Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 132(24)2022 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-36326820

RESUMO

The Hippo pathway nuclear effector Yes-associated protein (YAP) potentiates the progression of polycystic kidney disease (PKD) arising from ciliopathies. The mechanisms underlying the increase in YAP expression and transcriptional activity in PKD remain obscure. We observed that in kidneys from mice with juvenile cystic kidney (jck) ciliopathy, the aberrant hyperactivity of mechanistic target of rapamycin complex 1 (mTORC1), driven by ERK1/2 and PI3K/AKT cascades, induced ER proteotoxic stress. To reduce this stress by reprogramming translation, the protein kinase R-like ER kinase-eukaryotic initiation factor 2α (PERK/eIF2α) arm of the integrated stress response (ISR) was activated. PERK-mediated phosphorylation of eIF2α drove the selective translation of activating transcription factor 4 (ATF4), potentiating YAP expression. In parallel, YAP underwent K63-linked polyubiquitination by SCF S-phase kinase-associated protein 2 (SKP2) E3 ubiquitin ligase, a Hippo-independent, nonproteolytic ubiquitination that enhances YAP nuclear trafficking and transcriptional activity in cancer cells. Defective ISR cellular adaptation to ER stress in eIF2α phosphorylation-deficient jck mice further augmented YAP-mediated transcriptional activity and renal cyst growth. Conversely, pharmacological tuning down of ER stress/ISR activity and SKP2 expression in jck mice by administration of tauroursodeoxycholic acid (TUDCA) or tolvaptan impeded these processes. Restoring ER homeostasis and/or interfering with the SKP2-YAP interaction represent potential therapeutic avenues for stemming the progression of renal cystogenesis.


Assuntos
Proteínas Quinases Associadas a Fase S , Ubiquitina-Proteína Ligases , Camundongos , Animais , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas Quinases Associadas a Fase S/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fator de Iniciação 2 em Eucariotos/genética , Fator 4 Ativador da Transcrição/metabolismo , Fosforilação , Rim/metabolismo
2.
J Clin Endocrinol Metab ; 107(10): 2777-2783, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-35896139

RESUMO

CONTEXT: Autosomal recessive hypophosphatemic rickets (ARHR) are rare, heritable renal phosphate-wasting disorders that arise from overexpression of the bone-derived phosphaturic hormone fibroblast growth factor 23 (FGF23) leading to impaired bone mineralization (rickets and osteomalacia). Inactivating mutations of Dentin matrix protein 1 (DMP1) give rise to ARHR type 1 (ARHR1). Short stature, prominent bowing of the legs, fractures/pseudofractures, and severe enthesopathy are prominent in this patient population. Traditionally, treatment consists of oral phosphate replacement and the addition of calcitriol but this approach is limited by modest efficacy and potential renal and gastrointestinal side effects. OBJECTIVE: The advent of burosumab (Crysvita), a fully humanized monoclonal antibody to FGF23 for the treatment of X-linked hypophosphatemia and tumor-induced osteomalacia, offers a unique opportunity to evaluate its safety and efficacy in patients with ARHR1. RESULTS: Monthly administration of burosumab to 2 brothers afflicted with the disorder resulted in normalization of serum phosphate, healing of pseudofracture, diminished fatigue, less bone pain, and reduced incapacity arising from the extensive enthesopathy and soft tissue fibrosis/calcification that characterizes this disorder. No adverse effects were reported following burosumab administration. CONCLUSION: The present report highlights the beneficial biochemical and clinical outcomes associated with the use of burosumab in patients with ARHR1.


Assuntos
Doenças Ósseas Metabólicas , Entesopatia , Raquitismo Hipofosfatêmico Familiar , Osteomalacia , Raquitismo Hipofosfatêmico , Anticorpos Monoclonais Humanizados/uso terapêutico , Calcitriol/uso terapêutico , Raquitismo Hipofosfatêmico Familiar/tratamento farmacológico , Raquitismo Hipofosfatêmico Familiar/genética , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Hormônios/uso terapêutico , Humanos , Masculino , Osteomalacia/metabolismo , Fosfatos/metabolismo , Raquitismo Hipofosfatêmico/tratamento farmacológico , Raquitismo Hipofosfatêmico/genética
3.
Appl Opt ; 60(22): 6511-6519, 2021 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-34612888

RESUMO

We present an asymmetric encryption scheme for hyperspectral images using hybrid chaotic maps (HCMs) and an equal modulus decomposition tree (EMDT) structure in a discrete multiple-parameter fractional Fourier transform (dmpFrFT) domain. The original hyperspectral image was scrambled by an HCM and then encrypted into asymmetric ciphertext using the EMDT. In the EMDT, each pair of the band images of the scrambled hyperspectral image were regarded as leaf nodes, while the encryption modules using chaotic random phase mask, dmpFrFT, and improved equal modulus decomposition were regarded as branch nodes, and the encryption process was implemented along the paths from the leaf nodes to the topmost branch node. The EMDT structure could provide multiparameter encryption, real-valued output, and different pairs of band images with different secret keys and encryption/decryption paths. Compared with the previous optical encryption approaches for hyperspectral images, our asymmetric cryptosystem had larger key space, less data amount of storage and transmission, and stronger resistance to statistical attacks. Various numerical simulations verified the performance of our proposed asymmetric cryptosystem.

4.
Am J Physiol Renal Physiol ; 314(6): F1046-F1061, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29357413

RESUMO

Vascular calcification increases the risk of cardiovascular disease and death in patients with chronic kidney disease (CKD). Increased activity of mammalian target of rapamycin complex 1 (mTORC1) and endoplasmic reticulum (ER) stress-unfolded protein response (UPR) are independently reported to partake in the pathogenesis of vascular calcification in CKD. However, the association between mTORC1 activity and ER stress-UPR remains unknown. We report here that components of the uremic state [activation of the receptor for advanced glycation end products (RAGE) and hyperphosphatemia] potentiate vascular smooth muscle cell (VSMC) calcification by inducing persistent and exaggerated activity of mTORC1. This gives rise to prolonged and excessive ER stress-UPR as well as attenuated levels of sestrin 1 ( Sesn1) and Sesn3 feeding back to inhibit mTORC1 activity. Activating transcription factor 4 arising from the UPR mediates cell death via expression of CCAAT/enhancer-binding protein (c/EBP) homologous protein (CHOP), impairs the generation of pyrophosphate, a potent inhibitor of mineralization, and potentiates VSMC transdifferentiation to the osteochondrocytic phenotype. Short-term treatment of CKD mice with rapamycin, an inhibitor of mTORC1, or tauroursodeoxycholic acid, a bile acid that restores ER homeostasis, normalized mTORC1 activity, molecular markers of UPR, and calcium content of aortas. Collectively, these data highlight that increased and/or protracted mTORC1 activity arising from the uremic state leads to dysregulated ER stress-UPR and VSMC calcification. Manipulation of the mTORC1-ER stress-UPR pathway opens up new therapeutic strategies for the prevention and treatment of vascular calcification in CKD.


Assuntos
Doenças da Aorta/enzimologia , Estresse do Retículo Endoplasmático , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Músculo Liso Vascular/enzimologia , Resposta a Proteínas não Dobradas , Uremia/complicações , Calcificação Vascular/enzimologia , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Animais , Aorta/efeitos dos fármacos , Aorta/enzimologia , Aorta/patologia , Doenças da Aorta/tratamento farmacológico , Doenças da Aorta/etiologia , Doenças da Aorta/patologia , Morte Celular , Proliferação de Células , Transdiferenciação Celular , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HEK293 , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Camundongos Mutantes , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Osteogênese , Fosforilação , Receptor para Produtos Finais de Glicação Avançada/genética , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Proteínas S100/genética , Proteínas S100/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Ácido Tauroquenodesoxicólico/farmacologia , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Calcificação Vascular/tratamento farmacológico , Calcificação Vascular/etiologia , Calcificação Vascular/patologia
5.
J Clin Invest ; 126(2): 667-80, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26784541

RESUMO

CYP24A1 (hereafter referred to as CYP24) enzymatic activity is pivotal in the inactivation of vitamin D metabolites. Basal renal and extrarenal CYP24 is usually low but is highly induced by its substrate 1,25-dihydroxyvitamin D. Unbalanced high and/or long-lasting CYP24 expression has been proposed to underlie diseases like chronic kidney disease, cancers, and psoriasis that otherwise should favorably respond to supplemental vitamin D. Using genetically modified mice, we have shown that renal phosphate wasting hypophosphatemic states arising from high levels of fibroblast growth factor 23 (FGF23) are also associated with increased renal Cyp24 expression, suggesting that elevated CYP24 activity is pivotal to the pathophysiology of these disorders. We therefore crossed 2 mouse strains, each with distinct etiology for high levels of circulating FGF23, onto a Cyp24-null background. Specifically, we evaluated Cyp24 deficiency in Hyp mice, the murine homolog of X-linked dominant hypophosphatemic rickets, and transgenic mice that overexpress a mutant FGF23 (FGF23R176Q) that is associated with the autosomal dominant form of hypophosphatemic rickets. Loss of Cyp24 in these murine models of human disease resulted in near-complete recovery of rachitic/osteomalacic bony abnormalities in the absence of any improvement in the serum biochemical profile. Moreover, treatment of Hyp and FGF23R1760-transgenic mice with the CYP24 inhibitor CTA102 also ameliorated their rachitic bones. Our results link CYP24 activity to the pathophysiology of FGF23-dependent renal phosphate wasting states and implicate pharmacologic CYP24 inhibition as a therapeutic adjunct for their treatment.


Assuntos
Inibidores das Enzimas do Citocromo P-450/farmacologia , Fatores de Crescimento de Fibroblastos/metabolismo , Fosfatos/urina , Insuficiência Renal Crônica , Vitamina D3 24-Hidroxilase/antagonistas & inibidores , Síndrome de Emaciação , Animais , Modelos Animais de Doenças , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Humanos , Camundongos , Camundongos Knockout , Insuficiência Renal Crônica/tratamento farmacológico , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/patologia , Insuficiência Renal Crônica/urina , Vitamina D3 24-Hidroxilase/genética , Vitamina D3 24-Hidroxilase/metabolismo , Síndrome de Emaciação/tratamento farmacológico , Síndrome de Emaciação/genética , Síndrome de Emaciação/patologia , Síndrome de Emaciação/urina
6.
Endocrinology ; 153(12): 5906-17, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23038738

RESUMO

We have previously confirmed a paradoxical mineralizing enthesopathy as a hallmark of X-linked hypophosphatemia. X-linked hypophosphatemia is the most common of the phosphate-wasting disorders mediated by elevated fibroblast growth factor 23 (FGF23) and occurs as a consequence of inactivating mutations of the PHEX gene product. Despite childhood management of the disease, these complications of tendon and ligament insertion sites account for a great deal of the disease's morbidity into adulthood. It is unclear whether the enthesopathy occurs in other forms of renal phosphate-wasting disorders attributable to high FGF23 levels. Here we describe two patients with autosomal recessive hypophosphatemic rickets due to the Met1Val mutation in dentin matrix acidic phosphoprotein 1 (DMP1). In addition to the biochemical and skeletal features of long-standing rickets with elevated FGF23 levels, these individuals exhibited severe, debilitating, generalized mineralized enthesopathy. These data suggest that enthesophytes are a feature common to FGF23-mediated phosphate-wasting disorders. To address this possibility, we examined a murine model of FGF23 overexpression using a transgene encoding the secreted form of human FGF23 (R176Q) cDNA (FGF23-TG mice). We report that FGF23-TG mice display a similar mineralizing enthesopathy of the Achilles and plantar facial insertions. In addition, we examined the impact of standard therapy for phosphate-wasting disorders on enthesophyte progression. We report that fibrochondrocyte hyperplasia persisted in Hyp mice treated with oral phosphate and calcitriol. In addition, treatment had the untoward effect of further exacerbating the mineralization of fibrochondrocytes that define the bone spur of the Achilles insertion. These studies support the need for newer interventions targeted at limiting the actions of FGF23 and minimizing both the toxicities and potential morbidities associated with standard therapy.


Assuntos
Raquitismo Hipofosfatêmico Familiar/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Doenças Genéticas Ligadas ao Cromossomo X , Rim/metabolismo , Doenças Reumáticas/diagnóstico , Animais , Proteínas da Matriz Extracelular/genética , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Imuno-Histoquímica/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Genéticos , Mutação , Linhagem , Fosfatos/metabolismo , Fosfoproteínas/genética , Doenças Reumáticas/fisiopatologia , Transgenes , Regulação para Cima
7.
Brain Res ; 1412: 9-17, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21824606

RESUMO

Fibroblast growth factor-23 (FGF-23) is a potent circulating phosphaturic factor associated with renal phosphate wasting. Effects of FGF-23 on skeleton, phosphate homeostasis, and cardiovascular system have been investigated; however, the effect of FGF-23 on the central nervous system (CNS) is unknown. To assess whether FGF-23 influences the function and structure of the CNS and whether the effect of FGF-23 on the CNS is mediated by FGF receptors directly or by hypophosphatemia indirectly, FGF-23 transgenic mice and their wild-type littermates were fed a normal diet or a high-phosphate diet containing a normal diet plus 1.25% phosphate in drinking water from weaning for 5weeks and the phenotypes of the CNS were compared between FGF-23 transgenic mice and their wild-type littermates on the same diet. At the end of this time period, transgenic animals on the normal diet showed impaired spatial learning and memory. Furthermore, these mice exhibited the impairment of long-term potentiation in hippocampal CA1 region, and the reduction of hippocampal adenosine-triphosphate content and of choline acetyltransferase-positive neurons in basal forebrain, possibly as pathogenetic factors contributing to the cognitive deficit. The central nervous phenotypes of transgenic mice were rescued following improved hypophosphatemia by the high-phosphate diet intake. This study demonstrates that FGF-23 overexpression can result in abnormalities in the CNS mediated by the secondary severe hypophosphatemia.


Assuntos
Fatores de Crescimento de Fibroblastos/genética , Hipofosfatemia/genética , Potenciação de Longa Duração/genética , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/genética , Trifosfato de Adenosina/metabolismo , Animais , Colina O-Acetiltransferase/metabolismo , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Humanos , Hipofosfatemia/metabolismo , Hipofosfatemia/fisiopatologia , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Prosencéfalo/metabolismo
8.
Clin Exp Pharmacol Physiol ; 38(6): 395-402, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21488937

RESUMO

1. Though previous studies have shown that fibroblast growth factor 23 (FGF23) mRNA expression localizes in ameloblasts and odontoblasts in teeth, it is unclear what effect FGF23 overexpression has on dentin mineralization and dentinogenesis. Toward this end, the phenotypes of mandibles and teeth were compared between 6-week-old FGF23 transgenic mice and their wild-type littermates by radiography, microcomputed tomography scanning, histology, histochemistry and immunohistochemistry. 2. The mineral density was reduced in all teeth, including molars and incisors, and in the mandible, and the mineralized tooth volume in incisor and molars, and the mineralized cortical and alveolar bone volume in mandibles were decreased in FGF23 transgenic mice compared with their wild-type littermates. The dental volume, reparative dentin area, the expression of dentin sialoprotein in dentin, and the deposition of type I collagen and osteocalcin in the dental matrix were significantly reduced. However, the predentin volume and the expression of biglycan in dentin were increased in FGF23 transgenic mice compared with their wild-type littermates. 3. The results of the present study show that FGF23 overexpression plays a negative regulatory role on dentin mineralization and dentinogenesis.


Assuntos
Dentina/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Calcificação de Dente/fisiologia , Animais , Calcificação Fisiológica , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Genótipo , Mandíbula/anatomia & histologia , Camundongos , Camundongos Transgênicos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo
9.
Am J Physiol Heart Circ Physiol ; 297(4): H1514-20, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19684183

RESUMO

Fibroblast growth factor-23 (FGF-23) is a potent circulating phosphaturic factor associated with renal phosphate wasting. The effects of FGF-23 on skeletal and phosphate homeostasis have been investigated widely; however, the effect of FGF-23 on the cardiovascular system (CVS) is unknown. To assess whether FGF-23 influences the function and structure of the CVS and whether the effect of FGF-23 on the CVS is mediated by FGF receptors directly or indirectly by hypophosphatemia, FGF-23 transgenic mice and their wild-type littermates were fed a normal diet or a high-phosphate diet comprising a normal diet plus 1.25% phosphate in drinking water from weaning for 5 wk, and the phenotypes of the CVS were compared between FGF-23 transgenic mice and their wild-type littermates on the same diet. At the end of this time period, transgenic animals on the normal diet developed hypotension. The left ventricle was appropriately hypertrophic, and plasma catecholamine and renin-angiotensin system components were upregulated, indicating compensatory mechanisms in response to the hypotension. Transgenic mice also exhibited an impaired vascular reactivity and a downregulation of vasoconstrictor receptor gene expression, possibly as pathogenetic factors contributing to the hypotension. The high-phosphate diet improved the hypophosphatemia, resulting in a rescue of the cardiovascular phenotype. This study demonstrates that FGF-23 overexpression can result in abnormalities in the CVS and that the effect of FGF-23 overexpression on the CVS is mediated by the secondary severe hypophosphatemia.


Assuntos
Pressão Sanguínea , Fatores de Crescimento de Fibroblastos/biossíntese , Hipofosfatemia/complicações , Hipotensão/etiologia , Vasoconstrição , Animais , Pressão Sanguínea/efeitos dos fármacos , Catecolaminas/sangue , Dieta , Relação Dose-Resposta a Droga , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Humanos , Hipertrofia Ventricular Esquerda/etiologia , Hipertrofia Ventricular Esquerda/metabolismo , Hipertrofia Ventricular Esquerda/fisiopatologia , Hipofosfatemia/dietoterapia , Hipofosfatemia/metabolismo , Hipofosfatemia/fisiopatologia , Hipotensão/genética , Hipotensão/metabolismo , Hipotensão/fisiopatologia , Hipotensão/prevenção & controle , Masculino , Camundongos , Camundongos Transgênicos , Fenótipo , Fosfatos/administração & dosagem , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Angiotensina/genética , Receptores Adrenérgicos alfa 1/genética , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Sistema Renina-Angiotensina/genética , Regulação para Cima , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Função Ventricular Esquerda
10.
Am J Physiol Endocrinol Metab ; 296(1): E79-88, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18984852

RESUMO

Transgenic mice overexpressing fibroblast growth factor (FGF23) (R176Q) (F(Tg)) exhibit biochemical {hypophosphatemia, phosphaturia, abnormal 1,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)] metabolism} and skeletal (rickets and osteomalacia) abnormalities attributable to FGF23 action. In vitro studies now implicate the aging-related factor Klotho in the signaling mechanism of FGF23. In this study, we used a mouse genetic approach to validate in vivo the pivotal role of Klotho in the metabolic and skeletal derangements associated with FGF23 (R176Q) overexpression. To this end, we crossed mice heterozygous for the hypomorphic Klotho allele (Kl(+/-)) to F(Tg) mice and obtained F(Tg) transgenic mice homozygous for the Kl-hypomorphic allele (F(Tg)/Kl(-/-)). Mice were killed on postnatal day 50, and serum and tissues were procured for analysis and comparison with F(Tg), wild-type, and Kl(-/-) controls. From 4 wk onward, F(Tg)/Kl(-/-) mice were clearly distinguishable from F(Tg) mice and exhibited a striking phenotypic resemblance to the Kl(-/-) controls. Serum analysis for calcium, phosphorus, parathyroid hormone, 1,25(OH)(2)D(3), and alkaline phosphatase activity confirmed the biochemical similarity between the F(Tg)/Kl(-/-) and Kl(-/-) mice and their distinctness from the F(Tg) controls. The characteristic skeletal changes associated with FGF23 (R176Q) overexpression were also dramatically reversed by the absence of Klotho. Hence the wide, unmineralized growth plates and the osteomalacic abnormalities apparent in trabecular and cortical bone were completely reversed in the F(Tg)/Kl(-/-) mice. Nevertheless, independent actions of Klotho on bone were suggested as manifested by alterations in mineralized bone, and in cortical bone volume which were observed in both the Kl(-/-) and F(Tr)/Kl(-/-) mutants. In summary, our findings substantiate in vivo the essential role of Klotho in the mechanism of action of FGF23 in view of the fact that Klotho ablation converts the biochemical and skeletal manifestations resulting from FGF23 overexpression to a phenotype consistent with Klotho deficiency.


Assuntos
Remodelação Óssea/fisiologia , Fatores de Crescimento de Fibroblastos/metabolismo , Glucuronidase/deficiência , Osteomalacia/metabolismo , Fosfatase Ácida/sangue , Fosfatase Alcalina/sangue , Animais , Northern Blotting , Calcitriol/sangue , Cálcio/sangue , Cruzamentos Genéticos , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Fatores de Crescimento de Fibroblastos/genética , Glucuronidase/genética , Glucuronidase/metabolismo , Imuno-Histoquímica , Isoenzimas/sangue , Proteínas Klotho , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Osteomalacia/sangue , Osteomalacia/genética , Osteomalacia/patologia , Hormônio Paratireóideo/sangue , Fenótipo , Fósforo/sangue , Fosfatase Ácida Resistente a Tartarato
11.
Endocrinology ; 148(10): 4974-83, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17615144

RESUMO

Although increased circulating levels of PTH with mild hypocalcemia has been reported in Hyp mice, hyperparathyroidism in X-linked hypophosphatemic rickets is postulated to arise from the standard use of phosphate salts, which induce chronic stimulation of PTH secretion. In this study, we sought to examine the role of PTH in the metabolic derangements associated with Hyp by generating hemizygous hypophosphatemic (Hyp/Y) mice homozygous for the Pth-null allele (Pth(-/-);Hyp/Y). Early postnatal lethality was observed in the Pth(-/-);Hyp/Y mice. Within the first 6 h, postpartum serum phosphorus increased to levels comparable to those in the Pth(-/-) mice, whereas in Hyp mice, it decreased during the first 48 h after birth. Serum calcium concentration started low after birth and remained reduced in both Pth(-/-);Hyp/Y and Pth(-/-) mice although more profoundly so in the former group, whereas in Hyp/Y mice, the levels were initially lower than but reached wild-type levels by 24 h. Circulating PTH levels in Hyp/Y mice were higher than wild-type levels throughout the first 48 h after birth and continued to be so well into adulthood. Twice-daily administration of PTH 1-34 to Pth(-/-);Hyp/Y newborn mice increased serum calcium levels and prevented their early demise. The findings here indicate that the cause of death in the Pth(-/-);Hyp/Y mice is severe hypocalcemia. A potential role for fibroblast growth factor 23 in promoting secondary hyperparathyroidism by suppressing renal 25-hydroxyvitamin D(3)-1alpha-hydroxylase (Cyp27b1) activity while increasing that of renal 25-hydroxyvitamin D(3) 24-hydroxylase (Cyp24) is proposed. Hyperparathyroidism, therefore, is an integral component in the pathophysiology of Hyp, and likely X-linked hypophosphatemic rickets and serves to prevent severe hypocalcemia in mice and perhaps in patients afflicted with the disorder.


Assuntos
Deleção de Genes , Hipofosfatemia Familiar/genética , Hipofosfatemia Familiar/mortalidade , Hormônio Paratireóideo/genética , Animais , Animais Recém-Nascidos , Feminino , Hipocalcemia/genética , Hipocalcemia/mortalidade , Hipocalcemia/fisiopatologia , Hipofosfatemia Familiar/tratamento farmacológico , Hipofosfatemia Familiar/patologia , Masculino , Camundongos , Camundongos Knockout , Hormônio Paratireóideo/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , Fenótipo , Terapia de Salvação , Índice de Gravidade de Doença , Fatores Sexuais
12.
J Endocrinol ; 183(1): 203-16, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15525588

RESUMO

Thiazolidinediones (TZDs) increase peripheral tissue insulin sensitivity in patients with type 2 diabetes mellitus by activating the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma). In bone marrow stromal cell cultures and in vivo, activation of PPARgamma by high doses (20 mg/kg/day) of TZDs has been reported to alter stem cell differentiation by promoting commitment of progenitor cells to the adipocytic lineage while inhibiting osteoblastogenesis. Here, we have examined the in vivo effects of low-dose rosiglitazone (3 mg/kg/day) on bone, administered to mice by gavage for 90 days. Rosiglitazone-treated mice had increased weight when compared with controls, with no significant alterations in serum levels of glucose, calcium or parathyroid hormone (PTH). Bone mineral density (BMD) at the lumbar vertebrae (L1-L4), ilium/sacrum, and total body was diminished by rosiglitazone treatment. Histologically, bone was characterized by decreased trabecular bone volume and increased marrow space with no significant change in bone marrow adipocity. Decreased osteoblast number and activity due to increased apoptotic death of osteoblasts and osteocytes was apparent while osteoclast parameters and serum levels of osteocalcin, alkaline phosphatase activity, and leptin were unaltered by rosiglitazone treatment. Therefore, the imbalance in bone remodeling that follows rosiglitazone administration arises from increased apoptotic death of osteogenic cells and diminished bone formation leading to the observed decrease in trabecular bone volume and BMD. These novel in vivo effects of TZDs on bone are of clinical relevance as patients with type 2 diabetes mellitus and other insulin resistant states treated with these agents may potentially be at increased risk of osteoporosis.


Assuntos
Osso e Ossos/efeitos dos fármacos , Diabetes Mellitus/metabolismo , Hipoglicemiantes/efeitos adversos , Tiazolidinedionas/efeitos adversos , Animais , Apoptose/efeitos dos fármacos , Biomarcadores/sangue , Glicemia/metabolismo , Densidade Óssea/efeitos dos fármacos , Osso e Ossos/patologia , Cálcio/sangue , Células Cultivadas , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/patologia , Histocitoquímica , Hipoglicemiantes/uso terapêutico , Processamento de Imagem Assistida por Computador , Leptina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/efeitos dos fármacos , Osteoblastos/patologia , Osteocalcina/sangue , Osteócitos/efeitos dos fármacos , Osteócitos/patologia , Hormônio Paratireóideo/sangue , Rosiglitazona , Tiazolidinedionas/uso terapêutico
13.
Endocrinology ; 145(11): 5269-79, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15284207

RESUMO

Fibroblast growth factor 23 (FGF23) is a recently characterized protein likely involved in the regulation of serum phosphate homeostasis. Increased circulating levels of FGF23 have been reported in patients with renal phosphate-wasting disorders, but it is unclear whether FGF23 is the direct mediator responsible for the decreased phosphate transport at the proximal renal tubules and the altered vitamin D metabolism associated with these states. To examine this question, we generated transgenic mice expressing and secreting from the liver human FGF23 (R176Q), a mutant form that fails to be degraded by furin proteases. At 1 and 2 months of age, mice carrying the transgene recapitulated the biochemical (decreased urinary phosphate reabsorption, hypophosphatemia, low serum 1,25-dihydroxyvitamin D(3)) and skeletal (rickets and osteomalacia) alterations associated with these disorders. Unexpectantly, marked changes in parameters of calcium homeostasis were also observed, consistent with secondary hyperparathyroidism. Moreover, in the kidney the anticipated alterations in the expression of hydroxylases associated with vitamin D metabolism were not observed despite the profound hypophosphatemia and increased circulating levels of PTH, both major physiological stimuli for 1,25-dihydroxyvitamin D(3) production. Our findings strongly support the novel concept that high circulating levels of FGF23 are associated with profound disturbances in the regulation of phosphate and vitamin D metabolism as well as calcium homeostasis and that elevated PTH levels likely also contribute to the renal phosphate wasting associated with these disorders.


Assuntos
Fatores de Crescimento de Fibroblastos/genética , Hipofosfatemia Familiar/fisiopatologia , Osteomalacia/fisiopatologia , Hormônio Paratireóideo/fisiologia , Animais , Osso e Ossos/patologia , Osso e Ossos/fisiopatologia , Cálcio/sangue , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/sangue , Expressão Gênica , Humanos , Hipofosfatemia Familiar/sangue , Hipofosfatemia Familiar/patologia , Túbulos Renais Proximais/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos , Osteomalacia/sangue , Osteomalacia/patologia , Hormônio Paratireóideo/sangue , Fosfatos/sangue , Gravidez , Vitamina D/sangue
14.
Bone ; 34(4): 638-47, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15050894

RESUMO

X-linked hypophosphatemic rickets (HYP) in humans is caused by mutations in the PHEX gene. This gene mutation is also found in Hyp mice, the murine homologue of the human disease. At present, it is unknown why loss of Phex function leads to cartilage abnormalities in Hyp mice. In the present study, we compared in wild-type and Hyp mice Phex protein localization in cartilage of developing long bone as well as localization of skeletal matrix proteins and matrix metalloproteinase-9 (MMP-9). Also compared were chondrocyte apoptosis in the growth plate, mineralization and cartilage remnant retention in the metaphysis, and chondroclast/osteoclast characteristics in the primary spongiosa. Phex protein was detected in proliferating and hypertrophic chondrocytes in growth plate cartilage of wild-type mice, but not in Hyp mice. Hyp mice exhibited a widened and irregular hypertrophic zone in growth plate cartilage showing hypomineralization, increased cartilage remnants from the growth plate in both metaphyseal trabecular and cortical bone, and fewer and smaller chondroclasts/osteoclasts in the primary spongiosa. Increased link protein and C-propeptide of type II procollagen of Hyp mice reflected the increase in chondrocytes and matrix in the cartilaginous growth plate and in bone. In addition, growth plate osteocalcin and bone sialoprotein levels were decreased, while osteonectin was increased, in hypertrophic chondrocytes and cartilage matrix in Hyp mice. MMP-9 in hypertrophic chondrocytes was also reduced in Hyp mice and fewer apoptotic hypertrophic chondrocytes were detected. These findings suggest that Phex may control mineralization and removal of hypertrophic chondrocytes and cartilage matrix in growth plate by regulating the synthesis and deposition of certain bone matrix proteins and proteases such as MMP-9.


Assuntos
Cartilagem/anormalidades , Cartilagem/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Hipofosfatemia Familiar/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Proteínas/metabolismo , Animais , Apoptose , Osso e Ossos/metabolismo , Osso e Ossos/fisiopatologia , Calcificação Fisiológica , Cartilagem/crescimento & desenvolvimento , Cartilagem/fisiopatologia , Condrócitos/metabolismo , Condrócitos/patologia , Regulação da Expressão Gênica , Hiperostose/metabolismo , Hiperostose/patologia , Hipofosfatemia Familiar/genética , Hipofosfatemia Familiar/fisiopatologia , Imuno-Histoquímica , Camundongos , Osteocalcina/metabolismo , Endopeptidase Neutra Reguladora de Fosfato PHEX , Proteínas/genética
15.
Endocrinology ; 145(4): 2046-53, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14701672

RESUMO

We have examined the role of PTH in the postnatal state in a mouse model of PTH deficiency generated by targeting the Pth gene in embryonic stem cells. Mice homozygous for the ablated allele, when maintained on a normal calcium intake, developed hypocalcemia, hyperphosphatemia, and low circulating 1,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)] levels consistent with primary hypoparathyroidism. Bone turnover was reduced, leading to increased trabecular and cortical bone volume in PTH-deficient mice. When mutant mice were placed on a low-calcium diet, renal 25-hydroxyvitamin D 1 alpha-hydroxylase expression increased despite the absence of PTH, leading to a rise in circulating 1,25(OH)(2)D(3) levels, marked osteoclastogenesis, and profound bone resorption. These studies demonstrate the dependence of the skeletal phenotype in animals with genetically depleted PTH on the external environment as well as on internal hormonal and ionic circulatory factors. They also show that, although PTH action is the first defense against hypocalcemia, 1,25(OH)(2)D(3) can be mobilized, even in the absence of PTH, to guard against extreme calcium deficiency.


Assuntos
Osso e Ossos/fisiologia , Cálcio da Dieta/farmacologia , Homeostase/fisiologia , Hormônio Paratireóideo/fisiologia , 25-Hidroxivitamina D3 1-alfa-Hidroxilase/metabolismo , Animais , Animais Recém-Nascidos , Doenças Ósseas/etiologia , Doenças Ósseas/patologia , Doenças Ósseas/fisiopatologia , Remodelação Óssea/fisiologia , Reabsorção Óssea/etiologia , Cálcio da Dieta/administração & dosagem , Divisão Celular , Relação Dose-Resposta a Droga , Ergocalciferóis/sangue , Homozigoto , Hipoparatireoidismo/etiologia , Hipoparatireoidismo/patologia , Hipoparatireoidismo/fisiopatologia , Rim/metabolismo , Camundongos , Camundongos Knockout/genética , Osteoclastos/patologia , Glândulas Paratireoides/patologia , Hormônio Paratireóideo/deficiência , Hormônio Paratireóideo/genética
16.
J Biol Chem ; 278(11): 9843-9, 2003 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-12519781

RESUMO

Missense mutations in fibroblast growth factor 23 (FGF23) are the cause of autosomal dominant hypophosphatemic rickets (ADHR). The mutations (R176Q, R179W, and R179Q) replace Arg residues within a subtilisin-like proprotein convertase (SPC) cleavage site (RXXR motif), leading to protease resistance of FGF23. The goals of this study were to examine in vivo the biological potency of the R176Q mutant FGF23 form and to characterize alterations in homeostatic mechanisms that give rise to the phenotypic presentation of this disorder. For this, wild type and R176Q mutant FGF23 were overexpressed in the intact animals using a tumor-bearing nude mouse system. At comparable circulating levels, the mutant form was more potent in inducing hypophosphatemia, in decreasing circulating concentrations of 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)), and in causing rickets and osteomalacia in these animals compared with wild type FGF23. Parameters of calcium homeostasis were also altered, leading to secondary hyperparathyroidism and parathyroid gland hyperplasia. However, the raised circulating levels of parathyroid hormone were ineffective in normalizing the reduced 1,25(OH)(2)D(3) levels by increasing renal expression of 25(OH)D(3)-1alpha-hydroxylase (Cyp40) to promote its synthesis and by decreasing that of 25(OH)D(3)-24-hydroxylase (Cyp24) to prevent its catabolism. The findings provide direct in vivo evidence that missense mutations from ADHR kindreds are gain-of-function mutations that retain and increase the protein's biological potency. Moreover, for the first time, they define a potential role for FGF23 in dissociating parathyroid hormone actions on mineral fluxes and on vitamin D metabolism at the level of the kidney.


Assuntos
Arginina/química , Fatores de Crescimento de Fibroblastos/genética , Mutação , Raquitismo/genética , Animais , Northern Blotting , Células CHO , Calcitriol/farmacologia , Divisão Celular , Clonagem Molecular , Cricetinae , DNA Complementar/metabolismo , Fator de Crescimento de Fibroblastos 23 , Vetores Genéticos , Humanos , Hiperparatireoidismo/metabolismo , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Túbulos Renais/metabolismo , Camundongos , Camundongos Nus , Mutação de Sentido Incorreto , Osteomalacia/metabolismo , Fenótipo , Ribonucleases/metabolismo , Fatores de Tempo
17.
Mol Endocrinol ; 16(12): 2913-25, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12456809

RESUMO

Inactivating mutations and/or deletions of PHEX/Phex (phosphate-regulating gene with homologies to endopeptidases on the X chromosome) are responsible for X-linked hypophosphatemic rickets in humans and in the murine homolog Hyp. The predominant osteoblastic expression of Phex has implicated a primary metabolic osteoblast defect in the pathophysiology of this disorder. By targeting PHEX expression to osteoblasts in the Hyp genetic background, we aimed to correct the corresponding biochemical and morphological abnormalities and obtain information on their pathogenetic mechanism. When transgene Phex expression, driven by a mouse pro-alpha1(I) collagen gene promoter, was crossed into the Hyp background, it improved the defective mineralization of bone and teeth but failed to correct the hypophosphatemia and altered vitamin D metabolism associated with the disorder. Ex vivo bone marrow cultures confirmed the amelioration in the Hyp-associated matrix mineralization defect after Phex expression. These findings suggest that while the Hyp bone and teeth abnormalities partially correct after PHEX gene transfer, additional factors and/or sites of PHEX expression are likely critical for the elaboration of the appropriate molecular signals that alter renal phosphate handling and vitamin D metabolism in this disorder.


Assuntos
Expressão Gênica , Hipofosfatemia Familiar/terapia , Osteoblastos/metabolismo , Proteínas/genética , Fosfatase Alcalina/análise , Animais , Células da Medula Óssea/metabolismo , Calcificação Fisiológica , Cálcio/análise , Diferenciação Celular , Células Cultivadas , Marcação de Genes , Sialoproteína de Ligação à Integrina , Rim/metabolismo , Camundongos , Camundongos Transgênicos , Osteogênese , Endopeptidase Neutra Reguladora de Fosfato PHEX , Fenótipo , Fosfatos/sangue , Fosfatos/metabolismo , Sialoglicoproteínas/análise , Dente/química , Dente/metabolismo , Calcificação de Dente , Transfecção , Vitamina D/metabolismo , Vitronectina/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA