Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 337
Filtrar
1.
J Bone Miner Metab ; 42(2): 155-165, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38310177

RESUMEN

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


Asunto(s)
Raquitismo Hipofosfatémico Familiar , Síndrome de Fanconi , Hipofosfatemia Familiar , Osteomalacia , Raquitismo Hipofosfatémico , Humanos , Osteomalacia/genética , Raquitismo Hipofosfatémico Familiar/genética , Hipofosfatemia Familiar/genética , Hipofosfatemia Familiar/metabolismo , Síndrome de Fanconi/genética , Síndrome de Fanconi/metabolismo
2.
PLoS One ; 18(11): e0294791, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38015969

RESUMEN

Both mTOR and α-klotho play a role in the pathophysiology of renal disease, influence mineral metabolism and participate in the aging process. The influence of mTOR inhibition by rapamycin on renal α-klotho expression is unknown. Rats with normal (controls) and reduced (Nx) renal function were treated with rapamycin, 1.3 mg/kg/day, for 22 days. The experiments were conducted with rats fed 0.6% P diet (NP) and 0.2% P diet (LP). Treatment with rapamycin promoted phosphaturia in control and Nx rats fed NP and LP. A decrease in FGF23 was identified in controls after treatment with rapamycin. In rats fed NP, rapamycin decreased mRNA α-klotho/GADPH ratio both in controls, 0.6±0.1 vs 1.1±0.1, p = 0.001, and Nx, 0.3±0.1 vs 0.7±0.1, p = 0.01. At the protein level, a significant reduction in α-klotho was evidenced after treatment with rapamycin both by Western Blot: 0.6±0.1 vs 1.0±0.1, p = 0.01, in controls, 0.7±0.1 vs 1.1±0.1, p = 0.02, in Nx; and by immunohistochemistry staining. Renal α-klotho was inversely correlated with urinary P excretion (r = -0.525, p = 0.0002). The decrease in α-klotho after treatment with rapamycin was also observed in rats fed LP. In conclusion, rapamycin increases phosphaturia and down-regulates α-klotho expression in rats with normal and decreased renal function. These effects can be observed in animals ingesting normal and low P diet.


Asunto(s)
Hipofosfatemia Familiar , Sirolimus , Femenino , Ratas , Animales , Sirolimus/farmacología , Glucuronidasa/metabolismo , Proteínas Klotho , Riñón/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Hipofosfatemia Familiar/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo
3.
Sci Rep ; 13(1): 5794, 2023 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-37031318

RESUMEN

Fibroblast growth factor 23, parathyroid hormone, and 1,25-dihydroxyvitamin D are critical in phosphate homeostasis. Despite these factors' importance, regulators of phosphaturia in the acute postprandial phase remain largely unknown. This study investigated the mechanism of acute phosphate regulation in the postprandial phase in rats. Duodenal administration of radiolabeled phosphate (32P) showed that 32P levels in the inferior vena cava (IVC) blood were lower than those in the portal vein (PV) blood. Serum phosphate concentration transiently increased 5 min after phosphate solution administration through IVC, while it was maintained after the administration through PV. Phosphate administration through both IVC and PV resulted in increased fractional excretion of phosphate (FEPi) at 10 min without elevation of the known circulating factors, but urinary phosphate excretion during the period was 8% of the dose. Experiments using 32P or partial hepatectomy showed that the liver was one of the phosphate reservoirs. The elevation of FEPi and suppression of sodium-phosphate cotransporter 2a in the kidney at 10 min was attenuated in rats with SCH23390, hepatic denervation, or renal denervation, thus indicating that the liver communicated with the kidney via the nervous system to promote phosphaturia. These results revealed previously unknown mechanisms for serum phosphate maintenance.


Asunto(s)
Hipofosfatemia Familiar , Fosfatos , Ratas , Animales , Fosfatos/metabolismo , Vena Porta/metabolismo , Riñón/metabolismo , Hormona Paratiroidea , Homeostasis , Hipofosfatemia Familiar/metabolismo , Hígado/metabolismo
4.
J Cell Mol Med ; 24(9): 4931-4943, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32277576

RESUMEN

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


Asunto(s)
Ácidos Nucleicos Libres de Células , Análisis Mutacional de ADN , Neoplasias/complicaciones , Neoplasias/genética , Osteomalacia/complicaciones , Osteomalacia/genética , Síndromes Paraneoplásicos/complicaciones , Síndromes Paraneoplásicos/genética , Adulto , Biomarcadores de Tumor , Neoplasias Óseas/complicaciones , Neoplasias Óseas/genética , Estudios de Casos y Controles , Sistema Libre de Células , Femenino , Factor-23 de Crecimiento de Fibroblastos , Perfilación de la Expresión Génica , Biblioteca de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hipofosfatemia Familiar/metabolismo , Masculino , Complejo Mediador/genética , Persona de Mediana Edad , Mutación , Mutación Missense , Metástasis de la Neoplasia , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética
5.
Biochem J ; 477(4): 817-831, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32016357

RESUMEN

Inorganic phosphate (Pi) homeostasis is regulated by intestinal absorption via type II sodium-dependent co-transporter (Npt2b) and by renal reabsorption via Npt2a and Npt2c. Although we previously reported that vitamin A-deficient (VAD) rats had increased urine Pi excretion through the decreased renal expression of Npt2a and Npt2c, the effect of vitamin A on the intestinal Npt2b expression remains unclear. In this study, we investigated the effects of treatment with all-trans retinoic acid (ATRA), a metabolite of vitamin A, on the Pi absorption and the Npt2b expression in the intestine of VAD rats, as well as and the underlying molecular mechanisms. In VAD rats, the intestinal Pi uptake activity and the expression of Npt2b were increased, but were reduced by the administration of ATRA. The transcriptional activity of reporter plasmid containing the promoter region of the rat Npt2b gene was reduced by ATRA in NIH3T3 cells overexpressing retinoic acid receptor (RAR) and retinoid X receptor (RXR). On the other hand, CCAAT/enhancer-binding proteins (C/EBP) induced transcriptional activity of the Npt2b gene. Knockdown of the C/EBP gene and a mutation analysis of the C/EBP responsible element in the Npt2b gene promoter indicated that C/EBP plays a pivotal role in the regulation of Npt2b gene transcriptional activity by ATRA. EMSA revealed that the RAR/RXR complex inhibits binding of C/EBP to Npt2b gene promoter. Together, these results suggest that ATRA may reduce the intestinal Pi uptake by preventing C/EBP activation of the intestinal Npt2b gene.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Intestino Delgado/metabolismo , Riñón/metabolismo , Regiones Promotoras Genéticas , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIb/genética , Transcripción Genética/efectos de los fármacos , Tretinoina/farmacología , Animales , Antineoplásicos/farmacología , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Hipofosfatemia Familiar/metabolismo , Hipofosfatemia Familiar/patología , Hipofosfatemia Familiar/prevención & control , Intestino Delgado/efectos de los fármacos , Riñón/efectos de los fármacos , Masculino , Ratones , Células 3T3 NIH , Ratas , Ratas Wistar , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Receptores X Retinoide/genética , Receptores X Retinoide/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIb/metabolismo
6.
Adv Clin Chem ; 90: 133-196, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31122608

RESUMEN

Calcium kidney stones are common worldwide. Most are idiopathic and composed of calcium oxalate. Calcium phosphate is present in around 80% and may initiate stone formation. Stone production is multifactorial with a polygenic genetic contribution. Phosphaturia is found frequently among stone formers but until recently received scant attention. This review examines possible mechanisms for the phosphaturia and its relevance to stone formation from a wide angle. There is a striking lack of clinical data. Phosphaturia is associated, but not correlated, with hypercalciuria, increased 1,25 dihydroxy-vitamin D [1,25 (OH)2D], and sometimes evidence of disturbances in proximal renal tubular function. Phosphate reabsorption in the proximal renal tubules requires tightly regulated interaction of many proteins. Paracellular flow through intercellular tight junctions is the major route of phosphate absorption from the intestine and can be reduced therapeutically in hyperphosphatemic patients. In monogenic defects stones develop when phosphaturia is associated with hypercalciuria, generally explained by increased 1,25 (OH)2D production in response to hypophosphatemia. Calcification does not occur in disorders with increased FGF23 when phosphaturia occurs in isolation and 1,25 (OH)2D is suppressed. Candidate gene studies have identified mutations in the phosphate transporters, but in few individuals. One genome-wide study identified a polymorphism of the phosphate transporter gene SLC34A4 associated with stones. Others did not find mutations obviously linked to phosphate reabsorption. Future genetic studies should have a wide trawl and should focus initially on groups of patients with clearly defined phenotypes. The global data should be pooled.


Asunto(s)
Hipofosfatemia Familiar/metabolismo , Cálculos Renales/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Humanos , Concentración de Iones de Hidrógeno
7.
J Bone Miner Metab ; 37(4): 685-693, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30238432

RESUMEN

Congenital hypophosphataemia (CH) is a collection of disorders that cause defective bone mineralisation manifesting with rickets in childhood and osteomalacia in adulthood. Bone turnover markers (BTMs) are surrogate measures of metabolic bone disease severity. We explored the utility of BTMs in 27 adults with CH: 23 had X-linked hypophosphataemia (XLH), of whom 2 were hypoparathyroid post-total parathyroidectomy (PTx); 2 had autosomal dominant hypophosphataemic rickets (ADHR), and 2 had none of the known mutations. We measured the renal tubular maximum reabsorption rate of phosphate (TmP/GFR), C-terminal fibroblast growth factor 23 (FGF23), parathyroid hormone (PTH), ionised calcium, 1,25-dihydroxyvitamin D [1,25(OH)2D], and a panel of BTMs: serum bone-specific alkaline phosphatase (bone ALP), osteocalcin (Oc), total procollagen type I amino-terminal propeptide (PINP), and carboxy-terminal telopeptide of type I collagen (CTX); and urine amino-terminal telopeptides of type I collagen (uNTX). After excluding 2 patients with XLH and PTx, the frequency of abnormal elevation in BTMs was: bone ALP (96%); CTX (72%); PINP (52%); uNTX (48%); Oc (28%). The strongest association with bone ALP was TmP/GFR. Those patients receiving phosphate supplements and alfacalcidol had significant elevation in CTX. The 2 patients with XLH and PTx had normalisation of TmP/GFR and near normalisation of BTMs post-operatively, despite marked elevation in both C-terminal and intact FGF23. In conclusion, BTMs in our CH patients indicated that most have abnormalities consistent with osteomalacia and many have mild secondary hyperparathyroidism; and the normalisation of TmP/GFR after total PTx in 2 cases of XLH remains unexplained, but possible causes are speculated.


