101
|
Kallmeyer A, Pello A, Cánovas E, Aceña Á, González‐Casaus ML, Tarín N, Cristóbal C, Gutiérrez‐Landaluce C, Huelmos A, Rodríguez‐Valer A, González‐Lorenzo Ó, Alonso J, López‐Bescós L, Egido J, Mahillo I, Lorenzo Ó, Tuñón J. Fibroblast growth factor 23 independently predicts adverse outcomes after an acute coronary syndrome. ESC Heart Fail 2024; 11:240-250. [PMID: 37950429 PMCID: PMC10804179 DOI: 10.1002/ehf2.14568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/10/2023] [Accepted: 10/08/2023] [Indexed: 11/12/2023] Open
Abstract
AIMS Abnormalities of mineral metabolism (MM) have been related to cardiovascular disorders. There are no reports on the prognostic role of MM after an acute coronary syndrome (ACS). We aim to assess the prognostic role of MM after an ACS. METHODS AND RESULTS Plasma levels of components of MM [fibroblast growth factor 23 (FGF23), calcidiol, parathormone, klotho, and phosphate], high-sensitivity C-reactive protein, and N-terminal-pro-brain natriuretic peptide were measured in 1190 patients at discharge from an ACS. The primary outcome was a combination of acute ischaemic events, heart failure (HF) and death. Secondary outcomes were the separate components of the primary outcome. Age was 61.7 ± 12.2 years, and 77.1% were men. Median follow-up was 5.44 (3.03-7.46) years. Two hundred and ninety-four patients developed the primary outcome. At multivariable analysis FGF23 (hazard ratio, HR 1.18 [1.08-1.29], P < 0.001), calcidiol (HR 0.86 [0.74-1.00], P = 0.046), previous coronary or cerebrovascular disease, and hypertension were independent predictors of the primary outcome. The predictive power of FGF23 was homogeneous across different subgroups of population. FGF23 (HR 1.45 [1.28-1.65], P < 0.001) and parathormone (HR 1.06 1.01-1.12]; P = 0.032) resulted as independent predictors of HF. FGF23 (HR 1.21 [1.07-1.37], P = 0.002) and calcidiol (HR 0.72 [0.54-0.97), P = 0.028) were independent predictors of death. No biomarker predicted acute ischaemic events. FGF23 predicted independently the primary outcome in patients with estimated glomerular filtration rate > 60 mL/min/1.73 m2 . CONCLUSIONS FGF23 and other components of MM are independent predictors of HF and death after an ACS. This effect is homogeneous across different subgroups of population, and it is not limited to patients with chronic kidney disease.
Collapse
Affiliation(s)
- Andrea Kallmeyer
- Department of CardiologyIIS‐Fundación Jiménez DíazMadridSpain
- Faculty of MedicineAutónoma UniversityMadridSpain
| | - Ana Pello
- Department of CardiologyIIS‐Fundación Jiménez DíazMadridSpain
- Faculty of MedicineAutónoma UniversityMadridSpain
| | - Ester Cánovas
- Department of CardiologyIIS‐Fundación Jiménez DíazMadridSpain
| | - Álvaro Aceña
- Department of CardiologyIIS‐Fundación Jiménez DíazMadridSpain
- Faculty of MedicineAutónoma UniversityMadridSpain
| | | | - Nieves Tarín
- Department of CardiologyHospital Universitario de MóstolesMadridSpain
- Faculty of MedicineRey Juan Carlos UniversityMadridSpain
| | - Carmen Cristóbal
- Faculty of MedicineRey Juan Carlos UniversityMadridSpain
- Department of CardiologyHospital Universitario de FuenlabradaMadridSpain
| | | | - Ana Huelmos
- Department of CardiologyHospital Universitario Fundación AlcorcónMadridSpain
| | | | - Óscar González‐Lorenzo
- Department of CardiologyIIS‐Fundación Jiménez DíazMadridSpain
- Faculty of MedicineAutónoma UniversityMadridSpain
| | | | | | - Jesús Egido
- CIBERDEMMadridSpain
- Department of NephrologyIIS‐Fundación Jiménez DíazMadridSpain
- Laboratory of Vascular PathologyIIS‐Fundación Jiménez DíazMadridSpain
| | - Ignacio Mahillo
- Laboratory of EpidemiologyIIS‐Fundación Jiménez DíazMadridSpain
| | - Óscar Lorenzo
- Faculty of MedicineAutónoma UniversityMadridSpain
- CIBERDEMMadridSpain
- Laboratory of Vascular PathologyIIS‐Fundación Jiménez DíazMadridSpain
| | - José Tuñón
- Department of CardiologyIIS‐Fundación Jiménez DíazMadridSpain
- Faculty of MedicineAutónoma UniversityMadridSpain
- Laboratory of Vascular PathologyIIS‐Fundación Jiménez DíazMadridSpain
- CIBERCV, ISCIIIMadridSpain
| |
Collapse
|
102
|
Vinke JSJ, Kremer D, Knobbe TJ, Grote Beverborg N, Berger SP, Bakker SJ, de Borst MH, Eisenga MF. Iron Status and Cause-Specific Mortality After Kidney Transplantation. Kidney Med 2024; 6:100766. [PMID: 38375423 PMCID: PMC10874991 DOI: 10.1016/j.xkme.2023.100766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024] Open
Affiliation(s)
- Joanna Sophia J. Vinke
- Department of Nephrology, University Medical Center Groningen; Groningen, the Netherlands
| | - Daan Kremer
- Department of Nephrology, University Medical Center Groningen; Groningen, the Netherlands
| | - Tim J. Knobbe
- Department of Nephrology, University Medical Center Groningen; Groningen, the Netherlands
| | - Niels Grote Beverborg
- Department of Cardiology, University Medical Center Groningen; Groningen, the Netherlands
| | - Stefan P. Berger
- Department of Nephrology, University Medical Center Groningen; Groningen, the Netherlands
| | - Stephan J.L. Bakker
- Department of Nephrology, University Medical Center Groningen; Groningen, the Netherlands
| | - Martin H. de Borst
- Department of Nephrology, University Medical Center Groningen; Groningen, the Netherlands
| | - Michele F. Eisenga
- Department of Nephrology, University Medical Center Groningen; Groningen, the Netherlands
| |
Collapse
|
103
|
Abstract
Phosphorus is an essential mineral that is, in the form of inorganic phosphate (Pi), required for building cell membranes, DNA and RNA molecules, energy metabolism, signal transduction and pH buffering. In bone, Pi is essential for bone stability in the form of apatite. Intestinal absorption of dietary Pi depends on its bioavailability and has two distinct modes of active transcellular and passive paracellular absorption. Active transport is transporter mediated and partly regulated, while passive absorption depends mostly on bioavailability. Renal excretion controls systemic Pi levels, depends on transporters in the proximal tubule and is highly regulated. Deposition and release of Pi into and from soft tissues and bone has to be tightly controlled. The endocrine network coordinating intestinal absorption, renal excretion and bone turnover integrates dietary intake and metabolic requirements with renal excretion and is critical for bone stability and cardiovascular health during states of hypophosphataemia or hyperphosphataemia as evident from inborn or acquired diseases. This review provides an integrated overview of the biology of phosphate and Pi in mammals.
Collapse
Affiliation(s)
- Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
104
|
Wang X, Yu X, Gavardinas K, Dey A, Zhang HY, Porter G, Porras L, Yu L, Guo H, Reidy CA, Haas JV, Xu Y, Kowala MC, Jadhav PK, Wetterau JR. Effect of an NHE3 inhibitor in combination with an NPT2b inhibitor on gastrointestinal phosphate absorption in Rodent models. PLoS One 2024; 19:e0292091. [PMID: 38277356 PMCID: PMC10817170 DOI: 10.1371/journal.pone.0292091] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 09/12/2023] [Indexed: 01/28/2024] Open
Abstract
Many of the pathological consequences of chronic kidney disease can be attributed to an elevation in serum phosphate levels. Current therapies focused on decreasing intestinal phosphate absorption to treat hyperphosphatemia are inadequate. The most effective therapeutic strategy may be to target multiple absorptive pathways. In this study, the ability of a novel inhibitor of the intestinal sodium hydrogen exchanger 3 (NHE3), LY3304000, which inhibits paracellular, diffusional uptake of phosphate, to work in combination with an inhibitor of the active transporter, sodium dependent phosphate cotransporter 2b (NPT2b), LY3358966, was explored. LY3304000 modestly inhibited the acute uptake of phosphate into plasma of rats, while surprisingly, it doubled the rate of phosphate uptake in mice, an animal model dominated by NPT2b mediated acute phosphate uptake. In rats, LY3004000 and LY3358966 work in concert to inhibit acute phosphate uptake. On top of LY3358966, LY3304000 further decreased the acute uptake of phosphate into plasma. Studies measuring the recovery of radiolabeled phosphate in the intestine demonstrated LY3304000 and LY3358966 synergistically inhibited the absorption of phosphate in rats. We hypothesize the synergism is because the NHE3 inhibitor, LY3304000, has two opposing effects on intestinal phosphate absorption in rats, first it decreases diffusion mediated paracellular phosphate absorption, while second, it simultaneously increases phosphate absorption through the NPT2b pathway. NHE3 inhibition decreases proton export from enterocytes and raises the cell surface pH. In vitro, NPT2b mediated phosphate transport is increased at higher pHs. The increased NPT2b mediated transport induced by NHE3 inhibition is masked in rats which have relatively low levels of NPT2b mediated phosphate transport, by the more robust inhibition of diffusion mediated phosphate absorption. Thus, the inhibition of NPT2b mediated phosphate transport in rats in the presence of NHE3 inhibition has an effect that exceeds its effect in the absence of NHE3 inhibition, leading to the observed synergism on phosphate absorption between NPT2b and NHE3 inhibition.
Collapse
Affiliation(s)
- Xiaojun Wang
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Xiaohong Yu
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Kostas Gavardinas
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Asim Dey
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Hong Y. Zhang
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Gina Porter
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Leah Porras
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Lan Yu
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Haihong Guo
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Charles A. Reidy
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Joseph V. Haas
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Yanping Xu
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Mark C. Kowala
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Prabhakar K. Jadhav
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - John R. Wetterau
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| |
Collapse
|
105
|
Bouzemane A, Vignot E, Derain Dubourg L, De Mul A, Molin A, Chapurlat R, Fontanges E, Delsart D, Akbari A, Huang SHS, McIntyre CW, Bacchetta J, Lemoine S. Reassuring Data on the Cardiovascular Risk in Adults With X-linked Hypophosphatemia Receiving Conventional Therapy. J Clin Endocrinol Metab 2024; 109:e488-e494. [PMID: 37843399 DOI: 10.1210/clinem/dgad608] [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: 07/09/2023] [Revised: 09/04/2023] [Accepted: 10/11/2023] [Indexed: 10/17/2023]
Abstract
CONTEXT X-linked hypophosphatemia (XLH) is a rare genetic disorder that results in increased plasma levels of fibroblast growth factor 23 (FGF23). Several studies have demonstrated a direct association between FGF23 and cardiovascular mortality in cohorts of patients with chronic renal failure. However, in patients with XLH, studies on the cardiovascular impact of the disease are rare, with contradictory results. OBJECTIVE The aim was to assess whether the disease led to an increased cardiovascular risk. METHODS We conducted a single-center retrospective observational study on a local cohort of adult patients with XLH. The primary endpoint was a composite endpoint of the frequency of left ventricular hypertrophy (LVH) or presence of high blood pressure. Our secondary objectives were to assess echocardiographic, pulse wave velocity, and central blood pressure data as other markers of CV health. Independently of this cohort, tissue sodium content with magnetic resonance imaging was studied in 2 patients with XLH before and after burosumab. RESULTS Twenty-two patients were included. Median serum phosphate was 0.57 (0.47-0.72) mmol/L and FGF23 94 pg/L (58-2226). Median blood pressure was 124 (115-130)/68 (65-80) mm Hg, with only 9% of patients being hypertensive. A majority of patients (69%) had no LVH, only 1 had a left ventricular mass >100 g/m² and 25% of patients had left ventricular remodeling. Pulse wave velocity was normal in all patients. No differences in skin and muscle sodium content were observed before and after burosumab in the 2 patients who underwent sodium magnetic resonance imaging. CONCLUSION We found no elevated risk of developing hypertension or LVH in patients with XLH.
Collapse
Affiliation(s)
- Alexandre Bouzemane
- Hospices Civils de Lyon, Nephrology, hypertension renal and functional exploration, Hôpital Edouard Herriot, 69003, Lyon, France
| | | | - Laurence Derain Dubourg
- Hospices Civils de Lyon, Nephrology, hypertension renal and functional exploration, Hôpital Edouard Herriot, 69003, Lyon, France
| | - Aurélie De Mul
- Reference centre for rare calcium and phosphorus diseases, paediatric rheumatology and dermatology, rare diseases network, OSCAR, ORKID, ERKNet BOND, HFME, Bron 69029, France
| | - Arnaud Molin
- Genetic department, Centre Hospitalier Universitaire de Caen, Caen, 14033, France
| | - Roland Chapurlat
- Rheumatology Department, CHU Edouard-Herriot, 69003 Lyon, France
| | | | - Daphne Delsart
- Cardiology functional explorations, Hopital Edouard-Herriot, 69003 Lyon, France
| | - Alireza Akbari
- Canada Kidney clinical research unit, London Health Sciences Centre, East London, ON, N6A 5W9Canada
| | - Shih Han Susan Huang
- Canada Kidney clinical research unit, London Health Sciences Centre, East London, ON, N6A 5W9Canada
| | - Christopher W McIntyre
- Canada Kidney clinical research unit, London Health Sciences Centre, East London, ON, N6A 5W9Canada
| | - Justine Bacchetta
- Reference centre for rare calcium and phosphorus diseases, paediatric rheumatology and dermatology, rare diseases network, OSCAR, ORKID, ERKNet BOND, HFME, Bron 69029, France
- University of Lyon, CarMeN Laboratory, IRIS Team, INSERM, INSERM1033, INRA, INSA Lyon, 69100, Villeurbanne, France
- INSERM 1033, prevention of bone diseases, 69008 Lyon, France
| | - Sandrine Lemoine
- Hospices Civils de Lyon, Nephrology, hypertension renal and functional exploration, Hôpital Edouard Herriot, 69003, Lyon, France
- Reference centre for rare calcium and phosphorus diseases, paediatric rheumatology and dermatology, rare diseases network, OSCAR, ORKID, ERKNet BOND, HFME, Bron 69029, France
- University of Lyon, CarMeN Laboratory, IRIS Team, INSERM, INSERM1033, INRA, INSA Lyon, 69100, Villeurbanne, France
| |
Collapse
|
106
|
Hidaka N, Inoue K, Kato H, Hoshino Y, Koga M, Kinoshita Y, Takashi Y, Makita N, Fukumoto S, Nangaku M, Ito N. FGF-23, Left Ventricular Hypertrophy, and Mortality in Patients With CKD: A Revisit With Mediation Analysis. JACC. ADVANCES 2024; 3:100747. [PMID: 38939808 PMCID: PMC11198284 DOI: 10.1016/j.jacadv.2023.100747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 10/05/2023] [Accepted: 10/30/2023] [Indexed: 06/29/2024]
Abstract
Background In patients with chronic kidney disease (CKD), fibroblast growth factor (FGF)-23 is suspected to cause death or cardiovascular disease by inducing left ventricular hypertrophy (LVH). Objectives This study aims to quantify the mediational effect of LVH in the hypothetical causal pathway from FGF-23 to long-term adverse outcomes. Methods From 3,939 adults with CKD stages 2 to 4 enrolled in the CRIC (Chronic Renal Insufficiency Cohort) study, 2,368 participants with available data of FGF-23, left ventricular mass index at 1 year, and covariates were included. We employed linear and Cox proportional hazards regression models to investigate the association between FGF-23 and LVH, all-cause mortality, atrial fibrillation (AF), or congestive heart failure (CHF). Mediation analysis was used within a counterfactual framework to decompose the effect of FGF-23 into natural direct and indirect effects. Results Among 2,368 participants (mean age: 57.7 years, 1,252 males, median FGF-23 level: 138.8 RU/mL), left ventricular mass index was positively correlated with FGF-23. During a median of 12.0, 11.1, and 11.1 years, FGF-23 was associated with all-cause mortality (HR: 1.62, 95% CI: 1.24-2.12), AF (HR: 1.58, 95% CI: 1.12-2.24), and CHF (HR: 1.32, 95% CI: 0.95-1.84) when the highest quartile was compared to the lowest quartile. LVH mediated 7.4%, 11.2%, and 21.9% of the effect of FGF-23 on all-cause mortality, AF, and CHF, respectively. Conclusions In CKD patients, FGF-23 had a minor effect on the development of long-term adverse outcomes through LVH. Other potential mediators and the validity of negative effect of FGF-23 should be explored.
Collapse
Affiliation(s)
- Naoko Hidaka
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
| | - Kosuke Inoue
- Department of Social Epidemiology, Graduate School of Medicine, Kyoto University, Sakyo, Kyoto, Japan
| | - Hajime Kato
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
| | - Yoshitomo Hoshino
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
| | - Minae Koga
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
| | - Yuka Kinoshita
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
| | - Yuichi Takashi
- Department of Endocrinology and Diabetes Mellitus, Fukuoka University School of Medicine, Jonan, Fukuoka, Japan
| | - Noriko Makita
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
| | - Seiji Fukumoto
- Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
| | - Nobuaki Ito
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
| |
Collapse
|
107
|
Shaik SP, Karan HH, Singh A, Attuluri SK, Khan AAN, Zahid F, Patil D. HFpEF: New biomarkers and their diagnostic and prognostic value. Curr Probl Cardiol 2024; 49:102155. [PMID: 37866418 DOI: 10.1016/j.cpcardiol.2023.102155] [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: 10/01/2023] [Accepted: 10/14/2023] [Indexed: 10/24/2023]
Abstract
Heart failure characterized by preserved ejection fraction (HFpEF) poses a substantial challenge to healthcare systems worldwide and the diagnostic algorithms used currently mirror those utilized for reduced Ejection Fraction (HFrEF). This literature review aims to explore the diagnostic and prognostic credibility of numerous emerging biomarkers associated with HFpEF. We conducted a thorough analysis of the available medical literature and selected the biomarkers which yielded the maximum amount of published information. After reviewing the current literature we conclude that there are no biomarkers at present which are superior to natriuretic peptides in terms of diagnosis and prognosis of HFpEF. However biomarkers like Suppression of tumorigenicity2, Galectin3 and microRNAs are promising and can be researched further for future use. Although newer individual biomarkers may not be useful in diagnosing and prognosis of HFpEF, we believe that a specific biomarker profile may be identified in each phenotype,which can be used in future.
Collapse
Affiliation(s)
- Shahanaz Parveen Shaik
- Junior Resident, Internal Medicine, DR. Y.S.R University of Health Sciences, Andhra Pradesh, India.
| | - Hasnain Hyder Karan
- Resident, Internal Medicine, San Joaquin General Hospital,French Camp, CA, United States
| | - Arkaja Singh
- Junior Resident, Mahatma Gandhi Medical College and Hospital, Jaipur, India
| | - Sai Kiran Attuluri
- Junior Resident, Internal Medicine, DR. Y.S.R University of Health Sciences, Andhra Pradesh, India
| | - Afnan Akram Nawaz Khan
- Junior Resident, Internal Medicine, Vydehi Institute of Medical Sciences, Bangalore, India
| | - Fazila Zahid
- Resident, Internal Medicine, OSF St Francis Hospital, University of Illinois College of Medicine; IL; USA
| | - Dhrumil Patil
- Postdoctoral Research fellow, Cardiology department, Beth Israel Deaconess Medical Center, Harvard University, USA
| |
Collapse
|
108
|
Hagemann R, Watanabe MT, Hueb JC, Martín LC, Silva VDS, Caramori JDSCT. Assessment of atherosclerosis and endothelial dysfunction risk factors in patients with primary glomerulonephritis. J Bras Nefrol 2024; 46:29-38. [PMID: 36638246 PMCID: PMC10962406 DOI: 10.1590/2175-8239-jbn-2022-0116en] [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: 07/11/2022] [Accepted: 11/29/2022] [Indexed: 01/15/2023] Open
Abstract
INTRODUCTION Glomerulonephritis are the third cause of chronic kidney disease (CKD) requiring dialysis in Brazil. Mineral and bone disorder (MBD) is one of the complications of CKD and is already present in the early stages. Assessment of carotid intima-media thickness (CIMT) and flow-mediated vasodilatation (FMV) are non-invasive ways of assessing cardiovascular risk. HYPOTHESIS Patients with primary glomerulonephritis (PG) have high prevalence of atherosclerosis and endothelial dysfunction, not fully explained by traditional risk factors, but probably influenced by the early onset of MBD. OBJECTIVE To evaluate the main markers of atherosclerosis in patients with PG. METHOD Clinical, observational, cross-sectional and controlled study. Patients with PG were included and those under 18 years of age, pregnants, those with less than three months of follow-up and those with secondary glomerulonephritis were excluded. Those who, at the time of exams collection, had proteinuria higher than 6 grams/24 hours and using prednisone at doses higher than 0.2 mg/kg/day were also excluded. RESULTS 95 patients were included, 88 collected the exams, 1 was excluded and 23 did not undergo the ultrasound scan. Patients with PG had a higher mean CIMT compared to controls (0.66 versus 0.60), p = 0.003. After multivariate analysis, age and values for systolic blood pressure (SBP), FMV and GFR (p = 0.02); and FMV and serum uric acid (p = 0.048) remained statistically relevant. DISCUSSION AND CONCLUSION The higher cardiovascular risk in patients with PG was not explained by early MBD. Randomized and multicentric clinical studies are necessary to better assess this hypothesis.
Collapse
Affiliation(s)
- Rodrigo Hagemann
- Universidade Estadual Paulista Júlio de Mesquita Filho, Faculdade de
Medicina de Botucatu, Departamento de Clínica Médica, Botucatu, São Paulo,
Brazil
- Universidade Federal do Paraná, Complexo Hospital de Clínicas,
Departamento de Clínica Médica, Curitiba, Paraná, Brazil
| | - Marcela Tatiana Watanabe
- Universidade Estadual Paulista Júlio de Mesquita Filho, Faculdade de
Medicina de Botucatu, Departamento de Clínica Médica, Botucatu, São Paulo,
Brazil
| | - João Carlos Hueb
- Universidade Estadual Paulista Júlio de Mesquita Filho, Faculdade de
Medicina de Botucatu, Departamento de Clínica Médica, Botucatu, São Paulo,
Brazil
| | - Luis Cuadrado Martín
- Universidade Estadual Paulista Júlio de Mesquita Filho, Faculdade de
Medicina de Botucatu, Departamento de Clínica Médica, Botucatu, São Paulo,
Brazil
| | - Vanessa dos Santos Silva
- Universidade Estadual Paulista Júlio de Mesquita Filho, Faculdade de
Medicina de Botucatu, Departamento de Clínica Médica, Botucatu, São Paulo,
Brazil
| | | |
Collapse
|
109
|
Alake SE, Ice J, Robinson K, Price P, Hatter B, Wozniak K, Lin D, Chowanadisai W, Smith BJ, Lucas EA. Reduced estrogen signaling contributes to bone loss and cardiac dysfunction in interleukin-10 knockout mice. Physiol Rep 2024; 12:e15914. [PMID: 38217044 PMCID: PMC10787104 DOI: 10.14814/phy2.15914] [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: 08/28/2023] [Revised: 12/02/2023] [Accepted: 12/07/2023] [Indexed: 01/14/2024] Open
Abstract
Characterization of the interleukin (IL)-10 knockout (KO) mouse with chronic gut inflammation, cardiovascular dysfunction, and bone loss suggests a critical role for this cytokine in interorgan communication within the gut, bone, and cardiovascular axis. We sought to understand the role of IL-10 in the cross-talk between these systems. Six-week-old IL-10 KO mice and their wild type (WT) counterparts were maintained on a standard rodent diet for 3 or 6 months. Gene expression of proinflammatory markers and Fgf23, serum 17β-estradiol (E2), and cardiac protein expression were assessed. Ileal Il17a and Tnf mRNA increased while Il6 mRNA increased in the bone and heart by at least 2-fold in IL-10 KO mice. Bone Dmp1 and Phex mRNA were repressed at 6 months in IL-10 KO mice, resulting in increased Fgf23 mRNA (~4-fold) that contributed to increased fibrosis. In the IL-10 KO mice, gut bacterial β-glucuronidase activity and ovarian Cyp19a1 mRNA were lower (p < 0.05), consistent with reduced serum E2 and reduced cardiac pNOS3 (Ser1119 ) in these mice. Treatment of ileal lymphocytes with E2 reduced gut inflammation in WT but not IL-10 KO mice. In conclusion, our data suggest that diminished estrogen and defective bone mineralization increased FGF23 which contributed to cardiac fibrosis in the IL-10 KO mouse.
Collapse
Affiliation(s)
- Sanmi E. Alake
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - John Ice
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Kara Robinson
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Payton Price
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Bethany Hatter
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Karen Wozniak
- Department of Microbiology and Molecular GeneticsOklahoma State UniversityStillwaterOklahomaUSA
| | - Dingbo Lin
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Winyoo Chowanadisai
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| | - Brenda J. Smith
- Department of Obstetrics and GynecologyIndiana School of MedicineIndianapolisIndianaUSA
- Indiana Center for Musculoskeletal HealthIndiana School of MedicineIndianapolisIndianaUSA
| | - Edralin A. Lucas
- Department of Nutritional SciencesOklahoma State UniversityStillwaterOklahomaUSA
| |
Collapse
|
110
|
Dusso A, Bauerle KT, Zhang RM, Bernal-Mizrachi C. Vitamin D and renal disease. FELDMAN AND PIKE'S VITAMIN D 2024:587-618. [DOI: 10.1016/b978-0-323-91338-6.00029-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
111
|
Li H, Liu T, Yang L, Ma F, Wang Y, Zhan Y, Mao H. Knowledge landscapes and emerging trends of cardiorenal syndrome type 4: a bibliometrics and visual analysis from 2004 to 2022. Int Urol Nephrol 2024; 56:155-166. [PMID: 37422767 PMCID: PMC10776493 DOI: 10.1007/s11255-023-03680-4] [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: 03/21/2023] [Accepted: 06/14/2023] [Indexed: 07/11/2023]
Abstract
PURPOSE To evaluate the key topics and emerging trends in the field of cardiorenal syndrome type 4 (CRS-4) by bibliometrics and visual analysis. METHODS Citespace, VOSviewer, and Bibliometrix package were used to analyze the collected data from the Web of Science Core Collection, including publication trends, leading countries, active authors and institutions, co-cited references, journals, and keyword analysis. RESULTS Finally, 2267 articles were obtained. From 2004 to 2022, the number of publications was increasing year by year. A total of 735 authors from 543 institutions in 94 countries/regions participated in the publication of CRS-4 field, which were mostly from North America and Europe. Most of the co-cited references were reviews or guidelines from kidney/heart specialist journals or top journals. The journals concerning nephrology had a higher academic influence in this field. Oxidative stress and inflammation remained hot topics in CRS-4 research, as well as uremic toxins. Fibroblast growth factor 23 and klotho were emerging trends in recent years. Sodium glucose cotransporter 2 (SGLT2) inhibitors were the latest frontier hot spots. Future research advances may pay more attention to the prevention and prognosis assessment of CRS-4. CONCLUSION Our study provides some key information for scholars to determine the direction of future research.
Collapse
Affiliation(s)
- Han Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Tongtong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Liping Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Fang Ma
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yuyang Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yongli Zhan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Huimin Mao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
112
|
Ogata H, Sugawara H, Yamamoto M, Ito H. Phosphate and Coronary Artery Disease in Patients with Chronic Kidney Disease. J Atheroscler Thromb 2024; 31:1-14. [PMID: 37766573 PMCID: PMC10776333 DOI: 10.5551/jat.rv22012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/07/2023] [Indexed: 09/29/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in patients with chronic kidney disease (CKD). Both traditional and CKD-related factors are associated with CVD in CKD patients. Traditional factors that play an important role in the atherosclerotic process directly contribute to a higher risk of coronary artery disease in patients with early-stage CKD. Among CKD-related factors, CKD-mineral and bone disorder plays a critical role in the pathomechanism of nonatherosclerotic diseases, which increases the risk of cardiovascular morbidity and mortality in patients with advanced CKD. Higher serum phosphate levels were significantly associated with cardiovascular events and all-cause mortality in patients with or without CKD. An increased phosphate load, directly and indirectly, promotes arterial medial calcification and left ventricular hypertrophy, both of which predispose patients to coronary artery disease. Calciprotein particles that form in a hyperphosphatemic state promote the transformation of vascular smooth muscle cells (VSMCs) into osteoblastic cells, thereby providing a scaffold for medial calcification in the artery. Increases in fibroblast growth factor-23 and disturbed vitamin D metabolism induced by an excessive phosphate load play a significant role in the development of cardiomyocyte hypertrophy and cardiac fibrosis. Recently, hyperphosphatemia was reported to promote de novo cholesterol synthesis in VSMCs and macrophages, which is likely to contribute to statin resistance in patients with end-stage kidney disease. This review outlines the association between increased phosphate load and coronary artery disease in patients with CKD.
Collapse
Affiliation(s)
- Hiroaki Ogata
- Division of Nephrology, Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
- Department of Medical Education, Showa University School of Medicine, Tokyo, Japan
| | - Hirohito Sugawara
- Division of Nephrology, Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
| | - Masahiro Yamamoto
- Division of Nephrology, Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
| | - Hidetoshi Ito
- Division of Nephrology, Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
| |
Collapse
|
113
|
Edmonston D, Grabner A, Wolf M. FGF23 and klotho at the intersection of kidney and cardiovascular disease. Nat Rev Cardiol 2024; 21:11-24. [PMID: 37443358 DOI: 10.1038/s41569-023-00903-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/13/2023] [Indexed: 07/15/2023]
Abstract
Cardiovascular disease is the leading cause of death in patients with chronic kidney disease (CKD). As CKD progresses, CKD-specific risk factors, such as disordered mineral homeostasis, amplify traditional cardiovascular risk factors. Fibroblast growth factor 23 (FGF23) regulates mineral homeostasis by activating complexes of FGF receptors and transmembrane klotho co-receptors. A soluble form of klotho also acts as a 'portable' FGF23 co-receptor in tissues that do not express klotho. In progressive CKD, rising circulating FGF23 levels in combination with decreasing kidney expression of klotho results in klotho-independent effects of FGF23 on the heart that promote left ventricular hypertrophy, heart failure, atrial fibrillation and death. Emerging data suggest that soluble klotho might mitigate some of these effects via several candidate mechanisms. More research is needed to investigate FGF23 excess and klotho deficiency in specific cardiovascular complications of CKD, but the pathophysiological primacy of FGF23 excess versus klotho deficiency might never be precisely resolved, given the entangled feedback loops that they share. Therefore, randomized trials should prioritize clinical practicality over scientific certainty by targeting disordered mineral homeostasis holistically in an effort to improve cardiovascular outcomes in patients with CKD.
Collapse
Affiliation(s)
- Daniel Edmonston
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Alexander Grabner
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Myles Wolf
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
114
|
Chinnappa S, Maqbool A, Viswambharan H, Mooney A, Denby L, Drinkhill M. Beta Blockade Prevents Cardiac Morphological and Molecular Remodelling in Experimental Uremia. Int J Mol Sci 2023; 25:373. [PMID: 38203544 PMCID: PMC10778728 DOI: 10.3390/ijms25010373] [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: 11/21/2023] [Revised: 12/18/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Heart failure and chronic kidney disease (CKD) share several mediators of cardiac pathological remodelling. Akin to heart failure, this remodelling sets in motion a vicious cycle of progressive pathological hypertrophy and myocardial dysfunction in CKD. Several decades of heart failure research have shown that beta blockade is a powerful tool in preventing cardiac remodelling and breaking this vicious cycle. This phenomenon remains hitherto untested in CKD. Therefore, we set out to test the hypothesis that beta blockade prevents cardiac pathological remodelling in experimental uremia. Wistar rats had subtotal nephrectomy or sham surgery and were followed up for 10 weeks. The animals were randomly allocated to the beta blocker metoprolol (10 mg/kg/day) or vehicle. In vivo and in vitro cardiac assessments were performed. Cardiac tissue was extracted, and protein expression was quantified using immunoblotting. Histological analyses were performed to quantify myocardial fibrosis. Beta blockade attenuated cardiac pathological remodelling in nephrectomised animals. The echocardiographic left ventricular mass and the heart weight to tibial length ratio were significantly lower in nephrectomised animals treated with metoprolol. Furthermore, beta blockade attenuated myocardial fibrosis associated with subtotal nephrectomy. In addition, the Ca++- calmodulin-dependent kinase II (CAMKII) pathway was shown to be activated in uremia and attenuated by beta blockade, offering a potential mechanism of action. In conclusion, beta blockade attenuated hypertrophic signalling pathways and ameliorated cardiac pathological remodelling in experimental uremia. The study provides a strong scientific rationale for repurposing beta blockers, a tried and tested treatment in heart failure, for the benefit of patients with CKD.
Collapse
Affiliation(s)
- Shanmugakumar Chinnappa
- Department of Nephrology, Doncaster and Bassetlaw Teaching Hospitals NHS Trust, Doncaster DN2 5LT, UK
- Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), University of Leeds, Leeds LS2 9JT, UK; (A.M.); (H.V.)
| | - Azhar Maqbool
- Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), University of Leeds, Leeds LS2 9JT, UK; (A.M.); (H.V.)
| | - Hema Viswambharan
- Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), University of Leeds, Leeds LS2 9JT, UK; (A.M.); (H.V.)
| | - Andrew Mooney
- Department of Nephrology, Leeds Teaching Hospitals NHS Trust, Leeds LS9 7TF, UK;
| | - Laura Denby
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK;
| | - Mark Drinkhill
- Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), University of Leeds, Leeds LS2 9JT, UK; (A.M.); (H.V.)
| |
Collapse
|
115
|
Cepoi MR, Duca ST, Chetran A, Costache AD, Spiridon MR, Afrăsânie I, Leancă SA, Dmour BA, Matei IT, Miftode RS, Miftode L, Prepeliuc CS, Haba MȘC, Bădescu MC, Costache II. Chronic Kidney Disease Associated with Ischemic Heart Disease: To What Extent Do Biomarkers Help? Life (Basel) 2023; 14:34. [PMID: 38255650 PMCID: PMC10817293 DOI: 10.3390/life14010034] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/17/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Chronic kidney disease represents a complex and multifaceted pathology characterized by the presence of structural or functional renal anomalies associated with a persistent reduction in renal function. As the disease progresses, complications arise due to the chronic inflammatory syndrome, hydro-electrolytic disorders, and toxicity secondary to the uremic environment. Cardiovascular complications are the leading cause of death for these patients. Ischemic cardiac pathology can be both a consequence and complication of chronic kidney disease, highlighting the need to identify specific cardiorenal dysfunction biomarkers targeting pathophysiological mechanisms common to both conditions. This identification is crucial for establishing accurate diagnoses, prognoses, and risk stratifications for patients. This work is intended to elucidate the intricate relationship between chronic kidney disease and ischemic heart disease and to investigate the roles of cardiorenal biomarkers, including cardiac troponin, natriuretic peptides, galectin-3, copeptin, fibroblast growth factor 23 and its co-receptor Klotho, soluble suppression of tumorigenicity 2, and plasma growth differentiation factor 15.
Collapse
Affiliation(s)
- Maria-Ruxandra Cepoi
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Stefania Teodora Duca
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Adriana Chetran
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Alexandru Dan Costache
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iași, Romania
| | - Marilena Renata Spiridon
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Irina Afrăsânie
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Sabina Andreea Leancă
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Bianca-Ana Dmour
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of III Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Iulian Theodor Matei
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Radu Stefan Miftode
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Larisa Miftode
- Department of Infectious Diseases, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (L.M.); (C.S.P.)
- “St. Parascheva” Clinical Hospital of Infectious Diseases, 700116 Iași, Romania
| | - Cristian Sorin Prepeliuc
- Department of Infectious Diseases, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (L.M.); (C.S.P.)
- “St. Parascheva” Clinical Hospital of Infectious Diseases, 700116 Iași, Romania
| | - Mihai Ștefan Cristian Haba
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| | - Minerva Codruța Bădescu
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of III Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Irina Iuliana Costache
- Department of Internal Medicine, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania; (M.-R.C.); (S.T.D.); (A.C.); (I.A.); (S.A.L.); (B.-A.D.); (I.T.M.); (R.S.M.); (M.Ș.C.H.); (M.C.B.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania;
| |
Collapse
|
116
|
Fuchs MA, Burke EJ, Latic N, Murray S, Li H, Sparks M, Abraham D, Zhang H, Rosenberg P, Hänzelmann S, Hausmann F, Huber T, Erben R, Fisher-Wellman K, Bursac N, Wolf M, Grabner A. Fibroblast Growth Factor (FGF) 23 and FGF Receptor 4 promote cardiac metabolic remodeling in chronic kidney disease. RESEARCH SQUARE 2023:rs.3.rs-3705543. [PMID: 38196615 PMCID: PMC10775858 DOI: 10.21203/rs.3.rs-3705543/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Chronic kidney disease (CKD) is a global health epidemic that significantly increases mortality due to cardiovascular disease. Left ventricular hypertrophy (LVH) is an important mechanism of cardiac injury in CKD. High serum levels of fibroblast growth factor (FGF) 23 in patients with CKD may contribute mechanistically to the pathogenesis of LVH by activating FGF receptor (FGFR) 4 signaling in cardiac myocytes. Mitochondrial dysfunction and cardiac metabolic remodeling are early features of cardiac injury that predate development of hypertrophy, but these mechanisms of disease have been insufficiently studied in models of CKD. Wild-type mice with CKD induced by adenine diet developed LVH that was preceded by morphological changes in mitochondrial structure and evidence of cardiac mitochondrial and metabolic dysfunction. In bioengineered cardio-bundles and neonatal rat ventricular myocytes grown in vitro, FGF23-mediated activation of FGFR4 caused a mitochondrial pathology, characterized by increased bioenergetic stress and increased glycolysis, that preceded the development of cellular hypertrophy. The cardiac metabolic changes and associated mitochondrial alterations in mice with CKD were prevented by global or cardiac-specific deletion of FGFR4. These findings indicate that metabolic remodeling and eventually mitochondrial dysfunction are early cardiac complications of CKD that precede structural remodeling of the heart. Mechanistically, FGF23-mediated activation of FGFR4 causes mitochondrial dysfunction, suggesting that early pharmacologic inhibition of FGFR4 might serve as novel therapeutic intervention to prevent development of LVH and heart failure in patients with CKD.
Collapse
Affiliation(s)
- Michaela A. Fuchs
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Emily J. Burke
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Nejla Latic
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Susan Murray
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Hanjun Li
- Department of Biomedical Engineering, Duke University, Durham, USA
| | - Matthew Sparks
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Dennis Abraham
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Hengtao Zhang
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Paul Rosenberg
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Sonja Hänzelmann
- Division of Nephrology, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabian Hausmann
- Division of Nephrology, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Huber
- Division of Nephrology, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Reinhold Erben
- Ludwig Boltzmann Institute of Osteology, Hanusch Hospital, Vienna, Austria
| | - Kelsey Fisher-Wellman
- East Carolina Diabetes and Obesity Institute, Brody School of Medicine, Department of Physiology, East Carolina University, Greenville, North Carolina, USA
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, USA
- Duke Regeneration Center, Duke University, Durham, North Carolina, USA
| | - Myles Wolf
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Alexander Grabner
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Division of Nephrology, Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Duke Regeneration Center, Duke University, Durham, North Carolina, USA
| |
Collapse
|
117
|
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
|
118
|
van der Vaart A, Bakker SJL, Laverman GD, van Dijk PR, de Borst MH. NT-proBNP Mediates the Association Between FGF23 and All-Cause Mortality in Individuals With Type 2 Diabetes. J Am Heart Assoc 2023; 12:e031873. [PMID: 38014662 PMCID: PMC10727346 DOI: 10.1161/jaha.123.031873] [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: 07/20/2023] [Accepted: 09/27/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND FGF23 (fibroblast growth factor 23) is associated with a higher mortality risk in type 2 diabetes, but the mechanism is unclear. We aimed to study whether NT-proBNP (N-terminal pro-brain natriuretic peptide) mediates the association between FGF23 and mortality. METHODS AND RESULTS We analyzed C-terminal FGF23 and NT-proBNP levels in 399 patients with type 2 diabetes. Cox regression analyses were performed, followed by mediation analyses using Structural Equation Modeling. During follow-up of 9.2 [7.6-11.3] years, 117 individuals died. FGF23 was associated with all-cause mortality, independent of potential confounders (fully adjusted hazard ratio [HR], 2.32 [95% CI, 1.21-4.43], P=0.01). The association was lost upon further adjustment for NT-proBNP (HR, 1.84; 95% CI, 0.91-3.73). NT-proBNP accounted for 26% of the mediation effect between FGF23 and all-cause mortality. CONCLUSIONS These findings suggest that a higher FGF23 level is associated with increased mortality in individuals with type 2 diabetes through an effect on volume homeostasis.
Collapse
Affiliation(s)
- Amarens van der Vaart
- Department of Internal Medicine, Division of NephrologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
- Department of Internal Medicine, Division of EndocrinologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Stephan J. L. Bakker
- Department of Internal Medicine, Division of NephrologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Gozewijn D. Laverman
- Division of Nephrology, Department of Internal MedicineZiekenhuisgroep TwenteHengeloThe Netherlands
| | - Peter R. van Dijk
- Department of Internal Medicine, Division of EndocrinologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - Martin H. de Borst
- Department of Internal Medicine, Division of NephrologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| |
Collapse
|
119
|
Biruete A, Chen NX, Metzger CE, Srinivasan S, O'Neill K, Fallen PB, Fonseca A, Wilson HE, de Loor H, Evenepoel P, Swanson KS, Allen MR, Moe SM. The Dietary Fiber Inulin Slows Progression of Chronic Kidney Disease-Mineral Bone Disorder (CKD-MBD) in a Rat Model of CKD. JBMR Plus 2023; 7:e10837. [PMID: 38130753 PMCID: PMC10731114 DOI: 10.1002/jbm4.10837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/14/2023] [Accepted: 09/22/2023] [Indexed: 12/23/2023] Open
Abstract
Chronic kidney disease (CKD)-mineral bone disorder (CKD-MBD) leads to fractures and cardiovascular disease. Observational studies suggest beneficial effects of dietary fiber on both bone and cardiovascular outcomes, but the effect of fiber on CKD-MBD is unknown. To determine the effect of fiber on CKD-MBD, we fed the Cy/+ rat with progressive CKD a casein-based diet of 0.7% phosphate with 10% inulin (fermentable fiber) or cellulose (non-fermentable fiber) from 22 weeks to either 30 or 32 weeks of age (~30% and ~15% of normal kidney function; CKD 4 and 5). We assessed CKD-MBD end points of biochemistry, bone quantity and quality, cardiovascular health, and cecal microbiota and serum gut-derived uremic toxins. Results were analyzed by two-way analysis of variance (ANOVA) to evaluate the main effects of CKD stage and inulin, and their interaction. The results showed that in CKD animals, inulin did not alter kidney function but reduced the increase from stage 4 to 5 in serum levels of phosphate and parathyroid hormone, but not fibroblast growth factor-23 (FGF23). Bone turnover and cortical bone parameters were similarly improved but mechanical properties were not altered. Inulin slowed progression of aorta and cardiac calcification, left ventricular mass index, and fibrosis. To understand the mechanism, we assessed intestinal microbiota and found changes in alpha and beta diversity and significant changes in several taxa with inulin, together with a reduction in circulating gut derived uremic toxins such as indoxyl sulfate and short-chain fatty acids. In conclusion, the addition of the fermentable fiber inulin to the diet of CKD rats led to a slowed progression of CKD-MBD without affecting kidney function, likely mediated by changes in the gut microbiota composition and lowered gut-derived uremic toxins. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Annabel Biruete
- Department of Nutrition SciencePurdue UniversityWest LafayetteINUSA
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisINUSA
| | - Neal X. Chen
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisINUSA
| | - Corinne E. Metzger
- Department of Anatomy, Cell Biology, and PhysiologyIndiana University School of MedicineIndianapolisINUSA
| | - Shruthi Srinivasan
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisINUSA
| | - Kalisha O'Neill
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisINUSA
| | - Paul B. Fallen
- Department of Anatomy, Cell Biology, and PhysiologyIndiana University School of MedicineIndianapolisINUSA
| | - Austin Fonseca
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisINUSA
| | - Hannah E. Wilson
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisINUSA
| | - Henriette de Loor
- KU Leuven Department of Microbiology and ImmunologyNephrology and Renal Transplantation Research Group, KU LeuvenLeuvenBelgium
| | - Pieter Evenepoel
- KU Leuven Department of Microbiology and ImmunologyNephrology and Renal Transplantation Research Group, KU LeuvenLeuvenBelgium
- Department of Nephrology and Renal TransplantationUniversity Hospitals LeuvenLeuvenBelgium
| | - Kelly S. Swanson
- Department of Animal SciencesUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
| | - Matthew R. Allen
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisINUSA
- Department of Anatomy, Cell Biology, and PhysiologyIndiana University School of MedicineIndianapolisINUSA
| | - Sharon M. Moe
- Department of Medicine, Division of NephrologyIndiana University School of MedicineIndianapolisINUSA
- Department of Anatomy, Cell Biology, and PhysiologyIndiana University School of MedicineIndianapolisINUSA
| |
Collapse
|
120
|
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
|
121
|
Michon-Colin A, Metzger M, Bankir L, Gauci C, Brunel M, Baron S, Prot-Bertoye C, Stengel B, Thervet E, Haymann JP, Boffa JJ, Vrtovsnik F, Flamant M, Houillier P, Prie D, Courbebaisse M. Fibroblast growth factor 23 but not copeptin is independently associated with kidney failure and mortality in patients with chronic kidney disease. Clin Kidney J 2023; 16:2472-2481. [PMID: 38046034 PMCID: PMC10689138 DOI: 10.1093/ckj/sfad149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Indexed: 12/05/2023] Open
Abstract
Background Copeptin and intact fibroblast growth factor 23 (iFGF23) increase early during chronic kidney disease (CKD) and may be predictive of unfavourable outcomes. The aim of this study was to evaluate their respective associations with renal and vital outcomes in CKD patients. Methods We included CKD patients from the NephroTest cohort with concomitant measurements of plasma copeptin and iFGF23 concentrations and isotopic glomerular filtration rate measurement (mGFR). The primary endpoint was a composite outcome including kidney failure (KF) (dialysis initiation, pre-emptive transplantation or a 57% decrease of mGFR, corresponding to doubling of serum creatinine) or death before KF. Hazard ratios (HRs) of the primary endpoint associated with log-transformed copeptin and iFGF23 concentrations were estimated by Cox models. The slope of mGFR over time was analysed using a linear mixed model. Results A total of 329 CKD patients (243 men, mean age 60.3 ± 14.6 years) were included. Among them, 301 with an mGFR >15 ml/min/1.73 m2 were included in survival and mGFR slope analyses. During a median follow-up of 4.61 years (quartile 1-quartile 3: 3.72-6.07), 61 KFs and 32 deaths occurred. Baseline iFGF23 concentrations were associated with the composite outcome after multiple adjustments {HR 2.72 [95% confidence interval (CI) 1.85-3.99]}, whereas copeptin concentrations were not [HR 1.01 (95% CI 0.74-1.39)]. Neither copeptin nor iFGF23 were associated with mGFR slope over time. Conclusion Our study shows for the first time in population of CKD patients an independent association between iFGF23 and unfavourable renal and vital outcomes and shows no such association regarding copeptin, encouraging the integration of iFGF23 measurement into the follow-up of CKD.
Collapse
Affiliation(s)
- Arthur Michon-Colin
- Université Paris Cité, Paris, France
- Explorations Fonctionnelles Rénales – Physiologie, Hôpital Européen Georges-Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
- Centre de Référence des Maladies Rénales Héréditaires de l’Enfant et de l’Adulte, Paris, France
- Centre de Référence des Maladies Rares du Calcium et du Phosphate, Paris, France
| | - Marie Metzger
- INSERM UMRS 1018, Equipe d'Epidémiologie Clinique, CESP, Université Paris-Saclay, Villejuif, France
| | - Lise Bankir
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Paris, France
- CNRS, ERL 8228, Laboratory of Kidney Physiology and Tubulopathies, Paris, France
| | - Cédric Gauci
- Explorations Fonctionnelles Rénales – Physiologie, Hôpital Européen Georges-Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
- INSERM UMRS 1018, Equipe d'Epidémiologie Clinique, CESP, Université Paris-Saclay, Villejuif, France
| | - Mélanie Brunel
- Université Paris Cité, Paris, France
- Explorations Fonctionnelles Rénales – Physiologie, Hôpital Européen Georges-Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
- Centre de Référence des Maladies Rénales Héréditaires de l’Enfant et de l’Adulte, Paris, France
- Centre de Référence des Maladies Rares du Calcium et du Phosphate, Paris, France
| | - Stéphanie Baron
- Université Paris Cité, Paris, France
- Explorations Fonctionnelles Rénales – Physiologie, Hôpital Européen Georges-Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Paris, France
- Centre de Référence des Maladies Rénales Héréditaires de l’Enfant et de l’Adulte, Paris, France
- Centre de Référence des Maladies Rares du Calcium et du Phosphate, Paris, France
| | - Caroline Prot-Bertoye
- Explorations Fonctionnelles Rénales – Physiologie, Hôpital Européen Georges-Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Paris, France
- CNRS, ERL 8228, Laboratory of Kidney Physiology and Tubulopathies, Paris, France
- Centre de Référence des Maladies Rénales Héréditaires de l’Enfant et de l’Adulte, Paris, France
- Centre de Référence des Maladies Rares du Calcium et du Phosphate, Paris, France
| | - Bénédicte Stengel
- INSERM UMRS 1018, Equipe d'Epidémiologie Clinique, CESP, Université Paris-Saclay, Villejuif, France
| | - Eric Thervet
- Université Paris Cité, Paris, France
- Néphrologie et Hémodialyse, Hôpital Européen Georges-Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Jean-Philippe Haymann
- Explorations Fonctionnelles Multidisciplinaires, Sorbonne Université Paris, France
- Explorations Fonctionnelles Multidisciplinaires, Hôpital Tenon, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Jean-Jacques Boffa
- Explorations Fonctionnelles Multidisciplinaires, Sorbonne Université Paris, France
- Néphrologie et Dialyse, Hôpital Tenon, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - François Vrtovsnik
- Université Paris Cité, Paris, France
- Néphrologie, Hôpital Bichat, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Martin Flamant
- Université Paris Cité, Paris, France
- Explorations Fonctionnelles Multidisciplinaires, Hôpital Bichat, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Pascal Houillier
- Université Paris Cité, Paris, France
- Explorations Fonctionnelles Rénales – Physiologie, Hôpital Européen Georges-Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Paris, France
- Centre de Référence des Maladies Rénales Héréditaires de l’Enfant et de l’Adulte, Paris, France
- Centre de Référence des Maladies Rares du Calcium et du Phosphate, Paris, France
| | - Dominique Prie
- Université Paris Cité, Paris, France
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Département de Physiologie, Hôpital Necker, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Marie Courbebaisse
- Université Paris Cité, Paris, France
- Explorations Fonctionnelles Rénales – Physiologie, Hôpital Européen Georges-Pompidou, Assistance Publique – Hôpitaux de Paris, Paris, France
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
- Centre de Référence des Maladies Rénales Héréditaires de l’Enfant et de l’Adulte, Paris, France
- Centre de Référence des Maladies Rares du Calcium et du Phosphate, Paris, France
| |
Collapse
|
122
|
Balogh DB, Molnar A, Degi A, Toth A, Lenart L, Saeed A, Barczi A, Szabo AJ, Wagner LJ, Reusz G, Fekete A. Cardioprotective and Antifibrotic Effects of Low-Dose Renin-Angiotensin-Aldosterone System Inhibitors in Type 1 Diabetic Rat Model. Int J Mol Sci 2023; 24:17043. [PMID: 38069366 PMCID: PMC10707380 DOI: 10.3390/ijms242317043] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Diabetic cardiovascular complications are associated with up to 50% mortality, and current therapies are not effective enough. Renin-angiotensin-aldosterone system inhibitors (RAASis) are the standard of care for diabetic patients with hypertension and albuminuria. Based on our previous studies reporting the renoprotective effects of low-dose RAASis, here, we hypothesized that low-dose RAASi treatment has cardioprotective and antifibrotic benefits in type 1 diabetes mellitus (T1DM). After five weeks of T1DM, adult male Wistar rats received low doses of ramipril, losartan, or eplerenone for two weeks. Heart rate, blood pressure, and pulse wave velocity (PWV) were recorded. Aortic intima-media thickness (IMT), collagen accumulation, and myocardial fibrosis were assessed. All RAASis reduced PWV elevation, prevented the progression of myocardial fibrosis, and normalized B-type natriuretic peptide, troponin I, and fibroblast growth factor 23 levels without affecting blood pressure. Interestingly, only eplerenone reversed the decline in Klotho levels and reduced IMT and fibrosis in the media of the aorta. Our comparative analysis suggests that mineralocorticoid receptor antagonists, particularly eplerenone, may offer superior efficacy in halting both the arterial and the myocardial injuries in T1DM compared to angiotensin-converting enzyme inhibitors or angiotensin II type 1 receptor blockers.
Collapse
Affiliation(s)
- Dora B. Balogh
- MTA-SE Lendület “Momentum” Diabetes Research Group, 1083 Budapest, Hungary; (D.B.B.); (A.T.); (L.L.); (A.S.)
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary; (A.M.); (A.D.); (A.J.S.); (G.R.)
| | - Agnes Molnar
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary; (A.M.); (A.D.); (A.J.S.); (G.R.)
| | - Arianna Degi
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary; (A.M.); (A.D.); (A.J.S.); (G.R.)
| | - Akos Toth
- MTA-SE Lendület “Momentum” Diabetes Research Group, 1083 Budapest, Hungary; (D.B.B.); (A.T.); (L.L.); (A.S.)
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary; (A.M.); (A.D.); (A.J.S.); (G.R.)
| | - Lilla Lenart
- MTA-SE Lendület “Momentum” Diabetes Research Group, 1083 Budapest, Hungary; (D.B.B.); (A.T.); (L.L.); (A.S.)
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary; (A.M.); (A.D.); (A.J.S.); (G.R.)
| | - Adar Saeed
- MTA-SE Lendület “Momentum” Diabetes Research Group, 1083 Budapest, Hungary; (D.B.B.); (A.T.); (L.L.); (A.S.)
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary; (A.M.); (A.D.); (A.J.S.); (G.R.)
| | - Adrienn Barczi
- Medical Imaging Center, Semmelweis University, 1082 Budapest, Hungary;
| | - Attila J. Szabo
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary; (A.M.); (A.D.); (A.J.S.); (G.R.)
| | - Laszlo J. Wagner
- Department of Surgery, Transplantation and Gastroenterology, Semmelweis University, 1082 Budapest, Hungary;
| | - Gyorgy Reusz
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary; (A.M.); (A.D.); (A.J.S.); (G.R.)
| | - Andrea Fekete
- MTA-SE Lendület “Momentum” Diabetes Research Group, 1083 Budapest, Hungary; (D.B.B.); (A.T.); (L.L.); (A.S.)
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary; (A.M.); (A.D.); (A.J.S.); (G.R.)
| |
Collapse
|
123
|
Ryu JH, Jeon HJ, Han R, Jung HY, Kim MG, Huh KH, Park JB, Kang KP, Han S, Yang J. High pretransplant FGF23 level is associated with persistent vitamin D insufficiency and poor graft survival in kidney transplant patients. Sci Rep 2023; 13:19640. [PMID: 37949967 PMCID: PMC10638428 DOI: 10.1038/s41598-023-46889-0] [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: 08/08/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023] Open
Abstract
Vitamin D3 (25[OH]D3) insufficiency and fibroblast growth factor 23 (FGF23) elevation are usually attenuated after kidney transplantation (KT). However, elevated FGF23 may be associated with poor graft outcomes and vitamin D insufficiency after KT. This study investigated the effect of pretransplant FGF23 levels on post-KT 25(OH)D3 status and graft outcomes. Serum FGF23 levels from 400 participants of the KoreaN Cohort Study for Outcome in Patients With Kidney Transplantation were measured. Annual serum 25(OH)D3 levels, all-cause mortality, cardiovascular event, and graft survival were assessed according to baseline FGF23 levels. Serum 25(OH)D3 levels were initially increased 1 year after KT (12.6 ± 7.4 vs. 22.6 ± 6.4 ng/mL). However, the prevalence of post-KT vitamin D deficiency increased again after post-KT 3 years (79.1% at baseline, 30.8% and 37.8% at 3 and 6 years, respectively). Serum FGF23 level was decreased 3 years post-KT. When participants were categorized into tertiles according to baseline FGF23 level (low, middle, high), 25(OH)D3 level in the low FGF23 group was persistently low at a median follow-up of 8.3 years. Furthermore, high baseline FGF23 level was a risk factor for poor graft survival (HR 5.882, 95% C.I.; 1.443-23.976, P = 0.013). Elevated FGF23 levels are associated with persistently low post-transplant vitamin D levels and poor graft survival.
Collapse
Affiliation(s)
- Jung-Hwa Ryu
- Department of Internal Medicine, Ewha Womans University Seoul Hospital, Seoul, Republic of Korea
| | - Hee Jung Jeon
- Department of Internal Medicine, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Ro Han
- Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Hee-Yeon Jung
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Myung-Gyu Kim
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyu Ha Huh
- Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae Berm Park
- Department of Surgery, Seoul Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Kyung Pyo Kang
- Department of Internal Medicine, Jeonbuk National University Hospital, Jeonju, Republic of Korea
| | - Seungyeup Han
- Department of Internal Medicine, Dongsan Medical Center, Keimyung University, Daegu, Republic of Korea
| | - Jaeseok Yang
- Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
124
|
Junho CVC, Frisch J, Soppert J, Wollenhaupt J, Noels H. Cardiomyopathy in chronic kidney disease: clinical features, biomarkers and the contribution of murine models in understanding pathophysiology. Clin Kidney J 2023; 16:1786-1803. [PMID: 37915935 PMCID: PMC10616472 DOI: 10.1093/ckj/sfad085] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Indexed: 11/03/2023] Open
Abstract
The cardiorenal syndrome (CRS) is described as a multi-organ disease encompassing bidirectionally heart and kidney. In CRS type 4, chronic kidney disease (CKD) leads to cardiac injury. Different pathological mechanisms have been identified to contribute to the establishment of CKD-induced cardiomyopathy, including a neurohormonal dysregulation, disturbances in the mineral metabolism and an accumulation of uremic toxins, playing an important role in the development of inflammation and oxidative stress. Combined, this leads to cardiac dysfunction and cardiac pathophysiological and morphological changes, like left ventricular hypertrophy, myocardial fibrosis and cardiac electrical changes. Given that around 80% of dialysis patients suffer from uremic cardiomyopathy, the study of cardiac outcomes in CKD is clinically highly relevant. The present review summarizes clinical features and biomarkers of CKD-induced cardiomyopathy and discusses underlying pathophysiological mechanisms recently uncovered in the literature. It discloses how animal models have contributed to the understanding of pathological kidney-heart crosstalk, but also provides insights into the variability in observed effects of CKD on the heart in different CKD mouse models, covering both "single hit" as well as "multifactorial hit" models. Overall, this review aims to support research progress in the field of CKD-induced cardiomyopathy.
Collapse
Affiliation(s)
| | - Janina Frisch
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, Medical Faculty, Saarland University, Center for Human and Molecular Biology, Homburg/Saar, Germany
| | - Josefin Soppert
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
- Department of Anesthesiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Julia Wollenhaupt
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
125
|
Burstad KM, Cladis DP, Wiese GN, Butler M, Hill Gallant KM. Effects of Plant-Based Protein Consumption on Kidney Function and Mineral Bone Disorder Outcomes in Adults With Stage 3-5 Chronic Kidney Disease: A Systematic Review. J Ren Nutr 2023; 33:717-730. [PMID: 37116624 DOI: 10.1053/j.jrn.2023.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 02/13/2023] [Accepted: 04/07/2023] [Indexed: 04/30/2023] Open
Abstract
INTRODUCTION Plant-based protein is of growing interest for dietary management of chronic kidney disease (CKD) and is hypothesized to preserve kidney function and reduce CKD-mineral bone disorder (MBD) complications, among other benefits. This systematic review aimed to summarize the available clinical trial evidence for the effect of plant-based protein on kidney function and CKD-MBD outcomes in adults with stage 3-5 CKD not on dialysis. METHODS Searches of Medline, Embase, Agricola, CAB abstracts, Web of Science, Scopus, and hand searching were performed. Clinical trials with ≥8 participants ≥18 years of age with an estimated glomerular filtration rate <60 mL/min/1.73 m2 but not on dialysis were included. Additionally, only clinical trials with ≥1-week interventions with ≥50% dietary protein from plant-based sources and reported at least one outcome for both kidney function and CKD-MBD outcomes were included. Of the 10,962 identified abstracts, 32 met inclusion criteria and were assessed for risk of bias. RESULTS Results for kidney function and CKD-MBD outcomes were heterogenous, with most studies having suboptimal methodological quality. In most of the studies (27/32), protein source was altered only secondarily to low-protein diet interventions. Thus, data synthesis and interpretation were focused on a subset of five studies that investigated a change in protein source only (i.e., animal vs. plant). Of this subset, four studies reported no change in kidney function, while one study reported a decrease. Three studies reported no change in serum phosphorus, and one study reported lower serum phosphorus following a vegetarian diet. Further, limited data and inconclusive results were observed for phosphaturic hormones, parathyroid hormone, and fibroblast growth factor-23. CONCLUSION Current clinical trial evidence on plant-based protein interventions for preserving kidney function and preventing CKD-MBD is limited to inform clinical guidelines at this time. This systematic review emphasizes the ongoing need to research the effects of plant-based protein on kidney function and CKD-MBD outcomes.
Collapse
Affiliation(s)
- Kendal M Burstad
- Department of Food Science and Nutrition, University of Minnesota, Saint Paul, Minnesota
| | - Dennis P Cladis
- Department of Food Science and Nutrition, University of Minnesota, Saint Paul, Minnesota
| | | | - Mary Butler
- Division of Health Policy and Management, University of Minnesota School of Public Health, Minneapolis, Minnesota
| | - Kathleen M Hill Gallant
- Department of Food Science and Nutrition, University of Minnesota, Saint Paul, Minnesota; Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
126
|
Albrecht LV, Pereira RC, Salusky IB. All the might of the osteocyte: emerging roles in chronic kidney disease. Kidney Int 2023; 104:910-915. [PMID: 37648154 DOI: 10.1016/j.kint.2023.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/04/2023] [Accepted: 08/11/2023] [Indexed: 09/01/2023]
Abstract
Osteocytes are the most abundant type of bone cell and play crucial roles in bone health. Osteocytes sense mechanical stress and orchestrate osteoblasts and osteoclasts to maintain bone density and strength. Beyond this, osteocytes have also emerged as key regulators of organ crosstalk, and they function as endocrine organs via their roles in secreting factors that mediate signaling within their neighboring bone cells and in distant tissues. As such, osteocyte dysfunction has been associated with the bone abnormalities seen across a spectrum of chronic kidney disease. Specifically, dysregulated osteocyte morphology and signaling have been observed in the earliest stages of chronic kidney disease and have been suggested to contribute to kidney disease progression. More important, US Food and Drug Administration-approved inhibitors of osteocytic secreted proteins, such as fibroblast growth factor 23 and sclerostin, have been used to treat bone diseases. The present mini review highlights new research that links dysfunctional osteocytes to the pathogenesis of chronic kidney disease mineral and bone disorder.
Collapse
Affiliation(s)
- Lauren V Albrecht
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of California, Irvine, Irvine, California, USA.
| | - Renata C Pereira
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Isidro B Salusky
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA.
| |
Collapse
|
127
|
Biruete A, Hill Gallant KM, Lloyd L, Meade A, Moe SM, St-Jules DE, Kistler BM. 'Phos'tering a Clear Message: The Evolution of Dietary Phosphorus Management in Chronic Kidney Disease. J Ren Nutr 2023; 33:S13-S20. [PMID: 37343779 PMCID: PMC10728341 DOI: 10.1053/j.jrn.2023.05.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 04/21/2023] [Accepted: 05/04/2023] [Indexed: 06/23/2023] Open
Abstract
Phosphorus is a vital nutrient, but disturbances in phosphorus homeostasis are central to chronic kidney disease-mineral and bone disorder. To minimize disturbances, traditional dietary guidance focused on a numerical phosphorus target leading to the exclusion of many healthy foods and implementation challenges. Contemporary phosphorus guidance focuses on dietary source, avoiding additives, and emphasizing low-phosphorus bioaccessibility foods, leading to a more liberal approach. Additional work is needed to demonstrate the efficacy of these contemporary approaches and understand the influence of specific foods, processing, and cooking methods. Unfortunately, patient education using traditional and contemporary strategies may give mixed messages, particularly related to plant-based foods. Thus, greater clarity on the effects of specific foods and dietary patterns may improve phosphorus education. This review aims to discuss the evolution of dietary phosphorus management while highlighting areas for future research that can help move the field toward stronger evidence-based guidance to prevent and treat hyperphosphatemia.
Collapse
Affiliation(s)
- Annabel Biruete
- Department of Nutrition and Dietetics, Indiana University-Purdue University Indianapolis, Indianapolis; Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis; Department of Nutrition Science, Purdue University, West Lafayette, Indiana
| | - Kathleen M Hill Gallant
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis; Department of Food Science and Nutrition, University of Minnesota, St. Paul
| | - Lyn Lloyd
- Nutrition and Dietetics, Auckland City Hospital, Auckland, New Zealand
| | - Anthony Meade
- Central Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, Australia
| | - Sharon M Moe
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis
| | | | - Brandon M Kistler
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Department of Nutrition and Health Science, Ball State University, Muncie.
| |
Collapse
|
128
|
Leidner AS, Cai X, Zelnick LR, Lee J, Bansal N, Pasch A, Kansal M, Chen J, Anderson AH, Sondheimer JH, Lash JP, Townsend RR, Go AS, Feldman HI, Shah SJ, Wolf M, Isakova T, Mehta RC. Fibroblast Growth Factor 23 and Risk of Heart Failure Subtype: The CRIC (Chronic Renal Insufficiency Cohort) Study. Kidney Med 2023; 5:100723. [PMID: 37915961 PMCID: PMC10616385 DOI: 10.1016/j.xkme.2023.100723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023] Open
Abstract
Rationale & Objective Heart failure (HF) is an important cause of morbidity and mortality among individuals with chronic kidney disease (CKD). A large body of evidence from preclinical and clinical studies implicates excess levels of fibroblast growth factor 23 (FGF23) in HF pathogenesis in CKD. It remains unclear whether the relationship between elevated FGF23 levels and HF risk among individuals with CKD varies by HF subtype. Study Design Prospective cohort study. Settings & Participants A total of 3,502 participants were selected in the Chronic Renal Insufficiency Cohort study. Exposure Baseline plasma FGF23. Outcomes Incident HF by subtype and total rate of HF hospitalization. HF was categorized as HF with preserved ejection fraction (HFpEF, ejection fraction [EF] ≥ 50%), HF with reduced EF (HFrEF, EF < 50%) and HF with unknown EF (HFuEF). Analytical Approach Multivariable-adjusted cause-specific Cox proportional hazards models were used to investigate associations between FGF23 and incident hospitalizations for HF by subtype. The Lunn-McNeil method was used to compare hazard ratios across HF subtypes. Poisson regression models were used to evaluate the total rate of HF. Results During a median follow-up time of 10.8 years, 295 HFpEF, 242 HFrEF, and 156 HFuEF hospitalizations occurred. In multivariable-adjusted cause-specific Cox proportional hazards models, FGF23 was significantly associated with the incidence of HFpEF (HR, 1.41; 95% CI, 1.21-1.64), HFrEF (HR, 1.27; 95% CI, 1.05-1.53), and HFuEF (HR, 1.40; 95% CI, 1.13-1.73) per 1 standard deviation (SD) increase in the natural log of FGF23. The Lunn-McNeil method determined that the risk association was consistent across all subtypes. The rate ratio of total HF events increased with FGF23 quartile. In multivariable-adjusted models, compared with quartile 1, FGF23 quartile 4 had a rate ratio of 1.81 (95% CI, 1.28-2.57) for total HF events. Limitations Self-report of HF hospitalizations and possible lack of an echocardiogram at time of hospitalization. Conclusions In this large multicenter prospective cohort study, elevated FGF23 levels were associated with increased risks for all HF subtypes. Plain-Language Summary Heart failure (HF) is a prominent cause of morbidity and mortality in individuals with chronic kidney disease (CKD). Identifying potential pathways in the development of HF is essential in developing therapies to prevent and treat HF. In a large cohort of individuals with CKD, the Chronic Renal Insufficiency Cohort (N = 3,502), baseline fibroblast growth factor-23 (FGF23), a hormone that regulates phosphorous, was evaluated in relation to the development of incident and recurrent HF with reduced, preserved, and unknown ejection fraction. In this large multicenter prospective cohort study, elevated FGF23 levels were associated with increased risk of all HF subtypes. These findings demonstrate the need for further research into FGF23 as a target in preventing the development of HF in individuals with CKD.
Collapse
Affiliation(s)
| | - Xuan Cai
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Jungwha Lee
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | | | | | | | | | | | | | | | | | | | - Sanjiv J. Shah
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Myles Wolf
- Duke University School of Medicine, Durham, North Carolina
| | - Tamara Isakova
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Rupal C. Mehta
- Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
129
|
Singh M, Pushpakumar S, Zheng Y, Smolenkova I, Akinterinwa OE, Luulay B, Tyagi SC. Novel mechanism of the COVID-19 associated coagulopathy (CAC) and vascular thromboembolism. NPJ VIRUSES 2023; 1:3. [PMID: 38077924 PMCID: PMC10710223 DOI: 10.1038/s44298-023-00003-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/20/2023] [Indexed: 01/31/2024]
Abstract
Previous studies from our laboratory revealed that SARS-CoV-2 spike protein (SP) administration to a genetically engineered model expressing the human angiotensin-converting enzyme 2; ACE2 receptor (i.e., hACE2 humanized mouse) mimicked the coronavirus disease-19 (COVID-19) pathology. In humans the cause of high morbidity, and mortality is due to 'cytokine-storm' led thromboembolism; however, the exact mechanisms of COVID-19 associated coagulopathy (CAC) have yet to be discovered. Current knowledge suggests that CAC is distinct from the standard coagulopathy, in that the intrinsic and extrinsic thrombin-dependent coagulation factors, and the pathway(s) that are common to coagulopathy, are not recruited by SARS-CoV-2. Findings from patients revealed that there is little change in their partial thromboplastin, or the prothrombin time coupled with a significant decline in platelets. Further, there appears to be an endothelial dysfunction during COVID-19 suggesting an interaction of the endothelia with immune cells including neutrophils. There are also reports that inflammatory NGAL is elevated during COVID-19. Furthermore, the levels of NPT are also increased indicating an increase in inflammatory M1 macrophage iNOS which sequesters BH4; an essential enzyme co-factor that acts as a potent antioxidant thus causing damage to endothelia. SARS-CoV-2 entry into the host cells is facilitated by a co-operative action between TMPRSS2 and the main ACE2 receptor. Interestingly, after infection ADAMTS13; a von Willebrand factor; VWF cleaving enzyme is found to be decreased. Based on these facts, we hypothesize that vascular thromboembolism is associated with serine and metalloproteinase, and in that context, we opine that inhibition of iNOS might help mitigate COVID-19 harmful effects. To test this hypothesis, we administered SP to the hACE2 mice that were subsequently treated with amino guanidine (AG; a potent inhibitor of glycoxidation, lipoxidation and oxidative vicious cycles). Our results revealed increase in TMPRSS2, and NGAL by SP but treatment with AG mitigated their levels. Similarly, levels of MMP-2, and -9 were increased; however, AG treatment normalized these levels. Our findings suggest that occurrence of CAC is influenced by TMPRSS2, ADAMTS13, NGAL and MMP- 2, and -9 factors, and an intervention with iNOS blocker helped mitigate the CAC condition in experimental settings.
Collapse
Affiliation(s)
- Mahavir Singh
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
- These authors contributed equally: Mahavir Singh, Sathnur Pushpakumar
| | - Sathnur Pushpakumar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
- These authors contributed equally: Mahavir Singh, Sathnur Pushpakumar
| | - Yuting Zheng
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Irina Smolenkova
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Oluwaseun E. Akinterinwa
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Bana Luulay
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Suresh C. Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
130
|
Rostamzadeh F, Moosavi-Saeed Y, Yeganeh-Hajahmadi M. Interaction of Klotho and sirtuins. Exp Gerontol 2023; 182:112306. [PMID: 37804921 DOI: 10.1016/j.exger.2023.112306] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 09/30/2023] [Accepted: 10/02/2023] [Indexed: 10/09/2023]
Abstract
OBJECTIVE In this article, we review the articles that have reported the interaction between Klotho and sirtuins. RECENT FINDINGS Sirtuins are a family of histone deacetylase enzymes that are considered to be the main regulators of biological processes. This family is one of the essential factors for postponing aging and increasing the life span of organisms. Sirtuins play a role in regulating the function of various cellular processes such as cellular metabolism, oxidative stress, apoptosis, and inflammation. It has also been shown that various diseases are related to these enzymes. Klotho is an anti-aging protein that exists as a membrane protein as well as a soluble circulating form. The membrane type of this protein acts as a co-receptor of the FGF endocrine family. It has been shown that the Klotho gene is related to age-related diseases, including osteoporosis, coronary artery, brain diseases, diabetes, etc. At the same time, it is difficult to separate the actions of Klotho and endocrine FGFs. Several studies have shown that Klotho and sirtuins interact with each other at different regulatory levels. However, it is necessary to carry out more in-vivo investigations to create new windows towards the treatment or prevention of various diseases.
Collapse
Affiliation(s)
- Farzaneh Rostamzadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Yasamin Moosavi-Saeed
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahboobeh Yeganeh-Hajahmadi
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Science, Kerman, Iran.
| |
Collapse
|
131
|
Mehta A, Chandiramani R, Spirito A, Vogel B, Mehran R. Significance of Kidney Disease in Cardiovascular Disease Patients. Interv Cardiol Clin 2023; 12:453-467. [PMID: 37673491 DOI: 10.1016/j.iccl.2023.06.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Cardiorenal syndrome is a condition where is a bidirectional and mutually detrimental relationship between the heart and kidneys. The mechanisms underlying cardiorenal syndrome are multifactorial and complex. Patients with kidney disease exhibit increased cardiovascular risk, presenting as coronary and peripheral artery disease, structural heart disease, arrhythmias, heart failure, and sudden cardiac death, largely occurring because of a systemic proinflammatory state, causing myocardial and vascular remodeling, manifesting as atherosclerotic lesions, vascular and valvular calcification, and myocardial fibrosis, particularly among those with advanced disease. This review summarizes the current understanding and clinical implications of kidney disease in patients with cardiovascular disease.
Collapse
Affiliation(s)
- Adhya Mehta
- Department of Internal Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, 1400 Pelham Parkway South, Bronx, NY 10461, USA
| | - Rishi Chandiramani
- Department of Internal Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, 1400 Pelham Parkway South, Bronx, NY 10461, USA; The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029, USA
| | - Alessandro Spirito
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029, USA
| | - Birgit Vogel
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029, USA
| | - Roxana Mehran
- Center for Interventional Cardiovascular Research and Clinical Trials, The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1030, New York, NY 10029-6574, USA.
| |
Collapse
|
132
|
Jia Z, Liu Q, Xie Y, Wei J, Sun X, Meng F, Zhao B, Yu Z, Zhao L, Xing Z. Klotho/FGF23 Axis Regulates Cardiomyocyte Apoptosis and Cytokine Release through ERK/MAPK Pathway. Cardiovasc Toxicol 2023; 23:317-328. [PMID: 37704925 DOI: 10.1007/s12012-023-09805-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/24/2023] [Indexed: 09/15/2023]
Abstract
Coronary artery disease (CAD) as a major cardiovascular disease is the leading global cause of mortality, Klotho/FGF23 axis involved in development of cardiovascular disease, while the function and underlying mechanism of Klotho/FGF23 axis in CAD is unclear. Blood samples from 67 CAD patients with coronary artery bypass graft (CABG) surgery were collected, and the level of Klotho and FGF23 of those patients was measured by using an ELISA kit. Cardiomyocyte was isolated from 0 to 3 days Sprague Dawley (SD) rats. Expression of Klotho, FGF23 and the cardiomyocyte marker α-sarcomeric actin (α-SA), myosin heavy chain (MHC) and cardiac troponin I (cTnI) was assessed by immunofluorescence staining. Expression of Klotho and FGF23 mRNA was detected by qRT-PCR. Apoptosis and cell cycle were measured by flow cytometry. Cell viability was detected by using CCK-8. The protein expression of ERK/MAPK pathway related protein and cytokines production was measured by western blotting. The levels of Klotho in CAD patients increased after CABG surgery, while FGF23 decreased. Isolated cardiomyocyte morphology and structure were completed, and with stabilized beating within culture for 15 days, besides, α-SA, MHC, and cTnI proved positive. After transfected Lenti-Klotho and Lenti-FGF23 into isolated cardiomyocyte, fluorescence staining showed that the transfection was successful, and qRT-PCR results showed that the expression levels of Klotho and FGF23 mRNA significant increased compared with NEG (empty vector) group. Immunofluorescence staining results showed that compared with NEG group, there was a higher Klotho positive rate and lower FGF23 positive rate in Klotho overexpression (Klotho) group, while, there was a higher FGF23 positive rate and lower Klotho positive rate in FGF23 overexpression (FGF23) group. In addition, the expression of p-ERK1/2 and p-P38 increased in Klotho group but decreased in FGF23 group. Furthermore, overexpression of Klotho inhibited cardiomyocyte apoptosis, increased S phase fraction, promoted proliferation and elevated expression of transforming growth factor β1 (TGF-β1), nuclear factor-kappa B (NF-κB), angiotensin-II (AT-II), and activator protein-1 (AP-1), overexpression of FGF23 showed the opposite effect, however, ERK agonist (TPA) and inhibitor (U0126) reversed the effect caused by overexpression of Klotho and FGF23 separately. Klotho/FGF23 axis play a critical role in CAD progression through regulating ERK/MAPK pathway in Cardiomyocyte.
Collapse
Affiliation(s)
- Zheng Jia
- Department of Cardiovascular Surgery, Yan'an Hospital Affiliated to Kunming Medical University, NO. 245 Renmin East Road, Panlong District, Kunming, 650051, Yunnan, China
| | - Qian Liu
- Department of Geriatric Cardiovascular, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Ying Xie
- Department of Cardiovascular Surgery, Yan'an Hospital Affiliated to Kunming Medical University, NO. 245 Renmin East Road, Panlong District, Kunming, 650051, Yunnan, China
| | - Jie Wei
- Department of Cardiovascular Surgery, Yan'an Hospital Affiliated to Kunming Medical University, NO. 245 Renmin East Road, Panlong District, Kunming, 650051, Yunnan, China
| | - Xiaolin Sun
- Department of Cardiovascular Surgery, Yan'an Hospital Affiliated to Kunming Medical University, NO. 245 Renmin East Road, Panlong District, Kunming, 650051, Yunnan, China
| | - Fandi Meng
- Department of Cardiovascular Surgery, Yan'an Hospital Affiliated to Kunming Medical University, NO. 245 Renmin East Road, Panlong District, Kunming, 650051, Yunnan, China
| | - Bin Zhao
- Department of Cardiovascular Surgery, Yan'an Hospital Affiliated to Kunming Medical University, NO. 245 Renmin East Road, Panlong District, Kunming, 650051, Yunnan, China
| | - Zhenkun Yu
- Department of Cardiovascular Surgery, Yan'an Hospital Affiliated to Kunming Medical University, NO. 245 Renmin East Road, Panlong District, Kunming, 650051, Yunnan, China
| | - Li Zhao
- Department of Cardiovascular Ultrasound, Yan'an Hospital Affiliated to Kunming Medical University, NO. 245 Renmin East Road, Panlong District, Kunming, 650051, Yunnan, China.
| | - Zhengjiang Xing
- Department of Cardiovascular Surgery, Yan'an Hospital Affiliated to Kunming Medical University, NO. 245 Renmin East Road, Panlong District, Kunming, 650051, Yunnan, China.
| |
Collapse
|
133
|
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
|
134
|
Elder GJ. Current Status of Mineral and Bone Disorders in Transplant Recipients. Transplantation 2023; 107:2107-2119. [PMID: 36788445 DOI: 10.1097/tp.0000000000004538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Most patients with end-stage kidney disease undergoing kidney transplantation are affected by the chronic kidney disease-mineral and bone disorder. This entity encompasses laboratory abnormalities, calcification of soft tissues, and the bone abnormalities of renal osteodystrophy that together result in an increased risk of fracture, cardiovascular events, and mortality. Although many biochemical disturbances associated with end-stage kidney disease improve in the first year after transplantation, hyperparathyroidism commonly persists, and residual changes of renal osteodystrophy are slow to resolve. When superimposed on common, traditional risk factors, post-transplant glucocorticoid treatment, the possibility of tubular disturbances and post-transplant chronic kidney disease, rates of incident fracture remain high. This review examines hormonal and biochemical changes before and after kidney transplantation, fracture risk assessment tools and imaging modalities, a staged approach to management and concerns associated with antiresorptive and anabolic therapies. A multidisciplinary approach is proposed as the best means to improve patient-level outcomes.
Collapse
Affiliation(s)
- Grahame J Elder
- Department of Renal Medicine, Westmead Hospital, Sydney, Australia
- Skeletal Biology Program, Garvan Institute of Medical Research, Sydney, Australia
- School of Medicine, University of Notre Dame, Sydney, Australia
| |
Collapse
|
135
|
Benes J, Kroupova K, Kotrc M, Petrak J, Jarolim P, Novosadova V, Kautzner J, Melenovsky V. FGF-23 is a biomarker of RV dysfunction and congestion in patients with HFrEF. Sci Rep 2023; 13:16004. [PMID: 37749114 PMCID: PMC10520041 DOI: 10.1038/s41598-023-42558-4] [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: 04/03/2023] [Accepted: 09/12/2023] [Indexed: 09/27/2023] Open
Abstract
There is no biomarker reflecting right ventricular dysfunction in HFrEF patients used in clinical practice. We have aimed to look for a circulating marker of RV dysfunction employing a quantitative proteomic strategy. The Olink Proteomics Multiplex panels (Cardiovascular Disease II, III, Cardiometabolic, and Inflammation Target Panels) identified FGF-23 to be the most differentially abundant (more than 2.5-fold) in blood plasma of HF patients with severe RV dysfunction (n = 30) compared to those with preserved RV function (n = 31). A subsequent ELISA-based confirmatory analysis of circulating FGF-23 in a large cohort of patients (n = 344, 72.7% NYHA III/IV, LVEF 22.5%, 54.1% with moderate/severe RV dysfunction), followed by multivariable regression analysis, revealed that the plasma FGF-23 level was most significantly associated with RV dysfunction grade (p = 0.0004) and congestion in the systemic circulation (p = 0.03), but not with LV-ejection fraction (p = 0.69) or estimated glomerular filtration rate (eGFR, p = 0.08). FGF-23 was associated with the degree of RV dysfunction in both sub-cohorts (i.e. in patients with and without congestion, p < 0.0001). The association between FGF-23 and RV-dysfunction remained significant after the adjustment for BNP (p = 0.01). In contrast, when adjusted for BNP, FGF-23 was no longer associated with LV dysfunction (p = 0.59). The Cox proportional hazard model revealed that circulating FGF-23 was significantly associated with adverse outcomes even after adjusting for BNP, LVEF, RV dysfunction grade and eGFR. Circulating FGF-23 is thus a biomarker of right ventricular dysfunction in HFrEF patients regardless of congestion status.
Collapse
Affiliation(s)
- Jan Benes
- Department of Cardiology, Institute for Clinical and Experimental Medicine-IKEM, Videnska 1958/9, 140 21 Praha 4, Prague, Czech Republic.
| | - Katerina Kroupova
- Department of Cardiology, Institute for Clinical and Experimental Medicine-IKEM, Videnska 1958/9, 140 21 Praha 4, Prague, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martin Kotrc
- Department of Cardiology, Institute for Clinical and Experimental Medicine-IKEM, Videnska 1958/9, 140 21 Praha 4, Prague, Czech Republic
| | - Jiri Petrak
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Petr Jarolim
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vendula Novosadova
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Josef Kautzner
- Department of Cardiology, Institute for Clinical and Experimental Medicine-IKEM, Videnska 1958/9, 140 21 Praha 4, Prague, Czech Republic
| | - Vojtech Melenovsky
- Department of Cardiology, Institute for Clinical and Experimental Medicine-IKEM, Videnska 1958/9, 140 21 Praha 4, Prague, Czech Republic
| |
Collapse
|
136
|
Tabibi MA, Wilund KR, Salimian N, Nikbakht S, Soleymany M, Roshanaeian Z, Nazemi F, Ahmadi S. The effect of intradialytic exercise on calcium, phosphorus and parathyroid hormone: a randomized controlled trial. BMC Nephrol 2023; 24:276. [PMID: 37730530 PMCID: PMC10512624 DOI: 10.1186/s12882-023-03327-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/07/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND Patients with kidney failure experience derangements of circulating markers of mineral metabolism and dysregulation of skeletal and cardiovascular physiology which results in high mortality rate in these patients. This study aimed to evaluate the effect of intradialytic exercise on regulation of these abnormalities in patients receiving chronic hemodialysis (HD). METHODS In this randomized controlled trial conducted in an HD center in Iran, adult patients receiving chronic HD were randomized to intradialytic exercise (60 min) in the second hour of thrice weekly dialysis for 6 months (intervention) or no intradialytic exercise (control). The primary outcomes were serum calcium, serum phosphorous and parathyroid hormone levels. Secondary outcomes were serum alkaline phosphatase and calcium-phosphorous product. RESULTS The study included 44 participants randomized to intervention (n = 22) or control (n = 22). During the 6-month intervention period, significant between-group changes were observed in all primary and secondary outcomes between the intervention and control groups. Statistical analyses reveal a significant increase in serum calcium (P < 0.05) as well as a significant decrease in serum phosphorous, parathyroid hormone, alkaline phosphatase and calcium-phosphorous product (P < 0.05). CONCLUSION Intradialytic exercise performed for at least 60 min during thrice weekly dialysis sessions improves bone mineral metabolism in adult patients receiving HD. Further studies should focus on observing and comparing the effect of different types of exercise on bone mineral disorders and all-cause mortality in HD patients. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04916743, Registered on 08/06/2021. Registered trial name: The Effect of Intradialytic Exercise on Calcium, Phosphorous and Parathyroid Hormone: A Randomized Controlled Trial.
Collapse
Affiliation(s)
- Mohammad Ali Tabibi
- Department of Exercise Physiology, Pardis Specialized Wellness Institute, Isfahan, Iran.
| | - Kenneth R Wilund
- Department of Kinesiology and Community Health, Division of Nutritional Sciences, University of Illinois, Urbana-Champaign, USA
| | - Nasrin Salimian
- Department of Research and Development, Pardis Specialized Wellness Institute, Isfahan, Iran
| | - Saghar Nikbakht
- Department of Kinesiology, Pardis Specialized Wellness Institute, Isfahan, Iran
| | - Mahsa Soleymany
- Department of Exercise Physiology, Pardis Specialized Wellness Institute, Isfahan, Iran
| | - Zahra Roshanaeian
- Department of Sport Nutrition, Pardis Specialized Wellness Institute, Isfahan, Iran
| | - Farzad Nazemi
- Department of Exercise Physiology, Pardis Specialized Wellness Institute, Isfahan, Iran
| | - Saghar Ahmadi
- Department of Health and Palliative Care, Pardis Specialized Wellness Institute, Isfahan, Iran
| |
Collapse
|
137
|
Mattinzoli D, Turolo S, Ikehata M, Vettoretti S, Montini G, Agostoni C, Conti C, Benedetti M, Messa P, Alfieri CM, Castellano G. MCP1 Inverts the Correlation between FGF23 and Omega 6/3 Ratio: Is It Also True in Renal Transplantation? J Clin Med 2023; 12:5928. [PMID: 37762869 PMCID: PMC10532002 DOI: 10.3390/jcm12185928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
During chronic kidney disease (CKD) progression, an increase in fibroblast growth factor (FGF23) is present. In stage 5, a positive correlation between FGF23 and omega-6 (n-6) polyunsaturated fatty acids (PUFAs) emerges. Hypothesizing that the rising positive correlation between monocyte chemoattractant protein 1 (MCP1) and n-6 in stage 4 could be the cause, we previously explored FGF23 and MCP1's roles in dyslipidemia and cardiovascular risk in CKD. In the present paper, we retraced the study evaluating 40 kidney transplant patients (KTx), a cohort where several factors might modify the previous relationships found. An ELISA and gas chromatography assessed the MCP1, FGF23, and PUFA levels. Despite the FGF23 increase (p < 0.0001), low MCP1 levels were found. A decrease in the n-6/n-3 ratio (p = 0.042 CKD stage 4 vs. 5) lowered by the increase in both n-3 αlinolenic (p = 0.012) and docosapentaenoic acid (p = 0.049) was observed. A negative correlation between FGF23 and the n-6/n-3 ratio in CKD stage 4 (r2 -0.3 p = 0.043) and none with MCP1 appeared. According to our findings, different mechanisms in the relationship between FGF23, PUFAs, and MCP1 in CKD and KTx patients might be present, which is possibly related to the immunosuppressive status of the last. Future research will further clarify our hypothesis.
Collapse
Affiliation(s)
- Deborah Mattinzoli
- Renal Research Laboratory, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Stefano Turolo
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Masami Ikehata
- Renal Research Laboratory, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Simone Vettoretti
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Giovanni Montini
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Carlo Agostoni
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
- Pediatric-Immunorheumatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Costanza Conti
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Post-Graduate School of Specialization in Nephrology, University of Milan, 20157 Milan, Italy
| | - Matteo Benedetti
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Post-Graduate School of Specialization in Nephrology, University of Milan, 20157 Milan, Italy
| | - Piergiorgio Messa
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Carlo Maria Alfieri
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Giuseppe Castellano
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| |
Collapse
|
138
|
Miura M, Miura Y, Iwazu Y, Mukai H, Sugiura T, Suzuki Y, Kato M, Kano M, Nagata D, Shiizaki K, Kurosu H, Kuro-O M. Removal of calciprotein particles from the blood using an adsorption column improves prognosis of hemodialysis miniature pigs. Sci Rep 2023; 13:15026. [PMID: 37700060 PMCID: PMC10497634 DOI: 10.1038/s41598-023-42273-0] [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: 02/06/2023] [Accepted: 09/07/2023] [Indexed: 09/14/2023] Open
Abstract
Hyperphosphatemia is a major risk for poor prognosis in patients with end-stage renal disease. However, the molecular mechanism behind this link remains elusive. We and others have demonstrated that serum phosphorus levels correlate positively with circulating levels of calciprotein particles (CPPs). CPPs are colloidal mineral-protein complexes containing insoluble calcium-phosphate precipitates and have been reported to induce calcification in cultured vascular smooth muscle cells and inflammatory responses in cultured macrophages. Hence, we hypothesize that CPPs may be responsible for disorders associated with hyperphosphatemia. Using hyperphosphatemic miniature pigs receiving hemodialysis, here we show that removal of CPPs from the blood with a newly developed CPP adsorption column improves survival and alleviates complications including coronary artery calcification, vascular endothelial dysfunction, metastatic pulmonary calcification, left ventricular hypertrophy, and chronic inflammation. The present study identifies CPPs as an effective therapeutic target and justifies clinical trials to determine whether the CPP adsorption column may be useful as a medical device for improving clinical outcomes of hemodialysis patients.
Collapse
Affiliation(s)
- Marina Miura
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Yutaka Miura
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Yoshitaka Iwazu
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
- Department of Clinical Laboratory Medicine, Jichi Medical University, Tochigi, Japan
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
| | - Hideyuki Mukai
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | | | | | | | | | - Daisuke Nagata
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Tochigi, Japan
| | - Kazuhiro Shiizaki
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Hiroshi Kurosu
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Makoto Kuro-O
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| |
Collapse
|
139
|
Li X, Lozovatsky L, Tommasini SM, Fretz J, Finberg KE. Bone marrow sinusoidal endothelial cells are a site of Fgf23 upregulation in a mouse model of iron deficiency anemia. Blood Adv 2023; 7:5156-5171. [PMID: 37417950 PMCID: PMC10480544 DOI: 10.1182/bloodadvances.2022009524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 06/16/2023] [Accepted: 07/05/2023] [Indexed: 07/08/2023] Open
Abstract
Iron deficiency is a potent stimulator of fibroblast growth factor 23 (FGF23), a hormonal regulator of phosphate and vitamin D metabolism, that is classically thought to be produced by bone-embedded osteocytes. Here, we show that iron-deficient transmembrane serine protease 6 knockout (Tmprss6-/-) mice exhibit elevated circulating FGF23 and Fgf23 messenger RNA (mRNA) upregulation in the bone marrow (BM) but not the cortical bone. To clarify sites of Fgf23 promoter activity in Tmprss6-/- mice, we introduced a heterozygous enhanced green fluorescent protein (eGFP) reporter allele at the endogenous Fgf23 locus. Heterozygous Fgf23 disruption did not alter the severity of systemic iron deficiency or anemia in the Tmprss6-/- mice. Tmprss6-/-Fgf23+/eGFP mice showed green fluorescence in the vascular regions of BM sections and showed a subset of BM endothelial cells that were GFPbright by flow cytometry. Mining of transcriptomic data sets from mice with normal iron balance revealed higher Fgf23 mRNA in BM sinusoidal endothelial cells (BM-SECs) than that in other BM endothelial cell populations. Anti-GFP immunohistochemistry of fixed BM sections from Tmprss6-/-Fgf23+/eGFP mice revealed GFP expression in BM-SECs, which was more intense than in nonanemic controls. In addition, in mice with intact Tmprss6 alleles, Fgf23-eGFP reporter expression increased in BM-SECs following large-volume phlebotomy and also following erythropoietin treatment both ex vivo and in vivo. Collectively, our results identified BM-SECs as a novel site for Fgf23 upregulation in both acute and chronic anemia. Given the elevated serum erythropoietin in both anemic models, our findings raise the possibility that erythropoietin may act directly on BM-SECs to promote FGF23 production during anemia.
Collapse
Affiliation(s)
- Xiuqi Li
- Department of Pathology, Yale School of Medicine, New Haven, CT
| | | | - Steven M. Tommasini
- Department of Orthopaedics & Rehabilitation, Yale School of Medicine, New Haven, CT
| | - Jackie Fretz
- Department of Orthopaedics & Rehabilitation, Yale School of Medicine, New Haven, CT
| | | |
Collapse
|
140
|
Xie H, Bastepe I, Zhou W, Ay B, Ceraj Z, Portales-Castillo IA, Liu ES, Burnett-Bowie SAM, Jüppner H, Rhee EP, Bastepe M, Simic P. 1,25-Dihydroxyvitamin D3 regulates furin-mediated FGF23 cleavage. JCI Insight 2023; 8:e168957. [PMID: 37681408 PMCID: PMC10544208 DOI: 10.1172/jci.insight.168957] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 07/17/2023] [Indexed: 09/09/2023] Open
Abstract
Intact fibroblast growth factor 23 (iFGF23) is a phosphaturic hormone that is cleaved by furin into N-terminal and C-terminal fragments. Several studies have implicated vitamin D in regulating furin in infections. Thus, we investigated the effect of 1,25-dihydroxyvitamin D3 [1,25(OH)2D] and the vitamin D receptor (VDR) on furin-mediated iFGF23 cleavage. Mice lacking VDR (Vdr-/-) had a 25-fold increase in iFGF23 cleavage, with increased furin levels and activity compared with wild-type (WT) littermates. Inhibition of furin activity blocked the increase in iFGF23 cleavage in Vdr-/- animals and in a Vdr-knockdown osteocyte OCY454 cell line. Chromatin immunoprecipitation revealed VDR binding to DNA upstream of the Furin gene, with more transcription in the absence of VDR. In WT mice, furin inhibition reduced iFGF23 cleavage, increased iFGF23, and reduced serum phosphate levels. Similarly, 1,25(OH)2D reduced furin activity, decreased iFGF23 cleavage, and increased total FGF23. In a post hoc analysis of a randomized clinical trial, we found that ergocalciferol treatment, which increased serum 1,25(OH)2D, significantly decreased serum furin activity and iFGF23 cleavage, compared with placebo. Thus, 1,25(OH)2D inhibits iFGF23 cleavage via VDR-mediated suppression of Furin expression, thereby providing a mechanism by which vitamin D can augment phosphaturic iFGF23 levels.
Collapse
Affiliation(s)
- Han Xie
- Nephrology Division and
- Endocrine Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Isinsu Bastepe
- Nephrology Division and
- Endocrine Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Wen Zhou
- Nephrology Division and
- Endocrine Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Birol Ay
- Endocrine Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Zara Ceraj
- Nephrology Division and
- Endocrine Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ignacio A. Portales-Castillo
- Nephrology Division and
- Endocrine Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Eva S. Liu
- Endocrine Division, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Harald Jüppner
- Endocrine Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Pediatric Nephrology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Eugene P. Rhee
- Nephrology Division and
- Endocrine Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Murat Bastepe
- Endocrine Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Petra Simic
- Nephrology Division and
- Endocrine Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
141
|
Zoccali C, Mallamaci F, Adamczak M, de Oliveira RB, Massy ZA, Sarafidis P, Agarwal R, Mark PB, Kotanko P, Ferro CJ, Wanner C, Burnier M, Vanholder R, Wiecek A. Cardiovascular complications in chronic kidney disease: a review from the European Renal and Cardiovascular Medicine Working Group of the European Renal Association. Cardiovasc Res 2023; 119:2017-2032. [PMID: 37249051 PMCID: PMC10478756 DOI: 10.1093/cvr/cvad083] [Citation(s) in RCA: 103] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/29/2022] [Accepted: 01/09/2023] [Indexed: 05/31/2023] Open
Abstract
Chronic kidney disease (CKD) is classified into five stages with kidney failure being the most severe stage (stage G5). CKD conveys a high risk for coronary artery disease, heart failure, arrhythmias, and sudden cardiac death. Cardiovascular complications are the most common causes of death in patients with kidney failure (stage G5) who are maintained on regular dialysis treatment. Because of the high death rate attributable to cardiovascular (CV) disease, most patients with progressive CKD die before reaching kidney failure. Classical risk factors implicated in CV disease are involved in the early stages of CKD. In intermediate and late stages, non-traditional risk factors, including iso-osmotic and non-osmotic sodium retention, volume expansion, anaemia, inflammation, malnutrition, sympathetic overactivity, mineral bone disorders, accumulation of a class of endogenous compounds called 'uremic toxins', and a variety of hormonal disorders are the main factors that accelerate the progression of CV disease in these patients. Arterial disease in CKD patients is characterized by an almost unique propensity to calcification and vascular stiffness. Left ventricular hypertrophy, a major risk factor for heart failure, occurs early in CKD and reaches a prevalence of 70-80% in patients with kidney failure. Recent clinical trials have shown the potential benefits of hypoxia-inducible factor prolyl hydroxylase inhibitors, especially as an oral agent in CKD patients. Likewise, the value of proactively administered intravenous iron for safely treating anaemia in dialysis patients has been shown. Sodium/glucose cotransporter-2 inhibitors are now fully emerged as a class of drugs that substantially reduces the risk for CV complications in patients who are already being treated with adequate doses of inhibitors of the renin-angiotensin system. Concerted efforts are being made by major scientific societies to advance basic and clinical research on CV disease in patients with CKD, a research area that remains insufficiently explored.
Collapse
Affiliation(s)
- Carmine Zoccali
- Renal Research Institute, 315 E, 62nd St., New York, NY 10065, USA
- Associazione Ipertensione Nefrologia e Trapianto Renale (IPNET) c/o Nefrologia e CNR, Grande Ospedale Metropolitano, Contrada Camporeale, 83031 Ariano Irpino Avellino, Italy
| | - Francesca Mallamaci
- Nephrology and Transplantation Unit, Grande Ospedale Metropolitano Reggio Cal and CNR-IFC, Via Giuseppe Melacrino 21, 89124 Reggio Calabria, Italy
| | - Marcin Adamczak
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia in Katowice, Francuska 20-24 St. 40-027 Katowice, Poland
| | - Rodrigo Bueno de Oliveira
- Department of Internal Medicine (Nephrology), School of Medical Sciences, University of Campinas (Unicamp), Campinas, Brazil
| | - Ziad A Massy
- Ambroise Paré University Hospital, APHP, Boulogne Billancourt/Paris, and INSERM U-1018, Centre de recherche en épidémiologie et santé des populations (CESP), Equipe 5, Paris-Saclay University (PSU) and University of Paris Ouest-Versailles-Saint-Quentin-en-Yvelines (UVSQ), FCRIN INI-CRCT, Villejuif, France
| | - Pantelis Sarafidis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Rajiv Agarwal
- Indiana University School of Medicine and Richard L. Roudebush VA Medical Center, 1481 W 10th St, Indianapolis, IN 46202, USA
| | - Patrick B Mark
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Peter Kotanko
- Renal Research Institute, LLC Icahn School of Medicine at Mount Sinai, 315 East 62nd Street, 3rd Floor, New York, NY 10065, USA
| | - Charles J Ferro
- Department of Renal Medicine, University Hospitals Birmingham, Birmingham, UK
| | - Christoph Wanner
- Division of Nephrology, University Hospital of Würzburg, Würzburg, Germany
| | - Michel Burnier
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Raymond Vanholder
- Nephrology Section, Department of Internal Medicine and Pediatrics, University Hospital, Ghent, Belgium
| | - Andrzej Wiecek
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia in Katowice, Francuska 20-24 St. 40-027 Katowice, Poland
| |
Collapse
|
142
|
Cozzolino M, Maffei Faccioli F, Cara A, Boni Brivio G, Rivela F, Ciceri P, Magagnoli L, Galassi A, Barbuto S, Speciale S, Minicucci C, Cianciolo G. Future treatment of vascular calcification in chronic kidney disease. Expert Opin Pharmacother 2023; 24:2041-2057. [PMID: 37776230 DOI: 10.1080/14656566.2023.2266381] [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: 07/07/2023] [Accepted: 09/29/2023] [Indexed: 10/02/2023]
Abstract
INTRODUCTION Cardiovascular disease (CVD) is one of the global leading causes of morbidity and mortality in chronic kidney disease (CKD) patients. Vascular calcification (VC) is a major cause of CVD in this population and is the consequence of complex interactions between inhibitor and promoter factors leading to pathological deposition of calcium and phosphate in soft tissues. Different pathological landscapes are associated with the development of VC, such as endothelial dysfunction, oxidative stress, chronic inflammation, loss of mineralization inhibitors, release of calcifying extracellular vesicles (cEVs) and circulating calcifying cells. AREAS COVERED In this review, we examined the literature and summarized the pathophysiology, biomarkers and focused on the treatments of VC. EXPERT OPINION Even though there is no consensus regarding specific treatment options, we provide the currently available treatment strategies that focus on phosphate balance, correction of vitamin D and vitamin K deficiencies, avoidance of both extremes of bone turnover, normalizing calcium levels and reduction of inflammatory response and the potential and promising therapeutic approaches liketargeting cellular mechanisms of calcification (e.g. SNF472, TNAP inhibitors).Creating novel scores to detect in advance VC and implementing targeted therapies is crucial to treat them and improve the future management of these patients.
Collapse
Affiliation(s)
- Mario Cozzolino
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Federico Maffei Faccioli
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Anila Cara
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Giulia Boni Brivio
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Francesca Rivela
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Paola Ciceri
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Lorenza Magagnoli
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Andrea Galassi
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Simona Barbuto
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Serena Speciale
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Carlo Minicucci
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Giuseppe Cianciolo
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
143
|
Sunderraj A, Wong M, Gutiérrez OM, Wolf M, Akhabue E, Carnethon MR, Yancy CW, Isakova T. Associations of FGF23 with 10-Year Change in eGFR and UACR and with Incident CKD in the CARDIA Cohort. KIDNEY360 2023; 4:e1236-e1244. [PMID: 37265357 PMCID: PMC10547221 DOI: 10.34067/kid.0000000000000172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 05/12/2023] [Indexed: 06/03/2023]
Abstract
Key Points FGF23 has a nonlinear positive association with incident CKD among healthy, middle-aged adults. The relationship between FGF23 and incident CKD was only significant among participants with cFGF23 levels in the highest quartile. FGF23 is not associated with 10-year change in eGFR or 10-year change in UACR among healthy, middle-aged adults. Background The relationship of fibroblast growth factor 23 (FGF23) with incident CKD has been examined in older but not younger populations. Methods Linear regression models were used to examine the associations of c-terminal FGF23 (cFGF23) and intact FGF23 (iFGF23) with 10-year change (1995–96 to 2005–06) in eGFR and urine albumin-to-creatinine ratio (UACR) in the Coronary Artery Risk Development in Young Adults cohort. Cox proportional hazard models were used to assess the association of cFGF23 with incident CKD, defined as eGFR <60 ml/min per 1.73 m2 or UACR ≥30 mg/g. Multivariable models were adjusted for age, sex, race, education, field center, physical activity, body mass index, diabetes, smoking, and systolic BP. Results Among 2511 participants, the mean age was 45±3.6 years; mean eGFR was 96.5±14.0 ml/min per 1.73 m2; and median UACR was 4.3 (interquartile range, 3.0–6.7) mg/g. Most (62.6%) participants were nonsmokers; the prevalence of diabetes was low (6.6%); and median values for 10-year changes in eGFR and UACR were modest (−5.50 ml/min per 1.73 m2 and 0.70 mg/g, respectively). No consistent associations between cFGF23 and 10-year change in eGFR and UACR were observed. During a median follow-up of 9.98 years, incident CKD developed in 258 participants. There was a nonlinear association of cFGF23 with incident CKD, and relative to the lowest quartile of cFGF23, a significant relationship was detected only among participants in the highest quartile (hazard ratio, 1.58; 95% confidence interval, 1.09 to 2.27). Similar findings were observed for iFGF23. Conclusion Among middle-aged adults in the Coronary Artery Risk Development in Young Adults cohort, median eGFR and UACR changes were modest and cFGF23 and iFGF23 were not consistently associated with 10-year change in eGFR or UACR. A nonlinear relationship was observed between cFGF23 and incident CKD, with individuals with highest cFGF23 levels being at risk of developing CKD.
Collapse
Affiliation(s)
- Ashwin Sunderraj
- Department of Medicine and Clinical and Translational Immunocardiology Program, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Mandy Wong
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Orlando M. Gutiérrez
- Department of Medicine and Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Myles Wolf
- Department of Medicine and Duke Clinical Research Institute, Duke University, Durham, North Carolina
| | - Ehimare Akhabue
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Mercedes R. Carnethon
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Clyde W. Yancy
- Department of Medicine and Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Tamara Isakova
- Department of Medicine and Center for Translational Metabolism and Health, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
144
|
Vergaro G, Del Franco A, Aimo A, Gentile F, Castiglione V, Saponaro F, Masotti S, Prontera C, Fusari N, Emdin M, Passino C. Intact fibroblast growth factor 23 in heart failure with reduced and mildly reduced ejection fraction. BMC Cardiovasc Disord 2023; 23:433. [PMID: 37658340 PMCID: PMC10474676 DOI: 10.1186/s12872-023-03441-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 08/09/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND Fibroblast growth factor-23 (FGF23) has been associated to left ventricular (LV) hypertrophy and heart failure (HF) severity. We aimed to investigate the clinical correlates and prognostic value of intact FGF23 (iFGF23) in HF patients. METHODS Patients with stable HF and left ventricular ejection fraction (LVEF) < 50% were prospectively enrolled, managed according to current recommendations and followed over time. iFGF23 was measured at baseline with a fully automated immuno-chemiluminescent assay. RESULTS We enrolled 150 patients (82% males; median age 65 years). First, second, and third iFGF23 tertiles were < 35.2 pg/mL, 35.2-50.9 pg/mL, and > 50.9 pg/mL. LVEF decreased from the first iFGF23 tertile to the third tertile (p = 0.014). N-terminal pro-B-type natriuretic peptide (NT-proBNP) increased from the first to the third tertile (p = 0.001), while peak oxygen consumption decreased (p < 0.001). Thirty-five patients (23%) experienced the primary endpoint (all-cause death or HF hospitalization at 5 years), and 26 (17%) the secondary endpoint (all-cause death at 5 years). On multivariable analysis, iFGF23 independently predicted the primary endpoint on top of age, gender and LVEF (HR 4.6 [95% CI 2.1-10.3], p < 0.001), age, gender and eGFR (HR 4.1 [95% CI 1.6-10.3], p = 0.003), as well as age, gender and NT-proBNP (HR 3.6 [95% CI 1.6-8.2], p = 0.002). iFGF23 even reclassified patient risk on top of all the 3 models, with NRI values of 0.65 (95% CI 0.30-1.01), 0.55 (95% CI 0.25-0.88), and 0.60 (95% CI 0.24-0.96), respectively (both p < 0.001). CONCLUSIONS Circulating iFGF23 is associated with disease severity and outcome in HF patients with reduced and mildly reduced ejection fraction.
Collapse
Affiliation(s)
- Giuseppe Vergaro
- Division of Cardiology and Cardiovascular Medicine, Fondazione Toscana Gabriele Monasterio, Via Moruzzi, 1, Pisa, 56127, Italy.
- Health Science Interdisciplinary Center , Scuola Superiore Sant'Anna, Pisa, Italy.
| | - Annamaria Del Franco
- Division of Cardiology and Cardiovascular Medicine, Fondazione Toscana Gabriele Monasterio, Via Moruzzi, 1, Pisa, 56127, Italy
- Health Science Interdisciplinary Center , Scuola Superiore Sant'Anna, Pisa, Italy
| | - Alberto Aimo
- Division of Cardiology and Cardiovascular Medicine, Fondazione Toscana Gabriele Monasterio, Via Moruzzi, 1, Pisa, 56127, Italy
- Health Science Interdisciplinary Center , Scuola Superiore Sant'Anna, Pisa, Italy
| | - Francesco Gentile
- Division of Cardiology and Cardiovascular Medicine, Fondazione Toscana Gabriele Monasterio, Via Moruzzi, 1, Pisa, 56127, Italy
| | - Vincenzo Castiglione
- Division of Cardiology and Cardiovascular Medicine, Fondazione Toscana Gabriele Monasterio, Via Moruzzi, 1, Pisa, 56127, Italy
| | | | - Silvia Masotti
- Division of Cardiology and Cardiovascular Medicine, Fondazione Toscana Gabriele Monasterio, Via Moruzzi, 1, Pisa, 56127, Italy
| | - Concetta Prontera
- Division of Cardiology and Cardiovascular Medicine, Fondazione Toscana Gabriele Monasterio, Via Moruzzi, 1, Pisa, 56127, Italy
| | - Niccolò Fusari
- Division of Cardiology and Cardiovascular Medicine, Fondazione Toscana Gabriele Monasterio, Via Moruzzi, 1, Pisa, 56127, Italy
| | - Michele Emdin
- Division of Cardiology and Cardiovascular Medicine, Fondazione Toscana Gabriele Monasterio, Via Moruzzi, 1, Pisa, 56127, Italy
- Health Science Interdisciplinary Center , Scuola Superiore Sant'Anna, Pisa, Italy
| | - Claudio Passino
- Division of Cardiology and Cardiovascular Medicine, Fondazione Toscana Gabriele Monasterio, Via Moruzzi, 1, Pisa, 56127, Italy
- Health Science Interdisciplinary Center , Scuola Superiore Sant'Anna, Pisa, Italy
| |
Collapse
|
145
|
Yamada S, Nakano T. Role of Chronic Kidney Disease (CKD)-Mineral and Bone Disorder (MBD) in the Pathogenesis of Cardiovascular Disease in CKD. J Atheroscler Thromb 2023; 30:835-850. [PMID: 37258233 PMCID: PMC10406631 DOI: 10.5551/jat.rv22006] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 06/02/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in patients with chronic kidney disease (CKD). Multiple factors account for the increased incidence of cardiovascular morbidity and mortality in patients with CKD. Traditional risk factors for atherosclerosis and arteriosclerosis, including age, hypertension, dyslipidemia, diabetes mellitus, and smoking, are also risk factors for CKD. Non-traditional risk factors specific for CKD are also involved in CVD pathogenesis in patients with CKD. Recently, CKD-mineral and bone disorder (CKD-MBD) has emerged as a key player in CVD pathogenesis in the context of CKD. CKD-MBD manifests as hypocalcemia and hyperphosphatemia in the later stages of CKD; however, it initially develops much earlier in disease course. The initial step in CKD-MBD involves decreased phosphate excretion in the urine, followed by increased circulating concentrations of fibroblast growth factor 23 (FGF23) and parathyroid hormone (PTH), which increase urinary phosphate excretion. Simultaneously, the serum calcitriol concentration decreases as a result of FGF23 elevation. Importantly, FGF23 and PTH cause left ventricular hypertrophy, arrhythmia, and cardiovascular calcification. More recently, calciprotein particles, which are nanoparticles composed of calcium, phosphate, and fetuin-A, among other components, have been reported to cause inflammation, cardiovascular calcification, and other clinically relevant outcomes. CKD-MBD has become one of the critical therapeutic targets for the prevention of cardiovascular events and is another link between cardiology and nephrology. In this review, we describe the role of CKD-MBD in the pathogenesis of cardiovascular disorders and present the current treatment strategies for CKD-MBD.
Collapse
Affiliation(s)
- Shunsuke Yamada
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiaki Nakano
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Centers for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
146
|
Kee YK, Jeon HJ, Oh J, Cho A, Lee YK, Yoon JW, Kim H, Yoo TH, Shin DH. Fibroblast growth factor-23 and cardiovascular disease among prevalent hemodialysis patients focusing on residual kidney function. Front Endocrinol (Lausanne) 2023; 14:1099975. [PMID: 37501787 PMCID: PMC10368752 DOI: 10.3389/fendo.2023.1099975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 04/28/2023] [Indexed: 07/29/2023] Open
Abstract
Background In patients undergoing incident hemodialysis, increased fibroblast growth factor-23 (FGF-23) levels are associated with the development of cardiovascular disease (CVD), but the influence of residual kidney function (RFK) on this association is unclear. This study aimed to investigate the association between FGF-23 levels, RKF, and CVD in patients undergoing prevalent hemodialysis. Methods This cross-sectional and longitudinal observational study included 296 patients undergoing maintenance hemodialysis for at least three months who were followed up for a median of 44 months. RKF was defined as 24-h urine output >200 mL, left ventricular (LV) diastolic dysfunction as E/E' >15 on echocardiographic parameters. CVD was defined as hospitalization or emergency room visits due to cardiovascular causes, such as angina, myocardial infarction, or congestive heart failure. Results The median intact FGF-23 (iFGF-23) level was 423.8 pg/mL (interquartile range, 171-1,443). Patients with an FGF-23 level > 423.8 pg/mL significantly had a lower proportion of RKF (39.2% vs. 60.1%, P < 0.001) and a higher proportion of LV diastolic dysfunction (54. 1% vs. 29.1%, P < 0.001) than those with an iFGF-23 level ≤ 423.8 pg/mL. The odds ratio (OR) for LV diastolic dysfunction was significantly higher in patients with RFK (OR per one-unit increase in the natural log-transformed iFGF-23 levels, 1.80; 95% confidence interval [CI]: 1.11-2.93) than in patients without RKF (OR per one-unit increase in the natural log-transformed iFGF-23 levels: 1.42; 95% CI: 1.01-1.99) in multivariate analysis (p < 0.001). During the follow-up period, 55 patients experienced CVD. The hazard ratio (HR) for CVD development was also significantly higher in patients with RKF (HR per one-unit increase in the natural log-transformed iFGF-23 levels, 2.64; 95% CI: 1.29-5.40) than those without RKF (HR per one-unit increase in the natural log-transformed iFGF-23 levels: 1.44; 95% CI: 1.04-1.99) in multivariate analysis (p = 0.05). Conclusions Increased iFGF-23 levels were associated with LV diastolic dysfunction and CVD development in patients undergoing prevalent hemodialysis; however, the loss of RKF attenuated the magnitude of these associations. Therefore, in these patients, RKF strongly influenced the detrimental role of iFGF-23 in the development of CVD.
Collapse
Affiliation(s)
- Youn Kyung Kee
- Department of Internal Medicine, Kangdong Sacred Heart Hospital, Hallym University, College of Medicine, Seoul, Republic of Korea
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hee Jung Jeon
- Department of Internal Medicine, Kangdong Sacred Heart Hospital, Hallym University, College of Medicine, Seoul, Republic of Korea
| | - Jieun Oh
- Department of Internal Medicine, Kangdong Sacred Heart Hospital, Hallym University, College of Medicine, Seoul, Republic of Korea
| | - Ajin Cho
- Department of Internal Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Young-Ki Lee
- Department of Internal Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Jong-Woo Yoon
- Department of Internal Medicine, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, Gangwon-do, Republic of Korea
| | - Hyunsuk Kim
- Department of Internal Medicine, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, Gangwon-do, Republic of Korea
| | - Tae-Hyun Yoo
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dong Ho Shin
- Department of Internal Medicine, Kangdong Sacred Heart Hospital, Hallym University, College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
147
|
Courbon G, Thomas JJ, Martinez-Calle M, Wang X, Spindler J, Von Drasek J, Hunt-Tobey B, Mehta R, Isakova T, Chang W, Creemers JWM, Ji P, Martin A, David V. Bone-derived C-terminal FGF23 cleaved peptides increase iron availability in acute inflammation. Blood 2023; 142:106-118. [PMID: 37053547 PMCID: PMC10356820 DOI: 10.1182/blood.2022018475] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/24/2023] [Accepted: 04/06/2023] [Indexed: 04/15/2023] Open
Abstract
Inflammation leads to functional iron deficiency by increasing the expression of the hepatic iron regulatory peptide hepcidin. Inflammation also stimulates fibroblast growth factor 23 (FGF23) production by increasing both Fgf23 transcription and FGF23 cleavage, which paradoxically leads to excess in C-terminal FGF23 peptides (Cter-FGF23), rather than intact FGF23 (iFGF23) hormone. We determined that the major source of Cter-FGF23 is osteocytes and investigated whether Cter-FGF23 peptides play a direct role in the regulation of hepcidin and iron metabolism in response to acute inflammation. Mice harboring an osteocyte-specific deletion of Fgf23 showed a ∼90% reduction in Cter-FGF23 levels during acute inflammation. Reduction in Cter-FGF23 led to a further decrease in circulating iron in inflamed mice owing to excessive hepcidin production. We observed similar results in mice showing impaired FGF23 cleavage owing to osteocyte-specific deletion of Furin. We next showed that Cter-FGF23 peptides bind members of the bone morphogenetic protein (BMP) family, BMP2 and BMP9, which are established inducers of hepcidin. Coadministration of Cter-FGF23 and BMP2 or BMP9 prevented the increase in Hamp messenger RNA and circulating hepcidin levels induced by BMP2/9, resulting in normal serum iron levels. Finally, injection of Cter-FGF23 in inflamed Fgf23KO mice and genetic overexpression of Cter-Fgf23 in wild type mice also resulted in lower hepcidin and higher circulating iron levels. In conclusion, during inflammation, bone is the major source of Cter-FGF23 secretion, and independently of iFGF23, Cter-FGF23 reduces BMP-induced hepcidin secretion in the liver.
Collapse
Affiliation(s)
- Guillaume Courbon
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Jane Joy Thomas
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Marta Martinez-Calle
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Xueyan Wang
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Jadeah Spindler
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - John Von Drasek
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Bridget Hunt-Tobey
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Rupal Mehta
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Tamara Isakova
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Wenhan Chang
- Endocrine Research Unit, San Francisco Veterans Affairs Medical Center, University of California San Francisco, San Francisco, CA
| | | | - Peng Ji
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Aline Martin
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Valentin David
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
148
|
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
|
149
|
Kitase Y, Prideaux M. Regulation of the Osteocyte Secretome with Aging and Disease. Calcif Tissue Int 2023; 113:48-67. [PMID: 37148298 DOI: 10.1007/s00223-023-01089-w] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/21/2023] [Indexed: 05/08/2023]
Abstract
As the most numerous and long-lived of all bone cells, osteocytes have essential functions in regulating skeletal health. Through the lacunar-canalicular system, secreted proteins from osteocytes can reach cells throughout the bone. Furthermore, the intimate connectivity between the lacunar-canalicular system and the bone vasculature allows for the transport of osteocyte-secreted factors into the circulation to reach the entire body. Local and endocrine osteocyte signaling regulates physiological processes such as bone remodeling, bone mechanoadaptation, and mineral homeostasis. However, these processes are disrupted by impaired osteocyte function induced by aging and disease. Dysfunctional osteocyte signaling is now associated with the pathogenesis of many disorders, including chronic kidney disease, cancer, diabetes mellitus, and periodontitis. In this review, we focus on the targeting of bone and extraskeletal tissues by the osteocyte secretome. In particular, we highlight the secreted osteocyte proteins, which are known to be dysregulated during aging and disease, and their roles during disease progression. We also discuss how therapeutic or genetic targeting of osteocyte-secreted proteins can improve both skeletal and systemic health.
Collapse
Affiliation(s)
- Yukiko Kitase
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Matthew Prideaux
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
150
|
Abstract
PURPOSE OF REVIEW Fibroblast growth factor (FGF) 23 is a bone-derived hormone that regulates phosphate and vitamin D metabolism by targeting the kidney. When highly elevated, such as in chronic kidney disease (CKD), FGF23 can also target the heart and induce pathologic remodeling. Here we discuss the mechanisms that underlie the physiologic and pathologic actions of FGF23, with focus on its FGF receptors (FGFR) and co-receptors. RECENT FINDINGS Klotho is a transmembrane protein that acts as an FGFR co-receptor for FGF23 on physiologic target cells. Klotho also exists as a circulating variant, and recent studies suggested that soluble klotho (sKL) can mediate FGF23 effects in cells that do not express klotho. Furthermore, it has been assumed that the actions of FGF23 do not require heparan sulfate (HS), a proteoglycan that acts as a co-receptor for other FGF isoforms. However, recent studies revealed that HS can be part of the FGF23:FGFR signaling complex and modulate FGF23-induced effects. SUMMARY sKL and HS have appeared as circulating FGFR co-receptors that modulate the actions of FGF23. Experimental studies suggest that sKL protects from and HS accelerates CKD-associated heart injury. However, the in vivo relevance of these findings is still speculative.
Collapse
Affiliation(s)
- S Madison Thomas
- Division of Nephrology and Section of Mineral Metabolism, Department of Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | |
Collapse
|