8 ± 14.6 years; mean ejection fraction, 31.4% ± 14). New-onset AHF was reported in 43.3% of the population. Blood samples were measured at entry and on discharge from hospital. The main clinical outcomes assessed in this study were all-cause mortality or rehospitalization due to HF during a 3-year follow-up. Results: At admission sαKlotho, FGF-23, and NT-pro BNP levels, compared with discharge, were significantly higher at p < 0.001, p < 0.001, and p < 0.001 respectively. The 3-year Kaplan-Meier analysis, based on tertiles, revealed, for sαKlotho levels from Tertile 1 on admission and at discharge, a 2-fold higher rate of all-cause mortality or rehospitalization for HF compared with Tertile 3 (p = 0.006 and p = 0.028, respectively). One-third of patients showed an increase in FGF-23 and sαKlotho levels during hospitalization. Patients with the highest percentage increase in the levels of both biomarkers had an elevated risk of all-cause morality or hospitalization for HF (hazard ratio, 2.75; confidence interval, 1.19-6.35; p = 0.02). Conclusions: sαKlotho and FGF-23 levels are elevated during an episode of AHF. Low sαKlotho levels are associated with an increased risk of all-cause mortality or rehospitalization for HF. Increases in sαKlotho and FGF-23 values during hospitalization identify patients with poor prognosis.
Collapse
Affiliation(s)
- Joanna Płonka
- Department of Cardiology, University Hospital, Institute of Medical Sciences, University of Opole, 45-401 Opole, Poland; (N.W.); (L.R.); (K.D.); (K.N.); (M.G.)
| | - Agnieszka Olejnik
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry and Laboratory Haematology, Faculty of Pharmacy, Wrocław Medical University, 50-556 Wrocław, Poland; (A.O.); (I.B.-L.)
| | - Anna Klus
- Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, 45-401 Opole, Poland; (A.K.); (E.G.-D.)
| | - Ewa Gawrylak-Dryja
- Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, 45-401 Opole, Poland; (A.K.); (E.G.-D.)
| | - Natalia Wężyk
- Department of Cardiology, University Hospital, Institute of Medical Sciences, University of Opole, 45-401 Opole, Poland; (N.W.); (L.R.); (K.D.); (K.N.); (M.G.)
| | - Lidia Rzepiela
- Department of Cardiology, University Hospital, Institute of Medical Sciences, University of Opole, 45-401 Opole, Poland; (N.W.); (L.R.); (K.D.); (K.N.); (M.G.)
| | - Klaudia Dąbrowska
- Department of Cardiology, University Hospital, Institute of Medical Sciences, University of Opole, 45-401 Opole, Poland; (N.W.); (L.R.); (K.D.); (K.N.); (M.G.)
| | - Krzysztof Nalewajko
- Department of Cardiology, University Hospital, Institute of Medical Sciences, University of Opole, 45-401 Opole, Poland; (N.W.); (L.R.); (K.D.); (K.N.); (M.G.)
| | - Tomasz Porażko
- Department of Internal Medicine and Nephrology, Institute of Medical Sciences, University Hospital in Opole, 45-401 Opole, Poland;
| | - Iwona Bil-Lula
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry and Laboratory Haematology, Faculty of Pharmacy, Wrocław Medical University, 50-556 Wrocław, Poland; (A.O.); (I.B.-L.)
| | - Marek Gierlotka
- Department of Cardiology, University Hospital, Institute of Medical Sciences, University of Opole, 45-401 Opole, Poland; (N.W.); (L.R.); (K.D.); (K.N.); (M.G.)
| |
Collapse
|
4
|
Deng C, Wu Y. Vitamin D-Parathyroid Hormone-Fibroblast Growth Factor 23 Axis and Cardiac Remodeling. Am J Cardiovasc Drugs 2025; 25:25-36. [PMID: 39392562 DOI: 10.1007/s40256-024-00688-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/26/2024] [Indexed: 10/12/2024]
Abstract
Cardiac remodeling is a compensatory adaptive response to chronic heart failure (HF) altering the structure, function, and metabolism of the heart. Many nutritional and metabolic diseases can aggravate the pathophysiological development of cardiac remodeling. Vitamin D deficiency leads to cardiac remodeling by activating the renin-angiotensin-aldosterone system (RAAS), resulting in enhanced inflammation and directly promoting cardiac fibrosis and extracellular matrix deposition. Hyperparathyroidism upregulates protein kinase A or protein kinase C, enhances intracellular calcium influx, promotes oxidative stress, activates RAAS, and increases aldosterone levels, thereby aggravating cardiac remodeling. Besides, fibroblast growth factor 23 (FGF23) plays a direct role in the heart, resulting in ventricular hypertrophy and myocardial fibrosis. Vitamin D deficiency leads to hyperparathyroidism, which in turn increases the level of FGF23. Elevated levels of FGF23 further inhibit vitamin D synthesis. Evidence exists that vitamin D deficiency, hyperparathyroidism, and marked elevations in FGF23 concentration form a vicious cycle and are believed to contribute directly to cardiac remodeling. Therefore, the purpose of this article is to introduce the specific effects of the above substances on the heart and to explain the significance of understanding the vitamin D-parathyroid hormone-FGF23 axis in improving or even reversing cardiac remodeling, thus contributing to the treatment of patients with HF.
Collapse
Affiliation(s)
- Cuiyun Deng
- Special Demand Medical Care Ward, Beijing Anzhen Hospital Jilin Hospital (Changchun Central Hospital), Changchun, China
| | - Yihang Wu
- Interventional Center of Valvular Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.
| |
Collapse
|
5
|
Campos I, Richter B, Thomas SM, Czaya B, Yanucil C, Kentrup D, Fajol A, Li Q, Secor SM, Faul C. FGFR4 Is Required for Concentric Growth of Cardiac Myocytes during Physiologic Cardiac Hypertrophy. J Cardiovasc Dev Dis 2024; 11:320. [PMID: 39452290 PMCID: PMC11508992 DOI: 10.3390/jcdd11100320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
Fibroblast growth factor (FGF) 23 is a bone-derived hormone that promotes renal phosphate excretion. Serum FGF23 is increased in chronic kidney disease (CKD) and contributes to pathologic cardiac hypertrophy by activating FGF receptor (FGFR) 4 on cardiac myocytes, which might lead to the high cardiovascular mortality in CKD patients. Increases in serum FGF23 levels have also been observed following endurance exercise and in pregnancy, which are scenarios of physiologic cardiac hypertrophy as an adaptive response of the heart to increased demand. To determine whether FGF23/FGFR4 contributes to physiologic cardiac hypertrophy, we studied FGFR4 knockout mice (FGFR4-/-) during late pregnancy. In comparison to virgin littermates, pregnant wild-type and FGFR4-/- mice showed increases in serum FGF23 levels and heart weight; however, the elevation in myocyte area observed in pregnant wild-type mice was abrogated in pregnant FGFR4-/- mice. This outcome was supported by treatments of cultured cardiac myocytes with serum from fed Burmese pythons, another model of physiologic hypertrophy, where the co-treatment with an FGFR4-specific inhibitor abrogated the serum-induced increase in cell area. Interestingly, we found that in pregnant mice, the heart, and not the bone, shows elevated FGF23 expression, and that increases in serum FGF23 are not accompanied by changes in phosphate metabolism. Our study suggests that in physiologic cardiac hypertrophy, the heart produces FGF23 that contributes to hypertrophic growth of cardiac myocytes in a paracrine and FGFR4-dependent manner, and that the kidney does not respond to heart-derived FGF23.
Collapse
Affiliation(s)
- Isaac Campos
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Beatrice Richter
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Sarah Madison Thomas
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Brian Czaya
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Christopher Yanucil
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Dominik Kentrup
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Abul Fajol
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Qing Li
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| | - Stephen M. Secor
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA;
| | - Christian Faul
- Section of Mineral Metabolism, Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (I.C.); (B.R.); (S.M.T.); (B.C.); (C.Y.); (D.K.); (A.F.); (Q.L.)
| |
Collapse
|
6
|
Serban T, Hennings E, Strebel I, Knecht S, du Fay de Lavallaz J, Krisai P, Arnet R, Völlmin G, Osswald S, Sticherling C, Kühne M, Badertscher P. Biomarkers to predict improvement of left ventricular ejection fraction after atrial fibrillation ablation. Heart Rhythm 2024; 21:1581-1588. [PMID: 38614192 DOI: 10.1016/j.hrthm.2024.04.044] [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: 02/24/2024] [Revised: 03/27/2024] [Accepted: 04/04/2024] [Indexed: 04/15/2024]
Abstract
BACKGROUND Atrial fibrillation (AF) and heart failure frequently coexist. Prediction of left ventricular ejection fraction (LVEF) recovery after catheter ablation (CA) for AF remains difficult. OBJECTIVE The purpose of this study was to evaluate the value of biomarkers, alone and in combination with the Antwerp score, to predict LVEF recovery after CA for AF. METHODS Patients undergoing CA for AF with depressed LVEF (<50%) were included. Plasma levels of 13 biomarkers were measured immediately before CA. Patients were categorized into "responders" and "nonresponders" in a similar fashion to the Antwerp score performance derivation and validation cohorts. The predictive power of the biomarkers alone and combined in outcome prediction was evaluated. RESULTS A total of 208 patients with depressed LVEF were included (median age 63 years; 39-19% female; median indexed left atrial volume 42 (33-52) mL/m2; median LVEF 43 (38-46)%). At a median follow-up time of 30 (20-34) months, 161 (77%) were responders and 47 (23%) were nonresponders. Of 13 biomarkers, -4-angiopoietin 2 (ANG2), growth differentiation factor 15 (GDF15), fibroblast growth factor 23, and myosin binding protein C3-were significantly different between responders and nonresponders (P ≤ .001) and their combination could predict the end point with an area under the curve of 0.72 (95% confidence interval [CI] 0.64-0.81) overall, 0.69 (95% CI 0.59-0.78) in heart failure with mildly reduced ejection fraction, and 0.88 (95% CI 0.77-0.98) in heart failure with reduced ejection fraction. Only ANG2 and GDF15 remained significantly associated with LVEF recovery after adjustment for age, sex, and Antwerp score and significantly improved the accuracy of the Antwerp score predictions (P < .001). The area under the curve of the Antwerp score in the outcome prediction improved from 0.75 (95% CI 0.67-0.83) to 0.78 (95% CI 0.70-0.86). CONCLUSION A biomarker panel (ANG2 and GDF15) significantly improved the accuracy of the Antwerp score.
Collapse
Affiliation(s)
- Teodor Serban
- Cardiovascular Research Institute, Basel, Switzerland; Department of Cardiology, University Hospital of Basel, Switzerland
| | - Elisa Hennings
- Cardiovascular Research Institute, Basel, Switzerland; Department of Cardiology, University Hospital of Basel, Switzerland
| | - Ivo Strebel
- Cardiovascular Research Institute, Basel, Switzerland; Department of Cardiology, University Hospital of Basel, Switzerland
| | - Sven Knecht
- Cardiovascular Research Institute, Basel, Switzerland; Department of Cardiology, University Hospital of Basel, Switzerland
| | - Jeanne du Fay de Lavallaz
- Cardiovascular Research Institute, Basel, Switzerland; Department of Cardiology, University Hospital of Basel, Switzerland
| | - Philipp Krisai
- Cardiovascular Research Institute, Basel, Switzerland; Department of Cardiology, University Hospital of Basel, Switzerland
| | - Rebecca Arnet
- Cardiovascular Research Institute, Basel, Switzerland; Department of Cardiology, University Hospital of Basel, Switzerland
| | - Gian Völlmin
- Cardiovascular Research Institute, Basel, Switzerland; Department of Cardiology, University Hospital of Basel, Switzerland
| | - Stefan Osswald
- Cardiovascular Research Institute, Basel, Switzerland; Department of Cardiology, University Hospital of Basel, Switzerland
| | - Christian Sticherling
- Cardiovascular Research Institute, Basel, Switzerland; Department of Cardiology, University Hospital of Basel, Switzerland
| | - Michael Kühne
- Cardiovascular Research Institute, Basel, Switzerland; Department of Cardiology, University Hospital of Basel, Switzerland
| | - Patrick Badertscher
- Cardiovascular Research Institute, Basel, Switzerland; Department of Cardiology, University Hospital of Basel, Switzerland.
| |
Collapse
|
7
|
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
|
8
|
Meulendijks ER, Krul SPJ, Baalman SW, de Vries TAC, Wesselink R, Ernault AC, Kawasaki M, Al-Shama R, Neefs J, Limpens J, de Groot JR. Circulating adipose tissue proteins involved in atrial fibrillation: An explorative scoping review. Trends Cardiovasc Med 2024; 34:148-158. [PMID: 36538994 DOI: 10.1016/j.tcm.2022.12.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Obesity increases the risk of atrial fibrillation (AF), potentially through proteins secreted by adipose tissue (AT) that affect atrial electrical and structural remodeling. We aim to give a comprehensive overview of circulating AT proteins involved in inflammation and fibrosis, that are associated with prevalent AF (paroxysmal or persistent) and the risk on developing new-onset AF. These include adipokines, defined as proteins enriched in AT as adiponectin, but also proteins less specific to AT. We systematically performed an explorative search for studies reporting associations between proteins secreted from cells residing in the AT and AF, and additionally assessed the effect of obesity on these proteins by a secondary search. The AT proteins involved in inflammation were mostly increased in patients with prevalent and new-onset AF, and with obesity, while the AT enriched adipokines were mostly not associated with AF. This review provides insight into circulating adipose tissue proteins involved in AF substrate formation.
Collapse
Affiliation(s)
- Eva R Meulendijks
- Amsterdam UMC, University of Amsterdam, Heart Center, department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, the Netherlands.
| | - Sébastien P J Krul
- Amsterdam UMC, University of Amsterdam, Heart Center, department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105, the Netherlands
| | - Sarah W Baalman
- Amsterdam UMC, University of Amsterdam, Heart Center, department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105, the Netherlands
| | - Tim A C de Vries
- Amsterdam UMC, University of Amsterdam, Heart Center, department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, the Netherlands
| | - Robin Wesselink
- Amsterdam UMC, University of Amsterdam, Heart Center, department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, the Netherlands
| | - Auriane C Ernault
- Amsterdam UMC, University of Amsterdam, Heart Center, department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, the Netherlands
| | - Makiri Kawasaki
- Amsterdam UMC, University of Amsterdam, Heart Center, department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105, the Netherlands
| | - Rushd Al-Shama
- Amsterdam UMC, University of Amsterdam, Heart Center, department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, the Netherlands
| | - Jolien Neefs
- Amsterdam UMC, University of Amsterdam, Heart Center, department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105, the Netherlands
| | - Jacqueline Limpens
- Amsterdam UMC, University of Amsterdam, Heart Center, department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105, the Netherlands
| | - Joris R de Groot
- Amsterdam UMC, University of Amsterdam, Heart Center, department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam 1105, the Netherlands; Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, the Netherlands
| |
Collapse
|
9
|
King PH. Skeletal muscle as a molecular and cellular biomarker of disease progression in amyotrophic lateral sclerosis: a narrative review. Neural Regen Res 2024; 19:747-753. [PMID: 37843208 PMCID: PMC10664124 DOI: 10.4103/1673-5374.382226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/27/2023] [Accepted: 07/19/2023] [Indexed: 10/17/2023] Open
Abstract
Amyotrophic lateral sclerosis is a fatal multisystemic neurodegenerative disease with motor neurons being a primary target. Although progressive weakness is a hallmark feature of amyotrophic lateral sclerosis, there is considerable heterogeneity, including clinical presentation, progression, and the underlying triggers for disease initiation. Based on longitudinal studies with families harboring amyotrophic lateral sclerosis-associated gene mutations, it has become apparent that overt disease is preceded by a prodromal phase, possibly in years, where compensatory mechanisms delay symptom onset. Since 85-90% of amyotrophic lateral sclerosis is sporadic, there is a strong need for identifying biomarkers that can detect this prodromal phase as motor neurons have limited capacity for regeneration. Current Food and Drug Administration-approved therapies work by slowing the degenerative process and are most effective early in the disease. Skeletal muscle, including the neuromuscular junction, manifests abnormalities at the earliest stages of the disease, before motor neuron loss, making it a promising source for identifying biomarkers of the prodromal phase. The accessibility of muscle through biopsy provides a lens into the distal motor system at earlier stages and in real time. The advent of "omics" technology has led to the identification of numerous dysregulated molecules in amyotrophic lateral sclerosis muscle, ranging from coding and non-coding RNAs to proteins and metabolites. This technology has opened the door for identifying biomarkers of disease activity and providing insight into disease mechanisms. A major challenge is correlating the myriad of dysregulated molecules with clinical or histological progression and understanding their relevance to presymptomatic phases of disease. There are two major goals of this review. The first is to summarize some of the biomarkers identified in human amyotrophic lateral sclerosis muscle that have a clinicopathological correlation with disease activity, evidence of a similar dysregulation in the SOD1G93A mouse during presymptomatic stages, and evidence of progressive change during disease progression. The second goal is to review the molecular pathways these biomarkers reflect and their potential role in mitigating or promoting disease progression, and as such, their potential as therapeutic targets in amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Peter H. King
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, AL, USA; Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA
| |
Collapse
|
10
|
El‐Harasis MA, Quintana JA, Martinez‐Parachini JR, Jackson GG, Varghese BT, Yoneda ZT, Murphy BS, Crawford DM, Tomasek K, Su YR, Wells QS, Roden DM, Michaud GF, Saavedra P, Estrada JC, Richardson TD, Kanagasundram AN, Shen ST, Montgomery JA, Ellis CR, Crossley GH, Eberl M, Gillet L, Ziegler A, Shoemaker MB. Recurrence After Atrial Fibrillation Ablation and Investigational Biomarkers of Cardiac Remodeling. J Am Heart Assoc 2024; 13:e031029. [PMID: 38471835 PMCID: PMC11010019 DOI: 10.1161/jaha.123.031029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/23/2023] [Indexed: 03/14/2024]
Abstract
BACKGROUND Recurrence after atrial fibrillation (AF) ablation remains common. We evaluated the association between recurrence and levels of biomarkers of cardiac remodeling, and their ability to improve recurrence prediction when added to a clinical prediction model. METHODS AND RESULTS Blood samples collected before de novo catheter ablation were analyzed. Levels of bone morphogenetic protein-10, angiopoietin-2, fibroblast growth factor-23, insulin-like growth factor-binding protein-7, myosin-binding protein C3, growth differentiation factor-15, interleukin-6, N-terminal pro-brain natriuretic peptide, and high-sensitivity troponin T were measured. Recurrence was defined as ≥30 seconds of an atrial arrhythmia 3 to 12 months postablation. Multivariable logistic regression was performed using biomarker levels along with clinical covariates: APPLE score (Age >65 years, Persistent AF, imPaired eGFR [<60 ml/min/1.73m2], LA diameter ≥43 mm, EF <50%; which includes age, left atrial diameter, left ventricular ejection fraction, persistent atrial fibrillation, and estimated glomerular filtration rate), preablation rhythm, sex, height, body mass index, presence of an implanted continuous monitor, year of ablation, and additional linear ablation. A total of 1873 participants were included. A multivariable logistic regression showed an association between recurrence and levels of angiopoietin-2 (odds ratio, 1.08 [95% CI, 1.02-1.15], P=0.007) and interleukin-6 (odds ratio, 1.02 [95% CI, 1.003-1.03]; P=0.02). The area under the receiver operating characteristic curve of a model that only contained clinical predictors was 0.711. The addition of any of the 9 studied biomarkers to the predictive model did not result in a statistically significant improvement in the area under the receiver operating characteristic curve. CONCLUSIONS Higher angiopoietin-2 and interleukin-6 levels were associated with recurrence after atrial fibrillation ablation in multivariable modeling. However, the addition of biomarkers to a clinical prediction model did not significantly improve recurrence prediction.
Collapse
Affiliation(s)
- Majd A. El‐Harasis
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTN
| | - Joseph A. Quintana
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTN
| | | | - Gregory G. Jackson
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTN
| | - Bibin T. Varghese
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTN
| | - Zachary T. Yoneda
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTN
| | - Brittany S. Murphy
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTN
| | - Diane M. Crawford
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTN
| | - Kelsey Tomasek
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTN
| | - Yan Ru Su
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTN
| | - Quinn S. Wells
- Departments of Medicine, Pharmacology, and Biomedical InformaticsVanderbilt University Medical CenterNashvilleTN
| | - Dan M. Roden
- Departments of Medicine, Pharmacology, and Biomedical InformaticsVanderbilt University Medical CenterNashvilleTN
| | - Gregory F. Michaud
- Division of Cardiovascular Medicine, Massachusetts General HospitalBostonMA
| | - Pablo Saavedra
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTN
| | - Juan Carlos Estrada
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTN
| | - Travis D. Richardson
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTN
| | | | - Sharon T. Shen
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTN
| | - Jay A. Montgomery
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTN
| | - Christopher R. Ellis
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTN
| | - George H. Crossley
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTN
| | | | | | | | | |
Collapse
|
11
|
Oris C, Lautrette A, Dougé A, Bouraima F, Kahouadji S, Pickering ME, Garrouste C, Gagnière J, Guièze R, D'Ostrevy N, Futier E, Grobost V, Buisson A, Batisse M, Bouillon-Minois JB, Pereira B, Durif J, Sapin V, Bouvier D. Prevalence of FGF23 elevation in patients with hypophosphatemia. Clin Chim Acta 2024; 554:117782. [PMID: 38224930 DOI: 10.1016/j.cca.2024.117782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/28/2023] [Accepted: 01/10/2024] [Indexed: 01/17/2024]
Abstract
BACKGROUND AND AIMS To investigate the contribution of FGF23 in explaining the cases of hypophosphatemia observed in clinical practice, we aimed to determine for the first time the prevalence of FGF23 elevation in patients with hypophosphatemia and to describe the different mechanisms of FGF23-related hypophosphatemic disorders. MATERIALS AND METHODS We performed a prospective, observational, multicenter, cohort study of 260 patients with hypophosphatemia. Blood measurements (PTH, 1,25-dihydroxyvitamin D, bone alkaline phosphatase, 25-hydroxyvitamin D, and FGF23) were performed on a Liaison XL® (DiaSorin) analyzer. RESULTS Primary elevation of FGF23 (>95.4 pg/mL) was reported in 10.4% (95CI: 7.0-14.7) of patients (n = 27) with hypophosphatemia, suggesting that at least 1 in 10 cases of hypophosphatemia was erroneously attributed to an etiology other than FGF23 elevation. Patients with elevated blood FGF23 were grouped according to the etiology of the FGF23 elevation. Thus, 10 patients had a renal pathology, chronic kidney disease or post-renal transplantation condition. The remaining patients (n = 17) had the following etiologies: malignancies (n = 9), benign pancreatic tumor (n = 1), post-cardiac surgery (n = 4), cirrhosis (n = 2), and chronic obstructive pulmonary disease (n = 1). CONCLUSION In order to improve patient management, it seems essential to better integrate plasma FGF23 measurement into the routine evaluation of hypophosphatemia.
Collapse
Affiliation(s)
- Charlotte Oris
- Biochemistry and Molecular Genetics Department, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France; Clermont Auvergne University, CNRS, INSERM, iGReD, Clermont-Ferrand, France
| | - Alexandre Lautrette
- Department of Intensive Care Medicine, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Aurore Dougé
- Department of Oncology, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Farouk Bouraima
- Biochemistry and Molecular Genetics Department, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Samy Kahouadji
- Biochemistry and Molecular Genetics Department, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France; Clermont Auvergne University, CNRS, INSERM, iGReD, Clermont-Ferrand, France
| | - Marie-Eva Pickering
- Rheumatology Department, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Cyril Garrouste
- Department of Nephrology, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Johan Gagnière
- Department of Digestive and Hepatobiliary Surgery, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Romain Guièze
- Department of Clinical Hematology, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Nicolas D'Ostrevy
- Department of Cardiac Surgery, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Emmanuel Futier
- Department of Perioperative Medicine, Anesthesia & Critical Care, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Vincent Grobost
- Department of Internal Medicine, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Anthony Buisson
- Department of Hepato-Gastroenterology, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Marie Batisse
- Department of Oncology, Centre Jean Perrin, 63000 Clermont-Ferrand, France
| | | | - Bruno Pereira
- Biostatistics Unit (DRCI) Department, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Julie Durif
- Biochemistry and Molecular Genetics Department, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Vincent Sapin
- Biochemistry and Molecular Genetics Department, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France; Clermont Auvergne University, CNRS, INSERM, iGReD, Clermont-Ferrand, France
| | - Damien Bouvier
- Biochemistry and Molecular Genetics Department, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France; Clermont Auvergne University, CNRS, INSERM, iGReD, Clermont-Ferrand, France.
| |
Collapse
|
12
|
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
|
13
|
Feger M, Alber J, Strotmann J, Grund A, Leifheit-Nestler M, Haffner D, Föller M. Short-term fasting of mice elevates circulating fibroblast growth factor 23 (FGF23). Acta Physiol (Oxf) 2023; 239:e14049. [PMID: 37746883 DOI: 10.1111/apha.14049] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 08/24/2023] [Accepted: 09/04/2023] [Indexed: 09/26/2023]
Abstract
AIMS Phosphate and vitamin D homeostasis are controlled by fibroblast growth factor 23 (FGF23) from bone suppressing renal phosphate transport and enhancing 24-hydroxylase (Cyp24a1), thereby inactivating 1,25(OH)2 D3 . Serum FGF23 is correlated with outcomes in several diseases. Fasting stimulates the production of ketone bodies. We hypothesized that fasting can induce FGF23 synthesis through the production of ketone bodies. METHODS UMR106 cells and isolated neonatal rat ventricular myocytes (NRVM) were treated with ketone body β-hydroxybutyrate. Mice were fasted overnight, fed ad libitum, or treated with β-hydroxybutyrate. Proteins and further blood parameters were determined by enzyme-linked immunoassay (ELISA), western blotting, immunohistochemistry, fluorometric or colorimetric methods, and gene expression by quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS β-Hydroxybutyrate stimulated FGF23 production in UMR106 cells in a nuclear factor kappa-light-chain enhancer of activated B-cells (NFκB)-dependent manner, and in NRVMs. Compared to fed animals, fasted mice exhibited higher β-hydroxybutyrate and FGF23 serum levels (based on assays either detecting C-terminal or intact, biologically active FGF23 only), cardiac, pancreatic, and thymic Fgf23 and renal Cyp24a1 expression, and lower 1,25(OH)2 D3 serum concentration as well as renal Slc34a1 and αKlotho (Kl) expression. In contrast, Fgf23 expression in bone and serum phosphate, calcium, plasma parathyroid hormone (PTH) concentration, and renal Cyp27b1 expression were not significantly affected by fasting. CONCLUSION Short-term fasting increased FGF23 production, as did administration of β-hydroxybutyrate, effects possibly of clinical relevance in view of the increasing use of FGF23 as a surrogate parameter in clinical monitoring of diseases. The fasting state of patients might therefore affect FGF23 tests.
Collapse
Affiliation(s)
- Martina Feger
- Department of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Jana Alber
- Department of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Jörg Strotmann
- Department of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Andrea Grund
- 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
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Hannover, Germany
| | - Michael Föller
- Department of Physiology, University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
14
|
Tsuruda T, Funamoto T, Suzuki C, Yamamura Y, Nakai M, Chosa E, Kaikita K. Increasing baseline aortic valve peak flow velocity is associated with progression of aortic valve stenosis in osteoporosis patients-a possible link to low vitamin D status. Arch Osteoporos 2023; 18:129. [PMID: 37874407 PMCID: PMC10598115 DOI: 10.1007/s11657-023-01339-2] [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: 03/09/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023]
Abstract
PURPOSE The purpose of this study was to investigate the morphological characteristics of the aortic valve and identify factors associated with the progression of aortic valve stenosis (AS) in osteoporosis patients. METHODS In this single-center prospective cohort study, we recruited 10 patients (mean age: 75 ± 7 years, 90% female) who were taking anti-resorptive medicines at the outpatient clinic of University of Miyazaki Hospital, Japan. Baseline assessments, including transthoracic echocardiogram, blood sampling, and dual energy X-ray absorptiometry, were performed. Follow-up assessments were conducted at 6, 12, 18, and 24 months. RESULTS During the 2-year follow-up, three patients with aortic valve peak flow velocity (AV PFV) ≥2 m/s at baseline developed moderate AS, which is defined as AV PFV ≥3 m/s. However, seven patients with AV PFV <2 m/s did not exhibit any progression of AS. There were significant variations in terms of bone mineral density, T-score values, and biomarkers associated with bone turnover (i.e., bone alkaline phosphatase, tartrate-resistance acid phosphatase-5b) among the enrolled patients, but none of these factors were found to be associated with the progression of AS. All patients exhibited low vitamin D status, with a median level of 16.1 ng/mL (25th percentile, 9.7 ng/mL; 75th percentile, 23 ng/mL). The baseline levels of AV PFV values were negatively correlated with 25-hydroxyvitamin D levels, determined by univariate linear regression analysis (beta coefficient = -0.756, 95% confidence interval, -0.136 ̶ -0.023, p = 0.011). CONCLUSION Our data suggest that low vitamin D status might be a potential risk factor for the progression of AS in osteoporosis patients undergoing treatment with anti-resorptive medicines. Elderly patients with osteoporosis patients exhibited a subset of aortic valve stenosis. Our data suggest that the baseline aortic valve peak flow velocity predicts the progression of aortic valve stenosis, and there might be an association between the progression and the co-existing low vitamin D status in these patients.
Collapse
Affiliation(s)
- Toshihiro Tsuruda
- Cardiorenal Research Laboratory, Department of Vascular Advanced Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan.
- Division of Internal Medicine, Cardiovascular Medicine and Nephrology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan.
| | - Taro Funamoto
- Division of Orthopaedic Surgery, Department of Medicine of Sensory and Motor Organs, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Chiyoko Suzuki
- Clinical Laboratory, University of Miyazaki Hospital, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Yoshimasa Yamamura
- Division of Internal Medicine, Cardiovascular Medicine and Nephrology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Michikazu Nakai
- Clinical Research Support Center, University of Miyazaki Hospital, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Etsuo Chosa
- Division of Orthopaedic Surgery, Department of Medicine of Sensory and Motor Organs, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| | - Koichi Kaikita
- Division of Internal Medicine, Cardiovascular Medicine and Nephrology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
| |
Collapse
|
15
|
Widmann L, Keranov S, Jafari L, Liebetrau C, Keller T, Troidl C, Kriechbaum S, Voss S, Arsalan M, Richter MJ, Tello K, Gall H, Ghofrani HA, Guth S, Seeger W, Hamm CW, Dörr O, Nef H. Fibroblast growth factor 23 as a biomarker of right ventricular dysfunction in pulmonary hypertension. Clin Res Cardiol 2023; 112:1382-1393. [PMID: 36790465 PMCID: PMC10562503 DOI: 10.1007/s00392-023-02162-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 01/19/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND Fibroblast growth factor 23 (FGF-23) has been associated with left ventricular hypertrophy (LVH) and heart failure. However, its role in right ventricular (RV) remodeling and RV failure is unknown. This study analyzed the utility of FGF-23 as a biomarker of RV function in patients with pulmonary hypertension (PH). METHODS In this observational study, FGF-23 was measured in the plasma of patients with PH (n = 627), dilated cardiomyopathy (DCM, n = 59), or LVH with severe aortic stenosis (n = 35). Participants without LV or RV abnormalities served as controls (n = 36). RESULTS Median FGF-23 plasma levels were higher in PH patients than in healthy controls (p < 0.001). There were no significant differences between PH, DCM, and LVH patients. Analysis across tertiles of FGF-23 levels in PH patients revealed an association between higher FGF-23 levels and higher levels of NT-proBNP and worse renal function. Furthermore, patients in the high-FGF-23 tertile had a higher pulmonary vascular resistance (PVR), mean pulmonary artery pressure, and right atrial pressure and a lower cardiac index (CI) than patients in the low tertile (p < 0.001 for all comparisons). Higher FGF-23 levels were associated with higher RV end-diastolic diameter and lower tricuspid annular plane systolic excursions (TAPSE) and TAPSE/PASP. Receiver operating characteristic analysis revealed FGF-23 as a good predictor of RV maladaptation, defined as TAPSE < 17 mm and CI < 2.5 L/min/m2. Association of FGF-23 with parameters of RV function was independent of the glomerular filtration rate in regression analysis. CONCLUSION FGF-23 may serve as a biomarker for maladaptive RV remodeling in patients with PH.
Collapse
Affiliation(s)
- Laila Widmann
- Department of Cardiology and Angiology, University of Giessen, Klinikstr. 33, 35392, Giessen, Germany
| | - Stanislav Keranov
- Department of Cardiology and Angiology, University of Giessen, Klinikstr. 33, 35392, Giessen, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site RheinMain, Bad Nauheim, Germany.
| | - Leili Jafari
- Department of Cardiology, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
| | | | - Till Keller
- Department of Cardiology and Angiology, University of Giessen, Klinikstr. 33, 35392, Giessen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site RheinMain, Bad Nauheim, Germany
- Department of Cardiology, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
| | - Christian Troidl
- DZHK (German Center for Cardiovascular Research), Partner Site RheinMain, Bad Nauheim, Germany
- Department of Cardiology, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
| | - Steffen Kriechbaum
- DZHK (German Center for Cardiovascular Research), Partner Site RheinMain, Bad Nauheim, Germany
- Department of Cardiology, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
| | - Sandra Voss
- DZHK (German Center for Cardiovascular Research), Partner Site RheinMain, Bad Nauheim, Germany
- Department of Cardiology, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
| | - Mani Arsalan
- Department of Cardiology and Angiology, University of Giessen, Klinikstr. 33, 35392, Giessen, Germany
| | - Manuel J Richter
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Khodr Tello
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Henning Gall
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Hossein A Ghofrani
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Stefan Guth
- Department of Thoracic Surgery, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
| | - Werner Seeger
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Christian W Hamm
- Department of Cardiology and Angiology, University of Giessen, Klinikstr. 33, 35392, Giessen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site RheinMain, Bad Nauheim, Germany
- Department of Cardiology, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
| | - Oliver Dörr
- Department of Cardiology and Angiology, University of Giessen, Klinikstr. 33, 35392, Giessen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site RheinMain, Bad Nauheim, Germany
| | - Holger Nef
- Department of Cardiology and Angiology, University of Giessen, Klinikstr. 33, 35392, Giessen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site RheinMain, Bad Nauheim, Germany
| |
Collapse
|
16
|
Akwo EA, Robinson-Cohen C. Mendelian randomization and the association of fibroblast growth factor-23 with heart failure with preserved ejection fraction. Curr Opin Nephrol Hypertens 2023; 32:305-312. [PMID: 37016957 PMCID: PMC10313786 DOI: 10.1097/mnh.0000000000000888] [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] [Indexed: 04/06/2023]
Abstract
PURPOSE OF REVIEW Observational data provide compelling evidence for elevated fibroblast growth factor-23 (FGF23) as a risk factor for heart failure (HF), particularly heart failure with preserved ejection fraction (HFpEF). Given the limitations of observational studies, uncertainties persist regarding the causal role of FGF23 in the pathogenesis of HF and HFpEF. Recently, Mendelian randomization (MR) studies have been performed to examine causal associations between FGF23 and HF phenotypes. RECENT FINDINGS The current review describes the methodological basis of the MR techniques used to examine the causal role of FGF23 on HF phenotypes, highlighting the importance of large-scale multiomics data. The findings from most of the MR studies indicate an absence of evidence of a causal effect of FGF23 on the risk of HF in general population settings. However, analysis using individual-level data showed a strong association between genetically-predicted FGF23 and HFpEF in individuals with a genetic predisposition to low estimated glomerular filtration (eGFR). SUMMARY Evidence from MR analysis suggests a causal role of FGF23 in the pathogenesis of HFpEF in low eGFR settings - a finding supported by experimental, clinical, and epidemiological data. While future MR studies of FGF23 and HFpEF could provide further evidence, randomized trials of FGF23-lowering agents could provide the most definitive answers on the association in chronic kidney disease populations.
Collapse
Affiliation(s)
- Elvis A. Akwo
- Vanderbilt O’Brien Kidney Center, Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville TN
| | - Cassianne Robinson-Cohen
- Vanderbilt O’Brien Kidney Center, Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville TN
| |
Collapse
|
17
|
Marcellino A, Bloise S, Pirone C, Brandino G, Gizzone P, Fraternali R, Dilillo A, Del Giudice E, Martucci V, Sanseviero M, Rita LM, Ventriglia F, Lubrano R. Efficacy of Burosumab Every 2 Weeks in an Adult with X-Linked Hypophosphatemia: Should We Learn from Children? Monoclon Antib Immunodiagn Immunother 2023; 42:104-108. [PMID: 37343168 DOI: 10.1089/mab.2022.0026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2023] Open
Abstract
X-linked hypophosphatemia is a genetic condition that leads to fibroblast-growth-factor 23 (FGF23) increase, causing phosphate renal wasting. Since 2018, burosumab, an anti-FGF23 antibody, has been used for this disease with different dosage in children and adults. We report the case of burosumab administration every 2 weeks, as usually done in children. We retrospectively evaluated parathormone (PTH), alkaline phosphatase, serum phosphate, tubular reabsorption of phosphate (TRP), and 25OH vitamin D every 2 weeks in a 29-year-old man with nephrocalcinosis and tertiary hyperparathyroidism who did not respond to standard treatment with burosumab nor to maximum dosage and was treated with burosumab 90 mg every 2 weeks. His serum phosphate and TRP increased with this regimen compared with 4 weeks frequency (respectively 1.74 ± 0.26 mg/dL vs. 2.3 ± 0.19 mg/dL [p 0.0004] and 71.3% ± 4.8% vs. 83.9% ± 7.9% [p 0.01]) with decrease in PTH (183 ± 24.7 pg/mL vs. 109 ± 12.2 pg/mL [p 0.04]). Burosumab may be a good choice in adult patients with X-linked hypophosphatemia; new data are needed regarding the increase in dosage and/or frequency of administration as usually done in children, to achieve disease control.
Collapse
Affiliation(s)
- Alessia Marcellino
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, Latina, Italy
| | - Silvia Bloise
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, Latina, Italy
| | - Carmelo Pirone
- Department of Molecular Medicine, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Giulia Brandino
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, Latina, Italy
| | - Pietro Gizzone
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, Latina, Italy
| | - Roberta Fraternali
- Department of Emergency Medicine, University of Rome La Sapienza, Roma, Lazio, Italy
| | - Anna Dilillo
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, Latina, Italy
| | - Emanuela Del Giudice
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, Latina, Italy
| | - Vanessa Martucci
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, Latina, Italy
| | - Mariateresa Sanseviero
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, Latina, Italy
| | - Leone Maria Rita
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, Latina, Italy
| | - Flavia Ventriglia
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, Latina, Italy
| | - Riccardo Lubrano
- Pediatrics and Neonatology Unit, Maternal-Child Department, Santa Maria Goretti Hospital, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
18
|
Münz S, Feger M, Föller M. Oncostatin M is a regulator of fibroblast growth factor 23 (FGF23) in UMR106 osteoblast-like cells. Sci Rep 2023; 13:8420. [PMID: 37225713 DOI: 10.1038/s41598-023-34858-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 05/09/2023] [Indexed: 05/26/2023] Open
Abstract
Renal phosphate and vitamin D metabolism is under the control of fibroblast growth factor 23 (FGF23), an endocrine and paracrine factor predominantly produced in bone. FGF23 formation is stimulated by active vitamin D, or parathyroid hormone (PTH), which are further regulators of phosphate homeostasis. In renal, inflammatory, and other diseases, plasma FGF23 reflects disease stage and correlates with outcome. Oncostatin M is part of the interleukin-6 (IL-6) family and regulates remodeling and PTH effects in bone as well as cardiac FGF23 production in heart failure via glycoprotein gp130. Here, we studied whether oncostatin M is a regulator of FGF23 in bone cells. Experiments were performed in UMR106 osteoblast-like cells, Fgf23 mRNA was determined by qRT-PCR, FGF23 protein by Western Blotting and ELISA, and oncostatin M receptor and leukemia inhibitory factor (LIF) receptor gene knockout accomplished by siRNA. As a result, oncostatin M dose-dependently up-regulated Fgf23 expression and protein secretion. The oncostatin M effect on FGF23 was mediated by oncostatin M receptor and gp130 and involved, at least in part, STAT3 and MEK1/2. Taken together, oncostatin M is a regulator of FGF23 through oncostatin M receptor, gp130, as well as STAT3 and MEK1/2 in UMR106 osteoblasts.
Collapse
Affiliation(s)
- Sina Münz
- Department of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Martina Feger
- Department of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Michael Föller
- Department of Physiology, University of Hohenheim, Stuttgart, Germany.
| |
Collapse
|
19
|
Hofer F, Hammer A, Pailer U, Koller L, Kazem N, Steinacher E, Steinlechner B, Andreas M, Laufer G, Wojta J, Zelniker TA, Hengstenberg C, Niessner A, Sulzgruber P. Relationship of Fibroblast Growth Factor 23 With Hospitalization for Heart Failure and Cardiovascular Outcomes in Patients Undergoing Cardiac Surgery. J Am Heart Assoc 2023; 12:e027875. [PMID: 36802737 PMCID: PMC10111457 DOI: 10.1161/jaha.122.027875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Background Fibroblast growth factor 23 (FGF-23) is crucial in regulating phosphate and vitamin D metabolism and is moreover associated with an increased cardiovascular risk. The specific objective of this study was to investigate the influence of FGF-23 on cardiovascular outcomes, including hospitalization for heart failure (HHF), postoperative atrial fibrillation, and cardiovascular death, in an unselected patient population after cardiac surgery. Methods and Results Patients undergoing elective coronary artery bypass graft and/or cardiac valve surgery were prospectively enrolled. FGF-23 blood plasma concentrations were assessed before surgery. A composite of cardiovascular death/HHF was chosen as primary end point. A total of 451 patients (median age 70 years; 28.8% female) were included in the present analysis and followed over a median of 3.9 years. Individuals with higher FGF-23 quartiles showed elevated incidence rates of the composite of cardiovascular death/HHF (quartile 1, 7.1%; quartile 2, 8.6%; quartile 3, 15.1%; and quartile 4, 34.3%). After multivariable adjustment, FGF-23 modeled as a continuous variable (adjusted hazard ratio for a 1-unit increase in standardized log-transformed biomarker, 1.82 [95% CI, 1.34-2.46]) as well as using predefined risk groups and quartiles remained independently associated with the risk of cardiovascular death/HHF and the secondary outcomes, including postoperative atrial fibrillation. Reclassification analysis indicated that the addition of FGF-23 to N-terminal pro-B-type natriuretic peptide provides a significant improvement in risk discrimination (net reclassification improvement at the event rate, 0.58 [95% CI, 0.34-0.81]; P<0.001; integrated discrimination increment, 0.03 [95% CI, 0.01-0.05]; P<0.001). Conclusions FGF-23 is an independent predictor of cardiovascular death/HHF and postoperative atrial fibrillation in individuals undergoing cardiac surgery. Considering an individualized risk assessment, routine preoperative FGF-23 evaluation may improve detection of high-risk patients.
Collapse
Affiliation(s)
- Felix Hofer
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | - Andreas Hammer
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | | | - Lorenz Koller
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | - Niema Kazem
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | - Eva Steinacher
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | | | - Martin Andreas
- Department of Cardiac Surgery Medical University of Vienna Vienna Austria
| | - Günther Laufer
- Department of Cardiac Surgery Medical University of Vienna Vienna Austria
| | - Johann Wojta
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | - Thomas A Zelniker
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | - Christian Hengstenberg
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | - Alexander Niessner
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| | - Patrick Sulzgruber
- Division of Cardiology, Department of Internal Medicine II Medical University of Vienna Vienna Austria
| |
Collapse
|
20
|
FGF23 in Chronic Kidney Disease: Bridging the Heart and Anemia. Cells 2023; 12:cells12040609. [PMID: 36831276 PMCID: PMC9954184 DOI: 10.3390/cells12040609] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Fibroblast growth factor 23 (FGF23) is a phosphaturic hormone produced mainly in osteocytes. In chronic kidney disease (CKD) FGF23 levels increase due to higher production, but also as the result of impaired cleavage and reduced excretion from the body. FGF23 has a significant role in disturbed bone and mineral metabolism in CKD, which leads to a higher cardiovascular risk and mortality in these patients. Current research has emphasized the expression of FGF23 in cardiac myocytes, fibroblasts, and endothelial cells, and in addition to the effects on the kidney, its primary role is in cardiac remodeling in CKD patients. Recent discoveries found a significant link between increased FGF23 levels and anemia development in CKD. This review describes the FGF23 role in cardiac hypertrophy and anemia in the setting of CKD and discusses the best therapeutical approach for lowering FGF23 levels.
Collapse
|
21
|
Elzayat RS, Bahbah WA, Elzaiat RS, Elgazzar BA. Fibroblast growth factor 23 in children with or without heart failure: a prospective study. BMJ Paediatr Open 2023; 7:e001753. [PMID: 36828640 PMCID: PMC9972412 DOI: 10.1136/bmjpo-2022-001753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 02/01/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Elevated fibroblast growth factor 23 (FGF23) levels have been associated with mortality in adults with heart failure (HF), but data on FGF23 levels in paediatric HF are limited. In this prospective cohort study, we aimed to assess the prognostic value of FGF23 in children with chronic HF. METHODS We prospectively enrolled 40 children with chronic HF and 20 matched healthy controls. In each patient, a complete diagnostic workup was performed, including transthoracic echocardiography to evaluate cardiac systolic and diastolic functions. Serum FGF23, renal function tests, parathyroid hormone, serum calcium and phosphate were measured in patients and controls. N-terminal probrain natriuretic peptide (NT-proBNP) was measured in patients. The severity of symptoms was assessed using the modified Ross HF classification for children. Patients were followed for 1 year, and clinical worsening events such as death and HF hospitalisation were recorded. RESULTS Patients with HF had significantly higher FGF23 levels compared with controls (355.68±97.27 pg/mL and 60.20±11.04 pg/mL, respectively; p<0.001). Three patients died and 11 were admitted with HF. In comparison with patients without clinical worsening events, these 14 patients exhibited significantly higher FGF23 levels (320.04±89.56 pg/mL and 421.86±75.50 pg/mL, respectively; p<0.001). FGF23 was positively correlated with NT-proBNP and left ventricular end-diastolic diameter and negatively correlated with ejection fraction and fractional shortening. The ability of FGF23 to predict clinical worsening events in patients was analysed using a receiver operating characteristic curve. The optimal cut-off point was 375 pg/mL, with 85.71% sensitivity, 84.62% specificity, positive predictive value of 75.0, negative predictive value of 91.7 and area under the curve (AUC) of 0.878. Multivariable regression analysis revealed that FGF23 is the only independent predictor of clinical worsening events in children with chronic HF. CONCLUSION FGF23 levels were elevated in children with chronic HF and increased significantly as Ross score class increased. FGF23 levels increased in patients who experienced clinical worsening events.
Collapse
Affiliation(s)
| | - Wael Abbas Bahbah
- Pediatrics, Menoufia University Faculty of Medicine, Shebin El-Kom, Egypt
| | - Reham Salah Elzaiat
- Clinical Pathology, Menoufia University Faculty of Medicine, Shebin El-Kom, Egypt
| | | |
Collapse
|
22
|
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
|
23
|
Saito T, Mizobuchi M, Kato T, Ogata H, Koiwa F, Honda H. Fibroblast Growth Factor 23 Exacerbates Cardiac Fibrosis in Deoxycorticosterone Acetate-Salt Mice With Hypertension. J Transl Med 2023; 103:100003. [PMID: 36748187 DOI: 10.1016/j.labinv.2022.100003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/26/2022] [Accepted: 09/20/2022] [Indexed: 01/18/2023] Open
Abstract
Fibroblast growth factor 23 (FGF23) is associated with cardiovascular disease in patients with chronic kidney disease; however, the mechanisms underlying the effect of FGF23 on cardiac function remain to be investigated. Herein, we studied the effect of continuous intravenous (CIV) FGF23 loading in a deoxycorticosterone acetate (DOCA)-salt mouse model with mild chronic kidney disease and hypertension as well as heart failure with a preserved ejection fraction. Wild-type male mice were randomly allocated to 4 groups: normal control, vehicle-treated DOCA-salt mice, FGF23-treated DOCA-salt mice, and FGF23- and calcitriol-treated DOCA-salt mice. The DOCA-salt mice received the agents via the CIV route for 10 days using an infusion minipump. DOCA-salt mice that received FGF23 showed a marked increase in the serum FGF23 level, and echocardiography in these mice revealed heart failure with a preserved ejection fraction. These mice also showed exacerbation of myocardial fibrosis, concomitant with an inverse and significant correlation with Cyp27b1 expression. Calcitriol treatment attenuated FGF23-induced cardiac fibrosis and improved diastolic function via inhibition of transforming growth factor-β signaling. This effect was independent of the systemic and local levels of FGF23. These results suggest that CIV FGF23 loading exacerbates cardiac fibrosis and that locally abnormal vitamin D metabolism is involved in this mechanism. Calcitriol attenuates this exacerbation by mediating transforming growth factor-β signaling independently of the FGF23 levels.
Collapse
Affiliation(s)
- Tomohiro Saito
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Masahide Mizobuchi
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan.
| | - Tadashi Kato
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hiroaki Ogata
- Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
| | - Fumihiko Koiwa
- Division of Nephrology, Department of Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Hirokazu Honda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Cipriani C, Minisola S, Colangelo L, DE Martino V, Ferrone F, Biamonte F, Danese V, Sonato C, Santori R, Occhiuto M, Pepe J. FGF23 functions and disease. Minerva Endocrinol (Torino) 2022; 47:437-448. [PMID: 33792238 DOI: 10.23736/s2724-6507.21.03378-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The main function of fibroblast growth factor 23 (FGF23) is the regulation of phosphate metabolism through its action on the sodium-dependent phosphate cotransporters in the proximal renal tubules. Additionally, FGF23 interacts with vitamin D and parathyroid hormone in a complex metabolic pathway whose detailed mechanisms are still not clear in human physiology and disease. More recently, research has also focused on the understanding of mechanisms of FGF23 action on organs and system other than the kidneys and bone, as well as on its interaction with other metabolic pathways. Collectively, the new evidence are successfully used for the clinical evaluation and management of FGF23-related disorders, for which new therapies with many potential applications are now available.
Collapse
Affiliation(s)
- Cristiana Cipriani
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University, Rome, Italy -
| | - Salvatore Minisola
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | - Luciano Colangelo
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | - Viviana DE Martino
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | - Federica Ferrone
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | - Federica Biamonte
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | - Vittoria Danese
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | - Chiara Sonato
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | - Rachele Santori
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | - Marco Occhiuto
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | - Jessica Pepe
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University, Rome, Italy
| |
Collapse
|
25
|
Liu M, Xia P, Tan Z, Song T, Mei K, Wang J, Ma J, Jiang Y, Zhang J, Zhao Y, Yu P, Liu X. Fibroblast growth factor-23 and the risk of cardiovascular diseases and mortality in the general population: A systematic review and dose-response meta-analysis. Front Cardiovasc Med 2022; 9:989574. [PMID: 36407457 PMCID: PMC9669381 DOI: 10.3389/fcvm.2022.989574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/06/2022] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND In the past decade, fibroblast growth factor 23 (FGF23) has been recognized as an important biomarker of cardiovascular diseases. This study aimed to assess the relationship between FGF23 and the risk of cardiovascular diseases (CVDs) in general populations. METHODS The protocol was registered prospectively in PROSPERO (CRD42021281837) and two authors independently searched for relevant studies in the PubMed, EMBASE, and Cochrane Library databases. The random effects model was applied. RESULTS In total, 29 prospective studies involving 135,576 participants were included. In the general population, the category analysis revealed that elevated FGF23 levels were related to increased risks of myocardial infarction (MI) (RR: 1.40, 95%CI: 1.03-1.89), stroke (RR: 1.20, 95%CI: 1.02-1.43), heart failure (HF) (RR: 1.37, 95%CI: 1.23-1.52), CVD events (RR: 1.22, 95%CI: 0.99-1.51), cardiovascular mortality (RR: 1.46, 95%CI: 1.29-1.65), and all-cause mortality (RR: 1.50, 95%CI: 1.29-1.74). In the continuous analysis, per doubling of FGF23 was associated with increased risks of MI (RR: 1.08, 95%CI: 0.94-1.25), stroke (RR: 1.21, 95%CI: 0.99-1.48), HF (RR: 1.24, 95%CI: 1.14-1.35), CVD events (RR: 1.12, 95%CI: 0.99-1.27), cardiovascular mortality (RR: 1.43, 95%CI: 1.09-1.88), all-cause mortality (RR: 1.37, 95%CI: 1.15-1.62). Furthermore, the dose-response analysis demonstrated a potentially non-linear relationship between FGF23 and stroke, HF, and all-cause mortality. In contrast, a potentially linear relationship between FGF23 and cardiovascular mortality was observed (p for non-linearity = 0.73). CONCLUSION The present study suggests that increased serum FGF23 levels are positively related to CVD events and mortality in the general population. The clinical application of FGF23 levels to predict CVD risk requires further research.