Asunto(s)
Biomarcadores/metabolismo , Remodelación Ósea , Hipofosfatemia Familiar/metabolismo , Riñón/patología , Paratiroidectomía/efectos adversos , Fosfatos/metabolismo , Adolescente , Adulto , Femenino , Factor-23 de Crecimiento de Fibroblastos , Humanos , Hipofosfatemia Familiar/genética , Masculino , Persona de Mediana Edad , Adulto Joven
8.
Sci Rep ; 8(1): 13701, 2018 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-30209259

RESUMEN

In chronic kidney disease (CKD), high serum phosphate concentration is associated with cardiovascular disease and deterioration in renal function. In early CKD, the serum phosphate concentration is normal due to increased fractional excretion of phosphate. Our premise was that high phosphate intake even in patients with early CKD would result in an excessive load of phosphate causing tubular injury and accelerating renal function deterioration. In CKD 2-3 patients, we evaluated whether increased phosphaturia accelerates CKD progression. To have a uniform group of patients with early CKD, 95 patients with metabolic syndrome without overt proteinuria were followed for 2.7 ± 1.6 years. The median decline in eGFR was 0.50 ml/min/1.73 m2/year. Patients with a more rapid decrease in eGFR had greater phosphaturia. Moreover, the rate of decrease in eGFR inversely correlated with the degree of phosphaturia. Additionally, phosphaturia independently predicted renal function deterioration. In heminephrectomized rats, a high phosphate diet increased phosphaturia resulting in renal tubular damage associated with inflammation, oxidative stress and low klotho expression. Moreover, in rats with hyperphosphatemia and metabolic syndrome antioxidant treatment resulted in attenuation of renal lesions. In HEK-293 cells, high phosphate promoted oxidative stress while melatonin administration reduced ROS generation. Our findings suggest that phosphate loading in early CKD, results in renal damage and a more rapid decrease in renal function due to renal tubular injury.


Asunto(s)
Hipofosfatemia Familiar/fisiopatología , Riñón/fisiopatología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antioxidantes/metabolismo , Línea Celular , Femenino , Tasa de Filtración Glomerular/efectos de los fármacos , Tasa de Filtración Glomerular/fisiología , Glucuronidasa/metabolismo , Células HEK293 , Humanos , Hiperfosfatemia/metabolismo , Hiperfosfatemia/fisiopatología , Hipofosfatemia Familiar/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Proteínas Klotho , Masculino , Melatonina/farmacología , Síndrome Metabólico/metabolismo , Síndrome Metabólico/fisiopatología , Persona de Mediana Edad , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Fosfatos/metabolismo , Proteinuria/metabolismo , Proteinuria/fisiopatología , Ratas , Ratas Wistar , Ratas Zucker , Especies Reactivas de Oxígeno/metabolismo , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/fisiopatología , Adulto Joven
9.
Med Hypotheses ; 96: 66-67, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27959279

RESUMEN

The addition of phosphate groups is an essential requirement for the proper functioning of cyclin and cyclin dependent kinase which control various stages in the mitotic division of cancer cells. Thus limiting the availability of phosphate is likely to interfere with the metabolism of rapidly growing malignant cells. The human hormone glucagon and the anti metabolite mithramycin reduce serum phosphate by increasing phosphaturia and are both very effective in treating Paget's disease of bone, a precancerous condition. In this disorder large doses of glucagon given intravenously relieve bone pain and cause serum phosphate and alkaline phosphatase as well as urine hydroxyproline to fall, indicating a marked reduction in bone turnover. A constant iv infusion of glucagon was given to each of three patients all of whom had secondary malignant bone deposits. Two of the patients had primary prostate cancer and one had a squamous cell lung tumour. All three patients had relief of bone pain and a fall in serum alkaline phosphatase. Serum acid phosphatase also fell in the two patients with prostate cancer. It is proposed that the marked drop in serum phosphate due to glucagon causes intracellular phosphate to fall. This in turn disrupts the addition and removal of phosphate groups essential for the proper functioning of cyclin and cyclin dependent kinase. These two proteins control the transition from G1 to S (DNA synthesis phase) and G2 to M (mitotic phase) in the dividing cycle of malignant cells. Depriving a tumour of an essential ingredient used in phosphorylation reactions will disrupt its growth. It is also proposed that, by the same mechanism, glucagon induced hypophosphataemia renders malignant cells more sensitive to established chemotherapeutic agents and radiation waves. If this hypothesis proves to be correct, lowering intracellular phosphate may become an useful tool in cancer therapy. However extensive studies are necessary to determine whether mitosis in cancer cells can be advantageously disrupted by glucagon induced hypophosphataemia.


Asunto(s)
Mitosis , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Fosfatos/química , Fosfatasa Ácida/metabolismo , Fosfatasa Alcalina/metabolismo , Huesos/metabolismo , Carcinoma de Células Escamosas/tratamiento farmacológico , Glucagón/química , Glucagón/metabolismo , Humanos , Hidroxiprolina/orina , Hipofosfatemia Familiar/metabolismo , Insulina/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Masculino , Modelos Teóricos , Neoplasias/metabolismo , Osteítis Deformante/tratamiento farmacológico , Fosforilación , Neoplasias de la Próstata/tratamiento farmacológico
10.
PLoS One ; 11(7): e0159804, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27442254

RESUMEN

Contribution of salt wasting and volume depletion to the pathogenesis of hypercalciuria and hyperphosphaturia is poorly understood. Pendrin/NCC double KO (pendrin/NCC-dKO) mice display severe salt wasting under basal conditions and develop profound volume depletion, prerenal renal failure, and metabolic alkalosis and are growth retarded. Microscopic examination of the kidneys of pendrin/NCC-dKO mice revealed the presence of calcium phosphate deposits in the medullary collecting ducts, along with increased urinary calcium and phosphate excretion. Confirmatory studies revealed decreases in the expression levels of sodium phosphate transporter-2 isoforms a and c, increases in the expression of cytochrome p450 family 4a isotypes 12 a and b, as well as prostaglandin E synthase 1, and cyclooxygenases 1 and 2. Pendrin/NCC-dKO animals also had a significant increase in urinary prostaglandin E2 (PGE-2) and renal content of 20-hydroxyeicosatetraenoic acid (20-HETE) levels. Pendrin/NCC-dKO animals exhibit reduced expression levels of the sodium/potassium/2chloride co-transporter 2 (NKCC2) in their medullary thick ascending limb. Further assessment of the renal expression of NKCC2 isoforms by quantitative real time PCR (qRT-PCR) reveled that compared to WT mice, the expression of NKCC2 isotype F was significantly reduced in pendrin/NCC-dKO mice. Provision of a high salt diet to rectify volume depletion or inhibition of PGE-2 synthesis by indomethacin, but not inhibition of 20-HETE generation by HET0016, significantly improved hypercalciuria and salt wasting in pendrin/NCC dKO mice. Both high salt diet and indomethacin treatment also corrected the alterations in NKCC2 isotype expression in pendrin/NCC-dKO mice. We propose that severe salt wasting and volume depletion, irrespective of the primary originating nephron segment, can secondarily impair the reabsorption of salt and calcium in the thick ascending limb of Henle and/or proximal tubule, and reabsorption of sodium and phosphate in the proximal tubule via processes that are mediated by PGE-2.


Asunto(s)
Calcio/metabolismo , Dinoprostona/metabolismo , Nefronas/metabolismo , Fosfatos/metabolismo , Animales , Proteínas de Transporte de Anión/deficiencia , Dieta , Modelos Animales de Enfermedad , Hipercalciuria/etiología , Hipercalciuria/metabolismo , Hipercalciuria/patología , Hipofosfatemia Familiar/etiología , Hipofosfatemia Familiar/metabolismo , Hipofosfatemia Familiar/patología , Pruebas de Función Renal , Masculino , Ratones , Ratones Noqueados , Nefronas/patología , Fenotipo , Miembro 1 de la Familia de Transportadores de Soluto 12/deficiencia , Transportadores de Sulfato
11.
J Bone Miner Res ; 31(6): 1247-57, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26792657

RESUMEN

The transgenic and knockout (KO) animals involving Fgf23 have been highly informative in defining novel aspects of mineral metabolism, but are limited by shortened lifespan, inability of spatial/temporal FGF23 control, and infertility of the global KO. To more finely test the role of systemic and genetic influences in FGF23 production, a mouse was developed that carried a floxed ("f")-Fgf23 allele (exon 2 floxed) which demonstrated in vivo recombination when bred to global-Cre transgenic mice (eIIa-cre). Mice homozygous for the recombined allele ("Δ") had undetectable serum intact FGF23, elevated serum phosphate (p < 0.05), and increased kidney Cyp27b1 mRNA (p < 0.05), similar to global Fgf23-KO mice. To isolate cellular FGF23 responses during phosphate challenge, Fgf23(Δ/f) mice were mated with early osteoblast type Iα1 collagen 2.3-kb promoter-cre mice (Col2.3-cre) and the late osteoblast/early osteocyte Dentin matrix protein-1-cre (Dmp1-cre). Fgf23(Δ/f) /Col2.3-cre(+) and Fgf23(Δ/f) /Dmp1-cre(+) exhibited reduced baseline serum intact FGF23 versus controls. After challenge with high-phosphate diet Cre(-) mice had 2.1-fold to 2.5-fold increased serum FGF23 (p < 0.01), but Col2.3-cre(+) mice had no significant increase, and Dmp1-cre(+) mice had only a 37% increase (p < 0.01) despite prevailing hyperphosphatemia in both models. The Fgf23(Δ/f) /Col2.3-cre was bred onto the Hyp (murine X-linked hypophosphatemia [XLH] model) genetic background to test the contribution of osteoblasts and osteocytes to elevated FGF23 and Hyp disease phenotypes. Whereas Hyp mice maintained inappropriately elevated FGF23 considering their marked hypophosphatemia, Hyp/Fgf23(Δ/f) /Col2.3-cre(+) mice had serum FGF23 <4% of Hyp (p < 0.01), and this targeted restriction normalized serum phosphorus and ricketic bone disease. In summary, deleting FGF23 within early osteoblasts and osteocytes demonstrated that both cell types contribute to baseline circulating FGF23 concentrations, and that targeting osteoblasts/osteocytes for FGF23 production can modify systemic responses to changes in serum phosphate concentrations and rescue the Hyp genetic syndrome. © 2016 American Society for Bone and Mineral Research.


Asunto(s)
Huesos/metabolismo , Factores de Crecimiento de Fibroblastos/deficiencia , Hipofosfatemia Familiar/genética , Hipofosfatemia Familiar/metabolismo , Osteocitos/metabolismo , Fosfatos/metabolismo , Animales , Huesos/patología , Factor-23 de Crecimiento de Fibroblastos , Eliminación de Gen , Hipofosfatemia Familiar/patología , Ratones , Ratones Noqueados , Osteocitos/patología
12.
Med Mol Morphol ; 49(4): 203-209, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26122367

RESUMEN

Phosphaturic mesenchymal tumors (PMT) are the most common cause of tumor-induced osteomalacia (TIO) related to mesenchymal neoplasms. The lineage of differentiation of PMTs has not been elucidated in existing literature. Fourteen cases of PMT were analyzed for this study to elucidate its lineage. We used vascular and/or lymphatic endothelial markers for the immunohistochemical analysis, which included CD31, CD34, factor VIII-related antigen, podoplanin, Freund's leukemia integration site 1 (FLI1), and avian v-ets erythroblastosis virus E26 oncogene homolog (ERG). FLI1 and ERG were stained in all cases with proportion of immunopositive tumor cells largely more than 50 %; staining intensity was moderate or strong for both FLI1 and ERG. The tumor cells were stained with CD31 and/or CD34, with significantly less staining than observed for FLI1 and ERG. The tumor cells were completely immunonegative for factor VIII-related antigen and podoplanin. FLI1 and ERG are known to have considerable specificity to endothelial cells; ERG is more widely equipped in surgical pathology laboratories than FLI1. We concluded that ERG (or FLI1 if available) is useful marker for the diagnosis of PMT, and that PMTs may have an endothelial cell lineage.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Hipofosfatemia Familiar/patología , Mesodermo/patología , Neoplasias/metabolismo , Neoplasias/patología , Proteína Proto-Oncogénica c-fli-1/metabolismo , Adulto , Anciano , Femenino , Humanos , Hipofosfatemia Familiar/metabolismo , Inmunohistoquímica , Masculino , Mesodermo/metabolismo , Persona de Mediana Edad , Regulador Transcripcional ERG/metabolismo
13.
J Bone Miner Res ; 30(10): 1925-37, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25827490

RESUMEN

The Na(+) -dependent phosphate-cotransporter NaPi-IIb (SLC34A2) is widely expressed, with intestine, lung, and testis among the organs with highest levels of mRNA abundance. In mice, the intestinal expression of NaPi-IIb is restricted to the ileum, where the cotransporter localizes specifically at the brush border membrane (BBM) and mediates the active transport of inorganic phosphate (Pi). Constitutive full ablation of NaPi-IIb is embryonically lethal whereas the global but inducible removal of the transporter in young mice leads to intestinal loss of Pi and lung calcifications. Here we report the generation of a constitutive but intestinal-specific NaPi-IIb/Slc34a2-deficient mouse model. Constitutive intestinal ablation of NaPi-IIb results in viable pups with normal growth. Homozygous mice are characterized by fecal wasting of Pi and complete absence of Na/Pi cotransport activity in BBM vesicles (BBMVs) isolated from ileum. In contrast, the urinary excretion of Pi is reduced in these animals. The plasma levels of Pi are similar in wild-type and NaPi-IIb-deficient mice. In females, the reduced phosphaturia associates with higher expression of NaPi-IIa and higher Na/Pi cotransport activity in renal BBMVs, as well as with reduced plasma levels of intact FGF-23. A similar trend is found in males. Thus, NaPi-IIb is the only luminal Na(+) -dependent Pi transporter in the murine ileum and its absence is fully compensated for in adult females by a mechanism involving the bone-kidney axis. The contribution of this mechanism to the adaptive response is less apparent in adult males.


Asunto(s)
Íleon/metabolismo , Riñón/metabolismo , Microvellosidades/metabolismo , Fosfatos/sangre , Fosfatos/orina , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIb/metabolismo , Animales , Transporte Biológico Activo/genética , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/sangre , Hipofosfatemia Familiar/metabolismo , Íleon/patología , Riñón/patología , Masculino , Ratones , Ratones Noqueados , Microvellosidades/genética , Microvellosidades/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIb/genética
14.
J Am Soc Nephrol ; 25(12): 2730-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24854273

RESUMEN

The human response to acute phosphate (PO4) loading is poorly characterized, and it is unknown whether an intestinal phosphate sensor mechanism exists. Here, we characterized the human mineral and endocrine response to parenteral and duodenal acute phosphate loads. Healthy human participants underwent 36 hours of intravenous (IV; 1.15 [low dose] and 2.30 [high dose] mmol of PO4/kg per 24 hours) or duodenal (1.53 mmol of PO4/kg per 24 hours) neutral sodium PO4 loading. Control experiments used equimolar NaCl loads. Maximum PO4 urinary excretory responses occurred between 12 and 24 hours and were similar for low-dose IV and duodenal infusion. Hyperphosphatemic responses were also temporally and quantitatively similar for low-dose IV and duodenal PO4 infusion. Fractional renal PO4 clearance increased approximately 6-fold (high-dose IV group) and 4-fold (low-dose IV and duodenal groups), and significant reductions in plasma PO4 concentrations relative to peak values occurred by 36 hours, despite persistent PO4 loading. After cessation of loading, frank hypophosphatemia occurred. The earliest phosphaturic response occurred after plasma PO4 and parathyroid hormone concentrations increased. Plasma fibroblast growth factor-23 concentration increased after the onset of phosphaturia, followed by a decrease in plasma 1,25(OH)2D levels; α-Klotho levels did not change. Contrary to results in rodents, we found no evidence for intestinal-specific phosphaturic control mechanisms in humans. Complete urinary phosphate recovery in the IV loading groups provides evidence against any important extrarenal response to acute PO4 loads.


Asunto(s)
Fosfatos/sangre , Fosfatos/metabolismo , Administración Intravenosa , Adulto , Duodeno/efectos de los fármacos , Electrólitos/química , Sistema Endocrino/metabolismo , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Tasa de Filtración Glomerular , Glucuronidasa/metabolismo , Humanos , Hipocalcemia/metabolismo , Hipofosfatemia/metabolismo , Hipofosfatemia Familiar/metabolismo , Infusiones Intravenosas , Proteínas Klotho , Masculino , Hormona Paratiroidea/metabolismo , Fosfatos/orina , Factores de Tiempo , Adulto Joven
15.
PLoS One ; 9(4): e93840, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24710520

RESUMEN

Osteocytes express multiple genes involved in mineral metabolism including PHEX, FGF23, DMP1 and FAM20C. In Hyp mice, a murine model for X-linked hypophosphatemia (XLH), Phex deficiency results in the overproduction of FGF23 in osteocytes, which leads to hypophosphatemia and impaired vitamin D metabolism. In this study, to further clarify the abnormality in osteocytes of Hyp mice, we obtained detailed gene expression profiles in osteoblasts and osteocytes isolated from the long bones of 20-week-old Hyp mice and wild-type (WT) control mice. The expression of Fgf23, Dmp1, and Fam20c was higher in osteocytic cells than in osteoblastic cells in both genotypes, and was up-regulated in Hyp cells. Interestingly, the up-regulation of these genes in Hyp bones began before birth. On the other hand, the expression of Slc20a1 encoding the sodium/phosphate (Na+/Pi) co-transporter Pit1 was increased in osteoblasts and osteocytes from adult Hyp mice, but not in Hyp fetal bones. The direct effects of extracellular Pi and 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] on isolated osteoblastic and osteocytic cells were also investigated. Twenty-four-hour treatment with 10-8 M 1,25(OH)2D3 increased the expression of Fgf23 in WT osteoblastic cells but not in osteocytic cells. Dmp1 expression in osteocytic cells was increased due to the 24-hour treatment with 10 mM Pi and was suppressed by 10-8 M 1,25(OH)2D3 in WT osteocytic cells. We also found the up-regulation of the genes for FGF1, FGF2, their receptors, and Egr-1 which is a target of FGF signaling, in Hyp osteocytic cells, suggesting the activation of FGF/FGFR signaling. These results implicate the complex gene dysregulation in osteoblasts and osteocytes of Hyp mice, which might contribute to the pathogenesis.


Asunto(s)
Expresión Génica , Hipofosfatemia Familiar/genética , Osteoblastos/metabolismo , Osteocitos/metabolismo , Animales , Huesos/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica , Hipofosfatemia Familiar/metabolismo , Ratones , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo , Regulación hacia Arriba
16.
Kidney Int ; 84(4): 639-41, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24080872

RESUMEN

Fibroblast growth factor 23 (FGF23) is an important regulator of phosphate homeostasis, yet efforts to control its circulating levels by manipulating dietary phosphate intake in humans and animals with normal or impaired kidney function have yielded conflicting results. In the study by Zhang et al., severe phosphate restriction in a genetic mouse model of progressive kidney dysfunction failed to cause a uniform FGF23 reduction, again highlighting the complexity of FGF23 regulation.


Asunto(s)
Lesión Renal Aguda/sangre , Lesión Renal Aguda/fisiopatología , Factores de Crecimiento de Fibroblastos/sangre , Factores de Crecimiento de Fibroblastos/metabolismo , Hipofosfatemia Familiar/metabolismo , Hipofosfatemia Familiar/prevención & control , Nefritis Hereditaria/metabolismo , Fosfatos/administración & dosificación , Fosfatos/deficiencia , Insuficiencia Renal Crónica/metabolismo , Transducción de Señal/fisiología , Animales , Femenino , Factor-23 de Crecimiento de Fibroblastos , Humanos , Masculino
17.
Hum Pathol ; 44(12): 2711-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24060005

RESUMEN

Tumor-induced osteomalacia (TIO) is a paraneoplastic syndrome associated with tumors that secrete phosphaturic hormones, most notably fibroblast growth factor 23 (FGF23). The majority of tumors associated with this syndrome show stereotypical histological features and are now known as phosphaturic mesenchymal tumors (PMTs). We postulated that immunohistochemistry for somatostatin receptor 2A (SSTR2A) could be used to definitively identify PMTs or other tumors that cause TIO. Immunohistochemistry for FGF23 and SSTR2A was performed on 15 tumors from 14 patients with a definite diagnosis of TIO. All showed positive staining for both markers. While FGF23 staining was quite focal in some tumors, SSTR2A showed diffuse strong expression. In 40 control tumors not known to be associated with the clinical or biochemical features of TIO, FGF23 expression was found in 2 cases (one aneurysmal bone cyst and one osteosarcoma). SSTR2A expression was found in 9 control tumors (4 synovial sarcomas, 2 hemangiomas, 2 aneurysmal bone cysts and one osteosarcoma). Only one tumor (an aneurysmal bone cyst) showed positive staining for both FGF23 and SSTR2A. SSTR2A also commonly stained neoplastic and non-neoplastic endothelial cells. We conclude that neither FGF23 nor SSTR2A expression are specific for the diagnosis of PMT. However both stains are highly sensitive. Because of its diffuse strong expression and widespread availability, immunohistochemistry for SSTR2A is useful to confirm the diagnosis of PMT in an appropriate setting particularly if material is limited. Negative staining can serve as an excellent rule out test for this diagnosis.


Asunto(s)
Hipofosfatemia Familiar/diagnóstico , Mesenquimoma/diagnóstico , Neoplasias de Tejido Conjuntivo/diagnóstico , Osteomalacia/diagnóstico , Síndromes Paraneoplásicos/diagnóstico , Receptores de Somatostatina/metabolismo , Adulto , Medicamentos Herbarios Chinos , Eleutherococcus , Femenino , Factor-23 de Crecimiento de Fibroblastos , Humanos , Hipofosfatemia Familiar/metabolismo , Hipofosfatemia Familiar/patología , Masculino , Mesenquimoma/metabolismo , Mesenquimoma/patología , Persona de Mediana Edad , Neoplasias de Tejido Conjuntivo/metabolismo , Neoplasias de Tejido Conjuntivo/patología , Osteomalacia/metabolismo , Osteomalacia/patología , Síndromes Paraneoplásicos/metabolismo , Síndromes Paraneoplásicos/patología
18.
Contrib Nephrol ; 180: 124-37, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23652555

RESUMEN

Deregulated phosphate homeostasis can lead to a wide range of disorders, including myopathy, cardiac dysfunction, and skeletal abnormalities. Therefore, characterization of the molecular regulation of phosphate metabolism is of pathophysiological and clinical significance. Hyp mouse is the model for human X-linked hypophosphatemia which is due to mutations that inactivate the endopeptidases of the X chromosome (PHEX). PHEX inactivation leads to increased serum levels of fibroblast growth factor 23 (FGF23), a phosphaturic hormone that induces excessive renal phosphate excretion and severe hypophosphatemia. The expression of WNT signaling components is increased in Hyp mice. To determine the potential role of WNT signaling in FGF23-mediated hypophosphatemia, we cross-bred Hyp mice with mice deficient in the WNT coreceptor low-density lipoprotein receptor-related protein 6 (Lrp6) to generate Hyp and Lrp6 double mutant mice (Hyp/Lrp6). Like Hyp mice, Hyp/Lrp6 double mutants maintained high serum levels of FGF23, and accordingly exhibited hypophosphatemia to the same degree as the Hyp mice did, indicating that genetically reducing WNT signaling does not impact FGF23-induced phosphaturia. Moreover, similar to Hyp mice, the Hyp/Lrp6 double mutants also exhibited reduced mineralization of the bone, further supporting that reduced WNT signaling does not affect the chronic phosphate wasting caused by excess FGF23 in these mice. In further support of our finding, injection of bioactive FGF23 protein into Lrp6 mutant mice reduced serum phosphate levels to a similar degree as FGF23 injection into wild-type mice. Our in vivo studies provide genetic and pharmacological evidence for a WNT-independent function of FGF23 in the regulation of phosphate homeostasis.


Asunto(s)
Modelos Animales de Enfermedad , Raquitismo Hipofosfatémico Familiar/fisiopatología , Factores de Crecimiento de Fibroblastos/fisiología , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/deficiencia , Endopeptidasa Neutra Reguladora de Fosfato PHEX/fisiología , Vía de Señalización Wnt , Animales , Raquitismo Hipofosfatémico Familiar/diagnóstico por imagen , Raquitismo Hipofosfatémico Familiar/etiología , Raquitismo Hipofosfatémico Familiar/genética , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/sangre , Factores de Crecimiento de Fibroblastos/toxicidad , Homeostasis , Hipofosfatemia Familiar/genética , Hipofosfatemia Familiar/metabolismo , Riñón/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/fisiología , Masculino , Ratones , Ratones Noqueados , Endopeptidasa Neutra Reguladora de Fosfato PHEX/genética , Fosfatos/metabolismo , Radiografía , Proteínas Recombinantes/toxicidad , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo II/biosíntesis , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo II/genética
19.
Kidney Int ; 84(4): 713-21, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23698235

RESUMEN

Fibroblast growth factor 23 (FGF23) is a phosphaturic hormone that in end-stage renal disease is markedly increased in serum; however, the mechanisms responsible for this increase are unclear. Here, we tested whether phosphate retention in chronic kidney disease (CKD) is responsible for the elevation of FGF23 in serum using Col4α3 knockout mice, a murine model of Alport disease exhibiting CKD. We found a significant elevation in serum FGF23 in progressively azotemic 8- and 12-week-old CKD mice along with an increased fractional excretion of phosphorus. Both moderate and severe phosphate restriction reduced fractional excretion of phosphorus by 8 weeks, yet serum FGF23 levels remained strikingly elevated. By 12 weeks, FGF23 levels were further increased with moderate phosphate restriction, while severe phosphate restriction led to severe bone mineralization defects and decreased FGF23 production in bone. CKD mice on a control diet had low serum 1,25-dihydroxyvitamin D (1,25(OH)(2)D) levels and 3-fold higher renal Cyp24α1 gene expression compared to wild-type mice. Severe phosphate restriction increased 1,25(OH)(2)D levels in CKD mice by 8 weeks and lowered renal Cyp24α1 gene expression despite persistently elevated serum FGF23. Renal klotho gene expression declined in CKD mice on a control diet, but improved with severe phosphate restriction. Thus, dietary phosphate restriction reduces the fractional excretion of phosphorus independent of serum FGF23 levels in mice with CKD.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Hipofosfatemia Familiar/metabolismo , Hipofosfatemia Familiar/prevención & control , Nefritis Hereditaria/metabolismo , Fosfatos/administración & dosificación , Fosfatos/deficiencia , Insuficiencia Renal Crónica/metabolismo , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/metabolismo , Administración Oral , Animales , Autoantígenos/genética , Autoantígenos/metabolismo , Huesos/metabolismo , Colágeno Tipo IV/deficiencia , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Modelos Animales de Enfermedad , Femenino , Factor-23 de Crecimiento de Fibroblastos , Glucuronidasa/metabolismo , Riñón/metabolismo , Proteínas Klotho , Masculino , Ratones , Ratones Noqueados , Fosfatos/orina , Esteroide Hidroxilasas/metabolismo , Vitamina D/análogos & derivados , Vitamina D/sangre , Vitamina D3 24-Hidroxilasa
20.
J Bone Miner Res ; 28(6): 1378-85, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23325605

RESUMEN

Fibroblast growth factor 23 (FGF23) plays a crucial role in renal phosphate regulation, exemplified by the causal role of PHEX and DMP1 mutations in X-linked hypophosphatemic rickets and autosomal recessive rickets type 1, respectively. Using whole exome sequencing we identified compound heterozygous mutations in family with sequence similarity 20, member C (FAM20C) in two siblings referred for hypophosphatemia and severe dental demineralization disease. FAM20C mutations were not found in other undiagnosed probands of a national Norwegian population of familial hypophosphatemia. Our results demonstrate that mutations in FAM20C provide a putative new mechanism in human subjects leading to dysregulated FGF23 levels, hypophosphatemia, hyperphosphaturia, dental anomalies, intracerebral calcifications and osteosclerosis of the long bones in the absence of rickets.


Asunto(s)
Calcinosis , Exoma , Proteínas de la Matriz Extracelular , Factores de Crecimiento de Fibroblastos/metabolismo , Hipofosfatemia Familiar , Anomalías Dentarias , Adolescente , Adulto , Calcinosis/genética , Calcinosis/metabolismo , Calcinosis/patología , Quinasa de la Caseína I , Análisis Mutacional de ADN , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Humanos , Hipofosfatemia Familiar/genética , Hipofosfatemia Familiar/metabolismo , Hipofosfatemia Familiar/patología , Masculino , Mutación , Noruega , Osteosclerosis/genética , Osteosclerosis/metabolismo , Osteosclerosis/patología , Anomalías Dentarias/genética , Anomalías Dentarias/metabolismo , Anomalías Dentarias/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...