Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
Genes Chromosomes Cancer ; 61(12): 740-746, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35999193

RESUMO

Cutaneous skeletal hypophosphatemia syndrome (CSHS) is caused by somatic mosaic NRAS variants and characterized by melanocytic/sebaceous naevi, eye, and brain malformations, and FGF23-mediated hypophosphatemic rickets. The MEK inhibitor Trametinib, acting on the RAS/MAPK pathway, is a candidate for CSHS therapy. A 4-year-old boy with seborrheic nevus, eye choristoma, multiple hamartomas, brain malformation, pleural lymphangioma and chylothorax developed severe hypophosphatemic rickets unresponsive to phosphate supplementation. The c.182A > G;p.(Gln61Arg) somatic NRAS variant found in DNA from nevus biopsy allowed diagnosing CSHS. We administered Trametinib for 15 months investigating the transcriptional effects at different time points by whole blood RNA-seq. Treatment resulted in prompt normalization of phosphatemia and phosphaturia, catch-up growth, chylothorax regression, improvement of bone mineral density, reduction of epidermal nevus and hamartomas. Global RNA sequencing on peripheral blood mononucleate cells showed transcriptional changes under MEK inhibition consisting in a strong sustained downregulation of signatures related to RAS/MAPK, PI3 kinase, WNT and YAP/TAZ pathways, reverting previously defined transcriptomic signatures. CSHS was effectively treated with a MEK inhibitor with almost complete recovery of rickets and partial regression of the phenotype. We identified "core" genes modulated by MEK inhibition potentially serving as surrogate markers of Trametinib action.


Assuntos
Quilotórax , Hamartoma , Hipofosfatemia , Nevo Pigmentado , Nevo , Raquitismo Hipofosfatêmico , Neoplasias Cutâneas , DNA , GTP Fosfo-Hidrolases/genética , Humanos , Hipofosfatemia/diagnóstico , Hipofosfatemia/genética , Proteínas de Membrana/genética , Quinases de Proteína Quinase Ativadas por Mitógeno , Nevo Pigmentado/diagnóstico , Nevo Pigmentado/genética , Nevo Pigmentado/metabolismo , Fosfatos , Fosfatidilinositol 3-Quinases , Raquitismo Hipofosfatêmico/genética , Neoplasias Cutâneas/genética , Síndrome
3.
Hum Mutat ; 43(9): 1183-1200, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35475527

RESUMO

ENPP1 encodes ENPP1, an ectonucleotidase catalyzing hydrolysis of ATP to AMP and inorganic pyrophosphate (PPi), and an endogenous plasma protein physiologically preventing ectopic calcification of connective tissues. Mutations in ENPP1 have been reported in association with a range of human genetic diseases. In this mutation update, we provide a comprehensive review of all the pathogenic variants, likely pathogenic variants, and variants of unknown significance in ENPP1 associated with three autosomal recessive disorders-generalized arterial calcification of infancy (GACI), autosomal recessive hypophosphatemic rickets type 2 (ARHR2), and pseudoxanthoma elasticum (PXE), as well as with a predominantly autosomal dominant disorder-Cole disease. The classification of all variants is determined using the latest ACMG guidelines. A total of 140 ENPP1 variants were curated consisting of 133 previously reported variants and seven novel variants, with missense variants being the most prevalent (70.0%, 98/140). While the pathogenic variants are widely distributed in the ENPP1 gene of patientsgen without apparent genotype-phenotype correlation, eight out of nine variants associated with Cole disease are confined to the somatomedin-B-like (SMB) domains critical for homo-dimerization of the ENPP1 protein.


Assuntos
Hipopigmentação , Diester Fosfórico Hidrolases , Pirofosfatases , Raquitismo Hipofosfatêmico , Calcificação Vascular , Humanos , Hipopigmentação/genética , Mutação , Diester Fosfórico Hidrolases/genética , Pirofosfatases/genética , Raquitismo Hipofosfatêmico/complicações , Raquitismo Hipofosfatêmico/genética , Calcificação Vascular/genética
4.
Arch Dis Child Educ Pract Ed ; 107(2): 124-126, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-33127660