Collapse
Affiliation(s)
- Menglu Liu
- Department of Cardiology, Seventh People’s Hospital of Zhengzhou, Zhengzhou, China
| | - Panpan Xia
- Department of Endocrine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ziqi Tan
- Department of Endocrine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tiangang Song
- Department of Endocrine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Kaibo Mei
- Department of Anesthesiology, People’s Hospital of Shangrao, Shangrao, China
| | - Jingfeng Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jianyong Ma
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United Status
| | - Yuan Jiang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yujie Zhao
- Department of Cardiology, Seventh People’s Hospital of Zhengzhou, Zhengzhou, China
| | - Peng Yu
- Department of Endocrine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiao Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
26
|
Ward Z, Schmeier S, Pearson J, Cameron VA, Frampton CM, Troughton RW, Doughty RN, Richards AM, Pilbrow AP. Identifying Candidate Circulating RNA Markers for Coronary Artery Disease by Deep RNA-Sequencing in Human Plasma. Cells 2022; 11:3191. [PMID: 36291058 PMCID: PMC9599983 DOI: 10.3390/cells11203191] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2023] Open
Abstract
Advances in RNA sequencing (RNA-Seq) have facilitated transcriptomic analysis of plasma for the discovery of new diagnostic and prognostic markers for disease. We aimed to develop a short-read RNA-Seq protocol to detect mRNAs, long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) in plasma for the discovery of novel markers for coronary artery disease (CAD) and heart failure (HF). Circulating cell-free RNA from 59 patients with stable CAD (half of whom developed HF within 3 years) and 30 controls was sequenced to a median depth of 108 paired reads per sample. We identified fragments from 3986 messenger RNAs (mRNAs), 164 long non-coding RNAs (lncRNAs), 405 putative novel lncRNAs and 227 circular RNAs in plasma. Circulating levels of 160 mRNAs, 10 lncRNAs and 2 putative novel lncRNAs were altered in patients compared with controls (absolute fold change >1.2, p < 0.01 adjusted for multiple comparisons). The most differentially abundant transcripts were enriched in mRNAs encoded by the mitochondrial genome. We did not detect any differences in the plasma RNA profile between patients who developed HF compared with those who did not. In summary, we show that mRNAs, lncRNAs and circular RNAs can be reliably detected in plasma by deep RNA-Seq. Multiple coding and non-coding transcripts were altered in association with CAD, including several mitochondrial mRNAs, which may indicate underlying myocardial ischaemia and oxidative stress. If validated, circulating levels of these transcripts could potentially be used to help identify asymptomatic individuals with established CAD prior to an acute coronary event.
Collapse
Affiliation(s)
- Zoe Ward
- Christchurch Heart Institute, Department of Medicine, University of Otago—Christchurch, Christchurch 8140, New Zealand
| | - Sebastian Schmeier
- School of Natural and Computational Sciences, Massey University, Auckland 0632, New Zealand
- Evotec SE, Essener Bogen 7, 22419 Hamburg, Germany
| | - John Pearson
- Biostatistics and Computational Biology Unit, University of Otago—Christchurch, Christchurch 8140, New Zealand
| | - Vicky A Cameron
- Christchurch Heart Institute, Department of Medicine, University of Otago—Christchurch, Christchurch 8140, New Zealand
| | - Chris M Frampton
- Christchurch Heart Institute, Department of Medicine, University of Otago—Christchurch, Christchurch 8140, New Zealand
| | - Richard W Troughton
- Christchurch Heart Institute, Department of Medicine, University of Otago—Christchurch, Christchurch 8140, New Zealand
| | - Rob N Doughty
- Heart Health Research Group, University of Auckland, Auckland 1023, New Zealand
| | - A. Mark Richards
- Christchurch Heart Institute, Department of Medicine, University of Otago—Christchurch, Christchurch 8140, New Zealand
- Cardiovascular Research Institute, National University of Singapore, Singapore 119228, Singapore
| | - Anna P Pilbrow
- Christchurch Heart Institute, Department of Medicine, University of Otago—Christchurch, Christchurch 8140, New Zealand
| |
Collapse
|
27
|
Nandakumar M, Moin ASM, Ramanjaneya M, Qaissi AA, Sathyapalan T, Atkin SL, Butler AE. Severe iatrogenic hypoglycaemia modulates the fibroblast growth factor protein response. Diabetes Obes Metab 2022; 24:1483-1497. [PMID: 35415885 DOI: 10.1111/dom.14716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/31/2022] [Accepted: 04/08/2022] [Indexed: 11/30/2022]
Abstract
INTRODUCTION There is evidence that fibroblast growth factor (FGF) levels may be implicated in hypoglycaemia, with FGF19 being a potential contributor to insulin-independent pathways driving postprandial hypoglycaemia following bariatric surgery and basic FGF (FGF2) being elevated following mild hypoglycaemia occurring after the glucose tolerance test. However, their response following severe iatrogenic hypoglycaemia is unknown and therefore this pilot exploratory study was undertaken. METHODS A case-control study of aged-matched type 2 diabetes (T2D; n = 23) and control (n = 23) subjects who underwent a hyperinsulinaemic clamp, initially to euglycaemia in T2D (5 mmol/L; 90 mg/dl), and then to hypoglycaemia (<2 mmol/L; <36 mg/dl) with subsequent follow-up time course to 24 h. FGF and FGF receptor proteins were determined by Slow Off-rate Modified Aptamer (SOMA)-scan plasma protein measurement. RESULTS At baseline, FGF12 (p = .006) was higher and FGF20 (p = .004) was lower in T2D versus controls. At hypoglycaemia, FGF7 was lower in T2D. Post-hypoglycaemic levels of FGF18, FGF19, FGF20 and FGF23 were lower while FGF12 and FGF16 were higher in T2D versus control at different time points. No differences between T2D and controls were seen for FGF1, FGF2, FGF4, FGF6, FGF8, FGF9, FGF10, FGF21 or any of the FGF receptors. At 24 h post-hypoglycaemia, FGF20 (p = .01) differed between controls and T2D, while the levels for the other proteins measured returned to baseline. None of the FGF proteins altered from baseline to euglycaemia when clamped in T2D subjects. FGF23 negatively correlated with fasting blood glucose, but no FGFs correlated with body mass index in T2D. CONCLUSION Severe transient hypoglycaemia modulated FGF7, 16, 19, 20 and 23 (known to be associated with diabetes), together with FGF18 and 12, not previously reported to be associated with diabetes but that may be important in the pathophysiology of hypoglycaemia; FGF20 remained low at 24 h. Taken together, these data suggest that recurrent hypoglycaemia may contribute to the development of complications through changes in FGF proteins.
Collapse
Affiliation(s)
- Manjula Nandakumar
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Abu Saleh Md Moin
- Royal College of Surgeons in Ireland Bahrain, Adliya, Kingdom of Bahrain
| | - Manjunath Ramanjaneya
- Qatar Metabolic Institute, Hamad Medical Corporation, Doha, Qatar
- Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Ahmed Al Qaissi
- Academic Endocrinology, Diabetes and Metabolism, Hull York Medical School, Hull, UK
| | | | - Stephen L Atkin
- Royal College of Surgeons in Ireland Bahrain, Adliya, Kingdom of Bahrain
| | - Alexandra E Butler
- Royal College of Surgeons in Ireland Bahrain, Adliya, Kingdom of Bahrain
| |
Collapse
|
28
|
Akwo E, Pike MM, Ertuglu LA, Vartanian N, Farber-Eger E, Lipworth L, Perwad F, Siew E, Hung A, Bansal N, de Boer I, Kestenbaum B, Cox NJ, Ikizler TA, Wells Q, Robinson-Cohen C. Association of Genetically Predicted Fibroblast Growth Factor-23 with Heart Failure: A Mendelian Randomization Study. Clin J Am Soc Nephrol 2022; 17:1183-1193. [PMID: 35902130 PMCID: PMC9435988 DOI: 10.2215/cjn.00960122] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 05/31/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND OBJECTIVES Elevated fibroblast growth factor-23 (FGF23) has been consistently associated with heart failure, particularly heart failure with preserved ejection fraction, among patients with CKD and in the general population. FGF23 may directly induce cardiac remodeling and heart failure. However, biases affecting observational studies impede robust causal inferences. Mendelian randomization leverages genetic determinants of a risk factor to examine causality. We performed a two-sample Mendelian randomization to assess causal associations between FGF23 and heart failure. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Genetic instruments were genome-wide significant genetic variants associated with FGF23, including variants near PIP5K1B, RGS14, LINC01229, and CYP24A1. We analyzed data from the Heart Failure Molecular Epidemiology for Therapeutic Targets and BioVU biobanks to examine associations of the four variants with overall heart failure, heart failure with preserved ejection fraction, and heart failure with reduced and mid-range ejection fraction. We developed an eGFR polygenic risk score using summary statistics from the Chronic Kidney Disease Genetics Consortium (CKDGen) genome-wide association study of eGFR in >1 million individuals and performed stratified analyses across eGFR polygenic risk score strata. RESULTS Genetically determined FGF23 was not associated with overall heart failure in the Heart Failure Molecular Epidemiology for Therapeutic Targets consortium (odds ratio, 1.13; 95% confidence interval, 0.89 to 1.42 per unit higher genetically predicted log FGF23) and the full BioVU sample (odds ratio, 1.32; 95% confidence interval, 0.95 to 1.84). In stratified analyses in BioVU, higher FGF23 was associated with overall heart failure (odds ratio, 3.09; 95% confidence interval, 1.38 to 6.91) among individuals with low eGFR-polygenic risk score (<1 SD below the mean), but not those with high eGFR-polygenic risk score (P interaction = 0.02). Higher FGF23 was also associated with heart failure with preserved ejection fraction among all BioVU participants (odds ratio, 1.47; 95% confidence interval, 1.01 to 2.14) and individuals with low eGFR-polygenic risk score (odds ratio, 7.20; 95% confidence interval, 2.80 to 18.49), but not those high eGFR-polygenic risk score (P interaction = 2.25 × 10-4). No significant associations were observed with heart failure with reduced and midrange ejection fraction. CONCLUSION We found no association between genetically predicted FGF23 and heart failure in the Heart Failure Molecular Epidemiology for Therapeutic Targets consortium. In BioVU, genetically elevated FGF23 was associated with higher heart failure risk, specifically heart failure with preserved ejection fraction, particularly among individuals with low genetically predicted eGFR. PODCAST This article contains a podcast at https://www.asn-online.org/media/podcast/CJASN/2022_07_28_CJN00960122.mp3.
Collapse
Affiliation(s)
- Elvis Akwo
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mindy M. Pike
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee,Division of Cardiovascular Medicine, Division of Epidemiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lale A. Ertuglu
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Nicholas Vartanian
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Eric Farber-Eger
- Division of Cardiovascular Medicine, Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University Medical Center, Nashville, Tennessee,Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Loren Lipworth
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee,Division of Cardiovascular Medicine, Division of Epidemiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Farzana Perwad
- Division of Pediatric Nephrology, University of California San Francisco, San Francisco, California
| | - Edward Siew
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee,Division of Nephrology, Vanderbilt Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Adriana Hung
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee,Division of Nephrology, Vanderbilt Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Nisha Bansal
- Division of Nephrology, Vanderbilt Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Ian de Boer
- Division of Nephrology, University of Washington, Seattle, Washington
| | - Bryan Kestenbaum
- Division of Nephrology, University of Washington, Seattle, Washington
| | - Nancy J. Cox
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - T. Alp Ikizler
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Quinn Wells
- Division of Cardiovascular Medicine, Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University Medical Center, Nashville, Tennessee,Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | | |
Collapse
|
29
|
Ornitz DM, Itoh N. New developments in the biology of fibroblast growth factors. WIREs Mech Dis 2022; 14:e1549. [PMID: 35142107 PMCID: PMC10115509 DOI: 10.1002/wsbm.1549] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/28/2023]
Abstract
The fibroblast growth factor (FGF) family is composed of 18 secreted signaling proteins consisting of canonical FGFs and endocrine FGFs that activate four receptor tyrosine kinases (FGFRs 1-4) and four intracellular proteins (intracellular FGFs or iFGFs) that primarily function to regulate the activity of voltage-gated sodium channels and other molecules. The canonical FGFs, endocrine FGFs, and iFGFs have been reviewed extensively by us and others. In this review, we briefly summarize past reviews and then focus on new developments in the FGF field since our last review in 2015. Some of the highlights in the past 6 years include the use of optogenetic tools, viral vectors, and inducible transgenes to experimentally modulate FGF signaling, the clinical use of small molecule FGFR inhibitors, an expanded understanding of endocrine FGF signaling, functions for FGF signaling in stem cell pluripotency and differentiation, roles for FGF signaling in tissue homeostasis and regeneration, a continuing elaboration of mechanisms of FGF signaling in development, and an expanding appreciation of roles for FGF signaling in neuropsychiatric diseases. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology Congenital Diseases > Stem Cells and Development Cancer > Stem Cells and Development.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nobuyuki Itoh
- Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto, Japan
| |
Collapse
|
30
|
Nath M, Romaine SP, Koekemoer A, Hamby S, Webb TR, Nelson CP, Castellanos‐Uribe M, Papakonstantinou M, Anker SD, Lang CC, Metra M, Zannad F, Filippatos G, van Veldhuisen DJ, Cleland JG, Ng LL, May ST, Marelli‐Berg F, Voors AA, Timmons JA, Samani NJ. Whole blood transcriptomic profiling identifies molecular pathways related to cardiovascular mortality in heart failure. Eur J Heart Fail 2022; 24:1009-1019. [PMID: 35570197 PMCID: PMC9546237 DOI: 10.1002/ejhf.2540] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/08/2022] [Accepted: 03/18/2022] [Indexed: 12/01/2022] Open
Abstract
AIMS Chronic heart failure (CHF) is a systemic syndrome with a poor prognosis and a need for novel therapies. We investigated whether whole blood transcriptomic profiling can provide new mechanistic insights into cardiovascular (CV) mortality in CHF. METHODS AND RESULTS Transcriptome profiles were generated at baseline from 944 CHF patients from the BIOSTAT-CHF study, of whom 626 survived and 318 died from a CV cause during a follow-up of 21 months. Multivariable analysis, including adjustment for cell count, identified 1153 genes (6.5%) that were differentially expressed between those that survived or died and strongly related to a validated clinical risk score for adverse prognosis. The differentially expressed genes mainly belonged to five non-redundant pathways: adaptive immune response, proteasome-mediated ubiquitin-dependent protein catabolic process, T-cell co-stimulation, positive regulation of T-cell proliferation, and erythrocyte development. These five pathways were selectively related (RV coefficients >0.20) with seven circulating protein biomarkers of CV mortality (fibroblast growth factor 23, soluble ST2, adrenomedullin, hepcidin, pentraxin-3, WAP 4-disulfide core domain 2, and interleukin-6) revealing an intricate relationship between immune and iron homeostasis. The pattern of survival-associated gene expression matched with 29 perturbagen-induced transcriptome signatures in the iLINCS drug-repurposing database, identifying drugs, approved for other clinical indications, that were able to reverse in vitro the molecular changes associated with adverse prognosis in CHF. CONCLUSION Systematic modelling of the whole blood protein-coding transcriptome defined molecular pathways that provide a link between clinical risk factors and adverse CV prognosis in CHF, identifying both established and new potential therapeutic targets.
Collapse
Affiliation(s)
- Mintu Nath
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
- Institute of Applied Health SciencesUniversity of AberdeenAberdeenUK
| | - Simon P.R. Romaine
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
| | - Andrea Koekemoer
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
| | - Stephen Hamby
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
| | - Thomas R. Webb
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
| | - Christopher P. Nelson
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
| | | | - Manolo Papakonstantinou
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
| | - Stefan D. Anker
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Chim C. Lang
- Division of Molecular and Clinical Medicine, School of MedicineUniversity of DundeeDundeeUK
| | - Marco Metra
- Department of Medical and Surgical Specialties, Radiological Sciences and Public HealthUniversity of BresciaBresciaItaly
| | - Faiez Zannad
- Clinical Investigation Center 1433, Centre Hospitalier Regional et Universitaire de NancyVandoeuvre les NancyFrance
| | | | - Dirk J. van Veldhuisen
- Department of Cardiology, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - John G. Cleland
- National Heart and Lung Institute, Royal Brompton and Harefield Hospitals, Imperial College, London, UK and Robertson Centre for Biostatistics and Clinical TrialsUniversity of GlasgowGlasgowUK
| | - Leong L. Ng
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
| | - Sean T. May
- School of BiosciencesUniversity of Nottingham, Sutton Bonington CampusLoughboroughUK
| | | | - Adriaan A. Voors
- Department of Cardiology, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - James A. Timmons
- Barts & The London School of MedicineQueen Mary University of LondonLondonUK
- Augur Precision Medicine LtdStirling University Innovation ParkUK
| | - Nilesh J. Samani
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
| |
Collapse
|
31
|
Rabkin SW. Evaluating the adverse outcome of subtypes of heart failure with preserved ejection fraction defined by machine learning: A systematic review focused on defining high risk phenogroups. EXCLI JOURNAL 2022; 21:487-518. [PMID: 35391918 PMCID: PMC8983850 DOI: 10.17179/excli2021-4572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/12/2022] [Indexed: 11/24/2022]
Abstract
The ability to distinguish clinically meaningful subtypes of heart failure with preserved ejection fraction (HFpEF) has recently been examined by machine learning techniques but studies appear to have produced discordant results. The objective of this study is to synthesize the types of HFpEF by examining their features and relating them to phenotypes with adverse prognosis. A systematic search was conducted using the search terms "Diastolic Heart Failure" OR "heart failure with preserved ejection fraction" OR "heart failure with normal ejection fraction" OR "HFpEF" AND "machine learning" OR "artificial intelligence" OR 'computational biology'. Ten studies were identified and they varied in their prevalence of ten clinical variables: age, sex, body mass index (BMI) or obesity, hypertension, diabetes mellitus, coronary artery disease, atrial fibrillation, chronic kidney disease, chronic obstructive pulmonary disease or symptom severity (NYHA class or BNP). The clinical findings associated with the different phenotypes in > 85 % of studies were age, hypertension, atrial fibrillation, chronic kidney disease and worse symptoms severity; an adverse outcome was in 65 % to 85 % of studies identified diabetes mellitus and female sex and in less than 65 % of studies was body mass index or obesity, and coronary artery disease. COPD was a relevant factor in only 33 % of studies. Adverse clinical outcome - death or admission to hospital (for heart failure) defined phenogroups with the worst outcome. Combining the 4 studies that calculated the MAGGIC score showed a significant (p<0.05) linear relationship between MAGGIC score and outcome, using the one-year event rate. A new score based on strength of the evidence of the HFpEF studies analyzed here, using 9 variables (eliminating COPD), showed a significant (p<0.009) linear relationship with one-year event rate. Three studies examined biomarkers in detail and the ones most prominently related to outcome or consistently found in the studies were GDF15, FABP4, FGF23, sST2, renin and TNF. The dominant factors that identified phenotypes of HFpEF with adverse outcome were hypertension, atrial fibrillation, chronic kidney disease and worse symptoms severity. A new simplified score, based on clinical factors, was proposed to assess prognosis in HFpEF. Several biomarkers were consistently elevated in phenogroups with adverse outcomes and may indicate the underlying mechanism or pathophysiology specific for phenotypes with an adverse prognosis.
Collapse
Affiliation(s)
- Simon W. Rabkin
- University of British Columbia,*To whom correspondence should be addressed: Simon W. Rabkin, University of British Columbia, 9th Floor 2775 Laurel St., Vancouver, B.C., Canada V5Z 1M9; Phone: (604) 875 5847, Fax: (604) 875 5849, E-mail:
| |
Collapse
|
32
|
Alber J, Föller M. Lactic acid induces fibroblast growth factor 23 (FGF23) production in UMR106 osteoblast-like cells. Mol Cell Biochem 2022; 477:363-370. [PMID: 34731356 PMCID: PMC8800909 DOI: 10.1007/s11010-021-04287-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/23/2021] [Indexed: 01/04/2023]
Abstract
Endocrine and paracrine fibroblast growth factor 23 (FGF23) is a protein predominantly produced by bone cells with strong impact on phosphate and vitamin D metabolism by targeting the kidney. Plasma FGF23 concentration early rises in kidney and cardiovascular diseases correlating with progression and outcome. Lactic acid is generated in anaerobic glycolysis. Lactic acidosis is the consequence of various physiological and pathological conditions and may be fatal. Since FGF23 production is stimulated by inflammation and lactic acid induces pro-inflammatory signaling, we investigated whether and how lactic acid influences FGF23. Experiments were performed in UMR106 osteoblast-like cells, Fgf23 mRNA levels estimated from quantitative real-time polymerase chain reaction, and FGF23 protein determined by enzyme-linked immunosorbent assay. Lactic acid dose-dependently induced Fgf23 gene expression and up-regulated FGF23 synthesis. Also, Na+-lactate as well as formic acid and acetic acid up-regulated Fgf23. The lactic acid effect was significantly attenuated by nuclear factor kappa-light-chain enhancer of activated B-cells (NFκB) inhibitors wogonin and withaferin A. Lactic acid induces FGF23 production, an effect at least in part mediated by NFκB. Lactic acidosis may, therefore, be paralleled by a surge in plasma FGF23.
Collapse
Affiliation(s)
- Jana Alber
- Department of Physiology, University of Hohenheim, Garbenstraße 30, 70599, Stuttgart, Germany
| | - Michael Föller
- Department of Physiology, University of Hohenheim, Garbenstraße 30, 70599, Stuttgart, Germany.
| |
Collapse
|
33
|
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
|
34
|
Alehagen U, Aaseth J, Larsson A, Alexander J. Decreased Concentration of Fibroblast Growth Factor 23 (FGF-23) as a Result of Supplementation with Selenium and Coenzyme Q 10 in an Elderly Swedish Population: A Sub-Analysis. Cells 2022; 11:cells11030509. [PMID: 35159318 PMCID: PMC8834214 DOI: 10.3390/cells11030509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 12/14/2022] Open
Abstract
There is a reduced intake of selenium in many countries due to low levels of selenium in the soil. This results in an increased cardiovascular risk. Fibroblast growth factor 23 (FGF-23) is active mainly in the metabolism of vitamin D and phosphorus. However, there are indications that FGF-23 may also provide information both on cardiovascular function and prognosis. The aim of the study was to evaluate the effect of supplementation with selenium and coenzyme Q10 on the FGF-23 concentration in an elderly population with low concentrations of both selenium and coenzyme Q10 and in which the supplementation improved cardiac function and mortality. In a randomised double-blind placebo-controlled trial, FGF-23 was measured in 219 individuals at the start and after 48 months. Selenium yeast (200 µg/day) and coenzyme Q10 (200 mg/day) (n = 118) or placebo (n = 101) were given as a dietary supplement. The intervention time was 48 months. t-Tests, repeated measures of variance, and ANCOVA analyses were used to evaluate the differences in FGF-23 concentration. Following supplementation with selenium and coenzyme Q10, a significantly lower level of FGF-23 could be seen (p = 0.01). Applying 10 years of follow-up, those who later died a cardiovascular death had a significantly higher FGF-23 concentration after 48 months compared with those who survived (p = 0.036), and a significantly lower FGF-23 concentration could be seen in those with a normal renal function compared to those with an impaired renal function (p = 0.027). Supplementation with selenium and coenzyme Q10 to an elderly community-living population low in both substances prevented an increase of FGF-23 and also provided a reduced cardiovascular risk.
Collapse
Affiliation(s)
- Urban Alehagen
- Division of Cardiovascular Medicine, Department of Health, Medicine and Caring Sciences, Linköping University, 81 85 Linköping, Sweden
- Correspondence: ; Tel.: +46-10-103-0000
| | - Jan Aaseth
- Research Department, Innlandet Hospital Trust, 2381 Brumunddal, Norway;
| | - Anders Larsson
- Department of Medical Sciences, Uppsala University, 751 85 Uppsala, Sweden;
| | - Jan Alexander
- Norwegian Institute of Public Health, 0403 Oslo, Norway;
| |
Collapse
|
35
|
Kumari R, Dutta R, Ranjan P, Suleiman ZG, Goswami SK, Li J, Pal HC, Verma SK. ALKBH5 Regulates SPHK1-Dependent Endothelial Cell Angiogenesis Following Ischemic Stress. Front Cardiovasc Med 2022; 8:817304. [PMID: 35127873 PMCID: PMC8811170 DOI: 10.3389/fcvm.2021.817304] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/20/2021] [Indexed: 12/29/2022] Open
Abstract
Background Endothelial cells dysfunction has been reported in many heart diseases including acute myocardial infarction, and atherosclerosis. The molecular mechanism for endothelial dysfunction in the heart is still not clearly understood. We aimed to study the role of m6A RNA demethylase alkB homolog 5 (ALKBH5) in ECs angiogenesis during ischemic injury. Methods and Results ECs were treated with ischemic insults (lipopolysaccharide and 1% hypoxia) to determine the role of ALKBH5 in ECs angiogenesis. siRNA mediated ALKBH5 gene silencing was used for examining the loss of function. In this study, we report that ALKBH5 levels are upregulated following ischemia and are associated with maintaining ischemia-induced ECs angiogenesis. To decipher the mechanism of action, we found that ALKBH5 is required to maintain eNOS phosphorylation and SPHK1 protein levels. ALKBH5 silencing alone or with ischemic stress significantly increased SPHK1 m6A mRNA methylation. In contrast, METTL3 (RNA methyltransferase) overexpression resulted in the reduced expression of SPHK1. Conclusion We reported that ALKBH5 helps in the maintenance of angiogenesis in endothelial cells following acute ischemic stress via reduced SPHK1 m6A methylation and downstream eNOS-AKT signaling.
Collapse
Affiliation(s)
- Rajesh Kumari
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Roshan Dutta
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Prabhat Ranjan
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Zainab Gbongbo Suleiman
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sumanta Kumar Goswami
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jing Li
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Harish Chandra Pal
- Department of Pathology, Molecular and Cellular Pathology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Suresh Kumar Verma
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
- *Correspondence: Suresh Kumar Verma
| |
Collapse
|
36
|
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
|
37
|
Cardiac fibrosis and atrial fibrillation. POSTEP HIG MED DOSW 2022. [DOI: 10.2478/ahem-2022-0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Cardiac fibrosis is characterized by the imbalance of production and degradation of the extracellular matrix. The result of this process is an accumulation of scar tissue, which is associated with many pathological processes such as excessive mechanical stress on the heart, inflammation, ischemia, oxidative stress, or excessive neurohormonal activation. Fibrotic response results in damaged heart architecture and dysfunction of the heart. Cardiac fibrosis leads to increased stiffness of the left ventricle and arteries, promotes disorders of contraction and relaxation of the heart, disrupts electrophysiology of heart cells, and induces arrhythmias.
Atrial fibrillation is one of the most common arrhythmias. It is associated with a deterioration in the quality of life and more frequent use of medical assistance. It is also an instantaneous risk factor for many diseases, including stroke. The underlying cause of this arrhythmia is electrical and structural remodeling induced by cardiac fibrosis. Therefore, much attention is paid to the search for biochemical markers that would allow non-invasive determination of the degree of this fibrosis.
The promising markers include galectin-3, human epididymis protein 4 (HE4), serum soluble ST2, and adipose triglyceride lipase (ATGL). Studies have shown that plasma concentrations of these substances reflect the degree of myocardial fibrosis and are indirectly associated with AF.
There are high hopes for the use of these markers in patients undergoing arrhythmia ablation. More research is needed to confirm that these markers can be used to estimate the chance of maintaining sinus rhythm in patients after ablation.
Collapse
|
38
|
Nashawi M, Ahmed MS, Amin T, Abualfoul M, Chilton R. Cardiovascular benefits from SGLT2 inhibition in type 2 diabetes mellitus patients is not impaired with phosphate flux related to pharmacotherapy. World J Cardiol 2021; 13:676-694. [PMID: 35070111 PMCID: PMC8716977 DOI: 10.4330/wjc.v13.i12.676] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/02/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023] Open
Abstract
The beneficial cardiorenal outcomes of sodium-glucose cotransporter 2 inhibitors (SGLT2i) in patients with type 2 diabetes mellitus (T2DM) have been substantiated by multiple clinical trials, resulting in increased interest in the multifarious pathways by which their mechanisms act. The principal effect of SGLT2i (-flozin drugs) can be appreciated in their ability to block the SGLT2 protein within the kidneys, inhibiting glucose reabsorption, and causing an associated osmotic diuresis. This ameliorates plasma glucose elevations and the negative cardiorenal sequelae associated with the latter. These include aberrant mitochondrial metabolism and oxidative stress burden, endothelial cell dysfunction, pernicious neurohormonal activation, and the development of inimical hemodynamics. Positive outcomes within these domains have been validated with SGLT2i administration. However, by modulating the sodium-glucose cotransporter in the proximal tubule (PT), SGLT2i consequently promotes sodium-phosphate cotransporter activity with phosphate retention. Phosphatemia, even at physiologic levels, poses a risk in cardiovascular disease burden, more so in patients with type 2 diabetes mellitus (T2DM). There also exists an association between phosphatemia and renal impairment, the latter hampering cardiovascular function through an array of physiologic roles, such as fluid regulation, hormonal tone, and neuromodulation. Moreover, increased phosphate flux is associated with an associated increase in fibroblast growth factor 23 levels, also detrimental to homeostatic cardiometabolic function. A contemporary commentary concerning this notion unifying cardiovascular outcome trial data with the translational biology of phosphate is scant within the literature. Given the apparent beneficial outcomes associated with SGLT2i administration notwithstanding negative effects of phosphatemia, we discuss in this review the effects of phosphate on the cardiometabolic status in patients with T2DM and cardiorenal disease, as well as the mechanisms by which SGLT2i counteract or overcome them to achieve their net effects. Content drawn to develop this conversation begins with proceedings in the basic sciences and works towards clinical trial data.
Collapse
Affiliation(s)
- Mouhamed Nashawi
- Department of Internal Medicine, Baylor Scott and White All Saints Medical Center, Fort Worth, TX 76132, United States.
| | - Mahmoud S Ahmed
- Division of Medicine-Cardiology, UT Health San Antonio, San Antonio, TX 78229, United States
| | - Toka Amin
- Division of Medicine-Cardiology, UT Health San Antonio, San Antonio, TX 78229, United States
| | - Mujahed Abualfoul
- Department of Internal Medicine, Faculty of Medicine, Cairo University, Dallas, TX 75203, United States
| | - Robert Chilton
- Department of Internal Medicine, Methodist Dallas Medical Center, Dallas, TX 75203, United States
| |
Collapse
|
39
|
Münz S, Feger M, Edemir B, Föller M. Up-Regulation of Fibroblast Growth Factor 23 Gene Expression in UMR106 Osteoblast-like Cells with Reduced Viability. Cells 2021; 11:40. [PMID: 35011602 PMCID: PMC8750768 DOI: 10.3390/cells11010040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 02/07/2023] Open
Abstract
Fibroblast growth factor 23 (FGF23) controls vitamin D and phosphate homeostasis in the kidney and has additional paracrine effects elsewhere. As a biomarker, its plasma concentration is associated with progression of inflammatory, renal, and cardiovascular diseases. Major stimuli of FGF23 synthesis include active vitamin D and inflammation. Antineoplastic chemotherapy treats cancer by inducing cellular damage ultimately favoring cell death (apoptosis and necrosis) and causing inflammation. Our study explored whether chemotherapeutics and other apoptosis inducers impact on Fgf23 expression. Experiments were performed in osteoblast-like UMR106 cells, Fgf23 gene expression and protein synthesis were determined by qRT-PCR and ELISA, respectively. Viability was assessed by MTT assay and NFκB activity by Western Blotting. Antineoplastic drugs cisplatin and doxorubicin as well as apoptosis inducers procaspase-activating compound 1 (PAC-1), a caspase 3 activator, and serum depletion up-regulated Fgf23 transcripts while reducing cell proliferation and viability. The effect of cisplatin on Fgf23 transcription was paralleled by Il-6 up-regulation and NFκB activation and attenuated by Il-6 and NFκB signaling inhibitors. To conclude, cell viability-decreasing chemotherapeutics as well as apoptosis stimulants PAC-1 and serum depletion up-regulate Fgf23 gene expression. At least in part, Il-6 and NFκB may contribute to this effect.
Collapse
Affiliation(s)
- Sina Münz
- Department of Physiology, University of Hohenheim, 70599 Stuttgart, Germany; (S.M.); (M.F.)
| | - Martina Feger
- Department of Physiology, University of Hohenheim, 70599 Stuttgart, Germany; (S.M.); (M.F.)
| | - Bayram Edemir
- Department of Hematology and Oncology, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany;
| | - Michael Föller
- Department of Physiology, University of Hohenheim, 70599 Stuttgart, Germany; (S.M.); (M.F.)
| |
Collapse
|
40
|
Flores-Vergara R, Olmedo I, Aránguiz P, Riquelme JA, Vivar R, Pedrozo Z. Communication Between Cardiomyocytes and Fibroblasts During Cardiac Ischemia/Reperfusion and Remodeling: Roles of TGF-β, CTGF, the Renin Angiotensin Axis, and Non-coding RNA Molecules. Front Physiol 2021; 12:716721. [PMID: 34539441 PMCID: PMC8446518 DOI: 10.3389/fphys.2021.716721] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/26/2021] [Indexed: 11/20/2022] Open
Abstract
Communication between cells is a foundational concept for understanding the physiology and pathology of biological systems. Paracrine/autocrine signaling, direct cell-to-cell interplay, and extracellular matrix interactions are three types of cell communication that regulate responses to different stimuli. In the heart, cardiomyocytes, fibroblasts, and endothelial cells interact to form the cardiac tissue. Under pathological conditions, such as myocardial infarction, humoral factors released by these cells may induce tissue damage or protection, depending on the type and concentration of molecules secreted. Cardiac remodeling is also mediated by the factors secreted by cardiomyocytes and fibroblasts that are involved in the extensive reciprocal interactions between these cells. Identifying the molecules and cellular signal pathways implicated in these processes will be crucial for creating effective tissue-preserving treatments during or after reperfusion. Numerous therapies to protect cardiac tissue from reperfusion-induced injury have been explored, and ample pre-clinical research has attempted to identify drugs or techniques to mitigate cardiac damage. However, despite great success in animal models, it has not been possible to completely translate these cardioprotective effects to human applications. This review provides a current summary of the principal molecules, pathways, and mechanisms underlying cardiomyocyte and cardiac fibroblast crosstalk during ischemia/reperfusion injury. We also discuss pre-clinical molecules proposed as treatments for myocardial infarction and provide a clinical perspective on these potential therapeutic agents.
Collapse
Affiliation(s)
- Raúl Flores-Vergara
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile.,Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile
| | - Ivonne Olmedo
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile.,Red para el Estudio de Enfermedades Cardiopulmonares de alta letalidad (REECPAL), Universidad de Chile, Santiago de Chile, Chile
| | - Pablo Aránguiz
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andrés Bello, Viña del Mar, Chile
| | - Jaime Andrés Riquelme
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile.,Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago de Chile, Chile
| | - Raúl Vivar
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile
| | - Zully Pedrozo
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile.,Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile.,Red para el Estudio de Enfermedades Cardiopulmonares de alta letalidad (REECPAL), Universidad de Chile, Santiago de Chile, Chile
| |
Collapse
|
41
|
Insight into the Pro-inflammatory and Profibrotic Role of Macrophage in Heart Failure With Preserved Ejection Fraction. J Cardiovasc Pharmacol 2021; 76:276-285. [PMID: 32501838 DOI: 10.1097/fjc.0000000000000858] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The prevalence of heart failure (HF) with preserved ejection fraction (HFpEF) is higher than that of HF with reduced/midrange ejection fraction (HFrEF/HFmrEF). However, no evidence-based guidelines for managing HFpEF have been generated. The current body of knowledge indicates that fibrosis and inflammation are important components of the cardiac remodeling process in HFpEF. In addition, macrophages potentially play an important role in pro-inflammatory and profibrotic processes in HFpEF patients, whereas HFpEF comorbidities could be a driving force for systemic microvascular inflammation and endothelial dysfunction. Under such circumstances, macrophages reportedly contribute to inflammation and fibrosis through 3 phases namely, inflammation, repair, and resolution. Signal transduction pathway-targeted therapies using animal experiments have generated important discoveries and breakthroughs for understanding the underlying mechanisms of HFpEF. However, only a handful of studies have reported promising results using human trials. Further investigations are therefore needed to elucidate the exact mechanisms underlying HFpEF and immune-pathogenesis of cardiac fibrosis.
Collapse
|
42
|
Valtuille R. Potential Novel Benefits of Sodium Restriction in Chronic Kidney Disease. Curr Hypertens Rev 2021; 17:59-66. [PMID: 32538730 DOI: 10.2174/1573402116666200615152139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/20/2020] [Accepted: 04/30/2020] [Indexed: 11/22/2022]
Abstract
Chronic kidney disease is a global public health issue, and it has been considered as the epidemic of the 21st century. Therefore, all initiatives addressed to slow down the evolution and complications of this condition should be well received. While the effects of salt reduction on cardiovascular disease have some controversial issues, in chronic kidney disease, such a policy is beneficial in multiple aspects. In chronic kidney disease patients, dietary sodium restriction is regularly recommended to control extracellular fluid expansion, hypertension and cardiovascular risk. Instead, the effects of sodium reduction on chronic kidney disease progression are still controversial. In the last years, potentially beneficial effects of a low sodium diet on chronic kidney disease evolution have emerged. Firstly, recent magnetic resonance-based findings of increased Na depots in skin and muscle associated with renal function, ageing and sodium intake open a vast body of investigation as a potential tool for monitoring effects of sodium restriction. In this narrative review, we also discussed novel aspects of sodium restriction in chronic kidney disease to manage metabolic acidosis as well as renal effects on fibroblast growth factor 23 or gut microbiota. Beyond current evidence, these approaches showed that common findings of kidney failure environment such as sodium -sensitivity, micro-inflammation, arterial stiffness metabolic acidosis and sarcopenia could be delayed controlling dietary sodium. Additional studies are now needed in populations with chronic kidney disease to confirm these new findings, addressed to slow down the evolution and complications of this condition.
Collapse
Affiliation(s)
- Rodolfo Valtuille
- Department of Clinical Research, Universidad Nacional Guillermo Brown, 2289 Espora Avenue, Burzaco, B1852FZD Buenos Aires, Argentina
| |
Collapse
|
43
|
The Complexity of FGF23 Effects on Cardiomyocytes in Normal and Uremic Milieu. Cells 2021; 10:cells10051266. [PMID: 34065339 PMCID: PMC8161087 DOI: 10.3390/cells10051266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
Fibroblast growth factor-23 (FGF23) appears to be one of the most promising biomarkers and predictors of cardiovascular risk in patients with heart disease and normal kidney function, but moreover in those with chronic kidney disease (CKD). This review summarizes the current knowledge of FGF23 mechanisms of action in the myocardium in the physiological and pathophysiological state of CKD, as well as its cross-talk to other important signaling pathways in cardiomyocytes. In this regard, current therapeutic possibilities and future perspectives are also discussed.
Collapse
|
44
|
Kumari R, Ranjan P, Suleiman ZG, Goswami SK, Li J, Prasad R, Verma SK. mRNA modifications in cardiovascular biology and disease: with a focus on m6A modification. Cardiovasc Res 2021; 118:1680-1692. [PMID: 33956076 DOI: 10.1093/cvr/cvab160] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 05/04/2021] [Indexed: 12/23/2022] Open
Abstract
Among several known RNA modifications, N6-methyladenosine (m6A) is the most studied RNA epitranscriptomic modification and controls multiple cellular functions during development, differentiation, and disease. Current research advancements have made it possible to examine the regulatory mechanisms associated with RNA methylation and reveal its functional consequences in the pathobiology of many diseases, including heart failure. m6A methylation has been described both on coding (mRNA) and non-coding RNA species including rRNA, tRNA, small nuclear RNA and circular RNAs. The protein components which catalyze the m6A methylation are termed methyltransferase or "m6A writers." The family of proteins that recognize this methylation are termed "m6A readers" and finally the enzymes involved in the removal of a methyl group from RNA are known as demethylases or "m6A erasers." At the cellular level, different components of methylation machinery are tightly regulated by many factors to maintain the m6A methylation dynamics. The m6A methylation process impacts different stages of mRNA metabolism and the biogenesis of long non-coding RNA and miRNA. Although, mRNA methylation was initially described in the 1970s, its regulatory roles in various diseases, including cardiovascular diseases are broadly unexplored. Recent investigations suggest the important role of m6A mRNA methylation in both hypertrophic and ischemic heart diseases. In the present review, we evaluate the significance of m6A methylation in the cardiovascular system, in cardiac homeostasis and disease, all of which may help to improve therapeutic intervention for the treatment of heart failure.
Collapse
Affiliation(s)
- Rajesh Kumari
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Prabhat Ranjan
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Zainab Gbongbo Suleiman
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sumanta Kumar Goswami
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jing Li
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ram Prasad
- Department of Ophthalmology and Visual Sciences, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Suresh Kumar Verma
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
45
|
Myostatin regulates the production of fibroblast growth factor 23 (FGF23) in UMR106 osteoblast-like cells. Pflugers Arch 2021; 473:969-976. [PMID: 33895875 PMCID: PMC8164604 DOI: 10.1007/s00424-021-02561-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 03/01/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022]
Abstract
Myostatin is a signaling molecule produced by skeletal muscle cells (myokine) that inhibits muscle hypertrophy and has further paracrine and endocrine effects in other organs including bone. Myostatin binds to activin receptor type 2B which forms a complex with transforming growth factor-β type I receptor (TGF-βRI) and induces intracellular p38MAPK and NFκB signaling. Fibroblast growth factor 23 (FGF23) is a paracrine and endocrine mediator produced by bone cells and regulates phosphate and vitamin D metabolism in the kidney. P38MAPK and NFκB-dependent store-operated Ca2+ entry (SOCE) are positive regulators of FGF23 production. Here, we explored whether myostatin influences the synthesis of FGF23. Fgf23 gene expression was determined by qRT-PCR and FGF23 protein by ELISA in UMR106 osteoblast–like cells. UMR106 cells expressed activin receptor type 2A and B. Myostatin upregulated Fgf23 gene expression and protein production. The myostatin effect on Fgf23 was significantly attenuated by TGF-βRI inhibitor SB431542, p38MAPK inhibitor SB202190, and NFκB inhibitor withaferin A. Moreover, SOCE inhibitor 2-APB blunted the myostatin effect on Fgf23. Taken together, myostatin is a stimulator of Fgf23 expression in UMR106 cells, an effect at least partially mediated by downstream TGF-βRI/p38MAPK signaling as well as NFκB-dependent SOCE.
Collapse
|
46
|
George B, Amjesh R, Paul AM, Santhoshkumar TR, Pillai MR, Kumar R. Evidence of a dysregulated vitamin D endocrine system in SARS-CoV-2 infected patient's lung cells. Sci Rep 2021; 11:8570. [PMID: 33883570 PMCID: PMC8060306 DOI: 10.1038/s41598-021-87703-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/30/2021] [Indexed: 12/19/2022] Open
Abstract
Although a defective vitamin D endocrine system has been widely suspected to be associated in SARS-CoV-2 pathobiology, the status of the vitamin D endocrine system and vitamin D-modulated genes in lung cells of patients infected with SARS-CoV-2 remains unknown. To understand the significance of the vitamin D endocrine system in SARS-CoV-2 pathobiology, computational approaches were applied to transcriptomic datasets from bronchoalveolar lavage fluid (BALF) cells of such patients or healthy individuals. Levels of vitamin D receptor, retinoid X receptor, and CYP27A1 in BALF cells of patients infected with SARS-CoV-2 were found to be reduced. Additionally, 107 differentially expressed, predominantly downregulated genes, as potentially modulated by vitamin D endocrine system, were identified in transcriptomic datasets from patient's cells. Further analysis of differentially expressed genes provided eight novel genes with a conserved motif with vitamin D-responsive elements, implying the role of both direct and indirect mechanisms of gene expression by the dysregulated vitamin D endocrine system in SARS-CoV-2-infected cells. Protein-protein interaction network of differentially expressed vitamin D-modulated genes were enriched in the immune system, NF-κB/cytokine signaling, and cell cycle regulation as top predicted pathways that might be affected in the cells of such patients. In brief, the results presented here povide computational evidence to implicate a dysregulated vitamin D endocrine system in the pathobiology of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Bijesh George
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
- PhD Program, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Revikumar Amjesh
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| | - Aswathy Mary Paul
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
- PhD Program, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - T R Santhoshkumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| | | | - Rakesh Kumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India.
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, USA.
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, USA.
| |
Collapse
|
47
|
Vázquez-Sánchez S, Poveda J, Navarro-García JA, González-Lafuente L, Rodríguez-Sánchez E, Ruilope LM, Ruiz-Hurtado G. An Overview of FGF-23 as a Novel Candidate Biomarker of Cardiovascular Risk. Front Physiol 2021; 12:632260. [PMID: 33767635 PMCID: PMC7985069 DOI: 10.3389/fphys.2021.632260] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
Fibroblast growth factor-23 (FGF)-23 is a phosphaturic hormone involved in mineral bone metabolism that helps control phosphate homeostasis and reduces 1,25-dihydroxyvitamin D synthesis. Recent data have highlighted the relevant direct FGF-23 effects on the myocardium, and high plasma levels of FGF-23 have been associated with adverse cardiovascular outcomes in humans, such as heart failure and arrhythmias. Therefore, FGF-23 has emerged as a novel biomarker of cardiovascular risk in the last decade. Indeed, experimental data suggest FGF-23 as a direct mediator of cardiac hypertrophy development, cardiac fibrosis and cardiac dysfunction via specific myocardial FGF receptor (FGFR) activation. Therefore, the FGF-23/FGFR pathway might be a suitable therapeutic target for reducing the deleterious effects of FGF-23 on the cardiovascular system. More research is needed to fully understand the intracellular FGF-23-dependent mechanisms, clarify the downstream pathways and identify which could be the most appropriate targets for better therapeutic intervention. This review updates the current knowledge on both clinical and experimental studies and highlights the evidence linking FGF-23 to cardiovascular events. The aim of this review is to establish the specific role of FGF-23 in the heart, its detrimental effects on cardiac tissue and the possible new therapeutic opportunities to block these effects.
Collapse
Affiliation(s)
- Sara Vázquez-Sánchez
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Jonay Poveda
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - 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
| | - Elena Rodríguez-Sánchez
- 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
|
48
|
Zhou C, Shi Z, Ouyang N, Ruan X. Hyperphosphatemia and Cardiovascular Disease. Front Cell Dev Biol 2021; 9:644363. [PMID: 33748139 PMCID: PMC7970112 DOI: 10.3389/fcell.2021.644363] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Hyperphosphatemia or even serum phosphate levels within the “normal laboratory range” are highly associated with increased cardiovascular disease risk and mortality in the general population and patients suffering from chronic kidney disease (CKD). As the kidney function declines, serum phosphate levels rise and subsequently induce the development of hypertension, vascular calcification, cardiac valvular calcification, atherosclerosis, left ventricular hypertrophy and myocardial fibrosis by distinct mechanisms. Therefore, phosphate is considered as a promising therapeutic target to improve the cardiovascular outcome in CKD patients. The current therapeutic strategies are based on dietary and pharmacological reduction of serum phosphate levels to prevent hyperphosphatemia in CKD patients. Large randomized clinical trials with hard endpoints are urgently needed to establish a causal relationship between phosphate excess and cardiovascular disease (CVD) and to determine if lowering serum phosphate constitutes an effective intervention for the prevention and treatment of CVD.
Collapse
Affiliation(s)
- Chao Zhou
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhengyu Shi
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Nan Ouyang
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiongzhong Ruan
- John Moorhead Research Laboratory, Centre for Nephrology, University College London (UCL) Medical School, London, United Kingdom.,Centre for Lipid Research and Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
49
|
Abstract
The myocardium consists of different cell types, of which endothelial cells, cardiomyocytes, and fibroblasts are the most abundant. Communication between these different cell types, also called paracrine signaling, is essential for normal cardiac function, but also important in cardiac remodeling and heart failure. Systematic studies on the expression of ligands and their corresponding receptors in different cell types showed that for 60% of the expressed ligands in a particular cell, the receptor is also expressed. The fact that many ligand-receptor pairs are present in most cells, including the major cell types in the heart, indicates that autocrine signaling is a widespread phenomenon. Autocrine signaling in cardiac remodeling and heart failure is involved in all pathophysiological mechanisms generally observed: hypertrophy, fibrosis, angiogenesis, cell survival, and inflammation. Herein, we review ligand-receptor pairs present in the major cardiac cell types based on RNA-sequencing expression databases, and we review current literature on extracellular signaling proteins with an autocrine function in the heart; these include C-type natriuretic peptide, fibroblast growth factors 2, F21, and 23, macrophage migration inhibitory factor, heparin binding-epidermal growth factor, angiopoietin-like protein 2, leptin, adiponectin, follistatin-like 1, apelin, neuregulin 1, vascular endothelial growth factor, transforming growth factor β, wingless-type integration site family, member 1-induced secreted protein-1, interleukin 11, connective tissue growth factor/cellular communication network factor, and calcitonin gene‒related peptide. The large number of autocrine signaling factors that have been studied in the literature supports the concept that autocrine signaling is an essential part of myocardial biology and disease.
Collapse
Affiliation(s)
- Vincent F. M. Segers
- Laboratory of PhysiopharmacologyUniversity of AntwerpBelgium
- Department of CardiologyUniversity Hospital AntwerpEdegemBelgium
| | - Gilles W. De Keulenaer
- Laboratory of PhysiopharmacologyUniversity of AntwerpBelgium
- Department of CardiologyZNA HospitalAntwerpBelgium
| |
Collapse
|
50
|
Kidney function and the prognostic value of myocardial performance index. Int J Cardiovasc Imaging 2021; 37:1637-1647. [PMID: 33475871 DOI: 10.1007/s10554-020-02149-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 12/24/2020] [Indexed: 10/22/2022]
Abstract
A decreased glomerular filtration rate (GFR) is a risk factor for cardiovascular disease even after adjustment for conventional risk factors. The myocardial performance index (MPI) is defined as (isovolumetric relaxation time (IVRT) + isovolumetric contraction time (IVCT))/ejection time (ET). It has been shown to be an independent predictor of cardiovascular events. We hypothesized the MPI could prove valuable for assessing cardiac risk in subjects of the general population with decreased estimated GFR (eGFR). MPI was measured in 1915 subjects from a large general population prospective cohort study using color tissue Doppler imaging (TDI) M-mode through the mitral valve. We compared the prognostic capabilities of the MPI between subjects with eGFR ≥ 75 mL/min/1.73 m2 and subjects with eGFR < 75 mL/min/1.73 m2 using multivariable adjusted Cox regression models. The composite endpoint was heart failure, myocardial infarction or cardiovascular death. Mean age was 58 years (SD 16.2), 58% were women, 42% had hypertension and 8.3% diabetes. During a median follow-up time of 12.4 years [IQR 10.6-12.7 years] 269 participants reached the combined endpoint. eGFR modified the prognostic capability of MPI (p-value for interaction < 0.001): After multivariable adjustment, MPI remained an independent predictor of the composite endpoint only in participants with eGFR < 75 mL/min/1.73 m2: HR 1.18 (95% CI 1.02-1.38), p = 0.03, vs. in subjects with eGFR ≥ 75 mL/min/1.73 m2: HR 1.14 (95% CI 0.94-1.39), p = 0.17. These results suggest the MPI could be particularly valuable for identifying elevated cardiac risk in individuals from the general population with decreased eGFR.
Collapse
|