1
|
Dong DL, Jin GZ. Targeting Chondrocyte Hypertrophy as Strategies for the Treatment of Osteoarthritis. Bioengineering (Basel) 2025; 12:77. [PMID: 39851351 PMCID: PMC11760869 DOI: 10.3390/bioengineering12010077] [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: 11/28/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Osteoarthritis (OA) is a common joint disease characterized by pain and functional impairment, which severely impacts the quality of life of middle-aged and elderly individuals. During normal bone development, chondrocyte hypertrophy is a natural physiological process. However, in the progression of OA, chondrocyte hypertrophy becomes one of its key pathological features. Although there is no definitive evidence to date confirming that chondrocyte hypertrophy is the direct cause of OA, substantial experimental data indicate that it plays an important role in the disease's pathogenesis. In this review, we first explore the mechanisms underlying chondrocyte hypertrophy in OA and offer new insights. We then propose strategies for inhibiting chondrocyte hypertrophy from the perspectives of targeting signaling pathways and tissue engineering, ultimately envisioning the future prospects of OA treatment.
Collapse
Affiliation(s)
- Da-Long Dong
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea;
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Guang-Zhen Jin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea;
| |
Collapse
|
2
|
Chen X, Wei Y, Li Z, Zhou C, Fan Y. Distinct role of Klotho in long bone and craniofacial bone: skeletal development, repair and regeneration. PeerJ 2024; 12:e18269. [PMID: 39465174 PMCID: PMC11505971 DOI: 10.7717/peerj.18269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/17/2024] [Indexed: 10/29/2024] Open
Abstract
Bone defects are highly prevalent diseases caused by trauma, tumors, inflammation, congenital malformations and endocrine abnormalities. Ideally effective and side effect free approach to dealing with bone defects remains a clinical conundrum. Klotho is an important protein, which plays an essential role in regulating aging and mineral ion homeostasis. More recently, research revealed the function of Klotho in regulating skeleton development and regeneration. Klotho has been identified in mesenchymal stem cells, osteoblasts, osteocytes and osteoclasts in different skeleton regions. The specific function and regulatory mechanisms of Klotho in long bone and craniofacial bone vary due to their different embryonic development, ossification and cell types, which remain unclear and without conclusion. Moreover, studies have confirmed that Klotho is a multifunctional protein that can inhibit inflammation, resist cancer and regulate the endocrine system, which may further accentuate the potential of Klotho to be the ideal molecule in inducing bone restoration clinically. Besides, as an endogenous protein, Klotho has a promising potential for clinical therapy without side effects. In the current review, we summarized the specific function of Klotho in long bone and craniofacial skeleton from phenotype to cellular alternation and signaling pathway. Moreover, we illustrated the possible future clinical application for Klotho. Further research on Klotho might help to solve the existing clinical difficulties in bone healing and increase the life quality of patients with bone injury and the elderly.
Collapse
Affiliation(s)
- Xinyu Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yali Wei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zucen Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Fan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Zhang X, Xu J. Fibroblast growth factor 23-mediated regulation of osteoporosis: Assessed via Mendelian randomization and in vitro study. J Cell Mol Med 2024; 28:e18551. [PMID: 39054573 PMCID: PMC11272609 DOI: 10.1111/jcmm.18551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/17/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
Despite numerous investigations on the influence of fibroblast growth factor 23 (FGF23), α-Klotho and FGF receptor-1 (FGFR1) on osteoporosis (OP), there is no clear consensus. Mendelian randomization (MR) analysis was conducted on genome-wide association studies (GWASs)-based datasets to evaluate the causal relationship between FGF23, α-Klotho, FGFR1 and OP. The primary endpoint was the odds ratio (OR) of the inverse-variance weighted (IVW) approach. Furthermore, we stably transfected FGF23-mimic or siRNA-FGF23 into human bone marrow mesenchymal stem cells (hBMSCs) in culture and determined its cell proliferation and the effects on osteogenic differentiation. Using MR analysis, we demonstrated a strong correlation between serum FGF23 levels and Heel- and femoral neck-BMDs, with subsequent ORs of 0.919 (95% CI: 0.860-0.983, p = 0.014) and 0.751 (95% CI: 0.587-0.962; p = 0.023), respectively. The expression levels of FGF23 were significantly increased in femoral neck of patients with OP than in the control cohort (p < 0.0001). Based on our in vitro investigation, after overexpression of FGF23, compared to the control group, the BMSC's proliferation ability decreased, the expression level of key osteogenic differentiation genes (RUNX2, OCN and OSX) significantly reduced, mineralized nodules and ALP activity significantly decreased. After silencing FGF23, it showed a completely opposite trend. Augmented FGF23 levels are causally associated with increased risk of OP. Similarly, FGF23 overexpression strongly inhibits the osteogenic differentiation of hBMSCs, thereby potentially aggravating the pathological process of OP.
Collapse
Affiliation(s)
- Xiang Zhang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of EndocrinologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Shandong Key Laboratory of Endocrinology and Lipid MetabolismJinanShandongChina
- Shandong Institute of Endocrine and Metabolic DiseasesJinanShandongChina
| | - Jin Xu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of EndocrinologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Shandong Key Laboratory of Endocrinology and Lipid MetabolismJinanShandongChina
- Shandong Institute of Endocrine and Metabolic DiseasesJinanShandongChina
- “Chuangxin China” Innovation Base of stem cell and Gene Therapy for endocrine Metabolic diseasesJinanShandongChina
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic DiseasesJinanShandongChina
- Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic DiseasesJinanShandongChina
| |
Collapse
|
4
|
唐 帅, 杨 扬, 李 湘, 别 炳, 张 建. [Clinical study on growth impairment induced by oral glucocorticoids based on FGF23/Klotho homeostasis observations]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:269-274. [PMID: 38557379 PMCID: PMC10986375 DOI: 10.7499/j.issn.1008-8830.2309160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 02/02/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVES To observe the correlation between growth impairment induced by long-term oral glucocorticoids (GC) therapy and the ratio of FGF23/Klotho in children with primary nephrotic syndrome (PNS). METHODS A prospective study was conducted on 56 children with GC-sensitive PNS who had discontinued GC therapy for more than 3 months and revisited the Department of Pediatrics of the First Affiliated Hospital of Henan University of Traditional Chinese Medicine between June 2022 and December 2022. After monitoring qualitative and quantitative urine protein levels upon admission, the children with proteinuria relapse were treated with GC (GC group; n=29), while those without relapse did not receive GC treatment (non-GC group; n=27). In addition, 29 healthy children aged 3 to prepuberty were selected as the control group. Height, bone age, growth rate, and the FGF23/Klotho ratio were compared among the groups. The correlations of the FGF23/Klotho ratio with height, bone age, and growth rate were analyzed. RESULTS The FGF23/Klotho ratio in the GC group was significantly higher than that in the non-GC group after 1 month of GC therapy (P<0.05), and the height and bone age growth rates within 6 months were lower than those in the non-GC group (P<0.05). Correlation analysis showed significant negative correlations between the FGF23/Klotho ratio after 1 month of treatment and the growth rates of height and bone age within 6 months in children with PNS (r=-0.356 and -0.436, respectively; P<0.05). CONCLUSIONS The disturbance in FGF23/Klotho homeostasis is one of the mechanisms underlying the growth impairment caused by long-term oral GC therapy.
Collapse
Affiliation(s)
| | | | | | | | - 建 张
- 河南中医药大学第一附属医院儿科医院,河南郑州450000
| |
Collapse
|
5
|
Xue M, Huang N, Luo Y, Yang X, Wang Y, Fang M. Combined Transcriptomics and Metabolomics Identify Regulatory Mechanisms of Porcine Vertebral Chondrocyte Development In Vitro. Int J Mol Sci 2024; 25:1189. [PMID: 38256262 PMCID: PMC10816887 DOI: 10.3390/ijms25021189] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Porcine body length is closely related to meat production, growth, and reproductive performance, thus playing a key role in the profitability of the pork industry. Cartilage development is critical to longitudinal elongation of individual vertebrae. This study isolated primary porcine vertebral chondrocytes (PVCs) to clarify the complex mechanisms of elongation. We used transcriptome and target energy metabolome technologies to confirm crucial genes and metabolites in primary PVCs at different differentiation stages (0, 4, 8, and 12 days). Pairwise comparisons of the four stages identified 4566 differentially expressed genes (DEGs). Time-series gene cluster and functional analyses of these DEGs revealed four clusters related to metabolic processes, cartilage development, vascular development, and cell cycle regulation. We constructed a transcriptional regulatory network determining chondrocyte maturation. The network indicated that significantly enriched transcription factor (TF) families, including zf-C2H2, homeobox, TF_bZIP, and RHD, are important in cell cycle and differentiation processes. Further, dynamic network biomarker (DNB) analysis revealed that day 4 was the tipping point for chondrocyte development, consistent with morphological and metabolic changes. We found 24 DNB DEGs, including the TFs NFATC2 and SP7. Targeted energy metabolome analysis showed that most metabolites were elevated throughout chondrocyte development; notably, 16 differentially regulated metabolites (DRMs) were increased at three time points after cell differentiation. In conclusion, integrated metabolome and transcriptome analyses highlighted the importance of amino acid biosynthesis in chondrocyte development, with coordinated regulation of DEGs and DRMs promoting PVC differentiation via glucose oxidation. These findings reveal the regulatory mechanisms underlying PVC development and provide an important theoretical reference for improving pork production.
Collapse
Affiliation(s)
- Mingming Xue
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, Frontiers Science Center for Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (M.X.); (Y.L.); (X.Y.)
| | - Ning Huang
- Sanya Research Institute, China Agricultural University, Sanya 572025, China; (N.H.); (Y.W.)
| | - Yabiao Luo
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, Frontiers Science Center for Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (M.X.); (Y.L.); (X.Y.)
| | - Xiaoyang Yang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, Frontiers Science Center for Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (M.X.); (Y.L.); (X.Y.)
| | - Yubei Wang
- Sanya Research Institute, China Agricultural University, Sanya 572025, China; (N.H.); (Y.W.)
| | - Meiying Fang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Beijing Key Laboratory for Animal Genetic Improvement, State Key Laboratory of Animal Biotech Breeding, Frontiers Science Center for Molecular Design Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (M.X.); (Y.L.); (X.Y.)
- Sanya Research Institute, China Agricultural University, Sanya 572025, China; (N.H.); (Y.W.)
| |
Collapse
|
6
|
Sun T, Yu X. FGF23 Actions in CKD-MBD and other Organs During CKD. Curr Med Chem 2023; 30:841-856. [PMID: 35761503 DOI: 10.2174/0929867329666220627122733] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/26/2022] [Accepted: 04/12/2022] [Indexed: 02/08/2023]
Abstract
Fibroblast growth factor 23 (FGF23) is a new endocrine product discovered in the past decade. In addition to being related to bone diseases, it has also been found to be related to kidney metabolism and parathyroid metabolism, especially as a biomarker and a key factor to be used in kidney diseases. FGF23 is upregulated as early as the second and third stages of chronic kidney disease (CKD) in response to relative phosphorus overload. The early rise of FGF23 has a protective effect on the body and is essential for maintaining phosphate balance. However, with the decline in renal function, eGFR (estimated glomerular filtration rate) declines, and the phosphorus excretion effect caused by FGF23 is weakened. It eventually leads to a variety of complications, such as bone disease (Chronic Kidney Disease-Mineral and Bone Metabolism Disorder), vascular calcification (VC), and more. Monoclonal antibodies against FGF23 are currently used to treat genetic diseases with increased FGF23. CKD is also a state of increased FGF23. This article reviews the current role of FGF23 in CKD and discusses the crosstalk between various organs under CKD conditions and FGF23. Studying the effect of hyperphosphatemia on different organs of CKD is important. The prospect of FGF23 for therapy is also discussed.
Collapse
Affiliation(s)
- Ting Sun
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, Rare Disease Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, Rare Disease Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
7
|
André J, Zhukouskaya VV, Lambert AS, Salles JP, Mignot B, Bardet C, Chaussain C, Rothenbuhler A, Linglart A. Growth hormone treatment improves final height in children with X-linked hypophosphatemia. Orphanet J Rare Dis 2022; 17:444. [PMID: 36544157 PMCID: PMC9768884 DOI: 10.1186/s13023-022-02590-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND/AIM Despite optimal conventional treatment (oral phosphate supplements and active vitamin D analogs), about 40-50% of children with well-controlled X-linked hypophosphatemia (XLH) show linear growth failure, making them less likely to achieve an acceptable final height. Here, we studied the hypothesis that rhGH treatment improves final height in children with XLH and growth failure. METHODS Two cohorts of children with XLH were included in this retrospective longitudinal analysis: (1) a cohort treated with rhGH for short stature (n = 34) and (2) a cohort not treated with rhGH (n = 29). The mean duration of rhGH treatment was 4.4 ± 2.9 years. We collected the auxological parameters at various time points during follow-up until final height. RESULTS In rhGH-treated children, 2 years of rhGH therapy was associated with a significant increase in height from - 2.4 ± 0.9 to - 1.5 ± 0.7 SDS (p < 0.001). Their mean height at rhGH discontinuation was - 1.2 ± 0.9 SDS and at final height was - 1.3 ± 0.9 SDS corresponding to 165.5 ± 6.4 cm in boys and 155.5 ± 6.3 cm in girls. Notably, the two groups had similar final heights; i.e., the final height in children not treated with rhGH being - 1.2 ± 1.1 SDS (165.4 ± 6.8 cm in boys and 153.7 ± 7.8 cm in girls), p = 0.7. CONCLUSION Treatment with rhGH permits to improve final height in children with XLH and growth failure, despite optimal conventional treatment. We propose therefore that rhGH therapy could be considered as an option for short stature in the context of XLH.
Collapse
Affiliation(s)
- Julia André
- grid.413784.d0000 0001 2181 7253AP-HP, Endocrinology and Diabetes for Children, Reference Center for Rare Diseases of Calcium and Phosphate Metabolism, DMU SEA, OSCAR Filière, EndoRare and BOND ERN, Bicêtre Paris Saclay Hospital, 78 Rue du Général Leclerc, 94270 Le Kremlin Bicêtre, France
| | - Volha V. Zhukouskaya
- grid.413784.d0000 0001 2181 7253AP-HP, Endocrinology and Diabetes for Children, Reference Center for Rare Diseases of Calcium and Phosphate Metabolism, DMU SEA, OSCAR Filière, EndoRare and BOND ERN, Bicêtre Paris Saclay Hospital, 78 Rue du Général Leclerc, 94270 Le Kremlin Bicêtre, France ,grid.508487.60000 0004 7885 7602Laboratory Orofacial Pathologies, Imaging and Biotherapies URP2496 and FHU-DDS-Net, Dental School, and Plateforme d’Imagerie du Vivant (PIV), Université Paris Cité, Montrouge, France
| | - Anne-Sophie Lambert
- grid.413784.d0000 0001 2181 7253AP-HP, Endocrinology and Diabetes for Children, Reference Center for Rare Diseases of Calcium and Phosphate Metabolism, DMU SEA, OSCAR Filière, EndoRare and BOND ERN, Bicêtre Paris Saclay Hospital, 78 Rue du Général Leclerc, 94270 Le Kremlin Bicêtre, France ,grid.413784.d0000 0001 2181 7253AP-HP, Medicine for Adolescents, Bicêtre Paris Saclay Hospital, Le Kremlin Bicêtre, France
| | - Jean-Pierre Salles
- grid.508721.9Unit of Endocrinology and Bone Diseases, Children Hospital, Toulouse University Hospital, CHU de Toulouse, Université de Toulouse, ERN BOND, INSERM UMR 1291/CNRS 5051, INFINITY Center, Toulouse, France
| | - Brigitte Mignot
- grid.411158.80000 0004 0638 9213Department of Pediatrics, CHU of Besancon, Besançon, France
| | - Claire Bardet
- grid.508487.60000 0004 7885 7602Laboratory Orofacial Pathologies, Imaging and Biotherapies URP2496 and FHU-DDS-Net, Dental School, and Plateforme d’Imagerie du Vivant (PIV), Université Paris Cité, Montrouge, France
| | - Catherine Chaussain
- grid.413784.d0000 0001 2181 7253AP-HP, Endocrinology and Diabetes for Children, Reference Center for Rare Diseases of Calcium and Phosphate Metabolism, DMU SEA, OSCAR Filière, EndoRare and BOND ERN, Bicêtre Paris Saclay Hospital, 78 Rue du Général Leclerc, 94270 Le Kremlin Bicêtre, France ,grid.508487.60000 0004 7885 7602Laboratory Orofacial Pathologies, Imaging and Biotherapies URP2496 and FHU-DDS-Net, Dental School, and Plateforme d’Imagerie du Vivant (PIV), Université Paris Cité, Montrouge, France ,grid.50550.350000 0001 2175 4109AP-HP, Reference Center for Rare Disorders of the Calcium and Phosphate Metabolism, Dental Medicine Department, Bretonneau Hospital, GHN-Universite de Paris, Paris, France
| | - Anya Rothenbuhler
- grid.413784.d0000 0001 2181 7253AP-HP, Endocrinology and Diabetes for Children, Reference Center for Rare Diseases of Calcium and Phosphate Metabolism, DMU SEA, OSCAR Filière, EndoRare and BOND ERN, Bicêtre Paris Saclay Hospital, 78 Rue du Général Leclerc, 94270 Le Kremlin Bicêtre, France
| | - Agnès Linglart
- grid.413784.d0000 0001 2181 7253AP-HP, Endocrinology and Diabetes for Children, Reference Center for Rare Diseases of Calcium and Phosphate Metabolism, DMU SEA, OSCAR Filière, EndoRare and BOND ERN, Bicêtre Paris Saclay Hospital, 78 Rue du Général Leclerc, 94270 Le Kremlin Bicêtre, France ,grid.460789.40000 0004 4910 6535INSERM, Physiologie Et Physiopathologie Endocrinienne, Bicêtre Paris Saclay Hospital, Paris Saclay University, Le Kremlin Bicêtre, France
| |
Collapse
|
8
|
Chondrocyte Hypertrophy in Osteoarthritis: Mechanistic Studies and Models for the Identification of New Therapeutic Strategies. Cells 2022; 11:cells11244034. [PMID: 36552796 PMCID: PMC9777397 DOI: 10.3390/cells11244034] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/08/2022] [Indexed: 12/16/2022] Open
Abstract
Articular cartilage shows limited self-healing ability owing to its low cellularity and avascularity. Untreated cartilage defects display an increased propensity to degenerate, leading to osteoarthritis (OA). During OA progression, articular chondrocytes are subjected to significant alterations in gene expression and phenotype, including a shift towards a hypertrophic-like state (with the expression of collagen type X, matrix metalloproteinases-13, and alkaline phosphatase) analogous to what eventuates during endochondral ossification. Present OA management strategies focus, however, exclusively on cartilage inflammation and degradation. A better understanding of the hypertrophic chondrocyte phenotype in OA might give new insights into its pathogenesis, suggesting potential disease-modifying therapeutic approaches. Recent developments in the field of cellular/molecular biology and tissue engineering proceeded in the direction of contrasting the onset of this hypertrophic phenotype, but knowledge gaps in the cause-effect of these processes are still present. In this review we will highlight the possible advantages and drawbacks of using this approach as a therapeutic strategy while focusing on the experimental models necessary for a better understanding of the phenomenon. Specifically, we will discuss in brief the cellular signaling pathways associated with the onset of a hypertrophic phenotype in chondrocytes during the progression of OA and will analyze in depth the advantages and disadvantages of various models that have been used to mimic it. Afterwards, we will present the strategies developed and proposed to impede chondrocyte hypertrophy and cartilage matrix mineralization/calcification. Finally, we will examine the future perspectives of OA therapeutic strategies.
Collapse
|
9
|
Understanding the Stony Bridge between Osteoporosis and Vascular Calcification: Impact of the FGF23/Klotho axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7536614. [PMID: 34539972 PMCID: PMC8448600 DOI: 10.1155/2021/7536614] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 08/07/2021] [Indexed: 12/11/2022]
Abstract
A relationship between osteoporosis (OP) and vascular calcification (VC) is now proposed. There are common mechanisms underlying the regulation of them. Fibroblast growth factor- (FGF-) 23 and Klotho are hormones associated with the metabolic axis of osteovascular metabolism. Most recently, it was suggested that the FGF23-klotho axis is associated with increasing incidence of fractures and is potentially involved in the progression of the aortic-brachial stiffness ratio. Herein, we discussed the potential role of the FGF23/Klotho axis in the pathophysiology of OP and VC. We want to provide an update review in order to allow a better understanding of the potential role of the FGF23/Klotho axis in comorbidity of OP and VC. We believe that a better understanding of the relationship between both entities can help in proposing new therapeutic targets for reducing the increasing prevalence of OP and VC in the aging population.
Collapse
|
10
|
Qamar H, Perumal N, Papp E, Gernand AD, Al Mahmud A, Roth DE. Higher maternal parathyroid hormone concentration at delivery is not associated with smaller newborn size. Endocr Connect 2021; 10:345-357. [PMID: 33640873 PMCID: PMC8052570 DOI: 10.1530/ec-21-0056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 02/23/2021] [Indexed: 11/08/2022]
Abstract
Intrauterine growth restriction (IUGR) reflects inadequate growth in-utero and is prevalent in low resource settings. This study aimed to assess the association of maternal delivery parathyroid hormone (PTH) - a regulator of bone turnover and calcium homeostasis - with newborn anthropometry, to identify regulators of PTH, and to delineate pathways by which maternal PTH regulates birth size using path analysis. This was a cross-sectional analysis of data from participants (n = 537) enrolled in the Maternal Vitamin D for Infant Growth trial in Dhaka, Bangladesh. Primary exposures were maternal delivery intact PTH (iPTH) or whole PTH (wPTH) and outcomes were gestational age- and sex-standardized z-scores for birth length (LAZ), weight (WAZ), and head circumference (HCAZ). Hypothesized regulators of PTH included calcium and protein intake, vitamin D, magnesium, fibroblast-like growth factor-23 (FGF23), and C-reactive protein. Maternal iPTH was not associated with birth size in linear regression analyses; however, in path analysis models, every SD increase in log(iPTH) was associated with 0.08SD (95% CI: 0.002, 0.162) higher LAZ. In linear regression and path analysis models, wPTH was positively associated with WAZ. Vitamin D suppressed PTH, while FGF23 was positively associated with PTH. In path analysis models, higher magnesium was negatively associated with LAZ; FGF23 was positively associated and protein intake was negatively associated with LAZ, WAZ, and HCAZ. Higher maternal PTH in late pregnancy is unlikely to contribute to IUGR. Future studies should investigate maternal FGF23, magnesium and protein intake as regulators of fetal growth, particularly in settings where food insecurity and IUGR are public health problems.
Collapse
Affiliation(s)
- Huma Qamar
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Nandita Perumal
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Eszter Papp
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Alison D Gernand
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Abdullah Al Mahmud
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research (icddr,b), Dhaka, Bangladesh
| | - Daniel E Roth
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
- Correspondence should be addressed to D E Roth:
| |
Collapse
|
11
|
Muñoz-Castañeda JR, Rodelo-Haad C, Pendon-Ruiz de Mier MV, Martin-Malo A, Santamaria R, Rodriguez M. Klotho/FGF23 and Wnt Signaling as Important Players in the Comorbidities Associated with Chronic Kidney Disease. Toxins (Basel) 2020; 12:E185. [PMID: 32188018 PMCID: PMC7150840 DOI: 10.3390/toxins12030185] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/05/2020] [Accepted: 03/11/2020] [Indexed: 12/31/2022] Open
Abstract
Fibroblast Growth Factor 23 (FGF23) and Klotho play an essential role in the regulation of mineral metabolism, and both are altered as a consequence of renal failure. FGF23 increases to augment phosphaturia, which prevents phosphate accumulation at the early stages of chronic kidney disease (CKD). This effect of FGF23 requires the presence of Klotho in the renal tubules. However, Klotho expression is reduced as soon as renal function is starting to fail to generate a state of FGF23 resistance. Changes in these proteins directly affect to other mineral metabolism parameters; they may affect renal function and can produce damage in other organs such as bone, heart, or vessels. Some of the mechanisms responsible for the changes in FGF23 and Klotho levels are related to modifications in the Wnt signaling. This review examines the link between FGF23/Klotho and Wnt/β-catenin in different organs: kidney, heart, and bone. Activation of the canonical Wnt signaling produces changes in FGF23 and Klotho and vice versa; therefore, this pathway emerges as a potential therapeutic target that may help to prevent CKD-associated complications.
Collapse
Affiliation(s)
- Juan Rafael Muñoz-Castañeda
- Maimonides Institute for Biomedical Research (IMIBIC), 14005 Cordoba, Spain; (J.R.M.-C.); (C.R.-H.); (A.M.-M.); (R.S.); (M.R.)
- School of Medicine, Department of Medicine, University of Cordoba, 14005 Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, 14005 Cordoba, Spain
- Spanish Renal Research Network (REDinREN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Cristian Rodelo-Haad
- Maimonides Institute for Biomedical Research (IMIBIC), 14005 Cordoba, Spain; (J.R.M.-C.); (C.R.-H.); (A.M.-M.); (R.S.); (M.R.)
- School of Medicine, Department of Medicine, University of Cordoba, 14005 Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, 14005 Cordoba, Spain
- Spanish Renal Research Network (REDinREN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Maria Victoria Pendon-Ruiz de Mier
- Maimonides Institute for Biomedical Research (IMIBIC), 14005 Cordoba, Spain; (J.R.M.-C.); (C.R.-H.); (A.M.-M.); (R.S.); (M.R.)
- School of Medicine, Department of Medicine, University of Cordoba, 14005 Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, 14005 Cordoba, Spain
- Spanish Renal Research Network (REDinREN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Alejandro Martin-Malo
- Maimonides Institute for Biomedical Research (IMIBIC), 14005 Cordoba, Spain; (J.R.M.-C.); (C.R.-H.); (A.M.-M.); (R.S.); (M.R.)
- School of Medicine, Department of Medicine, University of Cordoba, 14005 Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, 14005 Cordoba, Spain
- Spanish Renal Research Network (REDinREN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Rafael Santamaria
- Maimonides Institute for Biomedical Research (IMIBIC), 14005 Cordoba, Spain; (J.R.M.-C.); (C.R.-H.); (A.M.-M.); (R.S.); (M.R.)
- School of Medicine, Department of Medicine, University of Cordoba, 14005 Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, 14005 Cordoba, Spain
- Spanish Renal Research Network (REDinREN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Mariano Rodriguez
- Maimonides Institute for Biomedical Research (IMIBIC), 14005 Cordoba, Spain; (J.R.M.-C.); (C.R.-H.); (A.M.-M.); (R.S.); (M.R.)
- School of Medicine, Department of Medicine, University of Cordoba, 14005 Cordoba, Spain
- Nephrology Service, Reina Sofia University Hospital, 14005 Cordoba, Spain
- Spanish Renal Research Network (REDinREN), Institute of Health Carlos III, 28029 Madrid, Spain
| |
Collapse
|
12
|
Bacchetta J, Bardet C, Prié D. Physiology of FGF23 and overview of genetic diseases associated with renal phosphate wasting. Metabolism 2020; 103S:153865. [PMID: 30664852 DOI: 10.1016/j.metabol.2019.01.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/08/2019] [Accepted: 01/16/2019] [Indexed: 12/31/2022]
Abstract
Phosphate is a cornerstone of several physiological pathways including skeletal development, bone mineralization, membrane composition, nucleotide structure, maintenance of plasma pH, and cellular signaling. The kidneys have a key role in phosphate homeostasis with three hormones having important functions in renal phosphate handling or intestinal absorption: parathyroid hormone (PTH), fibroblast growth factor 23 (FGF23), and 1-25-dihydroxyvitamin D (1,25(OH)2D). FGF23 is mainly synthesized by osteocytes; it is a direct phosphaturic factor that also inhibits 1,25(OH)2D and PTH. In addition to crucial effects on phosphate and calcium metabolism, FGF23 also has 'off-target' effects notably on the cardiovascular, immune and central nervous systems. Genetic diseases may affect the FGF23 pathway, resulting in either increased FGF23 levels leading to hypophosphatemia (such as in X-linked hypophosphatemia) or defective secretion/action of intact FGF23 inducing hyperphosphatemia (such as in familial tumoral calcinosis). The aim of this review is to provide an overview of FGF23 physiology and pathophysiology in X-linked hypophosphatemia, with a focus on FGF23-associated genetic diseases.
Collapse
Affiliation(s)
- Justine Bacchetta
- Reference Center for Rare Renal Disorders, Reference Center for Rare Disorders of Calcium and Phosphate Metabolism, Department of Pediatric Nephrology, Rheumatology and Dermatology, Femme Mère Enfant Hospital, Bron Cedex, France; Lyon-Est Medical School, Lyon 1 University, Lyon, France; INSERM 1033, LYOS, Bone Disorders Prevention, Lyon, France.
| | - Claire Bardet
- Paris Descartes University, EA2496, Faculty of Dental Surgery, Montrouge, France
| | - Dominique Prié
- Paris Descartes University of Medicine, Necker-Enfants Malades Institute, INSERM U1151, France; Functional Exploration Department, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| |
Collapse
|
13
|
Raimann A, Mindler GT, Kocijan R, Bekes K, Zwerina J, Haeusler G, Ganger R. Multidisciplinary patient care in X-linked hypophosphatemic rickets: one challenge, many perspectives. Wien Med Wochenschr 2020; 170:116-123. [PMID: 31993875 PMCID: PMC7098922 DOI: 10.1007/s10354-019-00732-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/17/2019] [Indexed: 11/29/2022]
Abstract
X‑linked hypophosphatemic rickets (XLH, OMIM #307800) is a rare genetic metabolic disorder caused by dysregulation of fibroblast-like growth factor 23 (FGF23) leading to profound reduction in renal phosphate reabsorption. Impaired growth, severe rickets and complex skeletal deformities are direct consequences of hypophosphatemia representing major symptoms of XLH during childhood. In adults, secondary complications including early development of osteoarthritis substantially impair quality of life and cause significant clinical burden. With the global approval of the monoclonal FGF23 antibody burosumab, a targeted treatment with promising results in phase III studies is available for children with XLH. Nevertheless, complete phenotypic rescue is rarely achieved and remaining multisystemic symptoms demand multidisciplinary specialist care. Coordination of patient management within the major medical disciplines is a mainstay to optimize treatment and reduce disease burden. This review aims to depict different perspectives in XLH patient care in the setting of a multidisciplinary centre of expertise for rare bone diseases.
Collapse
Affiliation(s)
- Adalbert Raimann
- Comprehensive Center for Pediatrics, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Pulmonology, Allergology and Endocrinology, Medical University of Vienna, Vienna, Austria. .,Vienna Bone and Growth Centre, Vienna, Austria.
| | - Gabriel T Mindler
- Department of Pediatric Orthopaedics, Orthopaedic Hospital Speising, Vienna, Austria.,Vienna Bone and Growth Centre, Vienna, Austria
| | - Roland Kocijan
- Hanusch Hospital of the WGKK and AUVA Trauma Center, 1st Medical Department at Hanusch Hospital, Ludwig Boltzmann Institute of Osteology, Vienna, Austria.,Vienna Bone and Growth Centre, Vienna, Austria
| | - Katrin Bekes
- Department of Pediatric Dentistry, School of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Jochen Zwerina
- Hanusch Hospital of the WGKK and AUVA Trauma Center, 1st Medical Department at Hanusch Hospital, Ludwig Boltzmann Institute of Osteology, Vienna, Austria.,Vienna Bone and Growth Centre, Vienna, Austria
| | - Gabriele Haeusler
- Comprehensive Center for Pediatrics, Department of Pediatrics and Adolescent Medicine, Division of Pediatric Pulmonology, Allergology and Endocrinology, Medical University of Vienna, Vienna, Austria.,Vienna Bone and Growth Centre, Vienna, Austria
| | - Rudolf Ganger
- Department of Pediatric Orthopaedics, Orthopaedic Hospital Speising, Vienna, Austria.,Vienna Bone and Growth Centre, Vienna, Austria
| |
Collapse
|
14
|
Delucchi Á, Toro L, Alzamora R, Barrientos V, González M, Andaur R, León P, Villanueva F, Galindo M, Las Heras F, Montecino M, Moena D, Lazcano A, Pinto V, Salas P, Reyes ML, Mericq V, Michea L. Glucocorticoids Decrease Longitudinal Bone Growth in Pediatric Kidney Transplant Recipients by Stimulating the FGF23/FGFR3 Signaling Pathway. J Bone Miner Res 2019; 34:1851-1861. [PMID: 31099911 DOI: 10.1002/jbmr.3761] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 05/05/2019] [Accepted: 05/11/2019] [Indexed: 12/11/2022]
Abstract
Renal transplantation (RTx) is an effective therapy to improve clinical outcomes in pediatric patients with terminal chronic kidney disease. However, chronic immunosuppression with glucocorticoids (GCs) reduces bone growth and BMD. The mechanisms causing GC-induced growth impairment have not been fully clarified. Fibroblast growth factor 23 (FGF23) is a peptide hormone that regulates phosphate homeostasis and bone growth. In pathological conditions, FGF23 excess or abnormal FGF receptors (FGFR) activity leads to bone growth impairment. Experimental data indicate that FGF23 expression is induced by chronic GC exposure. Therefore, we hypothesize that GCs impair bone growth by increasing FGF23 expression, which has direct effects on bone growth plate. In a post hoc analysis of a multicentric randomized clinical trial of prepubertal RTx children treated with early GC withdrawal or chronic GC treatment, we observed that GC withdrawal was associated with improvement in longitudinal growth and BMD, and lower plasma FGF23 levels as compared with a chronic GC group. In prepubertal rats, GC-induced bone growth retardation correlated with increased plasma FGF23 and bone FGF23 expression. Additionally, GC treatment decreased FGFR1 expression whereas it increased FGFR3 expression in mouse tibia explants. The GC-induced bone growth impairment in tibiae explants was prevented by blockade of FGF23 receptors using either a pan-FGFR antagonist (PD173074), a C-terminal FGF23 peptide (FGF23180-205) which blocks the binding of FGF23 to the FGFR-Klotho complex or a specific FGFR3 antagonist (P3). Finally, local administration of PD173074 into the tibia growth plate ameliorated cartilage growth impairment in GC-treated rats. These results show that GC treatment partially reduces longitudinal bone growth via upregulation of FGF23 and FGFR3 expression, thus suggesting that the FGF23/Klotho/FGFR3 axis at the growth plate could be a potential therapeutic target for the management of GC-induced growth impairment in children.
Collapse
Affiliation(s)
- Ángela Delucchi
- Division of Nephrology, Hospital Luis Calvo Mackenna, Santiago, Chile.,Division of Nephrology, Clínica Alemana de Santiago, Santiago, Chile
| | - Luis Toro
- Division of Nephrology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile.,Centro de Investigación Clínica Avanzada, Hospital Clínico Universidad de Chile, Santiago, Chile.,Clinica Las Condes, Santiago, Chile
| | - Rodrigo Alzamora
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Victor Barrientos
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Magdalena González
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Rodrigo Andaur
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Pablo León
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Francisco Villanueva
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mario Galindo
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy (MIII), Santiago, Chile
| | - Facundo Las Heras
- Clinica Las Condes, Santiago, Chile.,Department of Anatomic Pathology, Hospital Clínico Universidad de Chile, Santiago, Chile
| | - Martín Montecino
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andrés Bello, Santiago, Chile.,FONDAP Center for Genome Regulation, Universidad Andres Bello, Santiago, Chile
| | - Daniel Moena
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andrés Bello, Santiago, Chile.,FONDAP Center for Genome Regulation, Universidad Andres Bello, Santiago, Chile
| | - Andrea Lazcano
- Division of Nephrology, Clínica Alemana de Santiago, Santiago, Chile.,Division of Nephrology, Hospital de Niños Roberto del Río, Santiago, Chile
| | - Viola Pinto
- Clinica Las Condes, Santiago, Chile.,Pediatric Nephrology Unit, Hospital Doctor Exequiel González Cortés, Santiago, Chile
| | - Paulina Salas
- Pediatric Nephrology Unit, Hospital Doctor Exequiel González Cortés, Santiago, Chile
| | - María Loreto Reyes
- Pediatric Endocrinology Unit, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Verónica Mericq
- Clinica Las Condes, Santiago, Chile.,Institute of Maternal and Child Research, Universidad de Chile, Santiago, Chile
| | - Luis Michea
- Division of Nephrology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile.,Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy (MIII), Santiago, Chile
| |
Collapse
|
15
|
Ectopic expression of Klotho in fibroblast growth factor 23 (FGF23)-producing tumors that cause tumor-induced rickets/osteomalacia (TIO). Bone Rep 2018; 10:100192. [PMID: 30627598 PMCID: PMC6321977 DOI: 10.1016/j.bonr.2018.100192] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/28/2018] [Accepted: 12/27/2018] [Indexed: 02/03/2023] Open
Abstract
Tumor-induced rickets/osteomalacia (TIO) is a rare paraneoplastic syndrome caused by tumors that ectopically express fibroblast growth factor 23 (FGF23). FGF23 is a bone-derived hormone that regulates serum phosphate concentrations. Patients with TIO develop hypophosphatemic rickets/osteomalacia due to FGF23 excess and suffer from symptoms such as leg deformities, bone pain, skeletal muscle myopathy, and multiple fractures/pseudofractures. Usually, successful surgical removal of the causative tumors normalizes serum FGF23 and phosphate concentrations in patients with TIO. Most FGF23-producing tumors associated with TIO are histologically called phosphaturic mesenchymal tumor, mixed connective tissue variant (PMTMCT). The precise mechanism by which these tumors ectopically overproduce FGF23 outside of bone is yet to be clarified. Therefore, we performed an RNA sequencing analysis of a PMTMCT that was found in the left parotid gland of a patient with TIO. Among the upregulated genes, we focused on Klotho, the protein product of which is a single pass transmembrane protein that works along with an FGF receptor 1c as a receptor complex for FGF23. Subsequent histological analysis confirmed the ectopic expression of Klotho in other PMTMCTs. From these results, we assume that the ectopic expression of Klotho in PTMMCTs enables a positive feedback loop in FGF23 production via the activation of FGF receptor 1c and exacerbates disease manifestations in TIO. Klotho is ectopically expressed in the FGF23-producing mesenchymal tumors. Klotho enables the activation of the FGFR signaling pathway in PMTMCTs. Klotho enables the autocrine/paracrine effects of FGF23 in PMTMCTs.
Collapse
|
16
|
Wu Y, Zhang J, Chen F. The effects of elevated fibroblast growth factor 23 on mandibular growth in rats. Arch Oral Biol 2018; 95:156-164. [PMID: 30121527 DOI: 10.1016/j.archoralbio.2018.07.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/28/2018] [Accepted: 07/30/2018] [Indexed: 12/25/2022]
Abstract
OBJECTIVE The aim of this study is to elucidate the local effects of fibroblast growth factor 23 (FGF23) in on mandibular condylar growth in growing rats. DESIGN Growing Sprague-Dawley rats received intra-temporomandibular joint injections of phosphate buffer solution (PBS), adenovirus-mediated green fluorescent protein (Ad-GFP) or adenovirus-mediated fibroblast growth factor 23 (Ad-FGF23), which were marked as groups A, B, and C, respectively. In vitro, we treated rat mandibular cartilage chondrocytes with PBS, Ad-GFP, and Ad-FGF23. RESULTS The mandibular condyles in group C grew smaller sizes than those in the other control groups due to significant differences among the experimental and control groups with the value of C-D, Q-R (P ≤ 0.05), accompanied by diminished bone mass of sub-cartilage condyles via micro CT analysis. Histologically, the length of the hypertrophic zone was diminished and was associated with decreasing chondrocyte proliferation in group C. Quantitative real-time PCR indicated significant decreases in the expression of chondrogenesis marker genes, including Type X collagen (Col X) and SRY-type box 9 (Sox 9). Moreover, elevated Ad-FGF23 suppressed chondrocyte proliferation and the expression of the chondrogenic differentiation markers Col X and Sox 9 of in vitro. CONCLUSIONS Local injection of FGF23 suppressed the development and decreased the bone mass of condyles through the decreasing the formation of condylar cartilage, specifically by regulating condylar cartilage cell viability and chondrogenesis expression.
Collapse
Affiliation(s)
- Yanan Wu
- Department of Orthodontics, School & Hospital of Stomatology, Tongji University, Shanghai, China; Engineering Researching Center of Tooth Restoration and Regeneration, Shanghai 200082, China
| | - Jinkai Zhang
- Department of Orthodontics, School & Hospital of Stomatology, Tongji University, Shanghai, China; Engineering Researching Center of Tooth Restoration and Regeneration, Shanghai 200082, China
| | - Fengshan Chen
- Department of Orthodontics, School & Hospital of Stomatology, Tongji University, Shanghai, China; Engineering Researching Center of Tooth Restoration and Regeneration, Shanghai 200082, China.
| |
Collapse
|
17
|
|
18
|
Yashiro M, Ohya M, Mima T, Ueda Y, Nakashima Y, Kawakami K, Ishizawa Y, Yamamoto S, Kobayashi S, Yano T, Tanaka Y, Okuda K, Sonou T, Shoshihara T, Iwashita Y, Iwashita Y, Tatsuta K, Matoba R, Negi S, Shigematsu T. FGF23 modulates the effects of erythropoietin on gene expression in renal epithelial cells. Int J Nephrol Renovasc Dis 2018; 11:125-136. [PMID: 29670389 PMCID: PMC5894721 DOI: 10.2147/ijnrd.s158422] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background FGF23 plays an important role in calcium–phosphorus metabolism. Other roles of FGF23 have recently been reported, such as commitment to myocardium enlargement and immunological roles in the spleen. In this study, we aimed to identify the roles of FGF23 in the kidneys other than calcium–phosphorus metabolism. Methods DNA microarrays and bioinformatics tools were used to analyze gene expression in mIMCD3 mouse renal tubule cells following treatment with FGF23, erythropoietin and/or an inhibitor of ERK. Results Three protein-coding genes were upregulated and 12 were downregulated in response to FGF23. Following bioinformatics analysis of these genes, PPARγ and STAT3 were identified as candidate transcript factors for mediating their upregulation, and STAT1 as a candidate for mediating their downregulation. Because STAT1 and STAT3 also mediate erythropoietin signaling, we investigated whether FGF23 and erythropoietin might show interactive effects in these cells. Of the 15 genes regulated by FGF23, 11 were upregulated by erythropoietin; 10 of these were downregulated following cotreatment with FGF23. Inhibition of ERK, an intracellular mediator of FGF23, reversed the effects of FGF23. However, FGF23 did not influence STAT1 phosphorylation, suggesting that it impinges on erythropoietin signaling through other mechanisms. Conclusion Our results suggest cross talk between erythropoietin and FGF23 signaling in the regulation of renal epithelial cells.
Collapse
Affiliation(s)
- Mitsuru Yashiro
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Masaki Ohya
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Toru Mima
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Yumi Ueda
- DNA Chip Research Inc., Minato, Japan
| | - Yuri Nakashima
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Kazuki Kawakami
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | | | - Shuto Yamamoto
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Sou Kobayashi
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Takurou Yano
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Yusuke Tanaka
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Kouji Okuda
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Tomohiro Sonou
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | | | - Yuko Iwashita
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Yu Iwashita
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Kouichi Tatsuta
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | | | - Shigeo Negi
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | | |
Collapse
|
19
|
Ripmeester EGJ, Timur UT, Caron MMJ, Welting TJM. Recent Insights into the Contribution of the Changing Hypertrophic Chondrocyte Phenotype in the Development and Progression of Osteoarthritis. Front Bioeng Biotechnol 2018; 6:18. [PMID: 29616218 PMCID: PMC5867295 DOI: 10.3389/fbioe.2018.00018] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/08/2018] [Indexed: 12/31/2022] Open
Abstract
Osteoarthritis (OA) is an extremely prevalent age-related condition. The economic and societal burden due to the cost of symptomatic treatment, inability to work, joint replacement, and rehabilitation is huge and increasing. Currently, there are no effective medical therapies that delay or reverse the pathological manifestations of OA. Current treatment options are, without exception, focused on slowing down progression of the disease to postpone total joint replacement surgery for as long as possible and keeping the associated pain and joint immobility manageable. Alterations in the articular cartilage chondrocyte phenotype might be fundamental in the pathological mechanisms of OA development. In many ways, the changing chondrocyte phenotype in osteoarthritic cartilage resembles the process of endochondral ossification as seen, for instance, in developing growth plates. However, the relative contribution of endochondral ossification to the changing chondrocyte phenotype in the development and progression of OA remains poorly described. In this review, we will discuss the current knowledge regarding the cartilage endochondral phenotypic changes occurring during OA development and progression, as well as the molecular and environmental effectors driving these changes. Understanding how these molecular mechanisms determine the chondrocyte cell fate in OA will be essential in enabling cartilage regenerative approaches in future treatments of OA.
Collapse
Affiliation(s)
- Ellen G J Ripmeester
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Ufuk Tan Timur
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Marjolein M J Caron
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Tim J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| |
Collapse
|
20
|
Xiao L, Du E, Homer-Bouthiette C, Hurley MM. Inhibition of FGFR Signaling Partially Rescues Hypophosphatemic Rickets in HMWFGF2 Tg Male Mice. Endocrinology 2017; 158:3629-3646. [PMID: 28938491 PMCID: PMC5659690 DOI: 10.1210/en.2016-1617] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 08/08/2017] [Indexed: 12/13/2022]
Abstract
Transgenic mice harboring high molecular weight fibroblast growth factor (FGF)2 isoforms (HMWTg) in osteoblast lineage cells phenocopy human X-linked hypophosphatemic rickets (XLH) and Hyp murine model of XLH demonstrating increased FGF23/FGF receptor signaling and hypophosphatemic rickets. Because HMWFGF2 was upregulated in bones of Hyp mice and abnormal FGF receptor (FGFR) signaling is important in XLH, HMWTg mice were used to examine the effect of the FGFR inhibitor NVP-BGJ398, now in clinical trials for cancer therapy, on hypophosphatemic rickets. Short-term treatment with NVP-BGJ398 rescued abnormal FGFR signaling and hypophosphatemia in HMWTg. Long-term treatment with NVP-BGJ398 normalized tail, tibia, and femur length. Four weeks NVP-BGJ398 treatment significantly increased total body bone mineral density (BMD) and bone mineral content (BMC) in HMWTg mice; however, at 8 weeks, total body BMD and BMC was indistinguishable among groups. Micro-computed tomography revealed decreased vertebral bone volume, trabecular number, and increased trabecular spacing, whereas femur trabecular tissue density was increased; however, NVP-BGJ398 rescued defective cortical bone mineralization, increased thickness, reduced porosity, and increased endosteal perimeter and cortical tissue density in HMWTg. NVP-BGJ398 improved femur cancellous bone, cortical bone structure, growth plate, and double labeling in cortical bone and also increased femur trabeculae double labeled surface, mineral apposition rate, bone formation rate, and osteoclast number and surface in HMWTg. The decreased NPT2a protein that is important for renal phosphate excretion was rescued by NVP-BGJ398 treatment. We conclude that NVP-BGJ398 partially rescued hypophosphatemic rickets in HMWTg. However, long-term treatment with NVP-BGJ398 further increased serum FGF23 that could exacerbate the mineralization defect.
Collapse
Affiliation(s)
- Liping Xiao
- Department of Medicine, University of Connecticut School of Medicine, UConn Health, Farmington, Connecticut, 06030-052
| | - Erxia Du
- Department of Medicine, University of Connecticut School of Medicine, UConn Health, Farmington, Connecticut, 06030-052
| | - Collin Homer-Bouthiette
- Department of Medicine, University of Connecticut School of Medicine, UConn Health, Farmington, Connecticut, 06030-052
| | - Marja M. Hurley
- Department of Medicine, University of Connecticut School of Medicine, UConn Health, Farmington, Connecticut, 06030-052
| |
Collapse
|
21
|
Kaludjerovic J, Komaba H, Lanske B. Effects of klotho deletion from bone during chronic kidney disease. Bone 2017; 100:50-55. [PMID: 28232146 PMCID: PMC5474158 DOI: 10.1016/j.bone.2017.02.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 02/17/2017] [Accepted: 02/18/2017] [Indexed: 12/24/2022]
Abstract
Klotho is a type I transmembrane protein that acts as a permissive co-receptor for FGF23 and helps to maintain proper mineral metabolism. Mice carrying a loss-of-function mutation in either the Klotho or Fgf23 gene develop many similar phenotypes including osteoporosis. Based on these observations it was hypothesized that the bone phenotypes in Klotho- and Fgf23-null mice may be mediated through a common signaling pathway. Recent improvements in antibody specificity have shown that osteoblasts and osteocytes, which produce FGF23, also express low amount of membrane Klotho. But, the role of Klotho in bone is still largely unclear. In this review we summarize the literature and show that Klotho has an FGF23 dependent and independent effect in bone.
Collapse
Affiliation(s)
- Jovana Kaludjerovic
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Hirotaka Komaba
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Beate Lanske
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA; Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Olauson H, Mencke R, Hillebrands JL, Larsson TE. Tissue expression and source of circulating αKlotho. Bone 2017; 100:19-35. [PMID: 28323144 DOI: 10.1016/j.bone.2017.03.043] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 12/16/2022]
Abstract
αKlotho (Klotho), a type I transmembrane protein and a coreceptor for Fibroblast Growth Factor-23, was initially thought to be expressed only in a limited number of tissues, most importantly the kidney, parathyroid gland and choroid plexus. Emerging data may suggest a more ubiquitous Klotho expression pattern which has prompted reevaluation of the restricted Klotho paradigm. Herein we systematically review the evidence for Klotho expression in various tissues and cell types in humans and other mammals, and discuss potential reasons behind existing conflicting data. Based on current literature and tissue expression atlases, we propose a classification of tissues into high, intermediate and low/absent Klotho expression. The functional relevance of Klotho in organs with low expression levels remain uncertain and there is currently limited data on a role for membrane-bound Klotho outside the kidney. Finally, we review the evidence for the tissue source of soluble Klotho, and conclude that the kidney is likely to be the principal source of circulating Klotho in physiology.
Collapse
Affiliation(s)
- Hannes Olauson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| | - Rik Mencke
- Division of Pathology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan-Luuk Hillebrands
- Division of Pathology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Tobias E Larsson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
23
|
Quintero-Platt G, González-Reimers E, Rodríguez-Gaspar M, Martín-González C, Pérez-Hernández O, Romero-Acevedo L, Espelosín-Ortega E, Vega-Prieto MJDL, Santolaria-Fernández F. Alpha Klotho and Fibroblast Growth Factor-23 Among Alcoholics. Alcohol Alcohol 2017. [DOI: 10.1093/alcalc/agx041] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
24
|
Guibert M, Gasser A, Kempf H, Bianchi A. Fibroblast-growth factor 23 promotes terminal differentiation of ATDC5 cells. PLoS One 2017; 12:e0174969. [PMID: 28406928 PMCID: PMC5390990 DOI: 10.1371/journal.pone.0174969] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 03/17/2017] [Indexed: 01/01/2023] Open
Abstract
Objectives Fibroblast Growth Factor 23 (FGF23) is well documented as a crucial player in the systemic regulation of phosphate homeostasis. Moreover, loss-of-function experiments have revealed that FGF23 also has a phosphate-independent and local impact on skeletogenesis. Here, we used ATDC5 cell line to investigate the expression of FGF23 and the role it may play locally during the differentiation of these cells. Methods ATDC5 cells were differentiated in the presence of insulin, and treated with recombinant FGF23 (rFGF23), inorganic phosphate (Pi) and/or PD173074, an inhibitor of FGF receptors (FGFRs). The mRNA expressions of FGF23, FGFRs and markers of hypertophy, Col X and MMP13, were determined by qPCR analysis and FGF23 production was assessed by ELISA. FGFR activation was determined by immunoprecipitation and immunoblotting. Results FGF23 mRNA expression and production were increased during ATDC5 differentiation. At D28 in particular, rFGF23 stimulation increased hypertrophic markers expression, as Col X and MMP13, and mineralization. A synergic effect of Pi and rFGF23 stimulation was observed on these markers and on the mineralization process. The use of PD173074, a pan-FGFR inhibitor, decreased terminal differentiation of ATDC5 by preventing rFGF23 pro-hypertrophic effects. Conclusions Altogether, our results provide evidence that FGF23 plays an important role during differentiation of ATDC5 cell line, by promoting both hypertrophy and mineralization.
Collapse
Affiliation(s)
- Mathilde Guibert
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l’Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France
- * E-mail: (AB); (MG)
| | - Adeline Gasser
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l’Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France
| | - Hervé Kempf
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l’Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France
| | - Arnaud Bianchi
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l’Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France
- * E-mail: (AB); (MG)
| |
Collapse
|
25
|
Klotho preservation via histone deacetylase inhibition attenuates chronic kidney disease-associated bone injury in mice. Sci Rep 2017; 7:46195. [PMID: 28387374 PMCID: PMC5384196 DOI: 10.1038/srep46195] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/10/2017] [Indexed: 01/01/2023] Open
Abstract
Bone loss and increased fracture are the devastating outcomes of chronic kidney disease-mineral and bone disorder (CKD-MBD) resulting from Klotho deficit-related mineral disturbance and hyperparathyroidism. Because Klotho down-regulation after renal injury is presumably affected by aberrant histone deacetylase (HDAC) activities, here we assess whether HDAC inhibition prevents Klotho loss and attenuates the CKD-associated bone complication in a mouse model of CKD-MBD. Mice fed adenine-containing diet developed the expected renal damage, a substantial Klotho loss and the deregulated key factors causally affecting bone remodeling, which were accompanied by a marked reduction of bone mineral density. Intriguingly, administration of a potent HDAC inhibitor trichostatin A (TSA) impressively alleviated the Klotho deficit and the observed alterations of serum, kidney and bone. TSA prevented Klotho loss by increasing the promoter-associated histone acetylation, therefore increasing Klotho transcription. More importantly the mice lacking Klotho by siRNA interference largely abolished the TSA protections against the serum and renal abnormalities, and the deranged bone micro-architectures. Thus, our study identified Klotho loss as a key event linking HDAC deregulation to the renal and bone injuries in CKD-MBD mice and demonstrated the therapeutic potentials of endogenous Klotho restoration by HDAC inhibition in treating CKD and the associated extrarenal complications.
Collapse
|
26
|
Kaludjerovic J, Komaba H, Sato T, Erben RG, Baron R, Olauson H, Larsson TE, Lanske B. Klotho expression in long bones regulates FGF23 production during renal failure. FASEB J 2017; 31:2050-2064. [PMID: 28183805 DOI: 10.1096/fj.201601036r] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/17/2017] [Indexed: 12/24/2022]
Abstract
Circulating levels of bone-derived fibroblast growth factor 23 (FGF23) increase early during acute and chronic kidney disease and are associated with adverse outcomes. Membrane-bound Klotho acts as a permissive coreceptor for FGF23, and its expression was recently found in osteoblasts/osteocytes. We hypothesized that Klotho in bone cells is part of an autocrine feedback loop that regulates FGF23 expression during renal failure. Thus, we induced renal failure in mice with targeted deletion of Klotho in long bones. Uremic wild-type (KLfl/fl ) and knockout (Prx1-Cre;KLfl/fl ) mice both responded with reduced body weight, kidney atrophy, hyperphosphatemia, and increased bone turnover. Importantly, long bones of Prx1-Cre;KLfl/fl mice but not their axial skeleton failed to increase FGF23 expression as observed in uremic KLfl/fl mice. Consequently, Prx1-Cre;KLfl/fl mice had significantly lower serum FGF23 and parathyroid hormone levels, and higher renal 1-α-hydroxylase expression, serum 1,25-dihydroxyvitamin D, and calcium levels than KLfl/fl mice. These results were confirmed in two independent models of renal failure, adenine diet induced and 5/6 nephrectomy. Moreover, FGF23-treated bone cells required Klotho to increase FGF23 mRNA and ERK phosphorylation. In summary, our novel findings show that Klotho in bone is crucial for inducing FGF23 production upon renal failure. We propose the presence of an autocrine feedback loop in which Klotho senses the need for FGF23.-Kaludjerovic, J., Komaba, H., Sato, T., Erben, R. G., Baron, R., Olauson, H., Larsson, T. E., Lanske, B. Klotho expression in long bones regulates FGF23 production during renal failure.
Collapse
Affiliation(s)
- Jovana Kaludjerovic
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Hirotaka Komaba
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Tadatoshi Sato
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Reinhold G Erben
- Unit of Physiology, Pathophysiology, and Experimental Endocrinology, University of Veterinary Medicine, Vienna, Austria
| | - Roland Baron
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA.,Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Hannes Olauson
- Division of Renal Medicine, Department of Clinical Sciences, Intervention, and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Tobias E Larsson
- Division of Renal Medicine, Department of Clinical Sciences, Intervention, and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Beate Lanske
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA; .,Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
27
|
Nakashima Y, Mima T, Yashiro M, Sonou T, Ohya M, Masumoto A, Yamanaka S, Koreeda D, Tatsuta K, Hanba Y, Moribata M, Negi S, Shigematsu T. Expression and localization of fibroblast growth factor (FGF)23 and Klotho in the spleen: its physiological and functional implications. Growth Factors 2016; 34:196-202. [PMID: 28095739 DOI: 10.1080/08977194.2016.1273222] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The FGF23-Klotho signaling axis is known to exert anti-aging effects via calcium-phosphorus metabolism. In mice deficient in FGF23-Klotho signaling, however, the number of splenocytes is reduced. FGF23 is expressed in both bone and spleen, with regulation of its production differing in these organs. As FGF23-Klotho signaling may play an immunological role in the spleen, splenocytes in male C57BL/6J mice were assayed for expression of Klotho or FGF23 by flow cytometry and immunohistochemistry. Cells that expressed Klotho included CD45R/B220+ CD21/CD35+ CD1d+ CD43- marginal zone B cells. These cells also expressed FGF receptor 1, indicating that Klotho-positive B cells could respond to FGF23. Plasmacytoid dendritic cells (pDCs) with CD11c+ CD45R/B220+ CD11b- CD8α- were found to produce FGF23. Klotho-positive cells and FGF23-producing cells were present in close proximity to each other, suggesting that FGF23 produced by pDCs may act within a limited area. These findings indicate that FGF23-Klotho signaling could play a biological or immunological role in the spleen.
Collapse
Affiliation(s)
- Yuri Nakashima
- a Department of Nephrology , Wakayama Medical University , Wakayama , Japan
| | - Toru Mima
- a Department of Nephrology , Wakayama Medical University , Wakayama , Japan
| | - Mitsuru Yashiro
- a Department of Nephrology , Wakayama Medical University , Wakayama , Japan
| | - Tomohiro Sonou
- a Department of Nephrology , Wakayama Medical University , Wakayama , Japan
| | - Masaki Ohya
- a Department of Nephrology , Wakayama Medical University , Wakayama , Japan
| | - Asuka Masumoto
- a Department of Nephrology , Wakayama Medical University , Wakayama , Japan
| | - Shintaro Yamanaka
- a Department of Nephrology , Wakayama Medical University , Wakayama , Japan
| | - Daisuke Koreeda
- a Department of Nephrology , Wakayama Medical University , Wakayama , Japan
| | - Koichi Tatsuta
- a Department of Nephrology , Wakayama Medical University , Wakayama , Japan
| | - Yoshiyuki Hanba
- a Department of Nephrology , Wakayama Medical University , Wakayama , Japan
| | - Mari Moribata
- a Department of Nephrology , Wakayama Medical University , Wakayama , Japan
| | - Shigeo Negi
- a Department of Nephrology , Wakayama Medical University , Wakayama , Japan
| | - Takashi Shigematsu
- a Department of Nephrology , Wakayama Medical University , Wakayama , Japan
| |
Collapse
|
28
|
Bianchi A, Guibert M, Cailotto F, Gasser A, Presle N, Mainard D, Netter P, Kempf H, Jouzeau JY, Reboul P. Fibroblast Growth Factor 23 drives MMP13 expression in human osteoarthritic chondrocytes in a Klotho-independent manner. Osteoarthritis Cartilage 2016; 24:1961-1969. [PMID: 27307356 DOI: 10.1016/j.joca.2016.06.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 06/02/2016] [Accepted: 06/07/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Fibroblast Growth Factor 23 (FGF23) may represent an attractive candidate that could participate to the osteoarthritic (OA)-induced phenotype switch of chondrocytes. To address this hypothesis, we investigated the expression of FGF23, its receptors (FGFRs) and co-receptor (Klotho) in human cartilage and studied the effects of rhFGF23 on OA chondrocytes. METHOD Gene expression or protein levels were analysed by RT-PCR and immunohistochemistry. Collagenase 3 (MMP13) activity was measured by a fluorescent assay. MAPK signalling pathways were investigated by phosphoprotein array, immunoblotting and the use of selective inhibitors. RNA silencing was performed to confirm the respective contribution of FGFR1 and Klotho. RESULTS We showed that the expression of FGF23, FGFR1 and Klotho was up-regulated at both mRNA and protein levels in OA chondrocytes when compared to healthy ones. These overexpressions were markedly elevated in the damaged regions of OA cartilage. When stimulated with rhFGF23, OA chondrocytes displayed an extended expression of FGF23 and of markers of hypertrophy such as MMP13, COL10A1, and VEGF. We demonstrated that FGF23 auto-stimulation was both FGFR1-and Klotho-dependent, whereas the expression of markers of hypertrophy was mainly dependent on FGFR1 alone. Finally, we showed that FGF23-induced MMP13 expression was strongly regulated by the MEK/ERK cascade and to a lesser extent, by the PI-3K/AKT pathway. CONCLUSION These results demonstrate that FGF23 sustains differentiation of OA chondrocytes in a Klotho-independent manner.
Collapse
Affiliation(s)
- A Bianchi
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - M Guibert
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - F Cailotto
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - A Gasser
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - N Presle
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - D Mainard
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France; Département de Chirurgie Orthopédique et Traumatologique, Centre Hospitalier Universitaire, Nancy, France.
| | - P Netter
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France; Département de Pharmacologie Clinique et Toxicologie, Centre Hospitalier Universitaire, Nancy, France.
| | - H Kempf
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - J-Y Jouzeau
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France; Département de Pharmacologie Clinique et Toxicologie, Centre Hospitalier Universitaire, Nancy, France.
| | - P Reboul
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| |
Collapse
|
29
|
Clinkenbeard EL, Cass TA, Ni P, Hum JM, Bellido T, Allen MR, White KE. Conditional Deletion of Murine Fgf23: Interruption of the Normal Skeletal Responses to Phosphate Challenge and Rescue of Genetic Hypophosphatemia. J Bone Miner Res 2016; 31:1247-57. [PMID: 26792657 PMCID: PMC4891276 DOI: 10.1002/jbmr.2792] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 01/14/2016] [Accepted: 01/20/2016] [Indexed: 01/12/2023]
Abstract
The transgenic and knockout (KO) animals involving Fgf23 have been highly informative in defining novel aspects of mineral metabolism, but are limited by shortened lifespan, inability of spatial/temporal FGF23 control, and infertility of the global KO. To more finely test the role of systemic and genetic influences in FGF23 production, a mouse was developed that carried a floxed ("f")-Fgf23 allele (exon 2 floxed) which demonstrated in vivo recombination when bred to global-Cre transgenic mice (eIIa-cre). Mice homozygous for the recombined allele ("Δ") had undetectable serum intact FGF23, elevated serum phosphate (p < 0.05), and increased kidney Cyp27b1 mRNA (p < 0.05), similar to global Fgf23-KO mice. To isolate cellular FGF23 responses during phosphate challenge, Fgf23(Δ/f) mice were mated with early osteoblast type Iα1 collagen 2.3-kb promoter-cre mice (Col2.3-cre) and the late osteoblast/early osteocyte Dentin matrix protein-1-cre (Dmp1-cre). Fgf23(Δ/f) /Col2.3-cre(+) and Fgf23(Δ/f) /Dmp1-cre(+) exhibited reduced baseline serum intact FGF23 versus controls. After challenge with high-phosphate diet Cre(-) mice had 2.1-fold to 2.5-fold increased serum FGF23 (p < 0.01), but Col2.3-cre(+) mice had no significant increase, and Dmp1-cre(+) mice had only a 37% increase (p < 0.01) despite prevailing hyperphosphatemia in both models. The Fgf23(Δ/f) /Col2.3-cre was bred onto the Hyp (murine X-linked hypophosphatemia [XLH] model) genetic background to test the contribution of osteoblasts and osteocytes to elevated FGF23 and Hyp disease phenotypes. Whereas Hyp mice maintained inappropriately elevated FGF23 considering their marked hypophosphatemia, Hyp/Fgf23(Δ/f) /Col2.3-cre(+) mice had serum FGF23 <4% of Hyp (p < 0.01), and this targeted restriction normalized serum phosphorus and ricketic bone disease. In summary, deleting FGF23 within early osteoblasts and osteocytes demonstrated that both cell types contribute to baseline circulating FGF23 concentrations, and that targeting osteoblasts/osteocytes for FGF23 production can modify systemic responses to changes in serum phosphate concentrations and rescue the Hyp genetic syndrome. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Erica L. Clinkenbeard
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Taryn A. Cass
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Pu Ni
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Julia M. Hum
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Teresita Bellido
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Matthew R. Allen
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kenneth E. White
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
30
|
Sapir-Koren R, Livshits G. Bone mineralization is regulated by signaling cross talk between molecular factors of local and systemic origin: the role of fibroblast growth factor 23. Biofactors 2014; 40:555-68. [PMID: 25352227 DOI: 10.1002/biof.1186] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Accepted: 10/06/2014] [Indexed: 01/07/2023]
Abstract
Body phosphate homeostasis is regulated by a hormonal counter-balanced intestine-bone-kidney axis. The major systemic hormones involved in this axis are parathyroid hormone (PTH), 1,25-dihydroxyvitamin-D, and fibroblast growth factor-23 (FGF23). FGF23, produced almost exclusively by the osteocytes, is a phosphaturic hormone that plays a major role in regulation of the bone remodeling process. Remodeling composite components, bone mineralization and resorption cycles create a continuous influx-efflux loop of the inorganic phosphate (Pi) through the skeleton. This "bone Pi loop," which is formed, is controlled by local and systemic factors according to phosphate homeostasis demands. Although FGF23 systemic actions in the kidney, and for the production of PTH and 1,25-dihydroxyvitamin-D are well established, its direct involvement in bone metabolism is currently poorly understood. This review presents the latest available evidence suggesting two aspects of FGF23 bone local activity: (a) Regulation of FGF23 production by both local and systemic factors. The suggested local factors include extracellular levels of Pi and pyrophosphate (PPi), (the Pi/PPi ratio), and another osteocyte-derived protein, sclerostin. In addition, 1,25-dihydroxyvitamin-D, synthesized locally by bone cells, may contribute to regulation of FGF23 production. The systemic control is achieved via PTH and 1,25-dihydroxyvitamin-D endocrine functions. (b) FGF23 acts as a local agent, directly affecting bone mineralization. We support the assumption that under balanced physiological conditions, sclerostin, by para- autocrine signaling, upregulates FGF23 production by the osteocyte. FGF23, in turn, acts as a mineralization inhibitor, by stimulating the generation of the major mineralization antagonist-PPi.
Collapse
Affiliation(s)
- Rony Sapir-Koren
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | |
Collapse
|
31
|
Xie Y, Zhou S, Chen H, Du X, Chen L. Recent research on the growth plate: Advances in fibroblast growth factor signaling in growth plate development and disorders. J Mol Endocrinol 2014; 53:T11-34. [PMID: 25114206 DOI: 10.1530/jme-14-0012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Skeletons are formed through two distinct developmental actions, intramembranous ossification and endochondral ossification. During embryonic development, most bone is formed by endochondral ossification. The growth plate is the developmental center for endochondral ossification. Multiple signaling pathways participate in the regulation of endochondral ossification. Fibroblast growth factor (FGF)/FGF receptor (FGFR) signaling has been found to play a vital role in the development and maintenance of growth plates. Missense mutations in FGFs and FGFRs can cause multiple genetic skeletal diseases with disordered endochondral ossification. Clarifying the molecular mechanisms of FGFs/FGFRs signaling in skeletal development and genetic skeletal diseases will have implications for the development of therapies for FGF-signaling-related skeletal dysplasias and growth plate injuries. In this review, we summarize the recent advances in elucidating the role of FGFs/FGFRs signaling in growth plate development, genetic skeletal disorders, and the promising therapies for those genetic skeletal diseases resulting from FGFs/FGFRs dysfunction. Finally, we also examine the potential important research in this field in the future.
Collapse
Affiliation(s)
- Yangli Xie
- Department of Rehabilitation MedicineCenter of Bone Metabolism and Repair, Trauma Center, State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Siru Zhou
- Department of Rehabilitation MedicineCenter of Bone Metabolism and Repair, Trauma Center, State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Hangang Chen
- Department of Rehabilitation MedicineCenter of Bone Metabolism and Repair, Trauma Center, State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Xiaolan Du
- Department of Rehabilitation MedicineCenter of Bone Metabolism and Repair, Trauma Center, State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Lin Chen
- Department of Rehabilitation MedicineCenter of Bone Metabolism and Repair, Trauma Center, State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| |
Collapse
|
32
|
Su N, Jin M, Chen L. Role of FGF/FGFR signaling in skeletal development and homeostasis: learning from mouse models. Bone Res 2014; 2:14003. [PMID: 26273516 PMCID: PMC4472122 DOI: 10.1038/boneres.2014.3] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Revised: 11/29/2013] [Accepted: 12/03/2013] [Indexed: 01/06/2023] Open
Abstract
Fibroblast growth factor (FGF)/fibroblast growth factor receptor (FGFR) signaling plays essential roles in bone development and diseases. Missense mutations in FGFs and FGFRs in humans can cause various congenital bone diseases, including chondrodysplasia syndromes, craniosynostosis syndromes and syndromes with dysregulated phosphate metabolism. FGF/FGFR signaling is also an important pathway involved in the maintenance of adult bone homeostasis. Multiple kinds of mouse models, mimicking human skeleton diseases caused by missense mutations in FGFs and FGFRs, have been established by knock-in/out and transgenic technologies. These genetically modified mice provide good models for studying the role of FGF/FGFR signaling in skeleton development and homeostasis. In this review, we summarize the mouse models of FGF signaling-related skeleton diseases and recent progresses regarding the molecular mechanisms, underlying the role of FGFs/FGFRs in the regulation of bone development and homeostasis. This review also provides a perspective view on future works to explore the roles of FGF signaling in skeletal development and homeostasis.
Collapse
Affiliation(s)
- Nan Su
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University , Chongqing, 400042, China
| | - Min Jin
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University , Chongqing, 400042, China
| | - Lin Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University , Chongqing, 400042, China
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Disturbances in calcium-phosphate homeostasis play an important role in children with chronic kidney disease, and not only cause renal osteodystrophy but also result in increased cardiovascular morbidity and mortality. This review outlines the current aspects in the pathogenesis, diagnostic approach and treatment of renal osteodystrophy. RECENT FINDINGS The pathogenesis of renal osteodystrophy is under strong influence of the fibroblast growth factor 23/Klotho system, which is able to enhance phosphate excretion and reduce calcitriol synthesis in the kidney. Fibroblast growth factor 23 increases tissue calcinosis and is cardiotoxic, and is independently associated with mortality. Despite improvement in diagnostic imaging (bone density measurements), determination of biomarkers, mainly parathyroid hormone, still plays a central role. New treatment options resulted in improved bone health and also a reduction in mortality was achieved in adults with calcium-free phosphate binders. Substitution of active and inactive vitamin D is important and also has a beneficial effect on proteinuria. SUMMARY Knowledge about the biochemical and molecular mechanisms of renal osteodystrophy is increasing dramatically and has an impact not only to bone health but also overall morbidity and mortality. This will ultimately translate into further improved diagnostic approaches and novel treatment options.
Collapse
|
34
|
Penido MGMG, Alon US. Hypophosphatemic rickets due to perturbations in renal tubular function. Pediatr Nephrol 2014; 29:361-73. [PMID: 23636577 DOI: 10.1007/s00467-013-2466-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Revised: 03/03/2013] [Accepted: 03/04/2013] [Indexed: 12/13/2022]
Abstract
The common denominator for all types of rickets is hypophosphatemia, leading to inadequate supply of the mineral to the growing bone. Hypophosphatemia can result from insufficient uptake of the mineral from the gut or its disproportionate losses in the kidney, the latter being caused by either tubular abnormalities per se or the effect on the tubule of circulating factors like fibroblast growth factor-23 and parathyroid hormone (PTH). High serum levels of the latter result in most cases from abnormalities in vitamin D metabolism which lead to decreased calcium absorption in the gut and hypocalcemia, triggering PTH secretion. Rickets is a disorder of the growth plate and hence pediatric by definition. However, it is important to recognize that the effect of hypophosphatemia on other parts of the skeleton results in osteomalacia in both children and adults. This review addresses the etiology, pathophysiologic mechanisms, clinical manifestations and treatment of entities associated with hypophosphatemic rickets due to perturbations in renal tubular function.
Collapse
Affiliation(s)
- Maria Goretti M G Penido
- Pediatric Nephrology Unit, Clinics Hospital, School of Medicine, Federal University of Minas Gerais, Av. Professor Alfredo Balena, 190, CEP, 30130100, Belo Horizonte, MG, Brazil,
| | | |
Collapse
|
35
|
Li Y, He X, Olauson H, Larsson TE, Lindgren U. FGF23 affects the lineage fate determination of mesenchymal stem cells. Calcif Tissue Int 2013; 93:556-64. [PMID: 24068282 DOI: 10.1007/s00223-013-9795-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 07/30/2013] [Indexed: 12/16/2022]
Abstract
FGF23 is a bone-derived hormone that regulates mineral metabolism by inhibiting renal tubular phosphate reabsorption and suppressing circulating 1,25(OH)2D and PTH levels. These effects are mediated by FGF-receptor binding and activation in the presence of its coreceptor Klotho, which is expressed in the distal tubules of the kidney. Recently, expression of Klotho in skeletal tissues has been reported, indicating a direct, yet unclear, extrarenal effect of FGF23 on cells involved with bone development and remodeling. In the present study, we found that bone marrow stromal cells harvested from Klotho null mice developed fewer osteoblastic but more adipocytic colonies than cells from wild-type mice. The underlying mechanism was explored by experiments on mouse C3H10T1/2 cells. We found that Klotho was weakly expressed and that FGF23 dose-dependently affected the lineage fate determination. The effects of FGF23 on cell differentiation can be diminished by SU 5402, a specific tyrosine kinase inhibitor for FGF receptors. Our results indicate that FGF23 directly affects the differentiation of bone marrow stromal cells.
Collapse
Affiliation(s)
- Yan Li
- Division of Orthopedics and Biotechnology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Huddinge, 14186, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
36
|
Prié D, Forand A, Francoz C, Elie C, Cohen I, Courbebaisse M, Eladari D, Lebrec D, Durand F, Friedlander G. Plasma fibroblast growth factor 23 concentration is increased and predicts mortality in patients on the liver-transplant waiting list. PLoS One 2013; 8:e66182. [PMID: 23825530 PMCID: PMC3692511 DOI: 10.1371/journal.pone.0066182] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 05/02/2013] [Indexed: 02/06/2023] Open
Abstract
High plasma fibroblast growth factor-23 (FGF23) concentration predicts the risk of death and poor outcomes in patients with chronic kidney disease or chronic heart failure. We checked if FGF23 concentration could be modified in patients with end stage liver disease (ESLD) and predict mortality. We measured plasma FGF23 in 200 patients with ESLD registered on a liver transplant waiting list between January 2005 and October 2008. We found that median plasma FGF23 concentration was above normal values in 63% of the patients. Increased FGF23 concentration was not explained by its classical determinants: hyperphosphataemia, increased calcitriol concentration or decreased renal function. FGF23 concentration correlated with the MELD score, serum sodium concentration, and GFR. Forty-six patients died before being transplanted and 135 underwent liver transplantation. We analyzed the prognostic value of FGF23 levels. Mortality was significantly associated with FGF23 levels, the MELD score, serum sodium concentration and glomerular filtration rate. On multivariate analyses only FGF23 concentration was associated with mortality. FGF23 levels were independent of the cause of the liver disease. To determine if the damaged liver can produce FGF23 we measured plasma FGF23 concentration and liver FGF23 mRNA expression in control and diethyl-nitrosamine (DEN)-treated mice. FGF23 plasma levels increased with the apparition of liver lesions in DEN-treated mice and that FGF23 mRNA expression, which was undetectable in the liver of control mice, markedly increased with the development of liver lesions. The correlation between FGF23 plasma concentration and FGF23 mRNA expression in DEN-treated mice suggests that FGF23 production by the liver accounts for the increased plasma FGF23 concentration. In conclusion chronic liver lesions can induce expression of FGF23 mRNA leading to increased FGF23 concentration, which is associated with a higher mortality in patients on a liver-transplant waiting list. In these patients FGF23 concentration was the best predictor of mortality.
Collapse
Affiliation(s)
- Dominique Prié
- Université Paris Descartes, Faculté de Médecine, INSERM U845, Hôpital Necker-Enfants Malades, AP-HP, Paris, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|