RESUMO

A 7 year-old twin girl with hypophosphataemic rickets was evaluated for a recent onset of mild strabismus.She was a homozygous twin sister with hypophosphataemic rickets diagnosed at the age of 2 years, with a mutation in intron 21 of the PHEX gene, which was also present in her sister.The girls' clinical histories were remarkable for an important lower limb varus that progressively improved after starting phosphate supplementation with a galenical solution (Joulies solution 1 mmol phosphate/ml) and vitamin D 1,25 OH.During the examinations, both girls were in good general condition. Physical examinations were unremarkable, except for tibial varus, bilateral fifth finger clinodactyly and bilateral syndactyly of the third and fourth foot fingers. No major head shape abnormalities were noticeable except for a high forehead.One patient presented with a slight strabismus, normal isochoric isocyclic and reactive pupils, no signs of cranial nerve deficit, and no alterations in the rest of the neurological examination. An ophthalmological evaluation showed bilateral papilloedema. A cerebral MRI scan was then performed, suspecting elevated intracranial pressure (figure 1). The same examination was performed on the asymptomatic sister which also demonstrated papilloedema with similar findings on cranial MRI too.


Assuntos
Raquitismo Hipofosfatêmico Familiar , Papiledema , Raquitismo Hipofosfatêmico , Estrabismo , Criança , Pré-Escolar , Potenciais Evocados Visuais , Raquitismo Hipofosfatêmico Familiar/diagnóstico , Raquitismo Hipofosfatêmico Familiar/genética , Raquitismo Hipofosfatêmico Familiar/terapia , Feminino , Humanos , Masculino , Fosfatos , Raquitismo Hipofosfatêmico/diagnóstico , Raquitismo Hipofosfatêmico/genética , Raquitismo Hipofosfatêmico/terapia
5.
Development ; 145(15)2018 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-30002128

RESUMO

The bone tendon attachment site known as the enthesis comprises a transitional zone between bone and tendon, and plays an important role in enabling movement at this site. X-linked hypophosphatemia (XLH) is characterized by impaired activation of vitamin D, elevated serum FGF23 levels and low serum phosphate levels, which impair bone mineralization. Paradoxically, an important complication of XLH is mineralization of the enthesis (enthesopathy). Studies were undertaken to identify the cellular and molecular pathways important for normal post-natal enthesis maturation and to examine their role during the development of enthesopathy in mice with XLH (Hyp). The Achilles tendon entheses of Hyp mice demonstrate an expansion of hypertrophic-appearing chondrogenic cells by P14. Post-natally, cells in wild-type and Hyp entheses similarly descend from scleraxis- and Sox9-expressing progenitors; however, Hyp entheses exhibit an expansion of Sox9-expressing cells, and enhanced BMP and IHH signaling. These results support a role for enhanced BMP and IHH signaling in the development of enthesopathy in XLH.


Assuntos
Entesopatia/complicações , Entesopatia/genética , Raquitismo Hipofosfatêmico/complicações , Raquitismo Hipofosfatêmico/genética , Fosfatase Alcalina/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Proliferação de Células/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Modelos Animais de Doenças , Entesopatia/tratamento farmacológico , Entesopatia/patologia , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/farmacologia , Fatores de Crescimento de Fibroblastos/uso terapêutico , Proteínas Hedgehog/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Raquitismo Hipofosfatêmico/tratamento farmacológico , Raquitismo Hipofosfatêmico/patologia , Fatores de Transcrição SOX9/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Vitamina D/análogos & derivados , Vitamina D/farmacologia , Vitamina D/uso terapêutico
6.
Pediatr Dermatol ; 37(3): 541-544, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32157705

RESUMO

We report the case of a child who presented with a giant melanocytic nevus with numerous satellite nevi at birth and developed hypophosphatemic rickets due to excessive secretion of the FGF23 hormone. A NRAS c.182A>G (Q61R) mutation was identified in the lesional skin. The functional outcome was favorable with medical treatment.


Assuntos
Nevo Pigmentado , Nevo , Raquitismo Hipofosfatêmico , Neoplasias Cutâneas , Criança , Fator de Crescimento de Fibroblastos 23 , Humanos , Recém-Nascido , Mutação , Nevo Pigmentado/complicações , Nevo Pigmentado/genética , Raquitismo Hipofosfatêmico/complicações , Raquitismo Hipofosfatêmico/diagnóstico , Raquitismo Hipofosfatêmico/genética
7.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 35(5): 644-647, 2018 Oct 10.
Artigo em Zh | MEDLINE | ID: mdl-30298486

RESUMO

OBJECTIVE: To explore the molecular basis for three pedigrees affected with hypophosphatemia vitamin D resistant rickets (X-linked hypophosphatemia, XLH). METHODS: Peripheral blood samples from the three pedigrees were collected. Following DNA extraction, the 11 exons and flanking regions of the PHEX gene were subjected to PCR amplification and direct sequencing. Pathogenicity of identified mutations was evaluated through genotype-phenotype correlation. RESULTS: For pedigrees 1 and 2, pathogenic mutations were respectively identified in exon 8 (c.871C>T, p.R291X) and exon 15 (c.1601C>T, p.P534L) of the PHEX gene. For pedigree 3, a novel mutation (c.1234delA, p.S412Vfs*12) was found in exon 11 of the PHEX gene, which caused shift the reading frame and premature termination of protein translation. CONCLUSION: The three mutations probably account for the XLH in the affected pedigrees. The discovery of novel mutations has enriched the spectrum of PHEX gene mutations.


Assuntos
Endopeptidase Neutra Reguladora de Fosfato PHEX/genética , Raquitismo Hipofosfatêmico/genética , Adolescente , Adulto , Sequência de Bases , China , Análise Mutacional de DNA , Éxons , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Mutação , Linhagem , Raquitismo Hipofosfatêmico/diagnóstico
8.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 35(5): 638-643, 2018 Oct 10.
Artigo em Zh | MEDLINE | ID: mdl-30298485

RESUMO

OBJECTIVE: To detect potential mutations of PHEX gene in four pedigrees affected with hypophosphatemic rickets (HR) and provide prenatal diagnosis for a fetus at 13th gestational week. METHODS: The coding regions and exon/intron boundaries of PHEX, FGF23, DMP1, ENPP1, CLCN5 and SLC34A3 genes of the probands were analyzed by targeted next-generation sequencing (NGS). Suspected mutations were verified by Sanger sequencing among unaffected relatives and 200 unrelated healthy individuals. Deletions were confirmed by multiplex ligation-dependent probe amplification (MLPA) detection of probands, unaffected relatives and 20 unrelated healthy individuals. Prenatal diagnosis for a fetus with high risk was carried out through MLPA analysis. RESULTS: Four PHEX mutations were respectively detected in the pedigrees, which included c.850-3C>G, exon 11 deletion, exon 13 deletion and c.1753G>A (p.G585R). Among these, exon 11 deletion, exon 13 deletion and c.1753G>A (p.G585R) were novel mutations and not found among unaffected relatives and healthy controls. In pedigree 3, the same mutation was not found in the fetus. CONCLUSION: Mutations of the PHEX gene probably underlies the disease among the four pedigrees. NGS combined with Sanger sequencing and/or MLPA detection can ensure accurate diagnosis for this disease.


Assuntos
Raquitismo Hipofosfatêmico/genética , Adulto , Sequência de Bases , Pré-Escolar , Canais de Cloreto/genética , Análise Mutacional de DNA , Proteínas da Matriz Extracelular/genética , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Íntrons , Masculino , Dados de Sequência Molecular , Mutação , Linhagem , Fosfoproteínas/genética , Diester Fosfórico Hidrolases/genética , Gravidez , Pirofosfatases/genética , Raquitismo Hipofosfatêmico/diagnóstico , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/genética
9.
Clin Endocrinol (Oxf) ; 87(1): 103-112, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28383812

RESUMO

CONTEXT: Hypophosphataemic rickets (HR) is a group of rare hereditary renal phosphate wasting disorders caused by mutations in PHEX, FGF23, DMP1, ENPP1, CLCN5 or SLC34A3. OBJECTIVE: To investigate underlying genetic defects in patients with hypophosphataemic rickets. METHODS: We analysed genomic DNA from nine unrelated families for mutations in the entire coding region of PHEX, FGF23, DMP1, ENPP1, CLCN5 or SLC34A3 by PCR sequencing and copy number analysis. RESULTS: A total of 14 patients were studied. PHEX mutations were identified in 12 patients from seven families. Five of them were novel mutations present in eight patients: c.154G>T (p.E52*), c.401_402insGCCAAA (p.Q134_K135insPK), c.1600C>T (p.P534S), g.22016715_22056805del (40-kb deletion including promoter and exons 1-3) and c.2242_2243delCT (p.L748 fs*48). Four patients had previously reported mutations: c.1768+1G>A and c.1807G>A (p.W602*). Novel CLCN5 (c.1205G>A, p.W402*) and FGF23 (c.526C>G, p.R176G) mutations were found in two patients from the remaining two families. Many of the mutations were de novo: c.154G>T and c.2242_2243delCT in PHEX and c.526C>G in FGF23. Furthermore, we characterized the breakpoint of the novel PHEX g.22016715_22056805del and the c.2242_2243delCT, which is 6 bp from the stop codon, resulting in a frameshift and extension of the reading frame by 42 amino acids. CONCLUSIONS: Novel and de novo mutations are frequent and PHEX mutations are still the most common genetic defects in the Turkish population. Gene copy number analysis should be considered in patients with negative results by conventional PCR-based sequencing analysis. The current study further expands the mutation spectrum underlying HR.


Assuntos
Canais de Cloreto/genética , Análise Mutacional de DNA , Fatores de Crescimento de Fibroblastos/genética , Endopeptidase Neutra Reguladora de Fosfato PHEX/genética , Raquitismo Hipofosfatêmico/genética , Família , Feminino , Fator de Crescimento de Fibroblastos 23 , Dosagem de Genes , Humanos , Masculino , Linhagem , Turquia
10.
Clin Exp Dermatol ; 42(1): 75-79, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27900779

RESUMO

The association of hypophosphataemic rickets with verrucous epidermal naevus (EN) and elevated fibroblast growth factor 23 levels is known as cutaneous-skeletal hypophosphataemia syndrome (CSHS), and can be caused by somatic activating mutations in RAS genes. We report a unique patient with CSHS associated with giant congenital melanocytic naevus (CMN), neurocutaneous melanosis and EN syndrome, manifesting as facial linear sebaceous naevus, developmental delay and ocular dermoids. An activating mutation Q61R in the NRAS gene was found in affected skin and ocular tissue but not blood, implying that the disparate manifestations are due to a multilineage activating mutation (mosaic RASopathy). We speculate on the apparently rare association of CSHS with CMN compared with EN. We also report the favourable outcome of this patient at the age of 8 years after extensive neonatal curettage of the giant CMN and use of vitamin D and phosphate supplementation.


Assuntos
DNA de Neoplasias/genética , GTP Fosfo-Hidrolases/genética , Proteínas de Membrana/genética , Mosaicismo , Nevo Pigmentado/genética , Nevo/genética , Raquitismo Hipofosfatêmico/genética , Neoplasias Cutâneas/genética , Pele/patologia , Pré-Escolar , Análise Mutacional de DNA , GTP Fosfo-Hidrolases/metabolismo , Humanos , Masculino , Proteínas de Membrana/metabolismo , Mutação , Nevo/diagnóstico , Nevo/metabolismo , Nevo Pigmentado/diagnóstico , Nevo Pigmentado/metabolismo , Raquitismo Hipofosfatêmico/congênito , Raquitismo Hipofosfatêmico/diagnóstico , Neoplasias Cutâneas/diagnóstico , Neoplasias Cutâneas/metabolismo
11.
J Hum Genet ; 61(3): 223-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26559751

RESUMO

X-linked dominant hypophosphatemic rickets (XLHR), is characterized mainly by renal phosphate wasting with hypophosphatemia, short stature and abnormal bone mineralization. PHEX, located at Xp22.1-p22.2, is the gene causing XLHR. We aim to characterize the pathogenesis of a Chinese boy who is apparently 'heterozygous' in PHEX gene. Direct sequencing showed two peaks: one was a wild-type 'G' and the other was one base substitution to 'A', though the patient was a male. TA clone assay clearly showed each sequences and the ratios. The mutation effect was predicted via bioinformatics and validated by exon-trapping assay. Real-time PCR was applied to determine the copy number of PHEX. TA clone assay showed the frequency of normal (G) to mutant allele (A) as 19:13. Normal karyotype and real-time PCR results indicate the normal copy number of PHEX. This splice site mutation leads to 4 bp of exon 18 skipping out causing frame shift p.Gly590Glufs*28 that ends up with a loss of active site and Zn(2+)-binding site of PHEX, which probably interfere with renal phosphate reabsorption and bone mineralization. In conclusion, mutation at conserved splice acceptor site resulted in aberrant splicing, ending up with a damaged protein product. This novel mutation is de novo in mosaic pattern that may be induced during early postzygotic period. Taking mosaic somatic mutation of PHEX into consideration is strongly suggested in genetic counseling and etiology research for XLHR.


Assuntos
Mosaicismo , Mutação , Endopeptidase Neutra Reguladora de Fosfato PHEX/genética , Raquitismo Hipofosfatêmico/genética , Criança , Humanos , Cariotipagem , Masculino , Reação em Cadeia da Polimerase em Tempo Real
12.
Am J Med Genet A ; 170A(5): 1308-11, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26857895

RESUMO

Patients with generalized arterial calcification of infancy (GACI) develop vascular calcifications early in life. About half of them die within the first 6 months despite optimal medical care. A subset of those who survive eventually develop hypophosphatemic rickets. Since hypophosphatemia and hyperphosphaturia have been previously associated with increased survival in GACI patients, physicians often avoid phosphate repletion as treatment for rickets. As a consequence, GACI patients develop severe rachitic complications such as short stature and skeletal deformities. It appears that the recognition of hypophosphatemia later in life in some GACI patients is a consequence of having survived the first few months of life, and not the cause of their survival per se. Here, we report the long-term follow-up of a GACI patient who was phosphate-repleted for his rickets for more than 7 years without worsening of vascular calcification.


Assuntos
Diester Fosfórico Hidrolases/genética , Pirofosfatases/genética , Raquitismo Hipofosfatêmico/tratamento farmacológico , Calcificação Vascular/tratamento farmacológico , Adolescente , Adulto , Criança , Seguimentos , Humanos , Masculino , Mutação , Fosfatos/administração & dosagem , Raquitismo Hipofosfatêmico/genética , Raquitismo Hipofosfatêmico/fisiopatologia , Calcificação Vascular/genética , Calcificação Vascular/fisiopatologia
13.
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
14.
Am J Med Genet A ; 167A(10): 2430-4, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26059403

RESUMO

We report on a boy who presented with hypophosphatemic rickets with elevated serum fibroblast growth factor 23 (FGF23) and polyostotic osteolytic lesions at age 2 years. Tumor-induced hypophosphatemic rickets was suspected; however, bone biopsy for osteolytic changes revealed no tumorous change, except for irregularly dilated vessels associated with osteoclasts and fibrous proliferation. Venous sampling failed to point to FGF23-producing foci. After alfacalcidol and phosphate supplementation, the rachitic skeletal changes improved, but FGF23 increased and new osteolytic lesions developed. Serum levels of neopterin and a few cytokines, including plasma transforming growth factor-ß and soluble tumor necrosis factor receptor type II, were elevated. At age 4 years, high doses of phosphate resulted in increased serum phosphate levels, decreased neopterin and cytokines, decreased FGF23, and stabilization of osteolysis. We excluded germline mutations in PHEX, FGF23, DMP1, and ENPP1 (genes for hereditary hypophosphatemic rickets) and somatic mutations in the GNAS and HRAS/KRAS (the disease-causing genes for McCune-Albright syndrome and linear nevus sebaceous syndrome, respectively). We could not perform octreotide scintigraphy or fluorodeoxyglucose-positron emission tomography, and thus could not completely exclude occult FGF23-producing tumors. However, considering the course of the disease, it is intriguing to assume that dysregulation of osteoclast-macrophage lineage may have induced increased neopterin levels, increased cytokine levels, osteolytic process, and possibly FGF23 overproduction.


Assuntos
Fatores de Crescimento de Fibroblastos/genética , Displasia Fibrosa Poliostótica/genética , Osteólise/genética , Raquitismo Hipofosfatêmico/genética , Proliferação de Células , Pré-Escolar , Citocinas/sangue , Citocinas/genética , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Displasia Fibrosa Poliostótica/sangue , Displasia Fibrosa Poliostótica/tratamento farmacológico , Displasia Fibrosa Poliostótica/patologia , Regulação da Expressão Gênica , Humanos , Hidroxicolecalciferóis/uso terapêutico , Masculino , Neopterina/sangue , Neopterina/genética , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteólise/sangue , Osteólise/tratamento farmacológico , Osteólise/patologia , Fosfatos/uso terapêutico , Receptores Tipo II do Fator de Necrose Tumoral/sangue , Receptores Tipo II do Fator de Necrose Tumoral/genética , Raquitismo Hipofosfatêmico/sangue , Raquitismo Hipofosfatêmico/tratamento farmacológico , Raquitismo Hipofosfatêmico/patologia , Fator de Crescimento Transformador beta/sangue , Fator de Crescimento Transformador beta/genética
15.
Curr Osteoporos Rep ; 13(2): 88-97, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25620749

RESUMO

Fibroblast growth factor-23 (FGF23) regulates phosphate reabsorption in the kidney and therefore plays an essential role in phosphate balance in humans. There is a host of defects that ultimately lead to excess FGF23 levels and thereby cause renal phosphate wasting and hypophosphatemic rickets. We describe the genetic, pathophysiologic, and clinical aspects of this group of disorders with a focus on X-linked hypophosphatemia (XLH), the best characterized of these abnormalities. We also discuss autosomal dominant hypophosphatemic rickets (ADHR), autosomal recessive hypophosphatemic rickets (ARHR) and tumor-induced osteomalacia (TIO) in addition to other rarer FGF23-mediated conditions. We contrast the FGF23-mediated disorders with FGF23-independent hypophosphatemia, specifically hypophosphatemic rickets with hypercalciuria (HHRH). Errant diagnosis of hypophosphatemic disorders is common. This review aims to enhance the recognition and appropriate diagnosis of hypophosphatemia and to guide appropriate treatment.


Assuntos
Fatores de Crescimento de Fibroblastos/fisiologia , Homeostase/fisiologia , Fósforo/metabolismo , Raquitismo Hipofosfatêmico/fisiopatologia , Osso e Ossos/metabolismo , Calcitonina/uso terapêutico , Fator de Crescimento de Fibroblastos 23 , Humanos , Osteomalacia/fisiopatologia , Raquitismo Hipofosfatêmico/diagnóstico , Raquitismo Hipofosfatêmico/genética
16.
Pediatr Nephrol ; 29(3): 361-73, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23636577

RESUMO

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.


Assuntos
Lâmina de Crescimento/metabolismo , Túbulos Renais Proximais/metabolismo , Osteogênese , Fosfatos/metabolismo , Raquitismo Hipofosfatêmico/etiologia , Fatores Etários , Predisposição Genética para Doença , Lâmina de Crescimento/fisiopatologia , Humanos , Túbulos Renais Proximais/fisiopatologia , Prognóstico , Raquitismo Hipofosfatêmico/diagnóstico , Raquitismo Hipofosfatêmico/genética , Raquitismo Hipofosfatêmico/metabolismo , Raquitismo Hipofosfatêmico/fisiopatologia , Raquitismo Hipofosfatêmico/terapia , Fatores de Risco
17.
Curr Osteoporos Rep ; 12(3): 252-62, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24980542

RESUMO

Rapid and somewhat surprising advances have recently been made toward understanding the molecular mechanisms causing heritable disorders of hypophosphatemia. The results of clinical, genetic, and translational studies have interwoven novel concepts underlying the endocrine control of phosphate metabolism, with far-reaching implications for treatment of both rare Mendelian diseases as well as common disorders of blood phosphate excess such as chronic kidney disease (CKD). In particular, diseases caused by changes in the expression and proteolytic control of the phosphaturic hormone fibroblast growth factor-23 (FGF23) have come to the forefront in terms of directing new models explaining mineral metabolism. These hypophosphatemic disorders as well as others resulting from independent defects in phosphate transport or metabolism will be reviewed herein, and implications for emerging therapeutic strategies based upon these new findings will be discussed.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Hiperfosfatemia/metabolismo , Fosfatos/metabolismo , Raquitismo Hipofosfatêmico/metabolismo , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Homeostase , Humanos , Hiperfosfatemia/genética , Raquitismo Hipofosfatêmico/genética
19.
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
20.
Stem Cell Res ; 77: 103439, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38761687

RESUMO

Hypophosphatemic vitamin D-resistant rickets typically presents in infancy or early childhood with skeletal deformities and growth plate abnormalities. In this report, the SMUSHi005-A human induced pluripotent stem cell (hiPSC) line was successfully established from the PBMCs of a female patient carrying the PHEX mutation with c.1586-1586+1 delAG. The iPSC line has been confirmed to have a normal karyotype. The displayed cells clearly exhibit characteristics similar to embryonic stem cells, expressing pluripotency markers and demonstrating the ability to differentiate into three germ layers.


Assuntos
Células-Tronco Pluripotentes Induzidas , Mutação , Endopeptidase Neutra Reguladora de Fosfato PHEX , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Feminino , Endopeptidase Neutra Reguladora de Fosfato PHEX/genética , Linhagem Celular , Raquitismo Hipofosfatêmico Familiar/genética , Raquitismo Hipofosfatêmico Familiar/patologia , Diferenciação Celular , Raquitismo Hipofosfatêmico/genética , Vitamina D/análogos & derivados
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA