Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
J Clin Endocrinol Metab ; 108(9): e754-e768, 2023 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-36916904

RESUMO

CONTEXT: Kenny-Caffey syndrome (KCS) is a rare hereditary disorder characterized by short stature, hypoparathyroidism, and electrolyte disturbances. KCS1 and KCS2 are caused by pathogenic variants in TBCE and FAM111A, respectively. Clinically the phenotypes are difficult to distinguish. OBJECTIVE: The objective was to determine and expand the phenotypic spectrum of KCS1 and KCS2 in order to anticipate complications that may arise in these disorders. METHODS: We clinically and genetically analyzed 10 KCS2 patients from 7 families. Because we found unusual phenotypes in our cohort, we performed a systematic review of genetically confirmed KCS cases using PubMed and Scopus. Evaluation by 3 researchers led to the inclusion of 26 papers for KCS1 and 16 for KCS2, totaling 205 patients. Data were extracted following the Cochrane guidelines and assessed by 2 independent researchers. RESULTS: Several patients in our KCS2 cohort presented with intellectual disability (3/10) and chronic kidney disease (6/10), which are not considered common findings in KCS2. Systematic review of all reported KCS cases showed that the phenotypes of KCS1 and KCS2 overlap for postnatal growth retardation (KCS1: 52/52, KCS2: 23/23), low parathyroid hormone levels (121/121, 16/20), electrolyte disturbances (139/139, 24/27), dental abnormalities (47/50, 15/16), ocular abnormalities (57/60, 22/23), and seizures/spasms (103/115, 13/16). Symptoms more prevalent in KCS1 included intellectual disability (74/80, 5/24), whereas in KCS2 bone cortical thickening (1/18, 16/20) and medullary stenosis (7/46, 27/28) were more common. CONCLUSION: Our case series established chronic kidney disease as a new feature of KCS2. In the literature, we found substantial overlap in the phenotypic spectra of KCS1 and KCS2, but identified intellectual disability and the abnormal bone phenotype as the most distinguishing features.


Assuntos
Hiperostose Cortical Congênita , Hipoparatireoidismo , Deficiência Intelectual , Humanos , Deficiência Intelectual/genética , Deficiência Intelectual/diagnóstico , Hiperostose Cortical Congênita/genética , Fenótipo , Eletrólitos , Hipoparatireoidismo/genética
3.
Front Endocrinol (Lausanne) ; 13: 1008800, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36213261

RESUMO

Background: Hyperphosphatemic familial tumoral calcinosis (HFTC) is a rare disease characterized by hyperphosphatemia and ectopic calcification, predominantly at periarticular locations. This study was performed to characterize the clinical profile of tumoral calcinosis and to identify gene mutations associated with HFTC and elucidated its pathogenic role. Methods: The three subjects (two male and one female) were aged 30, 25 and 15 years, respectively. The clinical features, histopathological findings, and outcomes of three subjects with HFTC were retrospectively reviewed. The three subjects were analyzed for FGF23, GALNT3 and KL mutations. Function of mutant gene was analyzed by western blotting and wheat germ agglutinin affinity chromatography. Results: All subjects had hyperphosphatemia and elevated calcium-phosphorus product. Calcinosis positions included the left shoulder, left index finger, and right hip. Bone and joint damage were present in two cases and multiple foci influenced body growth in one case. The histopathological features were firm, rubbery masses comprising multiple nodules of calcified material bordered by the proliferation of mononuclear or multinuclear macrophages, osteoclastic-like giant cells, fibroblasts, and chronic inflammatory cells. The novel mutation c.484A>G (p.N162D) in exon 3 of FGF23 was identified in one subject and his family members. Measurement of circulating FGF23 in the subject confirmed low intact FGF23 and increased C-terminal fragment. In vitro experiments showed that the mutant FGF23 proteins had defective O-glycosylation and impaired protein proteolysis protection. Conclusion: We identified a novel FGF23 missense mutation, and confirmed its damaging role in FGF23 protein O-glycosylation. Our findings expand the current spectrum of FGF23 variations that influence phosphorus metabolism.


Assuntos
Calcinose , Hiperostose Cortical Congênita , Hiperfosfatemia , Calcinose/genética , Calcinose/patologia , Cálcio/metabolismo , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Glicosilação , Humanos , Hiperostose Cortical Congênita/genética , Hiperfosfatemia/complicações , Hiperfosfatemia/genética , Hiperfosfatemia/patologia , Masculino , Proteínas Mutantes/genética , Mutação , Fósforo , Estudos Retrospectivos , Aglutininas do Germe de Trigo/genética , Aglutininas do Germe de Trigo/metabolismo
4.
Sci Rep ; 12(1): 10211, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35715480

RESUMO

Autosomal dominant mutations in FAM111A are causative for Kenny-Caffey syndrome type 2. Patients with Kenny-Caffey syndrome suffer from severe growth retardation, skeletal dysplasia, hypoparathyroidism, hypocalcaemia, hyperphosphataemia and hypomagnesaemia. While recent studies have reported FAM111A to function in antiviral response and DNA replication, its role in regulating electrolyte homeostasis remains unknown. In this study, we assessed the role of FAM111A in the regulation of serum electrolyte balance using a Fam111a knockout (Fam111a-/-) C57BL/6 N mouse model. Fam111a-/- mice displayed normal weight and serum parathyroid hormone (PTH) concentration and exhibited unaltered magnesium, calcium and phosphate levels in serum and 24-hour urine. Expression of calciotropic (including Cabp28k, Trpv5, Klotho and Cyp24a1), magnesiotropic (including Trpm6, Trpm7, Cnnm2 and Cnnm4) and phosphotropic (Slc20a1, Slc20a2, Slc34a1 and Slc34a3) genes in the kidneys, duodenum and colon were not affected by Fam111a depletion. Only Slc34a2 expression was significantly upregulated in the duodenum, but not in the colon. Analysis of femurs showed unaffected bone morphology and density in Fam111a-/- mice. Kidney and parathyroid histology were also normal in Fam111a-/- mice. In conclusion, our study is the first to characterise the function of FAM111A in vivo and we report that mice lacking FAM111A exhibit normal electrolyte homeostasis on a standard diet.


Assuntos
Hiperostose Cortical Congênita , Hipocalcemia , Serina Proteases , Canais de Cátion TRPM , Animais , Humanos , Camundongos , Cálcio/metabolismo , Eletrólitos/metabolismo , Hiperostose Cortical Congênita/genética , Hipocalcemia/genética , Magnésio/metabolismo , Camundongos Endogâmicos C57BL , Hormônio Paratireóideo/metabolismo , Receptores Virais , Serina Proteases/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/metabolismo , Canais de Cátion TRPM/metabolismo , Equilíbrio Hidroeletrolítico
5.
Genes (Basel) ; 13(2)2022 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-35205306

RESUMO

Osteocraniostenosis (OCS, OMIM #602361) is a severe, usually lethal condition characterized by gracile bones with thin diaphyses, a cloverleaf-shaped skull and splenic hypo/aplasia. The condition is caused by heterozygous mutations in the FAM111A gene and is allelic to the non-lethal, dominant disorder Kenny-Caffey syndrome (KCS, OMIM #127000). Here we report two new cases of OCS, including one with a detailed pathological examination. We review the main diagnostic signs of OCS both before and after birth based on our observations and on the literature. We then review the current knowledge on the mutational spectrum of FAM111A associated with either OCS or KCS, including three novel variants, both from one of the OCS fetuses described here, and from further cases diagnosed at our centers. This report refines the previous knowledge on OCS and expands the mutational spectrum that results in either OCS or KCS.


Assuntos
Doenças do Desenvolvimento Ósseo , Hiperostose Cortical Congênita , Doenças do Desenvolvimento Ósseo/diagnóstico , Doenças do Desenvolvimento Ósseo/genética , Doenças do Desenvolvimento Ósseo/patologia , Anormalidades Craniofaciais , Feminino , Feto/diagnóstico por imagem , Feto/patologia , Humanos , Hiperostose Cortical Congênita/diagnóstico , Hiperostose Cortical Congênita/genética , Hiperostose Cortical Congênita/patologia , Recém-Nascido , Gravidez , Ultrassonografia Pré-Natal
6.
J Clin Res Pediatr Endocrinol ; 14(2): 239-243, 2022 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-33685073

RESUMO

Hyperphosphatemic familial tumoral calcinosis (HFTC) is a rare autosomal recessive disorder. Topical sodium thiosulfate (STS) and acetazolamide can be a safe and effective treatment for patients who do not respond to conventional therapy for ectopic calcifications. We report the successful treatment of deep soft-tissue calcifications with topical STS and acetazolamide in a boy diagnosed with HFTC due to a novel homozygous mutation of FGF23.


Assuntos
Acetazolamida , Hiperostose Cortical Congênita , Tiossulfatos , Acetazolamida/uso terapêutico , Calcinose , Fator de Crescimento de Fibroblastos 23/genética , Fatores de Crescimento de Fibroblastos/genética , Humanos , Hiperostose Cortical Congênita/diagnóstico , Hiperostose Cortical Congênita/tratamento farmacológico , Hiperostose Cortical Congênita/genética , Hiperfosfatemia , Masculino , Mutação , Tiossulfatos/uso terapêutico
7.
Am J Med Genet A ; 185(6): 1903-1907, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33750016

RESUMO

Kenny-Caffey syndrome type 2 (KCS2) and osteocraniostenosis (OCS) are allelic disorders caused by heterozygous pathogenic variants in the FAM111A gene. Both conditions are characterized by gracile bones, characteristic facial features, hypomineralized skull with delayed closure of fontanelles and hypoparathyroidism. OCS and KCS2 are often referred to as FAM111A-related syndromes as a group; although OCS presents with a more severe, perinatal lethal phenotype. We report a novel FAM111A mutation in a fetus with poorly ossified skull, proportionate long extremities with thin diaphysis, and hypoplastic spleen consistent with FAM111A-related syndromes. Trio whole exome sequencing identified a p.Y562S de novo missense variant in the FAM111A gene. The variant shows significant similarity to other reported pathogenic mutations fitting proposed pathophysiologic mechanism which provide sufficient evidence for classification as likely pathogenic. Our report contributed a novel variant to the handful of OCS and KCS2 cases reported with pathogenic variants.


Assuntos
Anormalidades Múltiplas/genética , Doenças do Desenvolvimento Ósseo/genética , Anormalidades Craniofaciais/genética , Nanismo/genética , Hiperostose Cortical Congênita/genética , Hipocalcemia/genética , Receptores Virais/genética , Anormalidades Múltiplas/diagnóstico por imagem , Anormalidades Múltiplas/patologia , Doenças do Desenvolvimento Ósseo/diagnóstico , Doenças do Desenvolvimento Ósseo/diagnóstico por imagem , Doenças do Desenvolvimento Ósseo/patologia , Anormalidades Cardiovasculares/diagnóstico , Anormalidades Cardiovasculares/genética , Anormalidades Cardiovasculares/patologia , Anormalidades Craniofaciais/diagnóstico , Anormalidades Craniofaciais/diagnóstico por imagem , Anormalidades Craniofaciais/patologia , Nanismo/diagnóstico , Nanismo/diagnóstico por imagem , Nanismo/patologia , Ossos Faciais/anormalidades , Ossos Faciais/patologia , Feminino , Feto , Predisposição Genética para Doença , Heterozigoto , Humanos , Hiperostose Cortical Congênita/diagnóstico , Hiperostose Cortical Congênita/diagnóstico por imagem , Hiperostose Cortical Congênita/patologia , Hipocalcemia/diagnóstico , Hipocalcemia/diagnóstico por imagem , Hipocalcemia/patologia , Masculino , Mutação/genética , Gravidez , Crânio/anormalidades , Crânio/patologia , Baço/anormalidades , Baço/diagnóstico por imagem , Sequenciamento do Exoma
8.
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
9.
Am J Med Genet A ; 182(12): 3029-3034, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33010201

RESUMO

Kenny-Caffey syndrome (KCS) is a rare hereditary skeletal disorder involving hypoparathyroidism. The autosomal dominant form (KCS2), caused by heterozygous pathogenic variants in the FAM111A gene, is distinguished from the autosomal recessive form (KCS1) and Sanjad-Sakati syndrome (SSS), both caused by pathogenic variants in the tubulin folding cofactor E (TBCE) gene, by the absence of microcephaly and intellectual disability. We present a patient with KCS2 caused by a de novo pathogenic variant c.1706G>A (p.Arg569His) in FAM111A gene, presenting intellectual disability and microcephaly, which are considered to be typical signs of SSS. We suggest that KCS1, KCS2, and SSS may not represent mutually exclusive clinical entities, but possibly an overlapping spectrum.


Assuntos
Anormalidades Múltiplas/patologia , Nanismo/patologia , Transtornos do Crescimento/patologia , Hiperostose Cortical Congênita/patologia , Hipocalcemia/patologia , Hipoparatireoidismo/patologia , Deficiência Intelectual/patologia , Mutação , Osteocondrodisplasias/patologia , Fenótipo , Receptores Virais/genética , Convulsões/patologia , Anormalidades Múltiplas/genética , Adolescente , Nanismo/complicações , Nanismo/genética , Transtornos do Crescimento/complicações , Transtornos do Crescimento/genética , Humanos , Hiperostose Cortical Congênita/complicações , Hiperostose Cortical Congênita/genética , Hipocalcemia/complicações , Hipocalcemia/genética , Hipoparatireoidismo/complicações , Hipoparatireoidismo/genética , Deficiência Intelectual/complicações , Deficiência Intelectual/genética , Masculino , Osteocondrodisplasias/complicações , Osteocondrodisplasias/genética , Convulsões/complicações , Convulsões/genética
10.
J Biol Chem ; 295(5): 1411-1425, 2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-31882545

RESUMO

The importance of the microbiome in health and its disruption in disease is continuing to be elucidated. However, the multitude of host and environmental factors that influence the microbiome are still largely unknown. Here, we examined UDP-GalNAc:polypeptide N-acetylgalactosaminyltransferase 3 (Galnt3)-deficient mice, which serve as a model for the disease hyperphosphatemic familial tumoral calcinosis (HFTC). In HFTC, loss of GALNT3 activity in the bone is thought to lead to altered glycosylation of the phosphate-regulating hormone fibroblast growth factor 23 (FGF23), resulting in hyperphosphatemia and subdermal calcified tumors. However, GALNT3 is expressed in other tissues in addition to bone, suggesting that systemic loss could result in other pathologies. Using semiquantitative real-time PCR, we found that Galnt3 is the major O-glycosyltransferase expressed in the secretory cells of salivary glands. Additionally, 16S rRNA gene sequencing revealed that the loss of Galnt3 resulted in changes in the structure, composition, and stability of the oral microbiome. Moreover, we identified the major secreted salivary mucin, Muc10, as an in vivo substrate of Galnt3. Given that mucins and their O-glycans are known to interact with various microbes, our results suggest that loss of Galnt3 decreases glycosylation of Muc10, which alters the composition and stability of the oral microbiome. Considering that oral findings have been documented in HFTC patients, our study suggests that investigating GALNT3-mediated changes in the oral microbiome may be warranted.


Assuntos
Calcinose/metabolismo , Calcinose/microbiologia , Hiperostose Cortical Congênita/metabolismo , Hiperostose Cortical Congênita/microbiologia , Hiperfosfatemia/metabolismo , Hiperfosfatemia/microbiologia , Microbiota/genética , N-Acetilgalactosaminiltransferases/metabolismo , Glândulas Salivares/metabolismo , Animais , Calcinose/genética , Feminino , Fator de Crescimento de Fibroblastos 23 , Glicosilação , Glicosiltransferases/metabolismo , Hiperostose Cortical Congênita/genética , Hiperfosfatemia/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucinas/química , Mucinas/metabolismo , N-Acetilgalactosaminiltransferases/genética , Polissacarídeos/metabolismo , RNA Ribossômico 16S/genética , Polipeptídeo N-Acetilgalactosaminiltransferase
12.
Turk J Pediatr ; 61(1): 130-133, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31559735

RESUMO

Albaramki J, Dmour H, Shboul M, Bonnard C, Venkatesh B, Odeh R. Recessive mutation in GALNT3 causes hyperphosphatemic familial tumoral calcinosis associated with chronic recurrent multifocal osteomyelitis. Turk J Pediatr 2019; 61: 130-133. Hyperphosphatemic familial tumoral calcinosis is a rare autosomal recessive disorder that is characterized by persistent hyperphosphatemia and extra-articular calcifications. Three cases were previously reported with hyperphosphatemic familial tumoral calcinosis that were associated with chronic recurrent multifocal osteomyelitis, an autoinflammatory disorder that is characterized by recurrent episodes of bone pain. We describe here an 11-year-old child who was diagnosed with these two conditions and was found to carry a splice site mutation c.1524+1G > A in the GALNT3 gene.


Assuntos
Calcinose/genética , Hiperostose Cortical Congênita/genética , Hiperfosfatemia/genética , Mutação , N-Acetilgalactosaminiltransferases/genética , Osteomielite/genética , Criança , Humanos , Masculino , Polipeptídeo N-Acetilgalactosaminiltransferase
13.
J Exp Med ; 216(8): 1944-1964, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-31196982

RESUMO

High bone mass (HBM) is usually caused by gene mutations, and its mechanism remains unclear. In the present study, we identified a novel mutation in the long noncoding RNA Reg1cp that is associated with HBM. Subsequent analysis in 1,465 Chinese subjects revealed that heterozygous Reg1cp individuals had higher bone density compared with subjects with WT Reg1cp Mutant Reg1cp increased the formation of the CD31hiEmcnhi endothelium in the bone marrow, which stimulated angiogenesis during osteogenesis. Mechanistically, mutant Reg1cp directly binds to Krüppel-like factor 3 (KLF3) to inhibit its activity. Mice depleted of Klf3 in endothelial cells showed a high abundance of CD31hiEmcnhi vessels and increased bone mass. Notably, we identified a natural compound, Ophiopogonin D, which functions as a KLF3 inhibitor. Administration of Ophiopogonin D increased the abundance of CD31hiEmcnhi vessels and bone formation. Our findings revealed a specific mutation in lncRNA Reg1cp that is involved in the pathogenesis of HBM and provides a new target to treat osteoporosis.


Assuntos
Hiperostose Cortical Congênita/genética , Hiperostose Cortical Congênita/metabolismo , Fatores de Transcrição Kruppel-Like/antagonistas & inibidores , Mutação , Osteopetrose/genética , Osteopetrose/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Densidade Óssea/genética , China , Estudos de Coortes , Células Progenitoras Endoteliais/metabolismo , Feminino , Heterozigoto , Humanos , Hiperostose Cortical Congênita/sangue , Hiperostose Cortical Congênita/patologia , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Neovascularização Fisiológica/genética , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Osteopetrose/sangue , Osteopetrose/patologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Saponinas/administração & dosagem , Saponinas/farmacologia , Sialoglicoproteínas/metabolismo , Espirostanos/administração & dosagem , Espirostanos/farmacologia , Adulto Jovem
15.
J Clin Res Pediatr Endocrinol ; 11(1): 94-99, 2019 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-30015621

RESUMO

Inactivating autosomal recessive mutations in fibroblast growth factor 23 (FGF23), klotho (KL) and polypeptide N-acetylgalactosaminotransferase 3 (GALNT3) genes lead to a rare disorder, hyperphosphatemic familial tumoral calcinosis (HFTC). Patients with HFTC present with hyperphosphatemia and tumor like soft tissue calcifications. Although 78% of patients develop their first symptoms between the ages of 2-13 years, diagnosis is usually delayed until adulthood. Some individuals with the same genetic defect develop a condition named hyperphosphatemic hyperostosis syndrome. Herein we report two siblings suffering from periarticular, warm, hard and tender subcutaneous masses. Subcutaneous calcifications were present on X-ray and biopsy results were consistent with calcinosis in both patients. Laboratory results showed marked hyperphosphatemia and elevated renal tubular phosphate reabsorption rates, normal renal function tests and normal serum 25-hydroxyvitamin D levels. Thus, we suspected HFTC and performed next generation sequencing for the GALNT3 gene, reported as the most frequent cause. A novel homozygote P85Rfs*6 (c.254_255delCT) mutation in GALNT3 was identified in both siblings. Our report adds two new patients to the literature about this rare genetic disease and suggests that small deletions in the GALNT3 gene may be related with HFTC phenotype.


Assuntos
Calcinose/genética , Predisposição Genética para Doença/genética , Hiperostose Cortical Congênita/genética , Hiperfosfatemia/genética , Mutação , N-Acetilgalactosaminiltransferases/genética , Irmãos , Calcinose/diagnóstico , Criança , Saúde da Família , Feminino , Fator de Crescimento de Fibroblastos 23 , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Hiperostose Cortical Congênita/diagnóstico , Hiperfosfatemia/diagnóstico , Linhagem , Turquia , Polipeptídeo N-Acetilgalactosaminiltransferase
16.
Osteoporos Int ; 29(9): 1987-2009, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29923062

RESUMO

Hyperphosphatemic familial tumoral calcinosis (HFTC), secondary to fibroblast growth factor 23 (FGF23) gene mutation, is a rare genetic disorder characterized by recurrent calcified masses. We describe young Lebanese cousins presenting with HFTC, based on a retrospective chart review and a prospective case study. In addition, we present a comprehensive review on the topic, based on a literature search conducted in PubMed and Google Scholar, in 2014 and updated in December 2017. While the patients had the same previously reported FGF23 gene mutation (homozygous c.G367T variant in exon 3 leading to a missense mutation), they presented with variable severity and age of disease onset (at 4 years in patient 1 and at 23 years in patient 2). A review of the literature revealed several potential patho-physiologic pathways of HFTC clinical manifestations, some of which may be independent of hyperphosphatemia. Most available treatment options aim at reducing serum phosphate level, by stimulating renal excretion or by inhibiting intestinal absorption. HFTC is a challenging disease. While the available medical treatment has a limited and inconsistent effect on disease symptomatology, surgical resection of calcified masses remains the last resort. Research is needed to determine the safety and efficacy of FGF23 replacement or molecular therapy, targeting the specific genetic aberration. Hyperphosphatemic familial tumoral calcinosis is a rare genetic disorder characterized by recurrent calcified masses, in addition to other visceral, skeletal, and vascular manifestations. It remains a very challenging disease.


Assuntos
Calcinose/genética , Fatores de Crescimento de Fibroblastos/genética , Hiperostose Cortical Congênita/genética , Hiperfosfatemia/genética , Mutação , Adolescente , Adulto , Densidade Óssea/genética , Calcinose/diagnóstico por imagem , Calcinose/patologia , Ecocardiografia , Feminino , Fator de Crescimento de Fibroblastos 23 , Humanos , Hiperostose Cortical Congênita/diagnóstico por imagem , Hiperostose Cortical Congênita/patologia , Hiperfosfatemia/diagnóstico por imagem , Hiperfosfatemia/patologia , Masculino , Linhagem , Estudos Prospectivos , Radiografia , Estudos Retrospectivos , Tomografia Computadorizada por Raios X
17.
Am J Kidney Dis ; 72(3): 457-461, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29548779

RESUMO

Primary tumoral calcinosis is a rare autosomal recessive disorder characterized by ectopic calcified tumoral masses. Mutations in 3 genes (GALNT3, FGF23, and KL) have been linked to this human disorder. We describe a case of a 28-year-old man with a history of painful firm masses over his right and left gluteal region, right clavicle region, knees, and left elbow. Biochemical analysis disclosed hyperphosphatemia (phosphate, 9.0 mg/dL) and normocalcemia (calcium, 4.8 mg/dL), with normal kidney function and fractional excretion of phosphate of 3%. Parathyroid hormone was suppressed (15 pg/mL), associated with a low-normal 25-hydroxyvitamin D (26 ng/mL) concentration but high 1,25-dihydroxyvitamin D concentration (92 pg/mL). Serum intact FGF-23 (fibroblast growth factor 23) was undetectable. Genetic analysis revealed tumoral calcinosis due to a compound heterozygous mutation in FGF23, c.201G>C (p.Gln67His) and c.466C>T (p.Gln156*). Due to lack of other treatment options and because the patient was facing severe vascular complications, we initiated a daily hemodialysis program even in the setting of normal kidney function. This unusual therapeutic option successful controlled hyperphosphatemia and reduced metastatic tumoral lesions. This is a report of a new mutation in FGF23 in which dialysis was an effective treatment option for tumoral calcinosis with normal kidney function.


Assuntos
Calcinose/genética , Calcinose/terapia , Fatores de Crescimento de Fibroblastos/genética , Hiperostose Cortical Congênita/genética , Hiperostose Cortical Congênita/terapia , Hiperfosfatemia/genética , Hiperfosfatemia/terapia , Rim/fisiologia , Mutação/genética , Diálise Renal , Adulto , Calcinose/diagnóstico por imagem , Fator de Crescimento de Fibroblastos 23 , Humanos , Hiperostose Cortical Congênita/diagnóstico por imagem , Hiperfosfatemia/diagnóstico por imagem , Masculino , Diálise Renal/métodos , Resultado do Tratamento
18.
Pediatr Nephrol ; 33(7): 1263-1267, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29594503

RESUMO

BACKGROUND: Hyperphosphatemic familial tumoral calcinosis (HFTC) is a rare autosomal recessive disease caused by mutations in genes encoding FGF23 or its regulators, and leading to functional deficiency or resistance to fibroblast growth factor 23 (FGF23). Subsequent biochemical features include hyperphosphatemia due to increased renal phosphate reabsorption, and increased or inappropriately normal 1,25-dihydroxyvitamin D (1,25-D) levels. CASE-DIAGNOSIS/TREATMENT: A 15-year-old girl was referred for a 1.2-kg-calcified mass of the thigh, with hyperphosphatemia (2.8 mmol/L); vascular impairment and soft tissue calcifications were already present. DNA sequencing identified compound heterozygous mutations in the FGF23 gene. Management with phosphate dietary restriction, phosphate binders (sevelamer, aluminum, nicotinamide), and acetazolamide moderately decreased serum phosphate levels; oral ketoconazole was secondary administered, leading to significantly decreased 1,25-D levels albeit only moderate additionally decreased phosphate levels. However, therapeutic compliance was questionable. Serum phosphate levels always remained far above the upper normal limit for age. The patient presented with two relapses of the thigh mass, requiring further surgery. CONCLUSIONS: We suggest that control of phosphate metabolism is crucial to prevent recurrences and vascular complications in HFTC; however, the medical management remains challenging.


Assuntos
Calcinose/terapia , Quelantes/uso terapêutico , Diuréticos/uso terapêutico , Fatores de Crescimento de Fibroblastos/genética , Hiperostose Cortical Congênita/terapia , Hiperfosfatemia/terapia , Fosfatos/metabolismo , Adolescente , Nádegas/diagnóstico por imagem , Nádegas/cirurgia , Calcinose/sangue , Calcinose/diagnóstico , Calcinose/genética , Terapia Combinada/métodos , Análise Mutacional de DNA , Feminino , Fator de Crescimento de Fibroblastos 23 , Heterozigoto , Humanos , Hiperostose Cortical Congênita/sangue , Hiperostose Cortical Congênita/diagnóstico , Hiperostose Cortical Congênita/genética , Hiperfosfatemia/sangue , Hiperfosfatemia/diagnóstico , Hiperfosfatemia/genética , Imageamento por Ressonância Magnética , Fosfatos/sangue , Resultado do Tratamento
19.
Curr Osteoporos Rep ; 15(4): 255-270, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28585220

RESUMO

PURPOSE OF REVIEW: We give an update on the etiology and potential treatment options of rare inherited monogenic disorders associated with arterial calcification and calcific cardiac valve disease. RECENT FINDINGS: Genetic studies of rare inherited syndromes have identified key regulators of ectopic calcification. Based on the pathogenic principles causing the diseases, these can be classified into three groups: (1) disorders of an increased extracellular inorganic phosphate/inorganic pyrophosphate ratio (generalized arterial calcification of infancy, pseudoxanthoma elasticum, arterial calcification and distal joint calcification, progeria, idiopathic basal ganglia calcification, and hyperphosphatemic familial tumoral calcinosis; (2) interferonopathies (Singleton-Merten syndrome); and (3) others, including Keutel syndrome and Gaucher disease type IIIC. Although some of the identified causative mechanisms are not easy to target for treatment, it has become clear that a disturbed serum phosphate/pyrophosphate ratio is a major force triggering arterial and cardiac valve calcification. Further studies will focus on targeting the phosphate/pyrophosphate ratio to effectively prevent and treat these calcific disease phenotypes.


Assuntos
Calcificação Vascular/genética , Anormalidades Múltiplas/tratamento farmacológico , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Doenças da Aorta/tratamento farmacológico , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Doenças dos Gânglios da Base/tratamento farmacológico , Doenças dos Gânglios da Base/genética , Doenças dos Gânglios da Base/metabolismo , Calcinose/tratamento farmacológico , Calcinose/genética , Calcinose/metabolismo , Doenças das Cartilagens/tratamento farmacológico , Doenças das Cartilagens/genética , Doenças das Cartilagens/metabolismo , Hipoplasia do Esmalte Dentário/tratamento farmacológico , Hipoplasia do Esmalte Dentário/genética , Hipoplasia do Esmalte Dentário/metabolismo , Difosfatos/metabolismo , Terapia de Reposição de Enzimas , Doença de Gaucher/tratamento farmacológico , Doença de Gaucher/genética , Doença de Gaucher/metabolismo , Deformidades Congênitas da Mão/tratamento farmacológico , Deformidades Congênitas da Mão/genética , Deformidades Congênitas da Mão/metabolismo , Humanos , Hiperostose Cortical Congênita/tratamento farmacológico , Hiperostose Cortical Congênita/genética , Hiperostose Cortical Congênita/metabolismo , Hiperfosfatemia/tratamento farmacológico , Hiperfosfatemia/genética , Hiperfosfatemia/metabolismo , Interferons/metabolismo , Metacarpo/anormalidades , Metacarpo/metabolismo , Doenças Musculares/tratamento farmacológico , Doenças Musculares/genética , Doenças Musculares/metabolismo , Odontodisplasia/tratamento farmacológico , Odontodisplasia/genética , Odontodisplasia/metabolismo , Osteoporose/tratamento farmacológico , Osteoporose/genética , Osteoporose/metabolismo , Fosfatos/metabolismo , Progéria/tratamento farmacológico , Progéria/genética , Progéria/metabolismo , Pseudoxantoma Elástico/tratamento farmacológico
20.
Horm Res Paediatr ; 86(1): 45-52, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27355663

RESUMO

BACKGROUND: Fibroblast growth factor-23 (FGF23) is a bone-derived hormone that regulates the homeostasis of phosphate and vitamin D. Three substitutions in the hormone are reported to cause autosomal dominant hypophosphatemic rickets and seven substitutions to cause autosomal recessive hyperphosphatemic familial tumoral calcinosis (HFTC). Both disorders are rare in the general population and occur most often in the Eastern Mediterranean region and Africa. None of the mutations could be identified using standard restriction fragment length polymorphism. The only technique currently available to confirm the clinical diagnosis is DNA sequencing. METHODS: Using a tri-primer ARMS-PCR, in vitro site-directed mutagenesis and DNA sequencing, we developed, verified and validated a rapid and reliable diagnostic test for the ten mutations in FGF23. RESULTS: We generated a test for all ten mutations and confirmed each test by DNA sequencing. We increased the specificity of the test by introducing a mismatch at position -2 in the 3'-terminus of the reverse primer of the normal and the mutant sequences. Finally, using DNA sequencing, we validated the technique for FGF23/S129F substitution by testing samples from 80 individuals from two unrelated Arab families harboring HFTC. CONCLUSIONS: This inexpensive and specific method could be adopted where DNA sequencing is not available or affordable.


Assuntos
Calcinose/genética , Fatores de Crescimento de Fibroblastos/genética , Hiperostose Cortical Congênita/genética , Hiperfosfatemia/genética , Mutação de Sentido Incorreto , Reação em Cadeia da Polimerase/métodos , Substituição de Aminoácidos , Análise Mutacional de DNA/métodos , Feminino , Fator de Crescimento de Fibroblastos 23 , Humanos , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA