1
|
Franco LP, Derakhshandeh-Rishehri SM, Nöthlings U, Hartmann MF, Herder C, Kalhoff H, Wudy SA, Remer T. Young adults' circulating FGF23 and α-klotho and their relationship with habitual dietary acid load and phosphorus intake during growth. Sci Rep 2024; 14:27784. [PMID: 39537770 PMCID: PMC11561314 DOI: 10.1038/s41598-024-79636-0] [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: 08/29/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024] Open
Abstract
The bone-derived hormone FGF23, primarily secreted by osteocytes, is a major player in the regulation of phosphate homeostasis. It becomes upregulated by increased circulating phosphate concentration, e.g. due to elevations in phosphorus intake (P-In) or alterations in habitual dietary acid load. The present study aimed to investigate whether long-term endogenous acid production or a habitual high phosphorus intake during childhood and adolescence may be prospectively related with altered adult levels of FGF23 and the FGF23-related metabolite α-klotho. Urinary phosphate excretion (PO4-Ex), net acid excretion (NAE), and potential renal acid load (uPRAL) were analyzed in 24-h urine samples (n = 3369) collected from 343 healthy 3-17 years old participants of the DONALD Study (Dortmund, Germany) to assess, biomarker-based, P-In and habitual dietary acid load. Circulating FGF23, α-klotho, and further blood parameters were additionally examined in young adulthood. Individual means of standard-deviation-scores were calculated for 24-h urinary biomarker excretions and anthropometrics longitudinally determined between ages 3-17 years. Multivariable linear regression was used to analyze the prospective relations of pre-adulthood PO4-Ex, NAE, and uPRAL with the adulthood outcomes FGF23 and α-klotho. After adjusting for growth period-related covariates and adulthood confounders only for P-In during growth, i.e., PO4-Ex, but not for NAE and uPRAL, a significant positive association (p = 0.03) with FGF23 and an inverse trend (p = 0.10) with the FGF23-α-klotho ratio were observed. Neither PO4-Ex, nor NAE or uPRAL were associated with soluble α-klotho levels in adulthood. The prospective relationships of long-term assessed 24-h phosphaturia and habitual dietary acid load during growth with adult circulating, phosphate-adjusted FGF23 strongly suggest that children´s habitually higher P-In does unfavorably affect adult FGF23-α-klotho axis.
Collapse
Affiliation(s)
- Luciana Peixoto Franco
- DONALD Study Center, Nutritional Epidemiology, Institute of Nutrition and Food Science, University of Bonn, Heinstück 11, 44225, Dortmund, Germany
| | | | - Ute Nöthlings
- Institute of Nutrition and Food Sciences, Nutritional Epidemiology, University of Bonn, Bonn, Germany
| | - Michaela F Hartmann
- Laboratory for Translational Hormone Analytics, Peptide Hormone & Immunoassay Unit, Pediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, München-Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Hermann Kalhoff
- Research Department of Child Nutrition, St. Josef-Hospital, University Hospital of Pediatrics and Adolescent Medicine, Ruhr-University Bochum, Bochum, Germany
- Pediatric Clinic Dortmund, Dortmund, Germany
| | - Stefan A Wudy
- Laboratory for Translational Hormone Analytics, Peptide Hormone & Immunoassay Unit, Pediatric Endocrinology & Diabetology, Center of Child and Adolescent Medicine, Justus Liebig University, Giessen, Germany
| | - Thomas Remer
- DONALD Study Center, Nutritional Epidemiology, Institute of Nutrition and Food Science, University of Bonn, Heinstück 11, 44225, Dortmund, Germany.
| |
Collapse
|
2
|
Lim SW, Lee KI, Cui S, Fang X, Shin YJ, Lee H, Lee JY, Chung BH, Yang CW. Generation of green fluorescent protein reporter knock-in iPSC line at the 3'UTR region of the KLOTHO locus. Stem Cell Res 2024; 80:103499. [PMID: 39111000 DOI: 10.1016/j.scr.2024.103499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/11/2024] [Accepted: 07/13/2024] [Indexed: 08/25/2024] Open
Abstract
We generated a human induced pluripotent stem cell (hiPSC) line (CMCi014-A-78) expressing a GFP reporter in the 3'-UTR region of the KLOTHO locus using CRISPR/Cas9-mediated homologous recombination to screen for candidates regulating KLOTHO. The established cell line exhibits a normal karyotype, typical stem cell morphology, expression of pluripotency markers, and the ability to differentiate into the three germ layers. Consequently, this hiPSC line could serve as a valuable resource for screening KLOTHO regulators in hiPSC-derived target cells or organoids.
Collapse
Affiliation(s)
- Sun Woo Lim
- Transplantation Research Center, College of Medcine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kang In Lee
- R&D Center, ToolGen, Inc., Seoul, Republic of Korea
| | - Sheng Cui
- Transplantation Research Center, College of Medcine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Xianying Fang
- Transplantation Research Center, College of Medcine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoo Jin Shin
- Transplantation Research Center, College of Medcine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hanbi Lee
- Transplantation Research Center, College of Medcine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | | | - Byung Ha Chung
- Transplantation Research Center, College of Medcine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Chul Woo Yang
- Transplantation Research Center, College of Medcine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Lederer ED, Sobh MM, Brier ME, Gaweda AE. Application of artificial intelligence to chronic kidney disease mineral bone disorder. Clin Kidney J 2024; 17:sfae143. [PMID: 38899159 PMCID: PMC11184350 DOI: 10.1093/ckj/sfae143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Indexed: 06/21/2024] Open
Abstract
The global derangement of mineral metabolism that accompanies chronic kidney disease (CKD-MBD) is a major driver of the accelerated mortality for individuals with kidney disease. Advances in the delivery of dialysis, in the composition of phosphate binders, and in the therapies directed towards secondary hyperparathyroidism have failed to improve the cardiovascular event profile in this population. Many obstacles have prevented progress in this field including the incomplete understanding of pathophysiology, the lack of clinical targets for early stages of chronic kidney disease, and the remarkably wide diversity in clinical manifestations. We describe in this review a novel approach to CKD-MBD combining mathematical modelling of biologic processes with machine learning artificial intelligence techniques as a tool for the generation of new hypotheses and for the development of innovative therapeutic approaches to this syndrome. Clinicians need alternative targets of therapy, tools for risk profile assessment, and new therapies to address complications early in the course of disease and to personalize therapy to each individual. The complexity of CKD-MBD suggests that incorporating artificial intelligence techniques into the diagnostic, therapeutic, and research armamentarium could accelerate the achievement of these goals.
Collapse
Affiliation(s)
- Eleanor D Lederer
- VA North Texas Health Care Services, Dallas TX, USA
- Department of Medicine and Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Medicine, University of Louisville Health Sciences Center, Louisville, KY, USA
| | - Mahmoud M Sobh
- Nephrology and Internal Medicine, Mansoura University, Mansoura, Egypt
| | - Michael E Brier
- Department of Medicine, University of Louisville Health Sciences Center, Louisville, KY, USA
- Robley Rex VA Medical Center, Louisville, KY, USA
| | - Adam E Gaweda
- Department of Medicine, University of Louisville Health Sciences Center, Louisville, KY, USA
- Robley Rex VA Medical Center, Louisville, KY, USA
| |
Collapse
|
4
|
Zhang L, Shi WY, Zhang LL, Sha Y, Xu JY, Shen LC, Li YH, Yuan LX, Qin LQ. Effects of selenium-cadmium co-enriched Cardamine hupingshanensis on bone damage in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116101. [PMID: 38359653 DOI: 10.1016/j.ecoenv.2024.116101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024]
Abstract
Selenium (Se) and cadmium (Cd) usually co-existed in soils, especially in areas with Se-rich soils in China. The potential health consequences for the local populations consuming foods rich in Se and Cd are unknown. Cardamine hupingshanensis (HUP) is Se and Cd hyperaccumulator plant that could be an ideal natural product to assess the protective effects of endogenous Se against endogenous Cd-caused bone damage. Male C57BL/6 mice were fed 5.22 mg/kg cadmium chloride (CdCl2) (Cd 3.2 mg/kg body weight (BW)), or HUP solutions containing Cd 3.2 mg/kg BW and Se 0.15, 0.29 or 0.50 mg/kg BW (corresponding to the HUP0, HUP1 and HUP2 groups) interventions. Se-enriched HUP1 and HUP2 significantly decreased Cd-induced femur microstructure damage and regulated serum bone osteoclastic marker levels and osteogenesis-related genes. In addition, endogenous Se significantly decreased kidney fibroblast growth factor 23 (FGF23) protein expression and serum parathyroid hormone (PTH) levels, and raised serum calcitriol (1,25(OH)2D3). Furthermore, Se also regulated gut microbiota involved in skeletal metabolism disorder. In conclusion, endogenous Se, especially with higher doses (the HUP2 group), positively affects bone formation and resorption by mitigating the damaging effects of endogenous Cd via the modulation of renal FGF23 expression, circulating 1,25(OH)2D3 and PTH and gut microbiota composition.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Wen-Yao Shi
- Department of Health and Environmental Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Li-Li Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Yu Sha
- Department of Medical Technology, Suzhou Vocational Health College, Suzhou, China
| | - Jia-Ying Xu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Le-Cheng Shen
- Jiangxi Center of Quality Supervision and Inspection for Selenium-enriched Products/Ganzhou General Inspection and Testing Institute, Ganzhou 341000, China
| | - Yun-Hong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China.
| | - Lin-Xi Yuan
- Department of Health and Environmental Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China.
| | - Li-Qiang Qin
- Department of Nutrition and Food Hygiene, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China.
| |
Collapse
|
5
|
Verlinden L, Li S, Veldurthy V, Carmeliet G, Christakos S. Relationship of the bone phenotype of the Klotho mutant mouse model of accelerated aging to changes in skeletal architecture that occur with chronological aging. Front Endocrinol (Lausanne) 2024; 15:1310466. [PMID: 38352710 PMCID: PMC10861770 DOI: 10.3389/fendo.2024.1310466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/09/2024] [Indexed: 02/16/2024] Open
Abstract
Introduction Due to the relatively long life span of rodent models, in order to expediate the identification of novel therapeutics of age related diseases, mouse models of accelerated aging have been developed. In this study we examined skeletal changes in the male and female Klotho mutant (kl/kl) mice and in male and female chronically aged mice to determine whether the accelerated aging bone phenotype of the kl/kl mouse reflects changes in skeletal architecture that occur with chronological aging. Methods 2, 6 and 20-23 month old C57BL/6 mice were obtained from the National Institute of Aging aged rodent colony and wildtype and kl/kl mice were generated as previously described by M. Kuro-o. Microcomputed tomography analysis was performed ex vivo to examine trabecular and cortical parameters from the proximal metaphyseal and mid-diaphyseal areas, respectively. Serum calcium and phosphate were analyzed using a colorimetric assay. The expression of duodenal Trpv6, which codes for TRPV6, a vitamin D regulated epithelial calcium channel whose expression reflects intestinal calcium absorptive efficiency, was analyzed by quantitative real-time PCR. Results and discussion Trabecular bone volume (BV/TV) and trabecular number decreased continuously with age in males and females. In contrast to aging mice, an increase in trabecular bone volume and trabecular number was observed in both male and female kl/kl mice. Cortical thickness decreased with advancing age and also decreased in male and female kl/kl mice. Serum calcium and phosphate levels were significantly increased in kl/kl mice but did not change with age. Aging resulted in a decline in Trpv6 expression. In the kl/kl mice duodenal Trpv6 was significantly increased. Our findings reflect differences in bone architecture as well as differences in calcium and phosphate homeostasis and expression of Trpv6 between the kl/kl mutant mouse model of accelerated aging and chronological aging. Although the Klotho deficient mouse has provided a new understanding of the regulation of mineral homeostasis and bone metabolism, our findings suggest that changes in bone architecture in the kl/kl mouse reflect in part systemic disturbances that differ from pathophysiological changes that occur with age including dysregulation of calcium homeostasis that contributes to age related bone loss.
Collapse
Affiliation(s)
- Lieve Verlinden
- Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Shanshan Li
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, NJ, United States
| | - Vaishali Veldurthy
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, NJ, United States
| | - Geert Carmeliet
- Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Sylvia Christakos
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
6
|
Levy-Shraga Y, Levi S, Regev R, Gal S, Brener A, Lebenthal Y, Gillis D, Strich D, Zung A, Cleper R, Borovitz Y, Bello R, Tenenbaum A, Zadik Z, Davidovits M, Zeitlin L, Tiosano D. Linear growth of children with X-linked hypophosphatemia treated with burosumab: a real-life observational study. Eur J Pediatr 2023; 182:5191-5202. [PMID: 37707589 DOI: 10.1007/s00431-023-05190-y] [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: 06/16/2023] [Revised: 08/30/2023] [Accepted: 09/02/2023] [Indexed: 09/15/2023]
Abstract
To assess the long-term efficacy of burosumab for pediatric patients with X-linked hypophosphatemia, focusing on linear growth. This multi-center retrospective study included 35 pediatric patients who began treatment with burosumab between January 2018 and January 2021. We collected clinical data, anthropometric measurements, laboratory results, and Rickets Severity Score (RSS), from 2 years prior to treatment initiation and up to 4 years after. Burosumab was initiated at a mean age of 7.5 ± 4.4 years (range 0.6-15.9), with a mean initial dose of 0.8 ± 0.3 mg/kg, which was subsequently increased to 1.1 ± 0.4 mg/kg. The patients were followed for 2.9 ± 1.4 years (range 1-4) after initiating burosumab. Serum phosphorus levels increased from 2.7 ± 0.8 mg/dl at burosumab initiation to 3.4 ± 0.6 mg/dl after 3 months and remained stable (p < 0.001). Total reabsorption of phosphorus increased from 82.0 ± 6.8 to 90.1 ± 5.3% after 12 months of treatment (p = 0.041). The RSS improved from 1.7 ± 1.0 at burosumab initiation to 0.5 ± 0.6 and 0.3 ± 0.6 after 12 and 24 months, respectively (p < 0.001). Both height z-score and weight z-score improved from burosumab initiation to the end of the study: from - 2.07 ± 1.05 to - 1.72 ± 1.04 (p < 0.001) and from - 0.51 ± 1.12 to - 0.11 ± 1.29 (p < 0.001), respectively. Eight children received growth hormone combined with burosumab treatment. Height z-score improved among those who received growth hormone (from - 2.33 ± 1.12 to - 1.94 ± 1.24, p = 0.042) and among those who did not (from - 2.01 ± 1.01 to - 1.66 ± 1.01, p = 0.001). CONCLUSION Burosumab treatment in a real-life setting improved phosphate homeostasis and rickets severity and enhanced linear growth. WHAT IS KNOWN • Compared to conventional therapy, burosumab treatment has been shown to increase serum phosphate levels and reduce the severity of rickets. • The effect of burosumab on growth is still being study. WHAT IS NEW • Height z-score improved between the start of burosumab treatment and the end of the study (-2.07 ± 1.05 vs. -1.72 ± 1.04, p < 0.001). • Eight children received burosumab combined with growth hormone treatment without side effects during the concomitant treatments.
Collapse
Affiliation(s)
- Yael Levy-Shraga
- Pediatric Endocrinology Unit, The Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, Tel Hashomer, 52621, Israel.
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Shelly Levi
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Pediatric Nephrology, Schneider Children's Medical Center, Petah Tikva, Israel
| | - Ravit Regev
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Endocrinology and Diabetes Unit, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Shoshana Gal
- Division of Pediatric Endocrinology, Ruth Rappaport Children's Hospital, Rambam Medical Center, Haifa, Israel
- Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Avivit Brener
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Endocrinology and Diabetes Unit, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Yael Lebenthal
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Endocrinology and Diabetes Unit, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - David Gillis
- Pediatric Endocrinology Unit, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Hadassah Medical School, Jerusalem, Israel
| | - David Strich
- Faculty of Medicine, Hebrew University of Jerusalem, Hadassah Medical School, Jerusalem, Israel
- Department of Pediatrics, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Amnon Zung
- Faculty of Medicine, Hebrew University of Jerusalem, Hadassah Medical School, Jerusalem, Israel
- Pediatrics Department, Kaplan Medical Center, Rehovot, Israel
| | - Roxana Cleper
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Nephrology Unit, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Yael Borovitz
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Pediatric Nephrology, Schneider Children's Medical Center, Petah Tikva, Israel
| | - Rachel Bello
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Shafer Institute for Endocrinology and Diabetes, Schneider Children's Medical Center, Petah Tikva, Israel
| | - Ariel Tenenbaum
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Shafer Institute for Endocrinology and Diabetes, Schneider Children's Medical Center, Petah Tikva, Israel
| | - Zvi Zadik
- Faculty of Medicine, Hebrew University of Jerusalem, Hadassah Medical School, Jerusalem, Israel
- Pediatrics Department, Kaplan Medical Center, Rehovot, Israel
| | - Miriam Davidovits
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Pediatric Nephrology, Schneider Children's Medical Center, Petah Tikva, Israel
| | - Leonid Zeitlin
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Orthopedic Department, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Dov Tiosano
- Division of Pediatric Endocrinology, Ruth Rappaport Children's Hospital, Rambam Medical Center, Haifa, Israel
- Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| |
Collapse
|
7
|
Sun F, Liang P, Wang B, Liu W. The fibroblast growth factor-Klotho axis at molecular level. Open Life Sci 2023; 18:20220655. [PMID: 37941788 PMCID: PMC10628560 DOI: 10.1515/biol-2022-0655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 06/03/2023] [Accepted: 06/10/2023] [Indexed: 11/10/2023] Open
Abstract
Klotho is a recently discovered protein that has positive effects on all systems of the body, for example, regulating calcium and phosphorus metabolism, protecting nerves, delaying aging and so on. Fibroblast growth factors (FGFs) are a group of polypeptides that function throughout the body by binding with cell surface FGF receptors (FGFRs). Endocrine FGFs require Klotho as a co-receptor for FGFRs. There is increasing evidence that Klotho participates in calcium and phosphorus regulation and metabolic regulation via the FGF-Klotho axis. Moreover, soluble Klotho can function as a separate hormone to regulate homeostasis on various ion channels and carrier channels on the cell surface. This review mainly explains the molecular basis of the membrane signaling mechanism of Klotho.
Collapse
Affiliation(s)
- Fuqiang Sun
- School of Anesthesiology, Weifang Medical University, Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, Weifang261053, Shandong, China
| | - Panpan Liang
- School of Basic Medical Sciences, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Bo Wang
- School of Anesthesiology, Weifang Medical University, Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, Weifang261053, Shandong, China
| | - Wenbo Liu
- Central Laboratory of the First Affiliated Hospital, Weifang Medical University, Weifang261000, Shandong, China
| |
Collapse
|
8
|
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
|
9
|
Guan G, Cai J, Zheng S, Xiang Y, Xia S, Zhang Y, Shi J, Wang J. Association between serum manganese and serum klotho in a 40–80-year-old American population from NHANES 2011–2016. FRONTIERS IN AGING 2023; 4:1120823. [PMID: 36970731 PMCID: PMC10031017 DOI: 10.3389/fragi.2023.1120823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 02/23/2023] [Indexed: 03/11/2023]
Abstract
Objectives: Manganese is one of the essential trace elements that are required by the human body. Klotho protein is a classic anti-aging marker. The association between the levels of serum manganese and serum klotho in individuals between the ages of 40–80 in the United States remains unclear.Methods: Data for this cross-sectional study was obtained from the National Health and Nutrition Examination Survey (NHANES 2011–2016) in the United States. We performed multiple linear regression analyses to investigate the association between the levels of serum manganese and serum klotho. Furthermore, we performed a fitted smoothing curve according to a restricted cubic spline (RCS). Stratification and subgroup analyses were performed for further verification of the results.Results: Weighted multivariate linear regression analysis showed that serum manganese levels were independently and positively associated with serum klotho levels (β = 6.30, 95% confidence interval: 3.30–9.40). Kruskal–Wallis test showed that participants with higher manganese quartiles had higher serum klotho levels (Q1: 808.54 ± 256.39 pg/mL; Q2: 854.56 ± 266.13 pg/mL; Q3: 865.13 ± 300.60 pg/mL; and Q4: 871.72 ± 338.85 pg/mL, p < 0.001). The RCS curve indicated that the association between the levels of serum manganese and serum klotho was non-linear. Furthermore, a significantly positive association was found between serum manganese and serum klotho levels in the majority of subgroups.Conclusion: A non-linear and positive association was found between the levels of serum manganese and serum klotho in individuals aged 40–80 in the United States according to the NHANES (2011–2016).
Collapse
Affiliation(s)
- Guoyu Guan
- Department of Geriatrics, Huadong Hospital, Shanghai Medical College Fudan University, Shanghai, China
| | - Jiasheng Cai
- Department of Cardiology, Huadong Hospital, Shanghai Medical College Fudan University, Shanghai, China
| | - Songbai Zheng
- Department of Geriatrics, Huadong Hospital, Shanghai Medical College Fudan University, Shanghai, China
- *Correspondence: Songbai Zheng,
| | - Yanzhen Xiang
- Department of General Practice, Huadong Hospital, Shanghai Medical College Fudan University, Shanghai, China
| | - Shijin Xia
- Department of Geriatrics, Huadong Hospital, Shanghai Medical College Fudan University, Shanghai, China
| | - Yixuan Zhang
- Department of Geriatrics, Huadong Hospital, Shanghai Medical College Fudan University, Shanghai, China
| | - Jiaqiang Shi
- Department of General Practice, Huadong Hospital, Shanghai Medical College Fudan University, Shanghai, China
| | - Jun Wang
- Department of General Practice, Huadong Hospital, Shanghai Medical College Fudan University, Shanghai, China
| |
Collapse
|
10
|
Zebrafish Models to Study Ectopic Calcification and Calcium-Associated Pathologies. Int J Mol Sci 2023; 24:ijms24043366. [PMID: 36834795 PMCID: PMC9967340 DOI: 10.3390/ijms24043366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/20/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
Ectopic calcification refers to the pathological accumulation of calcium ions in soft tissues and is often the result of a dysregulated action or disrupted function of proteins involved in extracellular matrix mineralization. While the mouse has traditionally been the go-to model organism for the study of pathologies associated with abnormal calcium deposition, many mouse mutants often have exacerbated phenotypes and die prematurely, limiting the understanding of the disease and the development of effective therapies. Since the mechanisms underlying ectopic calcification share some analogy with those of bone formation, the zebrafish (Danio rerio)-a well-established model for studying osteogenesis and mineralogenesis-has recently gained momentum as a model to study ectopic calcification disorders. In this review, we outline the mechanisms of ectopic mineralization in zebrafish, provide insights into zebrafish mutants that share phenotypic similarities with human pathological mineralization disorders, list the compounds capable of rescuing mutant phenotypes, and describe current methods to induce and characterize ectopic calcification in zebrafish.
Collapse
|
11
|
Detopoulou P, Papadopoulou SK, Voulgaridou G, Dedes V, Tsoumana D, Gioxari A, Gerostergios G, Detopoulou M, Panoutsopoulos GI. Ketogenic Diet and Vitamin D Metabolism: A Review of Evidence. Metabolites 2022; 12:metabo12121288. [PMID: 36557329 PMCID: PMC9788458 DOI: 10.3390/metabo12121288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/02/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
The ketogenic diet (KD), which is low in carbohydrates and high to normal in fat and protein, has been traditionally used in epilepsy for the last 100 years. More recently, its application in obesity has been introduced. The present review aimed to investigate the effects of the KD on vitamin D. In total, five studies were done in healthy adults, one in subjects with type 2 diabetes, and seven in subjects with epilepsy that assessed the levels of vitamin D pre- and post-intervention. In the majority of studies, increases in circulating vitamin D were reported. The relationship of the KD with vitamin D was explained through several mechanisms: ketone bodies, macronutrient intake, the status levels of other fat-soluble vitamins, weight loss, changes in the hormonal milieu, and effects on gut microbiota. Moreover, potential nutrient-gene-related interactions were discussed. There is still a need to conduct multiple arm studies to compare the effects of the KD versus other diets and better decipher the particular effects of the KD on vitamin D levels and metabolism. Moreover, differentiations of the diet's effects according to sex and genetic makeup should be investigated to prescribe KDs on a more personalized basis.
Collapse
Affiliation(s)
- Paraskevi Detopoulou
- Department of Clinical Nutrition, General Hospital Korgialenio Benakio, 11526 Athens, Greece
- Correspondence:
| | - Sousana K. Papadopoulou
- Department of Nutritional Science and Dietetics, International Hellenic University, 57400 Thessaloniki, Greece
| | - Gavriela Voulgaridou
- Department of Nutritional Science and Dietetics, International Hellenic University, 57400 Thessaloniki, Greece
| | - Vasileios Dedes
- Department of Nutritional Science and Dietetics, Faculty of Health Sciences, University of Peloponnese, 24100 Kalamata, Greece
| | - Despoina Tsoumana
- Department of Nutritional Science and Dietetics, International Hellenic University, 57400 Thessaloniki, Greece
| | - Aristea Gioxari
- Department of Nutritional Science and Dietetics, Faculty of Health Sciences, University of Peloponnese, 24100 Kalamata, Greece
| | - George Gerostergios
- Department of Clinical Nutrition, General Hospital Korgialenio Benakio, 11526 Athens, Greece
| | - Maria Detopoulou
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17676 Athens, Greece
| | - George I. Panoutsopoulos
- Department of Nutritional Science and Dietetics, Faculty of Health Sciences, University of Peloponnese, 24100 Kalamata, Greece
| |
Collapse
|
12
|
Bover J, Trinidad P, Jara A, Soler-Majoral J, Martín-Malo A, Torres A, Frazão J, Ureña P, Dusso A, Arana C, Graterol F, Romero-González G, Troya M, Samaniego D, D'Marco L, Valdivielso JM, Fernández E, Arenas MD, Torregrosa V, Navarro-González JF, Lloret MJ, Ballarín JA, Bosch RJ, Górriz JL, de Francisco A, Gutiérrez O, Ara J, Felsenfeld A, Canalejo A, Almadén Y. Silver jubilee: 25 years of the first demonstration of the direct effect of phosphate on the parathyroid cell. Nefrologia 2022; 42:645-655. [PMID: 36925324 DOI: 10.1016/j.nefroe.2023.02.008] [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: 12/27/2021] [Accepted: 12/30/2021] [Indexed: 06/18/2023] Open
Abstract
Although phosphorus is an essential element for life, it is not found in nature in its native state but rather combined in the form of inorganic phosphates (PO43-), with tightly regulated plasma levels that are associated with deleterious effects and mortality when these are out of bounds. The growing interest in the accumulation of PO43- in human pathophysiology originated in its attributed role in the pathogenesis of secondary hyperparathyroidism (SHPT) in chronic kidney disease. In this article, we review the mechanisms by which this effect was justified and we commemorate the important contribution of a Spanish group led by Dr. M. Rodríguez, just 25 years ago, when they first demonstrated the direct effect of PO43- on the regulation of the synthesis and secretion of parathyroid hormone by maintaining the structural integrity of the parathyroid glands in their original experimental model. In addition to demonstrating the importance of arachidonic acid (AA) and the phospholipase A2-AA pathway as a mediator of parathyroid gland response, these findings were predecessors of the recent description of the important role of PO43- on the activity of the calcium sensor-receptor, and also fueled various lines of research on the importance of PO43- overload not only for the pathophysiology of SHPT but also in its systemic pathogenic role.
Collapse
Affiliation(s)
- Jordi Bover
- Servicio de Nefrología, Hospital Universitario Germans Trias i Pujol, RICORS, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, Badalona (Barcelona), Spain.
| | - Pedro Trinidad
- Departamento de Nefrología, HECMN siglo XXI, IMSS, Ciudad de México, México
| | - Aquiles Jara
- Departamento de Nefrología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jordi Soler-Majoral
- Servicio de Nefrología, Hospital Universitario Germans Trias i Pujol, RICORS, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, Badalona (Barcelona), Spain
| | - Alejandro Martín-Malo
- Unidad de Gestión Clinica Nefrología, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Universidad de Córdoba, España. Red Nacional de Investigación en Nefrología (REDinREN), Instituto de Salud Carlos III, Spain
| | - Armando Torres
- Servicio de Nefrología, Hospital Universitario de Canarias, Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Tenerife, Spain
| | - João Frazão
- Department of Nephrology, Centro Hospitalar Universitário São João, Institute for Innovation and Health Research (I3S), Institute of Biomedical Engineering (INEB), Nephrology and Infectious Diseases Research Group, University of Porto, Porto, Portugal
| | - Pablo Ureña
- AURA Nord Saint Ouen Dialysis Service. Saint Ouen, France and Service d'Explorations Fonctionnelles Rénales, Hôpital Necker, Université Paris V, René Descartes, Paris, France
| | - Adriana Dusso
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St Louis, MO, USA
| | - Carolt Arana
- Departamento de Nefrología y Trasplante Renal, Hospital Clínic, Barcelona, España
| | - Fredzzia Graterol
- Servicio de Nefrología, Hospital Universitario Germans Trias i Pujol, RICORS, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, Badalona (Barcelona), Spain
| | - Gregorio Romero-González
- Servicio de Nefrología, Hospital Universitario Germans Trias i Pujol, RICORS, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, Badalona (Barcelona), Spain
| | - Maribel Troya
- Servicio de Nefrología, Hospital Universitario Germans Trias i Pujol, RICORS, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, Badalona (Barcelona), Spain
| | - Diana Samaniego
- Servicio de Nefrología, Hospital Universitario Germans Trias i Pujol, RICORS, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, Badalona (Barcelona), Spain
| | - Luis D'Marco
- CEU Cardenal Herrera University, Valencia, Spain
| | - José Manuel Valdivielso
- Vascular and Renal Translational Research Group, Biomedical Research Institute, IRBLLEIDA, Lleida, España. Red Nacional de Investigación en Nefrología (REDinREN, RETIC), Instituto de Salud Carlos III, Spain
| | - Elvira Fernández
- Vascular and Renal Translational Research Group, Biomedical Research Institute, IRBLLEIDA, Lleida, España. Red Nacional de Investigación en Nefrología (REDinREN, RETIC), Instituto de Salud Carlos III, Spain; Grupo de Investigación Traslacional Vascular y Renal, Fundación Renal Jaume Arnó, Lleida, Spain
| | | | - Vicente Torregrosa
- Departamento de Nefrología y Trasplante Renal, Hospital Clínic, Barcelona, España
| | - Juan F Navarro-González
- Unidad de Investigación y Servicio de Nefrología, Hospital Universitario Nuestra Señora de la Candelaria, Instituto Universitario de Tecnologías Biomédicas, Universidad de la Laguna, Santa Cruz de Tenerife, España. Red Nacional de Investigación en Nefrología (REDinREN, RICORS), Instituto de Salud Carlos III, Spain
| | - María Jesús Lloret
- Servicio de Nefrología, Fundació Puigvert, IIB Sant Pau, Barcelona, Spain
| | - J A Ballarín
- Servicio de Nefrología, Fundació Puigvert, IIB Sant Pau, Barcelona, Spain
| | - Ricardo J Bosch
- Unidad de Fisiología, Departamento de Biología de Sistemas, Facultad de Medicina, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| | - José L Górriz
- Servicio de Nefrología, Hospital Clínico Universitario, INCLIVA, Universidad de Valencia, Valencia, Spain
| | | | - Orlando Gutiérrez
- Division of Nephrology, Department of Medicine, Universidad de Alabama en Birmingham, Birmingham USA
| | - Jordi Ara
- Servicio de Nefrología, Hospital Universitario Germans Trias i Pujol, RICORS, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, Badalona (Barcelona), Spain
| | - Arnold Felsenfeld
- Department of Medicine, Veterans Affairs Greater Los Angeles Healthcare System and David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Antonio Canalejo
- Departamento de Ciencias Integradas/Centro de Investigación RENSMA, Facultad de Ciencias Experimentales, Universidad de Huelva. Huelva, Spain
| | - Yolanda Almadén
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, Universidad de Córdoba, Córdoba, Spain
| |
Collapse
|
13
|
Rubio-Aliaga I, Krapf R. Phosphate intake, hyperphosphatemia, and kidney function. Pflugers Arch 2022; 474:935-947. [PMID: 35511366 PMCID: PMC9338892 DOI: 10.1007/s00424-022-02691-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/25/2022]
Abstract
Phosphate is essential in living organisms and its blood levels are regulated by a complex network involving the kidneys, intestine, parathyroid glands, and the skeleton. The crosstalk between these organs is executed primarily by three hormones, calcitriol, parathyroid hormone, and fibroblast growth factor 23. Largely due to a higher intake of ultraprocessed foods, dietary phosphate intake has increased in the last decades. The average intake is now about twice the recommended dietary allowance. Studies investigating the side effect of chronic high dietary phosphate intake suffer from incomplete dietary phosphate assessment and, therefore, often make data interpretation difficult. Renal excretion is quickly adapted to acute and chronic phosphate intake. However, at the high ends of dietary intake, renal adaptation, even in pre-existing normal kidney function, apparently is not perfect. Experimental intervention studies suggest that chronic excess of dietary phosphate can result in sustained higher blood phosphate leading to hyperphosphatemia. Evidence exists that the price of the homeostatic response (phosphaturia in response to phosphate loading/hyperphosphatemia) is an increased risk for declining kidney function, partly due by intraluminal/tubular calcium phosphate particles that provoke renal inflammation. High dietary phosphate intake and hyperphosphatemia are progression factors for declining kidney function and are associated with higher cardiovascular disease and mortality risk. This is best established for pre-existing chronic kidney disease, but epidemiological and experimental data strongly suggest that this holds true for subjects with normal renal function as well. Here, we review the latest advances in phosphate intake and kidney function decline.
Collapse
Affiliation(s)
- Isabel Rubio-Aliaga
- Institute of Physiology, National Center of Competence in Research NCCR Kidney.CH, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
| | - Reto Krapf
- Synlab Suisse, 6002, Lucerne, Switzerland
- Department of Medicine, University of Basel, 4056, Basel, Switzerland
| |
Collapse
|
14
|
Yuan W, Song C. Crosstalk between bone and other organs. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:331-348. [PMID: 37724328 PMCID: PMC10471111 DOI: 10.1515/mr-2022-0018] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/06/2022] [Indexed: 09/20/2023]
Abstract
Bone has long been considered as a silent organ that provides a reservoir of calcium and phosphorus, traditionally. Recently, further study of bone has revealed additional functions as an endocrine organ connecting systemic organs of the whole body. Communication between bone and other organs participates in most physiological and pathological events and is responsible for the maintenance of homeostasis. Here, we present an overview of the crosstalk between bone and other organs. Furthermore, we describe the factors mediating the crosstalk and review the mechanisms in the development of potential associated diseases. These connections shed new light on the pathogenesis of systemic diseases and provide novel potential targets for the treatment of systemic diseases.
Collapse
Affiliation(s)
- Wanqiong Yuan
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| | - Chunli Song
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| |
Collapse
|
15
|
Hashimoto Y, Kato S, Kuro-O M, Miura Y, Itano Y, Ando M, Kuwatsuka Y, Maruyama S. Impact of etelcalcetide on fibroblast growth factor-23 and calciprotein particles in patients with secondary hyperparathyroidism undergoing hemodialysis. Nephrology (Carlton) 2022; 27:763-770. [PMID: 35749253 DOI: 10.1111/nep.14081] [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: 01/27/2022] [Revised: 05/02/2022] [Accepted: 06/21/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Recently, we demonstrated the efficacy of etelcalcetide in the control of secondary hyperparathyroidism (SHPT). This post hoc analysis aimed to evaluate changes in fibroblast growth factor-23 (FGF23) and calciprotein particles (CPPs) after treatment with calcimimetics. METHODS The DUET trial was a 12-week multicenter, open-label, parallel-group, randomized (1:1:1) study with patients treated with etelcalcetide plus active vitamin D (E+D group; n = 41), etelcalcetide plus oral calcium (E+Ca group; n = 41), or control (C group; n = 42) under maintenance hemodialysis. Serum levels of FGF23 and CPPs were measured at baseline, and 6 and 12 weeks after the start. RESULTS In the linear mixed model, serum levels of FGF23 in etelcalcetide users were significantly lower than those in non-users at week 6 (p < 0.001) and week 12 (p < 0.001). When compared the difference between the E+Ca group and the E+D group, serum levels of FGF23 in the E+Ca group were significantly lower than those in the E+D group at week 12 (p = 0.017). There were no significant differences in the serum levels of CPPs between etelcalcetide users and non-users at week 6 (p = 0.10) and week 12 (p = 0.18), while CPPs in the E+Ca group were significantly lower than those in the E+D group (p < 0.001) at week 12. CONCLUSION Etelcalcetide may be useful through suppression of FGF23 levels among hemodialysis patients with SHPT. When correcting hypocalcemia, loading oral calcium preparations could be more advantageous than active vitamin D for the suppression of both FGF23 and CPPs.
Collapse
Affiliation(s)
- Yusaku Hashimoto
- Department of Nephrology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Sawako Kato
- Department of Nephrology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Makoto Kuro-O
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Yutaka Miura
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Yuya Itano
- Department of Nephrology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Masahiko Ando
- Department of Advanced Medicine, Nagoya University Hospital, Aichi, Japan
| | - Yachiyo Kuwatsuka
- Department of Advanced Medicine, Nagoya University Hospital, Aichi, Japan
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Aichi, Japan
| |
Collapse
|
16
|
Yamada S, Tsuruya K, Kitazono T, Nakano T. Emerging cross-talks between chronic kidney disease-mineral and bone disorder (CKD-MBD) and malnutrition-inflammation complex syndrome (MICS) in patients receiving dialysis. Clin Exp Nephrol 2022; 26:613-629. [PMID: 35353283 PMCID: PMC9203392 DOI: 10.1007/s10157-022-02216-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/22/2022] [Indexed: 12/14/2022]
Abstract
Chronic kidney disease–mineral and bone disorder (CKD–MBD) is a systemic disorder that affects multiple organs and systems and increases the risk of morbidity and mortality in patients with CKD, especially those receiving dialysis therapy. CKD–MBD is highly prevalent in CKD patients, and its treatment is gaining attention from healthcare providers who manage these patients. Additional important pathologies often observed in CKD patients are chronic inflammation and malnutrition/protein-energy wasting (PEW). These two pathologies coexist to form a vicious cycle that accelerates the progression of various other pathologies in CKD patients. This concept is integrated into the term “malnutrition–inflammation–atherosclerosis syndrome” or “malnutrition–inflammation complex syndrome (MICS)”. Recent basic and clinical studies have shown that CKD–MBD directly induces inflammation as well as malnutrition/PEW. Indeed, higher circulating levels of inorganic phosphate, fibroblast growth factor 23, parathyroid hormone, and calciprotein particles, as markers for critical components and effectors of CKD–MBD, were shown to directly induce inflammatory responses, thereby leading to malnutrition/PEW, cardiovascular diseases, and clinically relevant complications. In this short review, we discuss the close interplay between CKD–MBD and MICS and emphasize the significance of simultaneous control of these two seemingly distinct pathologies in patients with CKD, especially those receiving dialysis therapy, for better management of the CKD/hemodialysis population.
Collapse
Affiliation(s)
- Shunsuke Yamada
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 8128582, Japan.
| | | | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 8128582, Japan
| | - Toshiaki Nakano
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 8128582, Japan
| |
Collapse
|
17
|
Bodas de plata: 25 años de la primera demostración del efecto directo del fósforo en la célula paratiroidea. Nefrologia 2022. [DOI: 10.1016/j.nefro.2021.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
18
|
Role of K + and Ca 2+-Permeable Channels in Osteoblast Functions. Int J Mol Sci 2021; 22:ijms221910459. [PMID: 34638799 PMCID: PMC8509041 DOI: 10.3390/ijms221910459] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/20/2022] Open
Abstract
Bone-forming cells or osteoblasts play an important role in bone modeling and remodeling processes. Osteoblast differentiation or osteoblastogenesis is orchestrated by multiple intracellular signaling pathways (e.g., bone morphogenetic proteins (BMP) and Wnt signaling pathways) and is modulated by the extracellular environment (e.g., parathyroid hormone (PTH), vitamin D, transforming growth factor β (TGF-β), and integrins). The regulation of bone homeostasis depends on the proper differentiation and function of osteoblast lineage cells from osteogenic precursors to osteocytes. Intracellular Ca2+ signaling relies on the control of numerous processes in osteoblast lineage cells, including cell growth, differentiation, migration, and gene expression. In addition, hyperpolarization via the activation of K+ channels indirectly promotes Ca2+ signaling in osteoblast lineage cells. An improved understanding of the fundamental physiological and pathophysiological processes in bone homeostasis requires detailed investigations of osteoblast lineage cells. This review summarizes the current knowledge on the functional impacts of K+ channels and Ca2+-permeable channels, which critically regulate Ca2+ signaling in osteoblast lineage cells to maintain bone homeostasis.
Collapse
|
19
|
Kuro-o M. Klotho and calciprotein particles as therapeutic targets against accelerated ageing. Clin Sci (Lond) 2021; 135:1915-1927. [PMID: 34374422 PMCID: PMC8355631 DOI: 10.1042/cs20201453] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 01/25/2023]
Abstract
The klotho gene, named after a Greek goddess who spins the thread of life, was identified as a putative 'ageing-suppressor' gene. Klotho-deficient mice exhibit complex ageing-like phenotypes including hypogonadism, arteriosclerosis (vascular calcification), cardiac hypertrophy, osteopenia, sarcopenia, frailty, and premature death. Klotho protein functions as the obligate co-receptor for fibroblast growth factor-23 (FGF23), a bone-derived hormone that promotes urinary phosphate excretion in response to phosphate intake. Thus, Klotho-deficient mice suffer not only from accelerated ageing but also from phosphate retention due to impaired phosphate excretion. Importantly, restoration of the phosphate balance by placing Klotho-deficient mice on low phosphate diet rescued them from premature ageing, leading us to the notion that phosphate accelerates ageing. Because the extracellular fluid is super-saturated in terms of phosphate and calcium ions, an increase in the phosphate concentration can trigger precipitation of calcium-phosphate. In the blood, calcium-phosphate precipitated upon increase in the blood phosphate concentration is adsorbed by serum protein fetuin-A to form colloidal nanoparticles called calciprotein particles (CPPs). In the urine, CPPs appear in the renal tubular fluid when FGF23 increases phosphate load excreted per nephron. CPPs can induce cell damage, ectopic calcification, and inflammatory responses. CPPs in the blood can induce arteriosclerosis and non-infectious chronic inflammation, whereas CPPs in the urine can induce renal tubular damage and interstitial inflammation/fibrosis. Thus, we propose that CPPs behave like a pathogen that accelerates ageing and should be regarded as a novel therapeutic target against age-related disorders including chronic kidney disease.
Collapse
Affiliation(s)
- Makoto Kuro-o
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| |
Collapse
|
20
|
Ogura Y, Kaneko R, Ujibe K, Wakamatsu Y, Hirata H. Loss of αklotho causes reduced motor ability and short lifespan in zebrafish. Sci Rep 2021; 11:15090. [PMID: 34301962 PMCID: PMC8302672 DOI: 10.1038/s41598-021-93909-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 06/28/2021] [Indexed: 11/09/2022] Open
Abstract
The klotho gene encodes a transmembrane protein αKlotho that interacts with a fibroblast growth factor (FGF) receptor in renal tubular epithelial cells and functions as a co-receptor for FGF23, which is an osteocytes-derived hormone. This bone-to-kidney signal promotes urinary phosphate excretion. Interestingly, αKlotho knockout mice show an accelerated aging and a shortened life span. Similarly, C. elegans lacking the αklotho homologue showed a short life span. However, the physiological basis of aging-related function of αklotho remain unclear. The αklotho-deficient vertebrate animals other than mice have been awaited as an alternative model of premature aging. We here employed zebrafish in our study and revealed that αklotho mutant zebrafish appeared to be normal at 3 months postfertilization (mpf) but eventually underwent premature death by 9 mpf, while normal zebrafish is known to survive for 42 months. We also assessed the motor ability of zebrafish in a forced swimming assay and found that αklotho mutant zebrafish displayed reduced swimming performance before their survival declined. A recent study also reported a similar finding that αklotho-deficient zebrafish exhibited a short life span and reduced spontaneous movements. Taken together, these results suggest that αKlotho mutant zebrafish show premature aging and are useful to investigate aging in vertebrates.
Collapse
Affiliation(s)
- Yurie Ogura
- Department of Chemistry and Biological Science, College of Science and Engineering, Aoyama Gakuin University, Sagamihara, 252-5258, Japan
| | - Ryoji Kaneko
- Department of Chemistry and Biological Science, College of Science and Engineering, Aoyama Gakuin University, Sagamihara, 252-5258, Japan
| | - Kota Ujibe
- Department of Chemistry and Biological Science, College of Science and Engineering, Aoyama Gakuin University, Sagamihara, 252-5258, Japan
| | - Yuma Wakamatsu
- Department of Chemistry and Biological Science, College of Science and Engineering, Aoyama Gakuin University, Sagamihara, 252-5258, Japan
| | - Hiromi Hirata
- Department of Chemistry and Biological Science, College of Science and Engineering, Aoyama Gakuin University, Sagamihara, 252-5258, Japan.
| |
Collapse
|
21
|
Gocho Y, Tanaka M, Sugawara H, Furuhashi M, Moniwa N, Yamashita T, Takizawa H, Mukai H, Ohno K, Maeda T, Osanami A, Ohnishi H, Komatsu H, Mori K, Miura T. Seasonal variation of serum 25-hydroxyvitamin D level in hemodialysis patients in the northernmost island of Japan. Clin Exp Nephrol 2021; 25:1360-1366. [PMID: 34251521 DOI: 10.1007/s10157-021-02104-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/21/2021] [Indexed: 01/27/2023]
Abstract
BACKGROUND Serum vitamin D level shows a seasonal variation, being lower in winter than in summer in healthy subjects. The aim of this study was to determine whether there is presence of such a seasonal variation in hemodialysis patients. METHODS A total of 102 patients on hemodialysis were enrolled in February 2017 (winter) for analyses of serum levels of 25-hydroxyvitamin D [25(OH)D] and 1,25-dihydroxyvitamin D [1,25(OH)2D] and treatments for chronic kidney disease-mineral and bone disorder (CKD-MBD). The examinations were repeated in August 2017 (summer). After exclusion of patients with malignancy, loss of follow-up and missing data, 78 patients contributed to the analyses. RESULTS Serum level of 25(OH)D, but not that of 1,25(OH)2D, was significantly lower in winter (14.0 ng/mL) than in summer (15.5 ng/mL), though there was no significant difference in regimen for CKD-MBD treatment including vitamin D receptor activators (VDRAs) between the two seasons. Serum intact parathyroid hormone level tended to be higher and alkaline phosphatase was significantly higher in winter than in summer. Linear mixed-effects model analysis showed that level of 25(OH)D, but not that of 1,25(OH)2D, was significantly associated with season (winter and summer) after adjustment of age, sex, dialysis vintage, albumin level and use of drugs for CKD-MBD. CONCLUSION Serum 25(OH)D has a seasonal variation, being lower in winter than in summer, independent of CKD-MBD treatment including treatment with VDRAs in Japanese hemodialysis patients. The impact of the seasonal variation on risk of vitamin D deficiency and its effect on prognosis remain to be investigated.
Collapse
Affiliation(s)
- Yufu Gocho
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 060-8543, Japan.,Department of Nephrology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Marenao Tanaka
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 060-8543, Japan. .,Tanaka Medical Clinic, Yoichi, Hokkaido, Japan.
| | - Hirohito Sugawara
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 060-8543, Japan.,Department of Nephrology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 060-8543, Japan
| | - Norihito Moniwa
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 060-8543, Japan
| | - Tomohisa Yamashita
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 060-8543, Japan.,Department of Nephrology, Asahikawa Red Cross Hospital, Asahikawa, Hokkaido, Japan
| | - Hideki Takizawa
- Department of Nephrology, Teine Keijinkai Hospital, Sapporo, Japan
| | | | - Kouhei Ohno
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 060-8543, Japan.,JR Sapporo Hospital, Sapporo, Japan
| | - Takuto Maeda
- Department of Nephrology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Arata Osanami
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 060-8543, Japan
| | - Hirofumi Ohnishi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 060-8543, Japan.,Department of Public Health, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroaki Komatsu
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 060-8543, Japan
| | - Kazuma Mori
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 060-8543, Japan.,Division of Internal Medicine, Japan Self-Defense Forces Sapporo Hospital, Sapporo, Japan
| | - Tetsuji Miura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 060-8543, Japan
| |
Collapse
|
22
|
Donate-Correa J, Martín-Núñez E, González-Luis A, Ferri CM, Luis-Rodríguez D, Tagua VG, Mora-Fernández C, Navarro-González JF. Pathophysiological Implications of Imbalances in Fibroblast Growth Factor 23 in the Development of Diabetes. J Clin Med 2021; 10:2583. [PMID: 34208131 PMCID: PMC8230948 DOI: 10.3390/jcm10122583] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/24/2021] [Accepted: 06/08/2021] [Indexed: 12/18/2022] Open
Abstract
Observational studies have associated the increase in fibroblast growth factor (FGF) 23 levels, the main regulator of phosphate levels, with the onset of diabetes. These studies open the debate on the plausible existence of undescribed diabetogenic mechanisms derived from chronic supraphysiological levels of FGF23, a prevalent condition in chronic kidney disease (CKD) and end-stage renal disease (ESRD) patients. These maladaptive and diabetogenic responses to FGF23 may occur at different levels, including a direct effect on the pancreatic ß cells, and an indirect effect derived from the stimulation of the synthesis of pro-inflammatory factors. Both mechanisms could be mediated by the binding of FGF23 to noncanonical receptor complexes with the subsequent overactivation of signaling pathways that leads to harmful effects. The canonical binding of FGF23 to the receptor complex formed by the receptor FGFR1c and the coreceptor αKlotho activates Ras/MAPK/ERK signaling. However, supraphysiological concentrations of FGF23 favor non-αKlotho-dependent binding of this molecule to other FGFRs, which could generate an undesired overactivation of the PLCγ/CN/NFAT pathway, as observed in cardiomyocytes and hepatocytes. Moreover, the decrease in αKlotho expression may constitute a contributing factor to the appearance of these effects by promoting the nonspecific activation of the PLCγ/CN/NFAT to the detriment of the αKlotho-dependent Ras/MAPK/ERK pathway. The description of these mechanisms would allow the development of new therapeutic targets susceptible to be modified by dietary changes or by pharmacological intervention.
Collapse
Affiliation(s)
- Javier Donate-Correa
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain; (J.D.-C.); (E.M.-N.); (A.G.-L.); (C.M.F.); (V.G.T.); (C.M.-F.)
- GEENDIAB (Grupo Español para el Estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, 39008 Santander, Spain
| | - Ernesto Martín-Núñez
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain; (J.D.-C.); (E.M.-N.); (A.G.-L.); (C.M.F.); (V.G.T.); (C.M.-F.)
- GEENDIAB (Grupo Español para el Estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, 39008 Santander, Spain
| | - Ainhoa González-Luis
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain; (J.D.-C.); (E.M.-N.); (A.G.-L.); (C.M.F.); (V.G.T.); (C.M.-F.)
- Escuela de Doctorado y Estudios de Posgrado, Universidad de La Laguna, 38200 San Cristóbal de La Laguna, Spain
| | - Carla M. Ferri
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain; (J.D.-C.); (E.M.-N.); (A.G.-L.); (C.M.F.); (V.G.T.); (C.M.-F.)
- Escuela de Doctorado y Estudios de Posgrado, Universidad de La Laguna, 38200 San Cristóbal de La Laguna, Spain
| | - Desirée Luis-Rodríguez
- Servicio de Nefrología, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain;
| | - Víctor G. Tagua
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain; (J.D.-C.); (E.M.-N.); (A.G.-L.); (C.M.F.); (V.G.T.); (C.M.-F.)
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, 38010 San Cristóbal de La Laguna, Spain
| | - Carmen Mora-Fernández
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain; (J.D.-C.); (E.M.-N.); (A.G.-L.); (C.M.F.); (V.G.T.); (C.M.-F.)
| | - Juan F. Navarro-González
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain; (J.D.-C.); (E.M.-N.); (A.G.-L.); (C.M.F.); (V.G.T.); (C.M.-F.)
- GEENDIAB (Grupo Español para el Estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, 39008 Santander, Spain
- Servicio de Nefrología, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain;
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, 38010 San Cristóbal de La Laguna, Spain
- REDINREN (Red de Investigación Renal-RD16/0009/0022), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
23
|
Jaschke N, Sipos W, Hofbauer LC, Rachner TD, Rauner M. Skeletal endocrinology: where evolutionary advantage meets disease. Bone Res 2021; 9:28. [PMID: 34050126 PMCID: PMC8163738 DOI: 10.1038/s41413-021-00149-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 02/16/2021] [Accepted: 02/23/2021] [Indexed: 02/06/2023] Open
Abstract
The regulation of whole-body homeostasis by the skeleton is mediated by its capacity to secrete endocrine signaling molecules. Although bone-derived hormones confer several adaptive benefits, their physiological functions also involve trade-offs, thus eventually contributing to disease. In this manuscript, we discuss the origins and functions of two of the best-studied skeletal mediators, fibroblast growth factor 23 and osteocalcin, in an evolutionary context. Moreover, we provide a theoretical framework seeking to explain the broad involvement of these two hormones in amniote physiology as well as their potential to fuel the development and progression of diseases. Vice versa, we outline which perturbations might be amenable to manipulation of these systems and discuss limitations and ongoing challenges in skeletal endocrine research. Finally, we summarize unresolved questions and potential future studies in this thriving field.
Collapse
Affiliation(s)
- Nikolai Jaschke
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Wolfgang Sipos
- Clinical Department for Farm Animals, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Lorenz C Hofbauer
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Tilman D Rachner
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
24
|
Chu C, Elitok S, Zeng S, Xiong Y, Hocher CF, Hasan AA, Krämer BK, Hocher B. C-terminal and intact FGF23 in kidney transplant recipients and their associations with overall graft survival. BMC Nephrol 2021; 22:125. [PMID: 33832449 PMCID: PMC8033679 DOI: 10.1186/s12882-021-02329-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/30/2021] [Indexed: 12/31/2022] Open
Abstract
Background Increased fibroblast growth factor 23 (FGF23) is a risk factor for mortality, cardiovascular disease, and progression of chronic kidney disease. Limited data exist comparing the association of either c-terminal FGF23 (cFGF23) or intact FGF23 (iFGF23) in kidney transplant recipients (KTRs) with overall (all-cause) graft loss. Methods We conducted a prospective observational cohort study in 562 stable kidney transplant recipients. Patients were followed for graft loss and all-cause mortality for a median follow-up of 48 months. Results During a median follow-up of 48 months, 94 patients had overall graft loss (primary graft loss or death with functioning graft). Both cFGF23 and iFGF23 concentrations were significantly higher in patients with overall graft loss than those without (24.59 [11.43–87.82] versus 10.67 [5.99–22.73] pg/ml; p < 0.0001 and 45.24 [18.63–159.00] versus 29.04 [15.23–60.65] pg/ml; p = 0.002 for cFGF23 and iFGF23, respectively). Time-dependent ROC analysis showed that cFGF23 concentrations had a better discriminatory ability than iFGF23 concentrations in predicting overall (all-cause) graft loss. Cox regression analyses adjusted for risk factors showed that cFGF23 (HR for one unit increase of log transformed cFGF23: 1.35; 95% CI, 1.01–1.79; p = 0.043) but not iFGF23 (HR for one unit increase of log transformed iFGF23: 0.97; 95% CI, 0.75–1.25; p = 0.794) was associated with the overall graft loss. Conclusion Elevated cFGF23 concentrations at baseline are independently associated with an increased risk of overall graft loss. iFGF23 measurements were not independently associated with overall graft loss. The cFGF23 ELISA might detect bioactive FGF23 fragments that are not detected by the iFGF23 ELISA. Supplementary Information The online version contains supplementary material available at 10.1186/s12882-021-02329-7.
Collapse
Affiliation(s)
- Chang Chu
- Fifth Department of Medicine (Nephrology/ Endocrinology/ Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.,Department of Nephrology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Saban Elitok
- Fifth Department of Medicine (Nephrology/ Endocrinology/ Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.,Clinic for Nephrology and Endocrinology, Klinikum Ernst von Bergmann, Potsdam, Germany
| | - Shufei Zeng
- Fifth Department of Medicine (Nephrology/ Endocrinology/ Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.,Department of Nephrology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Yingquan Xiong
- Department of Nephrology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany.,Institute of Pharmacy, Free University of Berlin, Berlin, Germany
| | - Carl-Friedrich Hocher
- Fifth Department of Medicine (Nephrology/ Endocrinology/ Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Ahmed A Hasan
- Fifth Department of Medicine (Nephrology/ Endocrinology/ Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.,Institute of Pharmacy, Free University of Berlin, Berlin, Germany.,Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Bernhard K Krämer
- Fifth Department of Medicine (Nephrology/ Endocrinology/ Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.,European Center for Angioscience, Medical Faculty Mannheim of the University of Heidelberg, Heidelberg, Germany.,Mannheim Institute for Innate Immunoscience, Medical Faculty Mannheim of the University of Heidelberg, Mannheim, Germany
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/ Endocrinology/ Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany. .,Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China. .,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China. .,IMD Institut für Medizinische Diagnostik Berlin-Potsdam GbR, Berlin, Germany.
| |
Collapse
|
25
|
Focus on the Possible Role of Dietary Sodium, Potassium, Phosphate, Magnesium, and Calcium on CKD Progression. J Clin Med 2021; 10:jcm10050958. [PMID: 33804573 PMCID: PMC7957473 DOI: 10.3390/jcm10050958] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/11/2021] [Accepted: 02/14/2021] [Indexed: 12/21/2022] Open
Abstract
The impressive estimated number of chronic kidney disease (CKD) patients in the world justifies any possible effort at implementing preventive measures of disease progression. Renal insufficiency is associated with significant changes in the electrolyte handling and body balance of sodium, potassium, phosphate, magnesium, and calcium, all of which are biologically vital molecules. Dietary habits could contribute significantly to the optimal management of possible derangements. In this review, we examined the available evidence recommending dietary prescriptions for these five elements aiming at reducing CKD progression. Clear evidence that specific dietary prescriptions may halt or reduce CKD progression is lacking. However, some practical recommendations are possible to prescribe the best possible therapy to the individual CKD patient.
Collapse
|
26
|
Kuro-O M. Phosphate as a Pathogen of Arteriosclerosis and Aging. J Atheroscler Thromb 2021; 28:203-213. [PMID: 33028781 PMCID: PMC8048948 DOI: 10.5551/jat.rv17045] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
During the evolution of skeletons, terrestrial vertebrates acquired strong bones made of calcium-phosphate. By keeping the extracellular fluid in a supersaturated condition regarding calcium and phosphate ions, they created the bone when and where they wanted simply by providing a cue for precipitation. To secure this strategy, they acquired a novel endocrine system to strictly control the extracellular phosphate concentration. In response to phosphate intake, fibroblast growth factor-23 (FGF23) is secreted from the bone and acts on the kidney through binding to its receptor Klotho to increase urinary phosphate excretion, thereby maintaining phosphate homeostasis. The FGF23-Klotho endocrine system, when disrupted in mice, results in hyperphosphatemia and vascular calcification. Besides, mice lacking Klotho or FGF23 suffer from complex aging-like phenotypes, which are alleviated by placing them on a low- phosphate diet, indicating that phosphate is primarily responsible for the accelerated aging. Phosphate acquires the ability to induce cell damage and inflammation when precipitated with calcium. In the blood, calcium-phosphate crystals are adsorbed by serum protein fetuin-A and prevented from growing into large precipitates. Consequently, nanoparticles that comprised calcium-phosphate crystals and fetuin-A, termed calciprotein particles (CPPs), are generated and dispersed as colloids. CPPs increase in the blood with an increase in serum phosphate and age. Circulating CPP levels correlate positively with vascular stiffness and chronic non-infectious inflammation, raising the possibility that CPPs may be an endogenous pro-aging factor. Terrestrial vertebrates with the bone made of calcium- phosphate may be destined to age due to calcium-phosphate in the blood.
Collapse
Affiliation(s)
- Makoto Kuro-O
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University
| |
Collapse
|
27
|
Zhang Y, Wang X, Huang X, Shen L, Zhang L, Shou D, Fan X. Transcriptome sequencing profiling identifies miRNA-331-3p as an osteoblast-specific miRNA in infected bone nonunion. Bone 2021; 143:115619. [PMID: 32858253 DOI: 10.1016/j.bone.2020.115619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/20/2020] [Accepted: 08/24/2020] [Indexed: 11/25/2022]
Abstract
Bone nonunion caused by bacterial infection accounts for bone fractures, bone trauma and bone transplantation surgeries. Severe consequences include delayed unions and amputation and result in functional limitations, work disability, and poor quality of life. However, the mechanism of bone nonunion remains unknown. In this study, we aimed to screen the miRNA biomarkers of bacterial bone infection and investigated whether miRNAs regulate the osteoblasts and thus contribute to bone nonunion. We established a miRNA-mRNA network based on high-throughput RNA sequencing to compare the model rabbits infected with Staphylococcus aureus with the control rabbits. After validation experiments, miRNA-331-3p and fibroblast growth factor 23 (FGF23) were found to be inversely correlated with the pathways of osteoblast mineralization and pathology of infected bone nonunion. In in vitro experiments, miRNA-331-3p was downregulated and FGF23 was upregulated in lipopolysaccharide (LPS)-induced mouse calvarial osteoblasts. Further studies of the mechanism showed that mutated of putative miRNA-331-3p can bind to FGF23 3'-untranslated region sites. MiRNA-331-3p acted as an osteoblast mineralization promoter by directly targeting FGF23. Downregulation of miRNA-331-3p led to inhibition of osteoblast mineralization by regulating the DKK1/β-catenin mediated signaling. Thus, we established an improved animal model and identified new bone-related biomarkers in the infected bone nonunion. The miRNA-331-3p biomarker was demonstrated to regulate osteoblast mineralization by targeting FGF23. The novel mechanism can be used as potential diagnostic biomarkers and therapeutic targets in the infected bone nonunion and other inflammatory bone disorders.
Collapse
Affiliation(s)
- Yang Zhang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China
| | - Xuping Wang
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China
| | - Xiaowen Huang
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China
| | - Lifeng Shen
- Department of Orthopaedic Surgery, Zhejiang Provincial Tongde Hospital, Hangzhou 310012, China
| | - Li Zhang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Dan Shou
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China.
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
28
|
Daryadel A, Ruiz PA, Gehring N, Stojanovic D, Ugrica M, Bettoni C, Sabrautzki S, Pastor‐Arroyo E, Frey‐Wagner I, Lorenz‐Depiereux B, Strom TM, Angelis MH, Rogler G, Wagner CA, Rubio‐Aliaga I. Systemic Jak1 activation provokes hepatic inflammation and imbalanced FGF23 production and cleavage. FASEB J 2021; 35:e21302. [DOI: 10.1096/fj.202002113r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 12/24/2022]
Affiliation(s)
- Arezoo Daryadel
- Institute of Physiology University of Zurich (UZH), and National Center of Competence in Research NCCR Kidney.CH Zurich Switzerland
| | - Pedro A. Ruiz
- Department of Gastroenterology and Hepatology University Hospital of Zurich, University of Zurich Zurich Switzerland
| | - Nicole Gehring
- Institute of Physiology University of Zurich (UZH), and National Center of Competence in Research NCCR Kidney.CH Zurich Switzerland
| | - Dragana Stojanovic
- Institute of Physiology University of Zurich (UZH), and National Center of Competence in Research NCCR Kidney.CH Zurich Switzerland
| | - Marko Ugrica
- Institute of Physiology University of Zurich (UZH), and National Center of Competence in Research NCCR Kidney.CH Zurich Switzerland
| | - Carla Bettoni
- Institute of Physiology University of Zurich (UZH), and National Center of Competence in Research NCCR Kidney.CH Zurich Switzerland
| | - Sibylle Sabrautzki
- Institute of Experimental Genetics German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH) Neuherberg85764Germany
| | - Eva‐Maria Pastor‐Arroyo
- Institute of Physiology University of Zurich (UZH), and National Center of Competence in Research NCCR Kidney.CH Zurich Switzerland
| | - Isabelle Frey‐Wagner
- Department of Gastroenterology and Hepatology University Hospital of Zurich, University of Zurich Zurich Switzerland
| | - Bettina Lorenz‐Depiereux
- Institute of Human Genetics, Helmholtz Zentrum München German Research Center for Environmental Health (GmbH) Neuherberg Germany
| | - Tim M. Strom
- Institut für Humangenetik Klinikum rechts der Isar der Technischen Universität München München Germany
| | - Martin Hrabě Angelis
- Institute of Experimental Genetics German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH) Neuherberg85764Germany
- Lehrstuhl für Experimentelle Genetik Technische Universität München Freising‐Weihenstephan Germany
- Member of German Center for Diabetes Research (DZD) Neuherberg Germany
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology University Hospital of Zurich, University of Zurich Zurich Switzerland
| | - Carsten A. Wagner
- Institute of Physiology University of Zurich (UZH), and National Center of Competence in Research NCCR Kidney.CH Zurich Switzerland
| | - Isabel Rubio‐Aliaga
- Institute of Physiology University of Zurich (UZH), and National Center of Competence in Research NCCR Kidney.CH Zurich Switzerland
| |
Collapse
|
29
|
Brener A, Lebenthal Y, Cleper R, Kapusta L, Zeitlin L. Body composition and cardiometabolic health of pediatric patients with X-linked hypophosphatemia (XLH) under burosumab therapy. Ther Adv Endocrinol Metab 2021; 12:20420188211001150. [PMID: 33796255 PMCID: PMC7970173 DOI: 10.1177/20420188211001150] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/12/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Burosumab, a recombinant anti-FGF23 monoclonal antibody, was recently introduced as a treatment for X-linked hypophosphatemia (XLH). Burosumab normalizes blood phosphate levels, thereby healing rickets, decreasing leg bowing, and reducing pain. We aimed to explore the body composition and cardiometabolic health of pediatric patients with XLH treated with burosumab. METHODS This observational real-life study was conducted on growing children and adolescents. The outcome measures included changes in sex- and age-adjusted anthropometric and body composition parameters [fat mass (FM), fat-free mass (FFM), appendicular skeletal muscle mass (ASMM), muscle-to-fat ratio (MFR)], blood pressure, laboratory evaluation, and radiographic rickets severity [Thacher Rickets Severity Score (TRSS)]. Body composition was assessed by bioelectrical impedance analysis (BIA). Percentiles for FFM% and ASMM% were calculated according to BIA pediatric reference curves. The delta variable was calculated as the variable at 12 months minus the variable at baseline. RESULTS A total of 15 pediatric patients with XLH are treated in our clinic; included in the analyses were 7 children and adolescents (3 males, mean age 8.7 ± 3.2 years) with XLH without comorbidities. Baseline BIA revealed an unfavorable physique, with increased body fat percentage in five patients and decreased muscle mass in six. Indices of lean body mass significantly increased after 6 and 12 months of treatment: FFM(kg) (p = 0.001, p = 0.046, respectively) and ASMM(kg) (p = 0.012, p = 0.034, respectively), without any significant change in FM(kg). The percentile of ASMM% increased significantly after 6 months of treatment (p = 0.006) and stabilized thereafter. TRSS improved significantly after 12 months of therapy (p = 0.005). Age was positively correlated with delta TRSS (r = 0.814, p = 0.026), and delta TRSS was negatively correlated with delta MFR (r = -0.826, p = 0.022). CONCLUSIONS There was a heretofore unrecognized improvement in body composition of growing children and adolescents with XLH who were treated with burosumab. These findings highlight the need to initiate burosumab treatment at a younger age when rickets is less severe.
Collapse
Affiliation(s)
| | - Yael Lebenthal
- Pediatric Endocrinology and Diabetes Unit,
Dana-Dwek Children’s Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv,
Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv,
Israel
| | - Roxana Cleper
- Pediatric Nephrology Unit, Dana-Dwek Children’s
Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv
University, Tel Aviv, Israel
| | - Livia Kapusta
- Pediatric Cardiology Unit, Dana-Dwek Children’s
Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv
University, Tel Aviv, Israel
- Department of Paediatric Cardiology, Amalia
Children’s Hospital, Radboud University Medical Centre, Nijmegen, The
Netherlands
| | - Leonid Zeitlin
- Pediatric Orthopedic Department, Dana-Dwek
Children’s Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv,
Israel
- Sackler Faculty of Medicine, Tel Aviv
University, Tel Aviv, Israel
| |
Collapse
|
30
|
Hildebrand S, Cunningham J. Is there a role for bisphosphonates in vascular calcification in chronic kidney disease? Bone 2021; 142:115751. [PMID: 33188959 DOI: 10.1016/j.bone.2020.115751] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 10/28/2020] [Accepted: 11/09/2020] [Indexed: 01/15/2023]
Abstract
Theoretically bisphosphonates could accelerate or retard vascular calcification. In subjects with low GFR, the position is further confounded by a combination of uncertain pharmacokinetics (GI absorption is poor and inconsistent at all levels of renal function and the effect of low GFR generally is to increase bioavailability) and a highly variable skeletal substrate with extremes of turnover that increase unpredictably further. Although bisphosphonates reduce bone formation by 70-90% in subjects with normal GFR and reduce the ability of bone to buffer exogenous calcium fluxes, in bisphosphonate treated postmenopausal women accelerated vascular calcification has not been documented. The kidneys assist with this buffering, but the capacity to modulate calcium excretion declines as GFR falls, increasing the risk of hypercalcaemia in the event of high calcium influx. In the ESRD patient, decreased buffering capacity substantially increases the risk of transient hypercalcaemia, especially in the setting of dialysis, and as such may promote vascular calcification which is highly prevalent in the CKD population. Low bone turnover may thus be less of a vascular problem in patients with preserved renal function and a bigger problem when the GFR is low. In patients with stage 4 and 5 CKD, adynamic bone disease associates with the severity and progression of arterial calcification, including coronary artery calcification, and further suppression of bone turnover by a bisphosphonate might exacerbate an already high predisposition to vascular calcification. No convincing signal of harm has emerged from clinical studies thus far. For example 51 individuals with CKD stage 3-4 treated with either alendronate 70 mg per week or placebo for 18 months showed no difference in the rate of vascular calcifications. Conversely an observational study of women with stage 3-4 CKD with pre-existing cardiovascular disease found an increased risk of mortality with a hazard ratio of 1.22 (1.04-1.42) in those given bisphosphonates. Direct suppression of vascular calcification by bisphosphonates is probably confined to etidronate - treatment of soft tissue calcification was a recognized indication for this drug and etidronate markedly reduced progression of vascular calcification in CKD patients. Bisphosphonates are analogues of pyrophosphate, a potent calcification inhibitor in bone and soft tissue. Thus the efficacy of etidronate as treatment for soft tissue calcification brought with it a problematic tendency to cause osteomalacia. In contrast, conventional doses of nitrogen-containing bisphosphonates fail to yield circulating concentrations sufficient to exert direct anti-calcifying effects, at least in patients with good renal function and studies using alendronate and ibandronate have yielded inconsistent vascular outcomes.
Collapse
Affiliation(s)
- S Hildebrand
- Centre for Nephrology, Royal Free Hospital, London, UK.
| | - J Cunningham
- Centre for Nephrology, Royal Free Hospital, London, UK
| |
Collapse
|
31
|
Role of the fibroblast growth factor 19 in the skeletal system. Life Sci 2020; 265:118804. [PMID: 33245964 DOI: 10.1016/j.lfs.2020.118804] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/10/2020] [Accepted: 11/18/2020] [Indexed: 02/05/2023]
Abstract
Fibroblast growth factor family (FGFs) is a kind of cytokine that plays an important role in growth, development, metabolism and disease. During bone development, multiple FGFs and fibroblast growth factor receptors (FGFRs) play important roles. Previous reports have elucidated the great importance of FGF1, 2, 4, 6, 7, 8, 9, 10, and 18 in bone development, and FGF21 and 23 in bone homeostasis and bone regulation. FGF19 was initially found in the human foetal brain, and its gene location is related to osteoporosis pseudoglioma syndrome. Presently, gene chip detection has repeatedly found that FGF19 shows spatiotemporal specificity of gene expression in bone development and bone-related diseases, as well as differences in the protein level, indicating that FGF19 affects the skeletal system. Considering the current insufficient understanding of FGF19 and its potential function in the skeletal system, this review aims to introduce the background of FGF19 in bone, summarise the research progress of FGF19 in the skeletal system, and discuss the role and therapeutic potential of FGF19 in bone development and bone-related diseases.
Collapse
|
32
|
Zeng S, Querfeld U, Feger M, Haffner D, Hasan AA, Chu C, Slowinski T, Bernd Dschietzig T, Schäfer F, Xiong Y, Zhang B, Rausch S, Horvathova K, Lang F, Karl Krämer B, Föller M, Hocher B. Relationship between GFR, intact PTH, oxidized PTH, non-oxidized PTH as well as FGF23 in patients with CKD. FASEB J 2020; 34:15269-15281. [PMID: 32964520 DOI: 10.1096/fj.202000596r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 08/07/2020] [Accepted: 09/08/2020] [Indexed: 12/20/2022]
Abstract
Fibroblast growth factor 23 (FGF23) and parathyroid hormone (PTH) are regulators of renal phosphate excretion and vitamin D metabolism. In chronic kidney disease (CKD), circulating FGF23 and PTH concentrations progressively increase as renal function declines. Oxidation of PTH at two methionine residues (positions 8 and 18) causes a loss of function. The impact of n-oxPTH and oxPTH on FGF23 synthesis, however, and how n-oxPTH and oxPTH concentrations are affected by CKD, is yet unknown. The effects of oxidized and non-oxidized PTH 1-34 on Fgf23 gene expression were analyzed in UMR106 osteoblast-like cells. Furthermore, we investigated the relationship between n-oxPTH and oxPTH, respectively, with FGF23 in two independent patients' cohorts (620 children with CKD and 600 kidney transplant recipients). While n-oxPTH stimulated Fgf23 mRNA synthesis in vitro, oxidation of PTH in particular at Met8 led to a markedly weaker stimulation of Fgf23. The effect was even stronger when both Met8 and Met18 were oxidized. In both clinical cohorts, n-oxPTH-but not oxPTH-was significantly associated with FGF23 concentrations, independent of known confounding factors. Moreover, with progressive deterioration of kidney function, intact PTH (iPTH) and oxPTH increased substantially, whereas n-oxPTH increased only moderately. In conclusion, n-oxPTH, but not oxPTH, stimulates Fgf23 gene expression. The increase in PTH with decreasing GFR is mainly due to an increase in oxPTH in more advanced stages of CKD.
Collapse
Affiliation(s)
- Shufei Zeng
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.,Department of Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Uwe Querfeld
- Division of Gastroenterology, Nephrology and Metabolic Diseases, Department of Pediatrics, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Martina Feger
- Department of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Hannover, Germany
| | - Ahmed A Hasan
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.,Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Chang Chu
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.,Department of Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Torsten Slowinski
- Department of Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Franz Schäfer
- Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Yingquan Xiong
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.,Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Bingbing Zhang
- Institute of Physiology, University of Tübingen, Tübingen, Germany.,College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Steffen Rausch
- Department of Physiology, University of Hohenheim, Stuttgart, Germany
| | | | - Florian Lang
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Bernhard Karl Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Michael Föller
- Department of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.,Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China.,Institute of Medical Diagnostics, IMD, Berlin, , Berlin, Germany
| |
Collapse
|
33
|
Buchanan S, Combet E, Stenvinkel P, Shiels PG. Klotho, Aging, and the Failing Kidney. Front Endocrinol (Lausanne) 2020; 11:560. [PMID: 32982966 PMCID: PMC7481361 DOI: 10.3389/fendo.2020.00560] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/09/2020] [Indexed: 12/11/2022] Open
Abstract
Klotho has been recognized as a gene involved in the aging process in mammals for over 30 years, where it regulates phosphate homeostasis and the activity of members of the fibroblast growth factor (FGF) family. The α-Klotho protein is the receptor for Fibroblast Growth Factor-23 (FGF23), regulating phosphate homeostasis and vitamin D metabolism. Phosphate toxicity is a hallmark of mammalian aging and correlates with diminution of Klotho levels with increasing age. As such, modulation of Klotho activity is an attractive target for therapeutic intervention in the diseasome of aging; in particular for chronic kidney disease (CKD), where Klotho has been implicated directly in the pathophysiology. A range of senotherapeutic strategies have been developed to directly or indirectly influence Klotho expression, with varying degrees of success. These include administration of exogenous Klotho, synthetic and natural Klotho agonists and indirect approaches, via modulation of the foodome and the gut microbiota. All these approaches have significant potential to mitigate loss of physiological function and resilience accompanying old age and to improve outcomes within the diseasome of aging.
Collapse
Affiliation(s)
- Sarah Buchanan
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Emilie Combet
- School of Medicine, Dentistry & Nursing, Human Nutrition, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Peter Stenvinkel
- Division of Renal Medicine M99, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Paul G. Shiels
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
34
|
Sakaguchi Y, Hamano T, Matsui I, Oka T, Yamaguchi S, Kubota K, Shimada K, Matsumoto A, Hashimoto N, Isaka Y. Low magnesium diet aggravates phosphate-induced kidney injury. Nephrol Dial Transplant 2020; 34:1310-1319. [PMID: 30535376 DOI: 10.1093/ndt/gfy358] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 10/11/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Magnesium is known to protect against phosphate-induced tubular cell injuries in vitro. We investigated in vivo effects of magnesium on kidney injuries and phosphate metabolism in mice exposed to a high phosphate diet. METHODS Heminephrectomized mice were maintained on a high phosphate/normal magnesium diet or a high phosphate/low magnesium diet for 6 weeks. We compared renal histology, phosphaturic hormones and renal α-Klotho expression between the two diet groups. RESULTS High phosphate diet-induced tubular injuries and interstitial fibrosis were remarkably aggravated by the low-magnesium diet. At 1 week after high phosphate feeding when serum creatinine levels were similar between the two groups, the low magnesium diet suppressed not only fecal phosphate excretion but also urinary phosphate excretion, resulting in increased serum phosphate levels. Parathyroid hormone (PTH) levels were not appropriately elevated in the low magnesium diet group despite lower 1,25-dihydroxyvitamin D and serum calcium levels compared with the normal magnesium diet group. Although fibroblast growth factor 23 (FGF23) levels were lower in the low magnesium diet group, calcitriol-induced upregulation of FGF23 could not restore the impaired urinary phosphate excretion. The low magnesium diet markedly downregulated α-Klotho expression in the kidney. This downregulation of α-Klotho occurred even when mice were fed the low phosphate diet. CONCLUSIONS A low magnesium diet aggravated high phosphate diet-induced kidney injuries. Impaired PTH secretion and downregulation of renal α-Klotho were likely to be involved in the blunted urinary phosphate excretion by the low magnesium diet. Increasing dietary magnesium may be useful to attenuate phosphate-induced kidney injury.
Collapse
Affiliation(s)
- Yusuke Sakaguchi
- Department of Inter-Organ Communication Research in Kidney Disease, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takayuki Hamano
- Department of Inter-Organ Communication Research in Kidney Disease, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Isao Matsui
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tatsufumi Oka
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Satoshi Yamaguchi
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Keiichi Kubota
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Karin Shimada
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ayumi Matsumoto
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Nobuhiro Hashimoto
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
35
|
Meng F, Bertucci C, Gao Y, Li J, Luu S, LeBoff MS, Glowacki J, Zhou S. Fibroblast growth factor 23 counters vitamin D metabolism and action in human mesenchymal stem cells. J Steroid Biochem Mol Biol 2020; 199:105587. [PMID: 32004706 DOI: 10.1016/j.jsbmb.2020.105587] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 11/15/2019] [Accepted: 01/08/2020] [Indexed: 02/07/2023]
Abstract
Chronic kidney disease (CKD) is associated with elevated circulating fibroblast growth factor 23 (FGF23), impaired renal biosynthesis of 1α,25-dihydroxyvitamin D (1α,25(OH)2D), low bone mass, and increased fracture risk. Our previous data with human mesenchymal stem cells (hMSCs) indicated that vitamin D metabolism in hMSCs is regulated as it is in the kidney and promotes osteoblastogenesis in an autocrine/paracrine manner. In this study, we tested the hypothesis that FGF23 inhibits vitamin D metabolism and action in hMSCs. hMSCs were isolated from discarded marrow during hip arthroplasty, including two subjects receiving hemodialysis and a series of 20 subjects (aged 49-83 years) with estimated glomerular filtration rate (eGFR) data. The direct in vitro effects of rhFGF23 on hMSCs were analyzed by RT-PCR, Western immunoblot, and biochemical assays. Ex vivo analyses showed positive correlations for both secreted and membrane-bound αKlotho gene expression in hMSCs with eGFR of the subjects from whom hMSCs were isolated. There was downregulated constitutive expression of αKlotho, but not FGFR1 in hMSCs obtained from two hemodialysis subjects. In vitro, rhFGF23 countered vitamin D-stimulated osteoblast differentiation of hMSCs by reducing the vitamin D receptor, CYP27B1/1α-hydroxylase, biosynthesis of 1α,25(OH)2D3, and signaling through BMP-7. These data demonstrate that dysregulated vitamin D metabolism in hMSCs may contribute to impaired osteoblastogenesis and altered bone and mineral metabolism in CKD subjects due to elevated FGF23. This supports the importance of intracellular vitamin D metabolism in autocrine/paracrine regulation of osteoblast differentiation in hMSCs.
Collapse
Affiliation(s)
- Fangang Meng
- Department of Orthopedic Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Christopher Bertucci
- Department of Orthopedic Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yuan Gao
- Department of Orthopedic Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jing Li
- Department of Orthopedic Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Endocrinology, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Simon Luu
- Department of Orthopedic Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Meryl S LeBoff
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Julie Glowacki
- Department of Orthopedic Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
36
|
Si J, Wang C, Zhang D, Wang B, Hou W, Zhou Y. Osteopontin in Bone Metabolism and Bone Diseases. Med Sci Monit 2020; 26:e919159. [PMID: 31996665 PMCID: PMC7003659 DOI: 10.12659/msm.919159] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 11/04/2019] [Indexed: 12/20/2022] Open
Abstract
Osteopontin (OPN), a secreted phosphoprotein, is a member of the small integrin-binding ligand N-linked glycoprotein (SIBLING) family of cell matrix proteins and participates in many biological activities. Studies have shown that OPN plays a role in bone metabolism and homeostasis. OPN not only is an important factor in neuron-mediated and endocrine-regulated bone mass, but also is involved in biological activities such as proliferation, migration, and adhesion of several bone-related cells, including bone marrow mesenchymal stem cells, hematopoietic stem cells, osteoclasts, and osteoblasts. OPN has been demonstrated to be closely related to the occurrence and development of many bone-related diseases, such as osteoporosis, rheumatoid arthritis, and osteosarcoma. As expected, the functions of OPN in the bone have become a research hotspot. In this article, we try to decipher the mechanism of OPN-regulated bone metabolism and bone diseases.
Collapse
Affiliation(s)
- Jinyan Si
- Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Chaowei Wang
- Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Denghui Zhang
- Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Bo Wang
- Shanxi Medical University, Taiyuan, Shanxi, P.R. China
| | - Weiwei Hou
- Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Yi Zhou
- Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
37
|
Abstract
The Klotho proteins, αKlotho and βKlotho, are essential components of endocrine fibroblast growth factor (FGF) receptor complexes, as they are required for the high-affinity binding of FGF19, FGF21 and FGF23 to their cognate FGF receptors (FGFRs). Collectively, these proteins form a unique endocrine system that governs multiple metabolic processes in mammals. FGF19 is a satiety hormone that is secreted from the intestine on ingestion of food and binds the βKlotho-FGFR4 complex in hepatocytes to promote metabolic responses to feeding. By contrast, under fasting conditions, the liver secretes the starvation hormone FGF21, which induces metabolic responses to fasting and stress responses through the activation of the hypothalamus-pituitary-adrenal axis and the sympathetic nervous system following binding to the βKlotho-FGFR1c complex in adipocytes and the suprachiasmatic nucleus, respectively. Finally, FGF23 is secreted by osteocytes in response to phosphate intake and binds to αKlotho-FGFR complexes, which are expressed most abundantly in renal tubules, to regulate mineral metabolism. Growing evidence suggests that the FGF-Klotho endocrine system also has a crucial role in the pathophysiology of ageing-related disorders, including diabetes, cancer, arteriosclerosis and chronic kidney disease. Therefore, targeting the FGF-Klotho endocrine axes might have therapeutic benefit in multiple systems; investigation of the crystal structures of FGF-Klotho-FGFR complexes is paving the way for the development of drugs that can regulate these axes.
Collapse
Affiliation(s)
- Makoto Kuro-O
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan. .,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
38
|
Wagner CA, Rubio-Aliaga I, Egli-Spichtig D. Fibroblast growth factor 23 in chronic kidney disease: what is its role in cardiovascular disease? Nephrol Dial Transplant 2019; 34:1986-1990. [PMID: 30903187 DOI: 10.1093/ndt/gfz044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 02/10/2019] [Indexed: 11/14/2022] Open
Affiliation(s)
- Carsten A Wagner
- National Center of Competence in Research Kidney, CH, Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Isabel Rubio-Aliaga
- National Center of Competence in Research Kidney, CH, Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Daniela Egli-Spichtig
- National Center of Competence in Research Kidney, CH, Institute of Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
39
|
Smith ER, Holt SG, Hewitson TD. αKlotho-FGF23 interactions and their role in kidney disease: a molecular insight. Cell Mol Life Sci 2019; 76:4705-4724. [PMID: 31350618 PMCID: PMC11105488 DOI: 10.1007/s00018-019-03241-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 07/09/2019] [Accepted: 07/22/2019] [Indexed: 12/16/2022]
Abstract
Following the serendipitous discovery of the ageing suppressor, αKlotho (αKl), several decades ago, a growing body of evidence has defined a pivotal role for its various forms in multiple aspects of vertebrate physiology and pathology. The transmembrane form of αKl serves as a co-receptor for the osteocyte-derived mineral regulator, fibroblast growth factor (FGF)23, principally in the renal tubules. However, compelling data also suggest that circulating soluble forms of αKl, derived from the same source, may have independent homeostatic functions either as a hormone, glycan-cleaving enzyme or lectin. Chronic kidney disease (CKD) is of particular interest as disruption of the FGF23-αKl axis is an early and common feature of disease manifesting in markedly deficient αKl expression, but FGF23 excess. Here we critically discuss recent findings in αKl biology that conflict with the view that soluble αKl has substantive functions independent of FGF23 signalling. Although the issue of whether soluble αKl can act without FGF23 has yet to be resolved, we explore the potential significance of these contrary findings in the context of CKD and highlight how this endocrine pathway represents a promising target for novel anti-ageing therapeutics.
Collapse
Affiliation(s)
- Edward R Smith
- Department of Nephrology, The Royal Melbourne Hospital, Melbourne, Australia.
- Department of Medicine, University of Melbourne, Grattan Street, Parkville, VIC, 3050, Australia.
| | - Stephen G Holt
- Department of Nephrology, The Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, University of Melbourne, Grattan Street, Parkville, VIC, 3050, Australia
| | - Tim D Hewitson
- Department of Nephrology, The Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, University of Melbourne, Grattan Street, Parkville, VIC, 3050, Australia
| |
Collapse
|
40
|
Bressendorff I, Hansen D, Pasch A, Holt SG, Schou M, Brandi L, Smith ER. The effect of increasing dialysate magnesium on calciprotein particles, inflammation and bone markers: post hoc analysis from a randomized controlled clinical trial. Nephrol Dial Transplant 2019; 36:713-721. [DOI: 10.1093/ndt/gfz234] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Indexed: 12/21/2022] Open
Abstract
Abstract
Background
The formation of calciprotein particles (CPPs) may be an important component of the humoral defences against ectopic calcification. Although magnesium (Mg) has been shown to delay the transition of amorphous calcium-/phosphate-containing primary CPP (CPP-1) to crystalline apatite-containing secondary CPP (CPP-2) ex vivo, effects on the endogenous CPP pool are unknown.
Methods
We used post hoc analyses from a randomized double-blind parallel-group controlled clinical trial of 28 days treatment with high dialysate Mg of 2.0 mEq/L versus standard dialysate Mg of 1.0 mEq/L in 57 subjects undergoing maintenance hemodialysis for end-stage kidney disease. CPP load, markers of systemic inflammation and bone turnover were measured at baseline and follow-up.
Results
After 28 days of treatment with high dialysate Mg, serum total CPP (−52%), CPP-1 (−42%) and CPP-2 (−68%) were lower in the high Mg group (all P < 0.001) but were unchanged in the standard dialysate Mg group. Tumour necrosis factor-α (−20%) and interleukin-6 (−22%) were also reduced with high dialysate Mg treatment (both P < 0.01). High dialysate Mg resulted in higher levels of bone-specific alkaline phosphatase (a marker of bone formation) (+17%) but lower levels of tartrate-resistant acid phosphatase 5 b (a marker of bone resorption; −33%) (both P < 0.01). Inflammatory cytokines and bone turnover markers were unchanged in the standard dialysate Mg group over the same period.
Conclusions
In this exploratory analysis, increasing dialysate Mg was associated with reduced CPP load and systemic inflammation and divergent changes in markers of bone formation and resorption.
Collapse
Affiliation(s)
- Iain Bressendorff
- Department of Endocrinology and Nephrology, Nordsjællands Hospital, Hillerød, Denmark
- Department of Nephrology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Ditte Hansen
- Department of Nephrology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Andreas Pasch
- Calciscon AG, Nidau, Switzerland
- Insitute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Stephen G Holt
- Department of Nephrology, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Morten Schou
- Department of Cardiology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Lisbet Brandi
- Department of Endocrinology and Nephrology, Nordsjællands Hospital, Hillerød, Denmark
| | - Edward R Smith
- Department of Nephrology, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
41
|
Gloux A, Le Roy N, Brionne A, Bonin E, Juanchich A, Benzoni G, Piketty ML, Prié D, Nys Y, Gautron J, Narcy A, Duclos MJ. Candidate genes of the transcellular and paracellular calcium absorption pathways in the small intestine of laying hens. Poult Sci 2019; 98:6005-6018. [PMID: 31328776 PMCID: PMC6771766 DOI: 10.3382/ps/pez407] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/24/2019] [Indexed: 12/31/2022] Open
Abstract
To meet the high calcium (Ca) demand during eggshell biomineralization (2 g of Ca per egg), laying hens develop specific metabolic regulations to maintain Ca homeostasis. The intake of Ca, its solubilization, and absorption capacity are enhanced at sexual maturity (SM). A better knowledge of the intestinal Ca transporters involved in their variations at this stage could indicate new nutritional strategies to enhance Ca digestive utilization. Transcellular Ca absorption pathway and its major player calbindin-D 28 K (CALB1) mediate a saturable transport, which has been extensively described in this model. Conversely, a contribution by the paracellular pathway involving non-saturable Ca transport through intercellular tight junction has also been suggested. The aim of the present study was to identify candidate genes of these two pathways and their patterns of expression, in immature pullets (12, 15, and 17 wk old) and mature laying hens (23 wk old) in the duodenum, jejunum, and ileum. Using RT-qPCR, this study identifies 3 new candidate genes for transcellular, and 9 for paracellular Ca transport. A total of 5 candidates of the transcellular pathway, transient receptor potential cation channels subfamily C member 1 (TRPC1) and M member 7 (TRPM7); CALB1 and ATPase plasma membrane Ca2+ transporting 1 (ATP2B1) and ATPase plasma membrane Ca2+ transporting 2 (ATP2B2) were enhanced with age or after SM in the duodenum, the jejunum or all 3 segments. A total of 4 candidates of the paracellular pathway Claudin 2 (CLDN2) and tight junction proteins 1, 2, and 3 (TJP1, TJP2 and TJP3) increased in the small intestine after SM. Additionally, CALB1, ATP2B2, and CLDN2 were overexpressed in the duodenum or the jejunum or both segments after SM. The enhanced expression of candidate genes of the paracellular Ca pathway after SM, supports that the non-saturable transport could be a mechanism of great importance when high concentrations of soluble Ca are observed in the intestinal content during eggshell formation. Both pathways may work cooperatively in the duodenum and jejunum, the main sites of Ca absorption in laying hens.
Collapse
Affiliation(s)
- A Gloux
- BOA, INRA, Université de Tours, 37380 Nouzilly, France
| | - N Le Roy
- BOA, INRA, Université de Tours, 37380 Nouzilly, France
| | - A Brionne
- BOA, INRA, Université de Tours, 37380 Nouzilly, France
| | - E Bonin
- GeT-PlaGe, INRA, Auzeville, 31326 Castanet-Tolosan, France
| | - A Juanchich
- BOA, INRA, Université de Tours, 37380 Nouzilly, France
| | - G Benzoni
- Prospective and Innovation department, Neovia, 56250 Saint-Nolff, France
| | - M-L Piketty
- Service des Explorations Fonctionnelles, G.H. Necker Enfants Malades, 75743 Paris Cedex 15, France, Université Paris Descartes Faculté de Médecine, INSERM U1151
| | - D Prié
- Service des Explorations Fonctionnelles, G.H. Necker Enfants Malades, 75743 Paris Cedex 15, France, Université Paris Descartes Faculté de Médecine, INSERM U1151
| | - Y Nys
- BOA, INRA, Université de Tours, 37380 Nouzilly, France
| | - J Gautron
- BOA, INRA, Université de Tours, 37380 Nouzilly, France
| | - A Narcy
- BOA, INRA, Université de Tours, 37380 Nouzilly, France
| | - M J Duclos
- BOA, INRA, Université de Tours, 37380 Nouzilly, France
| |
Collapse
|
42
|
Mazzaferro S, Cianciolo G, De Pascalis A, Guglielmo C, Urena Torres PA, Bover J, Tartaglione L, Pasquali M, La Manna G. Bone, inflammation and the bone marrow niche in chronic kidney disease: what do we know? Nephrol Dial Transplant 2019; 33:2092-2100. [PMID: 29733407 DOI: 10.1093/ndt/gfy115] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 04/02/2018] [Indexed: 02/06/2023] Open
Abstract
Recent improvements in our understanding of physiology have altered the way in which bone is perceived: no longer is it considered as simply the repository of divalent ions, but rather as a sophisticated endocrine organ with potential extraskeletal effects. Indeed, a number of pathologic conditions involving bone in different ways can now be reconsidered from a bone-centred perspective. For example, in metabolic bone diseases like osteoporosis (OP) and renal osteodystrophy (ROD), the association with a worse cardiovascular outcome can be tentatively explained by the possible derangements of three recently discovered bone hormones (osteocalcin, fibroblast growth factor 23 and sclerostin) and a bone-specific enzyme (alkaline phosphatase). Further, in recent years the close link between bone and inflammation has been better appreciated and a wide range of chronic inflammatory states (from rheumatoid arthritis to ageing) are being explored to discover the biochemical changes that ultimately lead to bone loss and OP. Also, it has been acknowledged that the concept of the bone-vascular axis may explain, for example, the relationship between bone metabolism and vessel wall diseases like atherosclerosis and arteriosclerosis, with potential involvement of a number of cytokines and metabolic pathways. A very important discovery in bone physiology is the bone marrow (BM) niche, the functional unit where stem cells interact, exchanging signals that impact on their fate as bone-forming cells or immune-competent haematopoietic elements. This new element of bone physiology has been recognized to be dysfunctional in diabetes (so-called diabetic mobilopathy), with possible clinical implications. In our opinion, ROD, the metabolic bone disease of renal patients, will in the future probably be identified as a cause of BM niche dysfunction. An integrated view of bone, which includes the BM niche, now seems necessary in order to understand the complex clinical entity of chronic kidney disease-mineral and bone disorders and its cardiovascular burden. Bone is thus becoming a recurrently considered paradigm for different inter-organ communications that needs to be considered in patients with complex diseases.
Collapse
Affiliation(s)
- Sandro Mazzaferro
- Department of Cardiovascular Respiratory Nephrologic Geriatric and Anesthetic Sciences, Sapienza University of Rome, Rome, Italy.,Nephrology Unit, Azienda Ospedaliero-Universitaria Policlinico Umberto I, Rome, Italy
| | - Giuseppe Cianciolo
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, St Orsola Hospital, University of Bologna, Bologna, Italy
| | - Antonio De Pascalis
- Nephrology, Dialysis and Renal Transplant Unit, Vito Fazzi Hospital, Lecce, Italy
| | - Chiara Guglielmo
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, St Orsola Hospital, University of Bologna, Bologna, Italy
| | - Pablo A Urena Torres
- Ramsay-Générale de Santé, Clinique du Landy, Department of Nephrology and Dialysis and Department of Renal Physiology, Necker Hospital, University of Paris Descartes, Paris, France
| | - Jordi Bover
- Fundació Puigvert, Department of Nephrology IIB Sant Pau, RedinRen, Barcelona, Catalonia, Spain
| | - Lida Tartaglione
- Department of Cardiovascular Respiratory Nephrologic Geriatric and Anesthetic Sciences, Sapienza University of Rome, Rome, Italy
| | - Marzia Pasquali
- Nephrology Unit, Azienda Ospedaliero-Universitaria Policlinico Umberto I, Rome, Italy
| | - Gaetano La Manna
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, St Orsola Hospital, University of Bologna, Bologna, Italy
| |
Collapse
|
43
|
Daryadel A, Natale L, Seebeck P, Bettoni C, Schnitzbauer U, Gassmann M, Wagner CA. Elevated FGF23 and disordered renal mineral handling with reduced bone mineralization in chronically erythropoietin over-expressing transgenic mice. Sci Rep 2019; 9:14989. [PMID: 31628396 PMCID: PMC6802194 DOI: 10.1038/s41598-019-51577-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 09/09/2019] [Indexed: 12/22/2022] Open
Abstract
Fibroblast Growth Factor 23 (FGF23) is a phosphaturic factor causing increased renal phosphate excretion as well as suppression of 1,25 (OH)2-vitamin D3. Highly elevated FGF23 can promote development of rickets and osteomalacia. We and others previously reported that acute application of erythropoietin (EPO) stimulates FGF23 production. Considering that EPO is clinically used as chronic treatment against anemia, we used here the Tg6 mouse model that constitutively overexpresses human EPO in an oxygen-independent manner, to examine the consequences of long-term EPO therapy on mineral and bone metabolism. Six to eight weeks old female Tg6 mice showed elevated intact and C-terminal fragment of FGF23 but normal plasma levels of PTH, calcitriol, calcium and phosphate. Renal function showed moderate alterations with higher urea and creatinine clearance and mild albuminuria. Renal phosphate excretion was normal whereas mild hypercalciuria was found. Renal expression of the key proteins TRPV5 and calbindin D28k involved in active calcium reabsorption was reduced in Tg6 mice. Plasma levels of the bone turnover marker osteocalcin were comparable between groups. However, urinary excretion of deoxypyridinoline (DPD) was lower in Tg6 mice. MicroCT analysis showed reduced total, cortical, and trabecular bone mineral density in femora from Tg6 mice. Our data reveal that chronic elevation of EPO is associated with high FGF23 levels and disturbed mineral homeostasis resulting in reduced bone mineral density. These observations imply the need to study the impact of therapeutically applied EPO on bone mineralization in patients, especially those suffering from chronic kidney disease.
Collapse
Affiliation(s)
- Arezoo Daryadel
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,National Centre for Competence in Research NCCR "Kidney.CH", Zurich, Switzerland
| | - Luciano Natale
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Petra Seebeck
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,Zurich Integrative Rodent Physiology (ZIRP), University of Zurich, Zurich, Switzerland
| | - Carla Bettoni
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,National Centre for Competence in Research NCCR "Kidney.CH", Zurich, Switzerland
| | - Udo Schnitzbauer
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,National Centre for Competence in Research NCCR "Kidney.CH", Zurich, Switzerland
| | - Max Gassmann
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland.,Universidad Peruana Cayetano Heredia (UPCH), Lima, Peru
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland. .,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland. .,National Centre for Competence in Research NCCR "Kidney.CH", Zurich, Switzerland.
| |
Collapse
|
44
|
Singh AP, Sosa MX, Fang J, Shanmukhappa SK, Hubaud A, Fawcett CH, Molind GJ, Tsai T, Capodieci P, Wetzel K, Sanchez E, Wang G, Coble M, Tang W, Cadena SM, Fishman MC, Glass DJ. αKlotho Regulates Age-Associated Vascular Calcification and Lifespan in Zebrafish. Cell Rep 2019; 28:2767-2776.e5. [DOI: 10.1016/j.celrep.2019.08.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/02/2019] [Accepted: 07/31/2019] [Indexed: 10/26/2022] Open
|
45
|
Alfieri C, Ruzhytska O, Vettoretti S, Caldiroli L, Cozzolino M, Messa P. Native Hypovitaminosis D in CKD Patients: From Experimental Evidence to Clinical Practice. Nutrients 2019; 11:E1918. [PMID: 31443249 PMCID: PMC6723756 DOI: 10.3390/nu11081918] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/07/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022] Open
Abstract
Native hypovitaminosis D (n-hVITD) is frequently found from the early stages of chronic kidney disease (CKD) and its prevalence increases with CKD progression. Even if the implications of n-hVITD in chronic kidney disease-mineral bone disorder (CKD-MBD) have been extensively characterized in the literature, there is a lot of debate nowadays about the so called "unconventional effects" of native vitamin D (25(OH)VitD) supplementation in CKD patients. In this review, highlights of the dimension of the problem of n-hVITD in CKD stages 2-5 ND patients will be presented. In addition, it will focus on the "unconventional effects" of 25(OH)VitD supplementation, the clinical impact of n-hVITD and the most significant interventional studies regarding 25(OH)VitD supplementation in CKD stages 2-5 ND.
Collapse
Affiliation(s)
- Carlo Alfieri
- 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
| | - Oksana Ruzhytska
- Department of Internal Medicine n3, Ternopil State Medical University, 46002 Ternopil, Ukraine
| | - Simone Vettoretti
- Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Lara Caldiroli
- Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Mario Cozzolino
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
- Renal Division, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, 20122 Milan, Italy
| | - Piergiorgio Messa
- 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
|
46
|
Imel EA, Biggin A, Schindeler A, Munns CF. FGF23, Hypophosphatemia, and Emerging Treatments. JBMR Plus 2019; 3:e10190. [PMID: 31485552 PMCID: PMC6715782 DOI: 10.1002/jbm4.10190] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 02/13/2019] [Accepted: 02/26/2019] [Indexed: 01/03/2023] Open
Abstract
FGF23 is an important hormonal regulator of phosphate homeostasis. Together with its co-receptor Klotho, it modulates phosphate reabsorption and both 1α-hydroxylation and 24-hydroxylation in the renal proximal tubules. The most common FGF23-mediated hypophosphatemia is X-linked hypophosphatemia (XLH), caused by mutations in the PHEX gene. FGF23-mediated forms of hypophosphatemia are characterized by phosphaturia and low or low-normal calcitriol concentrations, and unlike nutritional rickets, these cannot be cured with nutritional vitamin D supplementation. Autosomal dominant and autosomal recessive forms of FGF23-mediated hypophosphatemias show a similar pathophysiology, despite a variety of different underlying genetic causes. An excess of FGF23 activity has also been associated with a number of other conditions causing hypophosphatemia, including tumor-induced osteomalacia, fibrous dysplasia of the bone, and cutaneous skeletal hypophosphatemia syndrome. Historically phosphate supplementation and therapy using analogs of highly active vitamin D (eg, calcitriol, alfacalcidol, paricalcitol, eldecalcitol) have been used to manage conditions involving hypophosphatemia; however, recently a neutralizing antibody for FGF23 (burosumab) has emerged as a promising treatment agent for FGF23-mediated disorders. This review discusses the progression of clinical trials for burosumab for the treatment of XLH and its recent availability for clinical use. Burosumab may have potential for treating other conditions associated with FGF23 overactivity, but these are not yet supported by trial data. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Erik A Imel
- Division of EndocrinologyIndiana University School of Medicine, Indianapolis, INUSA
| | - Andrew Biggin
- The University of Sydney Children's Hospital Westmead Clinical School, University of SydneySydneyAustralia
- Department of EndocrinologyThe Children's Hospital at WestmeadWestmeadAustralia
| | - Aaron Schindeler
- The University of Sydney Children's Hospital Westmead Clinical School, University of SydneySydneyAustralia
- Orthopaedic Research Unit, The Children's Hospital at WestmeadWestmeadAustralia
| | - Craig F Munns
- The University of Sydney Children's Hospital Westmead Clinical School, University of SydneySydneyAustralia
- Department of EndocrinologyThe Children's Hospital at WestmeadWestmeadAustralia
| |
Collapse
|
47
|
Egli-Spichtig D, Imenez Silva PH, Glaudemans B, Gehring N, Bettoni C, Zhang MYH, Pastor-Arroyo EM, Schönenberger D, Rajski M, Hoogewijs D, Knauf F, Misselwitz B, Frey-Wagner I, Rogler G, Ackermann D, Ponte B, Pruijm M, Leichtle A, Fiedler GM, Bochud M, Ballotta V, Hofmann S, Perwad F, Föller M, Lang F, Wenger RH, Frew I, Wagner CA. Tumor necrosis factor stimulates fibroblast growth factor 23 levels in chronic kidney disease and non-renal inflammation. Kidney Int 2019; 96:890-905. [PMID: 31301888 DOI: 10.1016/j.kint.2019.04.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 03/11/2019] [Accepted: 04/05/2019] [Indexed: 01/03/2023]
Abstract
Fibroblast growth factor 23 (FGF23) regulates phosphate homeostasis, and its early rise in patients with chronic kidney disease is independently associated with all-cause mortality. Since inflammation is characteristic of chronic kidney disease and associates with increased plasma FGF23 we examined whether inflammation directly stimulates FGF23. In a population-based cohort, plasma tumor necrosis factor (TNF) was the only inflammatory cytokine that independently and positively correlated with plasma FGF23. Mouse models of chronic kidney disease showed signs of renal inflammation, renal FGF23 expression and elevated systemic FGF23 levels. Renal FGF23 expression coincided with expression of the orphan nuclear receptor Nurr1 regulating FGF23 in other organs. Antibody-mediated neutralization of TNF normalized plasma FGF23 and suppressed ectopic renal Fgf23 expression. Conversely, TNF administration to control mice increased plasma FGF23 without altering plasma phosphate. Moreover, in Il10-deficient mice with inflammatory bowel disease and normal kidney function, plasma FGF23 was elevated and normalized upon TNF neutralization. Thus, the inflammatory cytokine TNF contributes to elevated systemic FGF23 levels and also triggers ectopic renal Fgf23 expression in animal models of chronic kidney disease.
Collapse
Affiliation(s)
- Daniela Egli-Spichtig
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland; Department of Pediatrics, Division of Nephrology, University of California, San Francisco, San Francisco, California, USA
| | - Pedro Henrique Imenez Silva
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Bob Glaudemans
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Nicole Gehring
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Carla Bettoni
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Martin Y H Zhang
- Department of Pediatrics, Division of Nephrology, University of California, San Francisco, San Francisco, California, USA
| | - Eva M Pastor-Arroyo
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Désirée Schönenberger
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Michal Rajski
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - David Hoogewijs
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Felix Knauf
- Division of Nephrology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Benjamin Misselwitz
- University Hospital Zurich, Clinic for Gastroenterology and Hepatology, Zurich, Switzerland
| | - Isabelle Frey-Wagner
- University Hospital Zurich, Clinic for Gastroenterology and Hepatology, Zurich, Switzerland
| | - Gerhard Rogler
- University Hospital Zurich, Clinic for Gastroenterology and Hepatology, Zurich, Switzerland
| | - Daniel Ackermann
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Belen Ponte
- Department of Nephrology, University Hospital of Geneva (HUG), Geneva, Switzerland
| | - Menno Pruijm
- Department of Nephrology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Alexander Leichtle
- Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Georg-Martin Fiedler
- Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Murielle Bochud
- Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland; Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Virginia Ballotta
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Sandra Hofmann
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Farzana Perwad
- Department of Pediatrics, Division of Nephrology, University of California, San Francisco, San Francisco, California, USA
| | - Michael Föller
- Institute of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Florian Lang
- Institute of Physiology I, University of Tübingen, Tübingen, Germany
| | - Roland H Wenger
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Ian Frew
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland; Swiss National Center of Competence in Research NCCR-Kidney.CH, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
48
|
Huang X, Liu T, Zhao M, Fu H, Wang J, Xu Q. Protective Effects of Moderate Ca Supplementation against Cd-Induced Bone Damage under Different Population-Relevant Doses in Young Female Rats. Nutrients 2019; 11:E849. [PMID: 30991710 PMCID: PMC6521033 DOI: 10.3390/nu11040849] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/05/2019] [Accepted: 04/12/2019] [Indexed: 12/16/2022] Open
Abstract
Estimation of the skeleton-protective effects of Ca in Cd-induced bone damage is helpful in the assessment of Cd health risk. The aim of this study was to identify whether Ca supplementation during exposure to different population-relevant doses of Cd can prevent Cd-induced bone damage under the tolerable upper intake level of Ca supplementation. Young female Sprague-Dawley rats were given different population-relevant doses of Cd (1, 5, and 50 mg Cd/kg diet) and Ca supplementation (0.4% Ca supplementation) intervention. Ca supplementation significantly decreased Cd-induced bone microstructure damage, increased bone biomechanics (p < 0.05), serum bone formation marker level (p < 0.05) and expression of osteogenic gene markers exposure to the 5 and 50 mg Cd/kg diets. However, it had no impact on these indicators under the 1 mg Cd/kg diets, with the exception of expression of osteogenic marker genes. Ca supplementation significantly decreased serum Klotho level (p < 0.05), and fibroblast growth factor 23/Klotho-associated gene expression in the kidney and bone showed significant changes. In conclusion, Ca supplementation has a positive effect on bone formation and bone quality against the damaging impact of Cd, especially with exposure to the 5 mg and 50 mg Cd/kg diet, which may be related to its impact on the fibroblast growth factor 23/Klotho axis.
Collapse
Affiliation(s)
- Xiao Huang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, 210009 Nanjing, China.
| | - Teng Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, 210009 Nanjing, China.
| | - Meng Zhao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, 210009 Nanjing, China.
| | - Haowei Fu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, 210009 Nanjing, China.
| | - Jinming Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, 210009 Nanjing, China.
| | - Qian Xu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, 210009 Nanjing, China.
| |
Collapse
|
49
|
Structures of ligand-occupied β-Klotho complexes reveal a molecular mechanism underlying endocrine FGF specificity and activity. Proc Natl Acad Sci U S A 2019; 116:7819-7824. [PMID: 30944224 DOI: 10.1073/pnas.1822055116] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The three members of the endocrine fibroblast growth factor (FGF) family designated FGF19, FGF21, and FGF23 mediate their pleiotropic cellular effects by binding to and activating binary complexes composed of an FGF receptor (FGFR) bound to either α-Klotho or β-Klotho receptors. Structural analyses of ligand-occupied Klotho extracellular domains have provided important insights concerning mechanisms underlying the binding specificities of FGF21 and FGF23 to β-Klotho or α-Klotho, respectively. They have also demonstrated that Klotho proteins function as primary high-affinity receptors while FGFRs function as the catalytic subunits that mediate intracellular signaling. Here we describe the crystal structure the C-terminal tail of FGF19 (FGF19CT) bound to sKLB and demonstrate that FGF19CT and FGF21CT bind to the same binding site on sKLB, via a multiturn D-P motif to site 1 and via a S-P-S motif to the pseudoglycoside hydrolase region (site 2). Binding affinities to sKLB and cellular stimulatory activities of FGF19CT, FGF21CT, and a variety of chimeric mutants to cells expressing β-Klotho together with FGFR1c or FGFR4 were also analyzed. These experiments as well as detailed comparison of the structures of free and ligand-occupied sKLB to the structure of ligand-occupied sKLA reveal a general mechanism for recognition of endocrine FGFs by Klotho proteins and regulatory interactions with FGFRs that control their pleiotropic cellular responses.
Collapse
|
50
|
Bär L, Hase P, Föller M. PKC regulates the production of fibroblast growth factor 23 (FGF23). PLoS One 2019; 14:e0211309. [PMID: 30921339 PMCID: PMC6438472 DOI: 10.1371/journal.pone.0211309] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/14/2019] [Indexed: 12/20/2022] Open
Abstract
Serine/threonine protein kinase C (PKC) is activated by diacylglycerol that is released from membrane lipids by phospholipase C in response to activation of G protein-coupled receptors or receptor tyrosine kinases. PKC isoforms are particularly relevant for proliferation and differentiation of cells including osteoblasts. Osteoblasts/osteocytes produce fibroblast growth factor 23 (FGF23), a hormone regulating renal phosphate and vitamin D handling. PKC activates NFκB, a transcription factor complex controlling FGF23 expression. Here, we analyzed the impact of PKC on FGF23 synthesis. Fgf23 expression was analyzed by qRT-PCR in UMR106 osteoblast-like cells and in IDG-SW3 osteocytes, and FGF23 protein was measured by ELISA. Phorbol ester 12-O-tetradecanoylphorbol-13-acetate (PMA), a PKC activator, up-regulated FGF23 production. In contrast, PKC inhibitors calphostin C, Gö6976, sotrastaurin and ruboxistaurin suppressed FGF23 formation. NFκB inhibitor withaferin A abolished the stimulatory effect of PMA on Fgf23. PKC is a powerful regulator of FGF23 synthesis, an effect which is at least partly mediated by NFκB.
Collapse
Affiliation(s)
- Ludmilla Bär
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Philipp Hase
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Michael Föller
- Institute of Physiology, University of Hohenheim, Stuttgart, Germany
- * E-mail:
| |
Collapse
|