Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 188
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Calcif Tissue Int ; 114(2): 137-146, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37981601

RESUMO

Hereditary hypophosphatemic rickets with hypercalciuria (HHRH) is a rare disorder of phosphate homeostasis. We describe a single-center experience of genetically proven HHRH families and perform systematic review phenotype-genotype correlation in reported biallelic probands and their monoallelic relatives. Detailed clinical, biochemical, radiological, and genetic data were retrieved from our center and a systematic review of Pub-Med and Embase databases for patients and relatives who were genetically proven. Total of nine subjects (probands:5) carrying biallelic SLC34A3 mutations (novel:2) from our center had a spectrum from rickets/osteomalacia to normal BMD, with hypophosphatemia and hypercalciuria in all. We describe the first case of genetically proven HHRH with enthesopathy. Elevated FGF23 in another patient with hypophosphatemia, iron deficiency anemia, and noncirrhotic periportal fibrosis led to initial misdiagnosis as tumoral osteomalacia. On systematic review of 58 probands (with biallelic SLC34A3 mutations; 35 males), early-onset HHRH and renal calcification were present in ~ 70% and late-onset HHRH in 10%. c.575C > T p.(Ser192Leu) variant occurred in 53% of probands without skeletal involvement. Among 110 relatives harboring monoallelic SLC34A3 mutation at median age 38 years, renal calcification, hypophosphatemia, high 1,25(OH)2D, and hypercalciuria were observed in ~30%, 22.3%, 40%, and 38.8%, respectively. Renal calcifications correlated with age but were similar across truncating and non-truncating variants. Although most relatives were asymptomatic for bone involvement, 6/12(50%) had low bone mineral density. We describe the first monocentric HHRH case series from India with varied phenotypes. In a systematic review, frequent renal calcifications and low BMD in relatives with monoallelic variants (HHRH trait) merit identification.


Assuntos
Entesopatia , Raquitismo Hipofosfatêmico Familiar , Hipofosfatemia , Doenças Renais Císticas , Nefrocalcinose , Osteomalacia , Masculino , Humanos , Adulto , Raquitismo Hipofosfatêmico Familiar/complicações , Raquitismo Hipofosfatêmico Familiar/diagnóstico , Raquitismo Hipofosfatêmico Familiar/genética , Hipercalciúria/complicações , Hipercalciúria/genética , Osteomalacia/complicações , Osteomalacia/genética
2.
J Bone Miner Metab ; 42(2): 155-165, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38310177

RESUMO

INTRODUCTION: Fanconi renotubular syndromes (FRTS) are a rare group of inherited phosphaturic disorders with limited Indian as well as global data on this condition. Here, we describe the experience of a single Endocrinology center from Western India on FRTS. MATERIALS AND METHODS: Comprehensive clinical, biochemical, radiological, management, and genetic details of FRTS patients managed between 2010 and 2023 were collected and analyzed. RESULTS: FRTS probands had mutations (eight novel) in six genes [CLCN5 (n = 4), SLC2A2 (n = 2), GATM, EHHADH, HNF4A, and OCRL (1 each)]. Among 15 FRTS patients (11 families), rickets/osteomalacia was the most common (n = 14) presentation with wide inter- and intra-familial phenotypic variability. Delayed diagnosis (median: 8.8 years), initial misdiagnosis (8/11 probands), and syndrome-specific discriminatory features (8/11 probands) were commonly seen. Hypophosphatemia, elevated alkaline phosphatase, normal parathyroid hormone (median: 36 pg/ml), high-normal/elevated 1,25(OH)2D (median: 152 pg/ml), hypercalciuria (median spot urinary calcium to creatinine ratio: 0.32), and variable proximal tubular dysfunction(s) were observed. Elevated C-terminal fibroblast growth factor 23 in two probands was misleading, till the genetic diagnosis was reached. Novel observations in our FRTS cohort were preserved renal function (till sixth decade) and enthesopathy in FRTS1 and FRTS3 families, respectively. CONCLUSION: Our findings underscore frequent under- and misdiagnosis of FRTS; hence, a high index of suspicion for FRTS in phosphopenic rickets/osteomalacia, with early consideration of genetic testing is essential to ensure timely diagnosis of FRTS. The novel variants and phenotypic manifestations described here expand the disease spectrum of FRTS.


Assuntos
Raquitismo Hipofosfatêmico Familiar , Síndrome de Fanconi , Hipofosfatemia Familiar , Osteomalacia , Raquitismo Hipofosfatêmico , Humanos , Osteomalacia/genética , Raquitismo Hipofosfatêmico Familiar/genética , Hipofosfatemia Familiar/genética , Hipofosfatemia Familiar/metabolismo , Síndrome de Fanconi/genética , Síndrome de Fanconi/metabolismo
3.
J Cell Physiol ; 238(11): 2556-2569, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37698039

RESUMO

Family with sequence similarity 20-member C (FAM20C) is a kinase specific to most of the secreted phosphoproteome. FAM20C has been identified as the causative gene of Raine syndrome, initially characterized by lethal osteosclerosis bone dysplasia. However, since the identification of the cases of nonlethal Raine syndrome characterized by hypophosphatemia rickets, the previous definition of Raine syndrome has become debatable and raised a question about the role of mutations of FAM20C in controversial skeletal manifestation in the two forms of the disease. In this study, we aimed to investigate the influence of FAM20C mutations on skeletogenesis. We developed transgenic mice expressing Fam20c mutations mimicking those associated with human lethal and nonlethal Raine syndrome. The results revealed that transgenic mice expressing the mutant Fam20c found in the lethal (KO;G374R) and nonlethal (KO;D446N) Raine syndrome exhibited osteomalacia without osteosclerotic features. Additionally, both mutants significantly increased the expression of the Fgf23, indicating that Fam20c deficiency in skeletal compartments causes hypophosphatemia rickets. Furthermore, as FAM20C kinase activity catalyzes the phosphorylation of secreted proteomes other than those in the skeletal system, global FAM20C deficiency may trigger alterations in other systems resulting in osteosclerosis secondary to hypophosphatemia rickets. Together, the findings of this study suggest that FAM20C deficiency primarily causes hypophosphatemia rickets or osteomalacia; however, the heterogeneous skeletal manifestation in Raine syndrome was not determined solely by specific mutations of FAM20C. The findings also implicated that rickets or osteomalacia caused by FAM20C deficiency would deteriorate into osteosclerosis by the defects from other systems or environmental impacts.


Assuntos
Hipofosfatemia , Osteomalacia , Osteosclerose , Raquitismo , Camundongos , Animais , Humanos , Osteomalacia/complicações , Osteomalacia/genética , Osteosclerose/genética , Osteosclerose/complicações , Mutação/genética , Raquitismo/complicações , Camundongos Transgênicos , Hipofosfatemia/genética , Hipofosfatemia/complicações , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Proteínas de Ligação ao Cálcio/genética
4.
Proc Natl Acad Sci U S A ; 117(50): 31800-31807, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33257569

RESUMO

The three members of the endocrine-fibroblast growth factor (FGF) family, FGF19, 21, and 23 are circulating hormones that regulate critical metabolic processes. FGF23 stimulates the assembly of a signaling complex composed of α-Klotho (KLA) and FGF receptor (FGFR) resulting in kinase activation, regulation of phosphate homeostasis, and vitamin D levels. Here we report that the C-terminal tail of FGF23, a region responsible for KLA binding, contains two tandem repeats, repeat 1 (R1) and repeat 2 (R2) that function as two distinct ligands for KLA. FGF23 variants with a single KLA binding site, FGF23-R1, FGF23-R2, or FGF23-wild type (WT) with both R1 and R2, bind to KLA with similar binding affinity and stimulate FGFR1 activation and MAPK response. R2 is flanked by two cysteines that form a disulfide bridge in FGF23-WT; disulfide bridge formation in FGF23-WT is dispensable for KLA binding and for cell signaling via FGFRs. We show that FGF23-WT stimulates dimerization and activation of a chimeric receptor molecule composed of the extracellular domain of KLA fused to the cytoplasmic domain of FGFR and employ total internal reflection fluorescence microscopy to visualize individual KLA molecules on the cell surface. These experiments demonstrate that FGF23-WT can act as a bivalent ligand of KLA in the cell membrane. Finally, an engineered Fc-R2 protein acts as an FGF23 antagonist offering new pharmacological intervention for treating diseases caused by excessive FGF23 abundance or activity.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Glucuronidase/metabolismo , Multimerização Proteica/fisiologia , Sítios de Ligação , Calcinose/tratamento farmacológico , Calcinose/genética , Membrana Celular/metabolismo , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/uso terapêutico , Células HEK293 , Humanos , Hiperostose Cortical Congênita/tratamento farmacológico , Hiperostose Cortical Congênita/genética , Hiperfosfatemia/tratamento farmacológico , Hiperfosfatemia/genética , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Proteínas Klotho , Mutação , Osteomalacia/tratamento farmacológico , Osteomalacia/genética , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Domínios Proteicos , Multimerização Proteica/efeitos dos fármacos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/uso terapêutico , Raquitismo Hipofosfatêmico/tratamento farmacológico , Raquitismo Hipofosfatêmico/genética
5.
Curr Osteoporos Rep ; 19(6): 669-675, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34755323

RESUMO

PURPOSE OF REVIEW: X-linked hypophosphatemia and tumor-induced osteomalacia are diseases characterized by hypophosphatemia with impaired proximal tubular phosphate reabsorption. Complete resection of responsible tumors is the first-line therapy for patients with tumor-induced osteomalacia. In contrast, phosphate and active vitamin D have been used for patients with X-linked hypophosphatemia and inoperable ones with tumor-induced osteomalacia. The purpose of this review is to summarize the pathogenesis of these diseases and discuss about the new treatment. RECENT FINDINGS: Excessive FGF23 production has been shown to underline several kinds of hypophosphatemic rickets/osteomalacia including X-linked hypophosphatemia and tumor-induced osteomalacia. Burosumab, an anti-FGF23 monoclonal antibody, was approved for clinical use, while the indications of burosumab are different depending on countries. The inhibition of excessive FGF23 activity has been approved as a new therapy for several kinds of hypophosphatemic diseases. Further studies are necessary to clarify the long-term effects and safety of burosumab.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Raquitismo Hipofosfatêmico Familiar/tratamento farmacológico , Raquitismo Hipofosfatêmico Familiar/genética , Fator de Crescimento de Fibroblastos 23/genética , Osteomalacia/tratamento farmacológico , Osteomalacia/genética , Síndromes Paraneoplásicas/tratamento farmacológico , Síndromes Paraneoplásicas/genética , Animais , Doenças Genéticas Ligadas ao Cromossomo X/tratamento farmacológico , Doenças Genéticas Ligadas ao Cromossomo X/genética , Humanos , Camundongos
6.
Int J Mol Sci ; 22(6)2021 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-33809311

RESUMO

During the last two decades, the potential impact of vitamin D on the risk of cardiovascular disease (CVD) has been rigorously studied. Data regarding the effect of vitamin D on CVD risk are puzzling: observational data indicate an inverse nonlinear association between vitamin D status and CVD events, with the highest CVD risk at severe vitamin D deficiency; however, preclinical data and randomized controlled trials (RCTs) show several beneficial effects of vitamin D on the surrogate parameters of vascular and cardiac function. By contrast, Mendelian randomization studies and large RCTs in the general population and in patients with chronic kidney disease, a high-risk group for CVD events, largely report no significant beneficial effect of vitamin D treatment on CVD events. In patients with rickets and osteomalacia, cardiovascular complications are infrequently reported, except for an increased risk of heart failure. In conclusion, there is no strong evidence for beneficial vitamin D effects on CVD risk, either in the general population or in high-risk groups. Whether some subgroups such as individuals with severe vitamin D deficiency or a combination of low vitamin D status with specific gene variants and/or certain nutrition/lifestyle factors would benefit from vitamin D (metabolite) administration, remains to be studied.


Assuntos
Doenças Cardiovasculares/genética , Deficiência de Vitamina D/genética , Vitamina D/genética , Doenças Cardiovasculares/complicações , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/patologia , Suplementos Nutricionais , Humanos , Análise da Randomização Mendeliana , Osteomalacia/complicações , Osteomalacia/epidemiologia , Osteomalacia/genética , Raquitismo/complicações , Raquitismo/epidemiologia , Raquitismo/genética , Fatores de Risco , Deficiência de Vitamina D/complicações , Deficiência de Vitamina D/epidemiologia , Deficiência de Vitamina D/patologia
7.
J Cell Mol Med ; 24(9): 4931-4943, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32277576

RESUMO

Tumour-induced osteomalacia (TIO) is a very rare paraneoplastic syndrome with bone pain, fractures and muscle weakness, which is mostly caused by phosphaturic mesenchymal tumours (PMTs). Cell-free DNA (cfDNA) has been regarded as a non-invasive liquid biopsy for many malignant tumours. However, it has not been studied in benign tumours, which prompted us to adopt the targeted next-generation sequencing approach to compare cfDNAs of 4 TIO patients, four patients with bone metastasis (BM) and 10 healthy controls. The mutational landscapes of cfDNA in TIO and BM groups were similar in the spectrum of allele frequencies and mutation types. Markedly, deleterious missense mutations in FGFR1 and loss-of-function mutations in MED12 were found in 3/4 TIO patients but none of BM patients. The gene ontology analysis strongly supported that these mutated genes found in TIOs would play a potential role in PMTs' process. The genetic signatures and corresponding change in expression of FGFR1 and FGF23 were further validated in PMT tissues from a test cohort of another three TIO patients. In summary, we reported the first study of the mutational landscape and genetic signatures of cfDNA in TIO/PMTs.


Assuntos
Ácidos Nucleicos Livres , Análise Mutacional de DNA , Neoplasias/complicações , Neoplasias/genética , Osteomalacia/complicações , Osteomalacia/genética , Síndromes Paraneoplásicas/complicações , Síndromes Paraneoplásicas/genética , Adulto , Biomarcadores Tumorais , Neoplasias Ósseas/complicações , Neoplasias Ósseas/genética , Estudos de Casos e Controles , Sistema Livre de Células , Feminino , Fator de Crescimento de Fibroblastos 23 , Perfilação da Expressão Gênica , Biblioteca Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Hipofosfatemia Familiar/metabolismo , Masculino , Complexo Mediador/genética , Pessoa de Meia-Idade , Mutação , Mutação de Sentido Incorreto , Metástase Neoplásica , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética
8.
Pediatr Nephrol ; 35(10): 1843-1854, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31392510

RESUMO

Hypophosphatemic rickets is caused by renal phosphate wasting that is most commonly due to X-linked dominant mutations in PHEX. PHEX mutations cause hypophosphatemia indirectly, through the increased expression of fibroblast growth factor 23 (FGF23) by osteocytes. FGF23 decreases renal phosphate reabsorption and thereby increases phosphate excretion. The lack of phosphate leads to a mineralization defect at the level of growth plates (rickets), bone tissue (osteomalacia), and teeth, where the defect facilitates the formation of abscesses. The bone tissue immediately adjacent to osteocytes often remains unmineralized ("periosteocytic lesions"), highlighting the osteocyte defect in this disorder. Common clinical features of XLH include deformities of the lower extremities, short stature, enthesopathies, dental abscesses, as well as skull abnormalities such as craniosynostosis and Chiari I malformation. For the past four decades, XLH has been treated by oral phosphate supplementation and calcitriol, which improves rickets and osteomalacia and the dental manifestations, but often does not resolve all aspects of the mineralization defects. A newer treatment approach using inactivating FGF23 antibodies leads to more stable control of serum inorganic phosphorus levels and seems to heal rickets more reliably. However, the long-term benefits of FGF23 antibody treatment remain to be elucidated.


Assuntos
Raquitismo Hipofosfatêmico Familiar/patologia , Fatores de Crescimento de Fibroblastos/metabolismo , Osteomalacia/patologia , Endopeptidase Neutra Reguladora de Fosfato PHEX/genética , Fosfatos/metabolismo , Absorciometria de Fóton , Desenvolvimento Ósseo/efeitos dos fármacos , Desenvolvimento Ósseo/genética , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/patologia , Calcificação Fisiológica/efeitos dos fármacos , Calcificação Fisiológica/genética , Calcitriol/administração & dosagem , Raquitismo Hipofosfatêmico Familiar/diagnóstico , Raquitismo Hipofosfatêmico Familiar/tratamento farmacológico , Raquitismo Hipofosfatêmico Familiar/genética , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Humanos , Osteócitos/metabolismo , Osteomalacia/diagnóstico , Osteomalacia/tratamento farmacológico , Osteomalacia/genética , Endopeptidase Neutra Reguladora de Fosfato PHEX/metabolismo , Comunicação Parácrina/genética , Fosfatos/administração & dosagem , Fosfatos/sangue , Reabsorção Renal/efeitos dos fármacos , Reabsorção Renal/genética , Dente/crescimento & desenvolvimento , Dente/patologia , Resultado do Tratamento
9.
Biol Pharm Bull ; 40(4): 381-390, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28381792

RESUMO

Ezrin/radixin/moesin (ERM) proteins function as general cross-linkers between plasma membrane proteins and the actin cytoskeleton and are involved in the functional expression of membrane proteins on the cell surface. They also integrate Rho guanosine 5'-triphosphatase (GTPase) signaling to regulate cytoskeletal organization by sequestering Rho-related proteins. They act as protein kinase A (PKA)-anchoring proteins and sequester PKA close to its target proteins for their effective phosphorylation and functional regulation. Therefore, ERM proteins seem to play important roles in the membrane transport of electrolytes by ion channels and transporters. In this review, we focus on the pathophysiological roles of ERM proteins in in vivo studies and introduce the phenotypes of their knockout and knockdown mice.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Animais , Membrana Celular/metabolismo , Membrana Celular/patologia , Colestase/genética , Colestase/metabolismo , Colestase/fisiopatologia , Proteínas do Citoesqueleto/deficiência , Proteínas do Citoesqueleto/genética , Humanos , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Osteomalacia/genética , Osteomalacia/metabolismo , Osteomalacia/fisiopatologia , Transporte Proteico/fisiologia , Distribuição Tecidual/fisiologia , Quinases Associadas a rho/metabolismo
10.
Gene Ther ; 23(6): 500-9, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26934099

RESUMO

Our previous work showed that a Sca-1(+) cell-based FGF2 therapy was capable of promoting robust increases in trabecular bone formation and connectivity on the endosteum of long bones. Past work reported that administration of FGF2 protein promoted bone formation in red marrow but not in yellow marrow. The issue as to whether the Sca-1(+) cell-based FGF2 therapy is effective in yellow marrow is highly relevant to its clinical potential for osteoporosis, as most red marrows in a person of an advanced age are converted to yellow marrows. Accordingly, this study sought to compare the osteogenic effects of this stem cell-based FGF2 therapy on red marrow-filled lumbar vertebrae with those on yellow marrow-filled caudal vertebrae of young adult W(41)/W(41) mice. The Sca-1(+) cell-based FGF2 therapy drastically increased trabecular bone formation in lumbar vertebrae, but the therapy not only did not promote bone formation but instead caused substantial loss of trabecular bone in caudal vertebrae. The lack of an osteogenic response was not due to insufficient engraftment of FGF2-expressing Sca-1(+) cells or inadequate FGF2 expression in caudal vertebrae. Previous studies have demonstrated that recipient mice of this stem cell-based FGF2 therapy developed secondary hyperparathyroidism and increased bone resorption. Thus, the loss of bone mass in caudal vertebrae might in part be due to an increase in resorption without a corresponding increase in bone formation. In conclusion, the Sca-1(+) cell-based FGF2 therapy is osteogenic in red marrow but not in yellow marrow.


Assuntos
Antígenos Ly/genética , Antígenos Ly/metabolismo , Fator 2 de Crescimento de Fibroblastos/genética , Terapia Genética/métodos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Animais , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea/métodos , Osso Esponjoso/citologia , Osso Esponjoso/transplante , Caspase 3/genética , Feminino , Fator 2 de Crescimento de Fibroblastos/administração & dosagem , Fator 2 de Crescimento de Fibroblastos/biossíntese , Fator 2 de Crescimento de Fibroblastos/sangue , Humanos , Vértebras Lombares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteogênese/genética , Osteomalacia/etiologia , Osteomalacia/genética , Transplante de Células-Tronco/métodos
11.
Hum Mol Genet ; 23(2): 397-407, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24006476

RESUMO

Pathologically elevated serum levels of fibroblast growth factor-23 (FGF23), a bone-derived hormone that regulates phosphorus homeostasis, result in renal phosphate wasting and lead to rickets or osteomalacia. Rarely, elevated serum FGF23 levels are found in association with mosaic cutaneous disorders that affect large proportions of the skin and appear in patterns corresponding to the migration of ectodermal progenitors. The cause and source of elevated serum FGF23 is unknown. In those conditions, such as epidermal and large congenital melanocytic nevi, skin lesions are variably associated with other abnormalities in the eye, brain and vasculature. The wide distribution of involved tissues and the appearance of multiple segmental skin and bone lesions suggest that these conditions result from early embryonic somatic mutations. We report five such cases with elevated serum FGF23 and bone lesions, four with large epidermal nevi and one with a giant congenital melanocytic nevus. Exome sequencing of blood and affected skin tissue identified somatic activating mutations of HRAS or NRAS in each case without recurrent secondary mutation, and we further found that the same mutation is present in dysplastic bone. Our finding of somatic activating RAS mutation in bone, the endogenous source of FGF23, provides the first evidence that elevated serum FGF23 levels, hypophosphatemia and osteomalacia are associated with pathologic Ras activation and may provide insight in the heretofore limited understanding of the regulation of FGF23.


Assuntos
Fatores de Crescimento de Fibroblastos/sangue , GTP Fosfo-Hidrolases/genética , Hipofosfatemia/genética , Proteínas de Membrana/genética , Nevo Pigmentado/genética , Osteomalacia/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Neoplasias Cutâneas/genética , Adolescente , Criança , Exoma , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hipofosfatemia/sangue , Hipofosfatemia/patologia , Masculino , Mutação , Nevo , Nevo Pigmentado/sangue , Nevo Pigmentado/patologia , Osteomalacia/sangue , Osteomalacia/patologia , Análise de Sequência de DNA , Pele/metabolismo , Pele/patologia , Neoplasias Cutâneas/sangue , Neoplasias Cutâneas/patologia
12.
J Am Acad Dermatol ; 75(2): 420-7, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27444071

RESUMO

BACKGROUND: We recently demonstrated multilineage somatic mosaicism in cutaneous skeletal hypophosphatemia syndrome (CSHS), which features epidermal or melanocytic nevi, elevated fibroblast growth factor (FGF)-23, and hypophosphatemia, finding identical RAS mutations in affected skin and bone. OBJECTIVE: We sought to: (1) provide an updated overview of CSHS; (2) review its pathobiology; (3) present a new patient with CSHS; and (4) discuss treatment modalities. METHODS: We searched PubMed for "nevus AND rickets," and "nevus AND hypophosphatemia," identifying cases of nevi with hypophosphatemic rickets or elevated serum FGF-23. For our additional patient with CSHS, we performed histopathologic and radiographic surveys of skin and skeletal lesions, respectively. Sequencing was performed for HRAS, KRAS, and NRAS to determine causative mutations. RESULTS: Our new case harbored somatic activating HRAS p.G13 R mutation in affected tissue, consistent with previous findings. Although the mechanism of FGF-23 dysregulation is unknown in CSHS, interaction between FGF and MAPK pathways may provide insight into pathobiology. Anti-FGF-23 antibody KRN-23 may be useful in managing CSHS. LIMITATIONS: Multilineage RAS mutation in CSHS was recently identified; further studies on mechanism are unavailable. CONCLUSION: Patients with nevi in association with skeletal disease should be evaluated for serum phosphate and FGF-23. Further studies investigating the role of RAS in FGF-23 regulation are needed.


Assuntos
Genes ras/genética , Mosaicismo , Nevo Pigmentado/genética , Osteomalacia/genética , Raquitismo Hipofosfatêmico/genética , Neoplasias Cutâneas/genética , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Humanos , Nevo Pigmentado/tratamento farmacológico , Osteomalacia/tratamento farmacológico , Raquitismo Hipofosfatêmico/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Síndrome
13.
Subcell Biochem ; 76: 155-66, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26219711

RESUMO

To-date, the function of tissue-nonspecific alkaline phosphatase (TNAP) has largely been defined through studies in patients and mice affected by hypophosphatasia (HPP), a rare inborn-error-of-metabolism caused by mutation(s) in the TNAP gene (ALPL). The skeletal disease in HPP can be explained by alterations in the Pi/PPi ratio, with accumulation in the concentration of the mineralization inhibitor PPi as the culprit in preventing propagation of mineralization onto the collagenous extracellular matrix in bones and teeth. Accumulation of phosphorylated osteopontin increases the severity of HPP, at least in mice. Disruption in the metabolism of vitamin B6 leads to intracellular deficiency of pyridoxal, and this causes vitamin B6-responsive seizures in patients with the severe forms of the disease. Recent findings also implicate TNAP in the metabolism of ATP, in the production of adenosine and in the dephosphorylation of the bacterial toxin lipopolysaccharide, all molecules known to be involved in inflammation. The role of TNAP in establishing the ATP/adenosine ratio is important for purinergic signaling, and these mechanisms could be significant in determining axonal growth in the brain. Finally, the potential involvement of TNAP in dephosphorylating tau protein and its role in the pathogenesis of Alzheimer's disease is intriguing.


Assuntos
Fosfatase Alcalina/fisiologia , Neurônios/enzimologia , Fosfatase Alcalina/metabolismo , Animais , Calcificação Fisiológica/genética , Humanos , Hipofosfatasia/genética , Camundongos , Neurônios/metabolismo , Osteomalacia/genética , Osteomalacia/metabolismo , Raquitismo/genética , Distribuição Tecidual
14.
Nat Genet ; 38(11): 1310-5, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17033621

RESUMO

The osteocyte, a terminally differentiated cell comprising 90%-95% of all bone cells, may have multiple functions, including acting as a mechanosensor in bone (re)modeling. Dentin matrix protein 1 (encoded by DMP1) is highly expressed in osteocytes and, when deleted in mice, results in a hypomineralized bone phenotype. We investigated the potential for this gene not only to direct skeletal mineralization but also to regulate phosphate (P(i)) homeostasis. Both Dmp1-null mice and individuals with a newly identified disorder, autosomal recessive hypophosphatemic rickets, manifest rickets and osteomalacia with isolated renal phosphate-wasting associated with elevated fibroblast growth factor 23 (FGF23) levels and normocalciuria. Mutational analyses showed that autosomal recessive hypophosphatemic rickets family carried a mutation affecting the DMP1 start codon, and a second family carried a 7-bp deletion disrupting the highly conserved DMP1 C terminus. Mechanistic studies using Dmp1-null mice demonstrated that absence of DMP1 results in defective osteocyte maturation and increased FGF23 expression, leading to pathological changes in bone mineralization. Our findings suggest a bone-renal axis that is central to guiding proper mineral metabolism.


Assuntos
Proteínas da Matriz Extracelular/genética , Minerais/metabolismo , Osteócitos/fisiologia , Osteomalacia/genética , Fosfoproteínas/genética , Raquitismo/genética , Adulto , Animais , Osso e Ossos/patologia , Calcificação Fisiológica/genética , Calcificação Fisiológica/fisiologia , Células Cultivadas , Análise Mutacional de DNA , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Humanos , Rim/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteócitos/patologia , Osteomalacia/sangue , Osteomalacia/patologia , Fosfatos/metabolismo , Raquitismo/sangue , Raquitismo/patologia
15.
Proc Natl Acad Sci U S A ; 108(46): E1146-55, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22006328

RESUMO

Autosomal dominant hypophosphatemic rickets (ADHR) is unique among the disorders involving Fibroblast growth factor 23 (FGF23) because individuals with R176Q/W and R179Q/W mutations in the FGF23 (176)RXXR(179)/S(180) proteolytic cleavage motif can cycle from unaffected status to delayed onset of disease. This onset may occur in physiological states associated with iron deficiency, including puberty and pregnancy. To test the role of iron status in development of the ADHR phenotype, WT and R176Q-Fgf23 knock-in (ADHR) mice were placed on control or low-iron diets. Both the WT and ADHR mice receiving low-iron diet had significantly elevated bone Fgf23 mRNA. WT mice on a low-iron diet maintained normal serum intact Fgf23 and phosphate metabolism, with elevated serum C-terminal Fgf23 fragments. In contrast, the ADHR mice on the low-iron diet had elevated intact and C-terminal Fgf23 with hypophosphatemic osteomalacia. We used in vitro iron chelation to isolate the effects of iron deficiency on Fgf23 expression. We found that iron chelation in vitro resulted in a significant increase in Fgf23 mRNA that was dependent upon Mapk. Thus, unlike other syndromes of elevated FGF23, our findings support the concept that late-onset ADHR is the product of gene-environment interactions whereby the combined presence of an Fgf23-stabilizing mutation and iron deficiency can lead to ADHR.


Assuntos
Raquitismo Hipofosfatêmico Familiar/genética , Fatores de Crescimento de Fibroblastos/genética , Deficiências de Ferro , Anemia Ferropriva/complicações , Animais , Raquitismo Hipofosfatêmico Familiar/fisiopatologia , Feminino , Fator de Crescimento de Fibroblastos 23 , Interação Gene-Ambiente , Glucuronidase/metabolismo , Hipofosfatemia/genética , Proteínas Klotho , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Transgênicos , Osteócitos/citologia , Osteomalacia/genética , Fenótipo , Estrutura Terciária de Proteína , Ratos
16.
Proc Jpn Acad Ser B Phys Biol Sci ; 90(10): 405-12, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25504229

RESUMO

Ossification of the posterior longitudinal ligament of the spine (OPLL) is a common disease after the middle age. OPLL frequently causes serious neurological problems due to compression of the spinal cord and/or nerve roots. OPLL occurs in patients with monogenic metabolic diseases including rickets/osteomalacia and hypoparathyroidism; however most of OPLL is idiopathic and is considered as a multi-factorial (polygenic) disease influenced by genetic and environmental factors. Genomic studies for the genetic factors of OPLL have been conducted, mainly in Japan, including linkage and association studies. This paper reviews the recent progress in the genomic study of OPLL and comments on its future direction.


Assuntos
Estudo de Associação Genômica Ampla , Herança Multifatorial , Ossificação do Ligamento Longitudinal Posterior/genética , Animais , Humanos , Hipoparatireoidismo/complicações , Hipoparatireoidismo/genética , Hipoparatireoidismo/patologia , Japão , Ossificação do Ligamento Longitudinal Posterior/patologia , Osteomalacia/complicações , Osteomalacia/genética , Osteomalacia/patologia , Medula Espinal/patologia
17.
Clin Calcium ; 24(2): 169-75, 2014 Feb.
Artigo em Japonês | MEDLINE | ID: mdl-24473349

RESUMO

Calcium and phosphate are the constituents of biomineral, and their levels in circulation are regulated by various hormones. Disorders in calcium/phosphate metabolism are often associated with skeletal demineralization and pathological calcification. Some of the hypophosphatemic rickets/osteomalacia are caused by the increased bioactivity of FGF23, and classified into FGF23-mediated hypophosphatemic rickets/osteomalacia. In the adult patients with FGF23-mediated hypophosphatemic rickets/osteomalacia, mineralizing enthesopathy is an often observed complication. In addition, mutations of the same genes can be responsible for both rickets/osteomalacia and ectopic calcification.


Assuntos
Cálcio/metabolismo , Raquitismo Hipofosfatêmico Familiar/metabolismo , Fosfatos/metabolismo , Animais , Calcificação Fisiológica/fisiologia , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/metabolismo , Humanos , Osteomalacia/etiologia , Osteomalacia/genética , Osteomalacia/metabolismo
18.
Clin Calcium ; 24(6): 879-84, 2014 Jun.
Artigo em Japonês | MEDLINE | ID: mdl-24870839

RESUMO

FGF23 is an endocrine FGF produced by osteocytes, which increases excretion of phosphate and suppresses the production of 1,25 (OH) 2D. Excessive action of FGF23 causes various forms of hypophosphatemic rickets/osteomalacia, while the loss of function of FGF23 results in the condition called familial hyperphosphatemic tumoral calcinosis. 1,25 (OH) 2D stimulates the production of FGF23, and the interaction between FGF23 and 1,25 (OH) 2D plays a central role in mineral homeostasis. In addition to the its roles in mineral homeostasis, recent studies have suggested the direct action of FGF23 on osteoblasts and chondrocytes.


Assuntos
Osso e Ossos/metabolismo , Cartilagem/metabolismo , Fatores de Crescimento de Fibroblastos/fisiologia , Animais , Calcificação Fisiológica/genética , Calcinose/genética , Condrócitos , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/biossíntese , Fatores de Crescimento de Fibroblastos/deficiência , Homeostase/genética , Homeostase/fisiologia , Humanos , Hiperostose Cortical Congênita/genética , Hiperfosfatemia/genética , Osteoblastos , Osteócitos/metabolismo , Osteomalacia/genética , Fosfatos/metabolismo , Raquitismo Hipofosfatêmico/genética , Vitamina D/análogos & derivados , Vitamina D/biossíntese , Vitamina D/fisiologia
19.
Bone ; 182: 117057, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38412893

RESUMO

Excessive actions of FGF23 cause several kinds of hypophosphatemic rickets/osteomalacia. It is possible that there still remain unknown causes or mechanisms for FGF23-related hypophosphatemic diseases. We report two male cousins who had been suffering form FGF23-related hypophosphatemic osteomalacia. Sequencing of exons and exon-intron junctions of known causative genes for FGF23-related hypophosphatemic diseases and whole genome sequencing were conducted. Luciferase assay was used to evaluate the effect of a detected nucleotide change on mRNA stability. Two cousins showed hypophosphatemia with impaired proximal tubular phosphate reabsorption and high FGF23. Serum phosphate of their mothers was within the reference range. Exome sequencing of the proband detected no mutations. Whole genome sequencing of the patients and their mothers identified a nucleotide change in the 3'-UTR of phosphate-regulating gene with homologies to endopeptidases on the X chromosome (PHEX) gene (c.*1280_*1287dupGTGTGTGT) which is heterozygous in the mothers and hemizygous in the patients. While sixteen is the most prevalent number of GT repeats, this family had twenty repeats. Luciferase assay indicated that mRNA with 3'-UTR of PHEX with 20 GT repeats was more unstable than that with 16 repeats. Sequencing of exons and exon-intron junctions of known causative genes for FGF23-related hypophosphatemic diseases cannot identify all the genetic causes. Our results strongly suggest that changes of PHEX expression by a nucleotide change in the 3'-UTR is a novel mechanism of FGF23-related hypophosphatemic osteomalacia.


Assuntos
Doenças Genéticas Ligadas ao Cromossomo X , Osteomalacia , Endopeptidase Neutra Reguladora de Fosfato PHEX , Adulto , Humanos , Masculino , Análise Mutacional de DNA , Raquitismo Hipofosfatêmico Familiar/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Doenças Genéticas Ligadas ao Cromossomo X/genética , Hipofosfatemia , Luciferases/genética , Nucleotídeos , Osteomalacia/genética , Endopeptidase Neutra Reguladora de Fosfato PHEX/genética , Fosfatos
20.
Endocrine ; 84(1): 76-91, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38117452

RESUMO

INTRODUCTION: X-linked hypophosphatemia is an orphan disease of genetic origin and multisystem involvement. It is characterized by a mutation of the PHEX gene which results in excess FGF23 production, with abnormal renal and intestinal phosphorus metabolism, hypophosphatemia and osteomalacia secondary to chronic renal excretion of phosphate. Clinical manifestations include hypophosphatemic rickets leading to growth abnormalities and osteomalacia, myopathy, bone pain and dental abscesses. The transition of these patients to adult life continues to pose challenges to health systems, medical practitioners, patients and families. For this reason, the aim of this consensus is to provide a set of recommendations to facilitate this process and ensure adequate management and follow-up, as well as the quality of life for patients with X-linked hypophosphatemia as they transition to adult life. MATERIALS AND METHODS: Eight Latin American experts on the subject participated in the consensus and two of them were appointed as coordinators. The consensus work was done in accordance with the nominal group technique in 6 phases: (1) question standardization, (2) definition of the maximum number of choices, (3) production of individual solutions or answers, (4) individual question review, (5) analysis and synthesis of the information and (6) synchronic meetings for clarification and voting. An agreement was determined to exist with 80% votes in favor in three voting cycles. RESULTS AND DISCUSSION: Transition to adult life in patients with hypophosphatemia is a complex process that requires a comprehensive approach, taking into consideration medical interventions and associated care, but also the psychosocial components of adult life and the participation of multiple stakeholders to ensure a successful process. The consensus proposes a total of 33 recommendations based on the evidence and the knowledge and experience of the experts. The goal of the recommendations is to optimize the management of these patients during their transition to adulthood, bearing in mind the need for multidisciplinary management, as well as the most relevant medical and psychosocial factors in the region.


Assuntos
Raquitismo Hipofosfatêmico Familiar , Hipofosfatemia , Osteomalacia , Adulto , Humanos , Raquitismo Hipofosfatêmico Familiar/genética , Osteomalacia/genética , Osteomalacia/metabolismo , Consenso , Qualidade de Vida , Hipofosfatemia/genética , Hipofosfatemia/metabolismo , Fatores de Crescimento de Fibroblastos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA