Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 44(3): 584-602, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38205639

RESUMEN

Hyperphosphatemia is a common feature in patients with impaired kidney function and is associated with increased risk of cardiovascular disease. This phenomenon extends to the general population, whereby elevations of serum phosphate within the normal range increase risk; however, the mechanism by which this occurs is multifaceted, and many aspects are poorly understood. Less than 1% of total body phosphate is found in the circulation and extracellular space, and its regulation involves multiple organ cross talk and hormones to coordinate absorption from the small intestine and excretion by the kidneys. For phosphate to be regulated, it must be sensed. While mostly enigmatic, various phosphate sensors have been elucidated in recent years. Phosphate in the circulation can be buffered, either through regulated exchange between extracellular and cellular spaces or through chelation by circulating proteins (ie, fetuin-A) to form calciprotein particles, which in themselves serve a function for bulk mineral transport and signaling. Either through direct signaling or through mediators like hormones, calciprotein particles, or calcifying extracellular vesicles, phosphate can induce various cardiovascular disease pathologies: most notably, ectopic cardiovascular calcification but also left ventricular hypertrophy, as well as bone and kidney diseases, which then propagate phosphate dysregulation further. Therapies targeting phosphate have mostly focused on intestinal binding, of which appreciation and understanding of paracellular transport has greatly advanced the field. However, pharmacotherapies that target cardiovascular consequences of phosphate directly, such as vascular calcification, are still an area of great unmet medical need.


Asunto(s)
Enfermedades Cardiovasculares , Hiperfosfatemia , Insuficiencia Renal Crónica , Calcificación Vascular , Humanos , Fosfatos/metabolismo , Enfermedades Cardiovasculares/metabolismo , Hiperfosfatemia/tratamiento farmacológico , Calcificación Vascular/etiología , Hormonas/uso terapéutico
2.
Int J Mol Sci ; 22(11)2021 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-34071837

RESUMEN

Phosphate homeostasis is essential for health and is achieved via interaction between the bone, kidney, small intestine, and parathyroid glands and via intricate processes involving phosphate transporters, phosphate sensors, and circulating hormones. Numerous genetic and acquired disorders are associated with disruption in these processes and can lead to significant morbidity and mortality. The role of the kidney in phosphate homeostasis is well known, although it is recognized that the cellular mechanisms in murine models and humans are different. Intestinal phosphate transport also appears to differ in humans and rodents, with recent studies demonstrating a dominant role for the paracellular pathway. The existence of phosphate sensing has been acknowledged for decades; however, the underlying molecular mechanisms are poorly understood. At least three phosphate sensors have emerged. PiT2 and FGFR1c both act as phosphate sensors controlling Fibroblast Growth Factor 23 secretion in bone, whereas the calcium-sensing receptor controls parathyroid hormone secretion in response to extracellular phosphate. All three of the proposed sensors are expressed in the kidney and intestine but their exact function in these organs is unknown. Understanding organ interactions and the mechanisms involved in phosphate sensing requires significant research to develop novel approaches for the treatment of phosphate homeostasis disorders.


Asunto(s)
Homeostasis , Proteínas de Transporte de Fosfato/metabolismo , Fosfatos/metabolismo , Transducción de Señal , Animales , Fenómenos Fisiológicos Celulares , Susceptibilidad a Enfermedades , Humanos , Modelos Animales , Especificidad de Órganos
3.
J Biol Chem ; 293(6): 2102-2114, 2018 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-29233890

RESUMEN

Extracellular phosphate (Pi) can act as a signaling molecule that directly alters gene expression and cellular physiology. The ability of cells or organisms to detect changes in extracellular Pi levels implies the existence of a Pi-sensing mechanism that signals to the body or individual cell. However, unlike in prokaryotes, yeasts, and plants, the molecular players involved in Pi sensing in mammals remain unknown. In this study, we investigated the involvement of the high-affinity, sodium-dependent Pi transporters PiT1 and PiT2 in mediating Pi signaling in skeletal cells. We found that deletion of PiT1 or PiT2 blunted the Pi-dependent ERK1/2-mediated phosphorylation and subsequent gene up-regulation of the mineralization inhibitors matrix Gla protein and osteopontin. This result suggested that both PiTs are necessary for Pi signaling. Moreover, the ERK1/2 phosphorylation could be rescued by overexpressing Pi transport-deficient PiT mutants. Using cross-linking and bioluminescence resonance energy transfer approaches, we found that PiT1 and PiT2 form high-abundance homodimers and Pi-regulated low-abundance heterodimers. Interestingly, in the absence of sodium-dependent Pi transport activity, the PiT1-PiT2 heterodimerization was still regulated by extracellular Pi levels. Of note, when two putative Pi-binding residues, Ser-128 (in PiT1) and Ser-113 (in PiT2), were substituted with alanine, the PiT1-PiT2 heterodimerization was no longer regulated by extracellular Pi These observations suggested that Pi binding rather than Pi uptake may be the key factor in mediating Pi signaling through the PiT proteins. Taken together, these results demonstrate that Pi-regulated PiT1-PiT2 heterodimerization mediates Pi sensing independently of Pi uptake.


Asunto(s)
Fosfatos/metabolismo , Multimerización de Proteína , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo , Animales , Transporte Biológico , Sistema de Señalización de MAP Quinasas , Mamíferos , Fosfatos/fisiología , Fosforilación , Unión Proteica , Transducción de Señal
4.
Pflugers Arch ; 471(1): 175-184, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30511265

RESUMEN

Under normal physiological condition, the biomineralization process is limited to skeletal tissues and teeth and occurs throughout the individual's life. Biomineralization is an actively regulated process involving the progressive mineralization of the extracellular matrix secreted by osteoblasts in bone or odontoblasts and ameloblasts in tooth. Although the detailed molecular mechanisms underlying the formation of calcium-phosphate apatite crystals are still debated, it is suggested that calcium and phosphate may need to be transported across the membrane of the mineralizing cell, suggesting a pivotal role of phosphate transporters in bone and tooth mineralization. In this context, this short review describes the current knowledge on the role of Slc34 Na+-phosphate transporters in skeletal and tooth mineralization.


Asunto(s)
Huesos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo II/genética , Diente/metabolismo , Animales , Biomineralización , Humanos , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo II/metabolismo
5.
Cell Physiol Biochem ; 35(3): 841-57, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25632940

RESUMEN

BACKGROUND/AIMS: Multipotent stem/stromal cells (MSC) are considered promising for cartilage tissue engineering. However, chondrogenic differentiation of MSC can ultimately lead to the formation of hypertrophic chondrocytes responsible for the calcification of cartilage. To prevent the production of this calcified matrix at the articular site, the late hypertrophic differentiation of MSCs must be carefully controlled. Given that articular cartilage is avascular, we hypothesized that in addition to its stimulatory role in the early differentiation of chondrogenic cells, hypoxia may prevent their late hypertrophic conversion. METHODS: Early and late chondrogenic differentiation were evaluated using human adipose MSC and murine ATDC5 cells cultured under either normoxic (21%O2) or hypoxic (5%O2) conditions. To investigate the effect of hypoxia on late chondrogenic differentiation, the transcriptional activity of hypoxia-inducible factor-1alpha (HIF-1α) and HIF-2α were evaluated using the NoShift DNA-binding assay and through modulation of their activity (chemical inhibitor, RNA interference). Results : Our data demonstrate that low oxygen tension not only stimulates the early chondrogenic commitment of two complementary models of chondrogenic cells, but also inhibits their hypertrophic differentiation. Conclusion : These results suggest that hypoxia can be used as an instrumental tool to prevent the formation of a calcified matrix in MSC-based cartilage tissue engineering.


Asunto(s)
Cartílago Articular/crecimiento & desarrollo , Diferenciación Celular/genética , Células Madre Mesenquimatosas/citología , Ingeniería de Tejidos , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/biosíntesis , Calcinosis/genética , Calcinosis/patología , Cartílago Articular/citología , Hipoxia de la Célula , Condrocitos/citología , Condrogénesis/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Células Madre Mesenquimatosas/metabolismo , Ratones , Oxígeno/metabolismo
6.
Blood ; 121(4): 666-78, 2013 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-23190530

RESUMEN

The PIT1/SLC20A1 protein, a well-described sodium/phosphate cotransporter and retrovirus receptor, has been identified recently as a modular of proliferation and apoptosis in vitro. The targeted deletion of the PIT1 gene in mice revealed a lethal phenotype due to severe anemia attributed to defects in liver development. However, the presence of immature erythroid cells associated with impaired maturation of the globin switch led us to investigate the role of PIT1 in hematopoietic development. In the present study, specific deletion of PIT1 in the hematopoietic system and fetal liver transplantation experiments demonstrated that anemia was associated with an erythroid cell- autonomous defect. Moreover, anemia was not due to RBC destruction but rather to maturation defects. Because Erythroid Krüppel-like Factor (EKLF)-knockout mice showed similar maturation defects, we investigated the functional link between PIT1 and EKLF. We demonstrated that EKLF increases PIT1 expression during RBC maturation by binding to its promoter in vivo and that shRNA-driven depletion of either PIT1 or EKLF impairs erythroid maturation of G1E cells in vitro, whereas reexpression of PIT1 in EKLF-depleted G1E cells partially restores erythroid maturation. This is the first demonstration of a physiologic involvement of PIT1 in erythroid maturation in vivo.


Asunto(s)
Células Eritroides/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Factor de Transcripción Pit-1/genética , Animales , Secuencia de Bases , Diferenciación Celular , Ensayo de Unidades Formadoras de Colonias , Células Eritroides/citología , Eritropoyesis/genética , Eliminación de Gen , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hígado/embriología , Hígado/metabolismo , Ratones , Datos de Secuencia Molecular , Fenotipo , Regiones Promotoras Genéticas , Alineación de Secuencia , Activación Transcripcional
7.
Biochim Biophys Acta ; 1828(11): 2399-409, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23791703

RESUMEN

Several Cl(-) channels have been described in the native renal tubule, but their correspondence with ClC-K1 and ClC-K2 channels (orthologs of human ClC-Ka and ClC-Kb), which play a major role in transcellular Cl(-) absorption in the kidney, has yet to be established. This is partly because investigation of heterologous expression has involved rat or human ClC-K models, whereas characterization of the native renal tubule has been done in mice. Here, we investigate the electrophysiological properties of mouse ClC-K1 channels heterologously expressed in Xenopus laevis oocytes and in HEK293 cells with or without their accessory Barttin subunit. Current amplitudes and plasma membrane insertion of mouse ClC-K1 were enhanced by Barttin. External basic pH or elevated calcium stimulated currents followed the anion permeability sequence Cl(-)>Br(-)>NO3(-)>I(-). Single-channel recordings revealed a unit conductance of ~40pS. Channel activity in cell-attached patches increased with membrane depolarization (voltage for half-maximal activation: ~-65mV). Insertion of the V166E mutation, which introduces a glutamate in mouse ClC-K1, which is crucial for channel gating, reduced the unit conductance to ~20pS. This mutation shifted the depolarizing voltage for half-maximal channel activation to ~+25mV. The unit conductance and voltage dependence of wild-type and V166E ClC-K1 were not affected by Barttin. Owing to their strikingly similar properties, we propose that the ClC-K1/Barttin complex is the molecular substrate of a chloride channel previously detected in the mouse thick ascending limb (Paulais et al., J Membr. Biol, 1990, 113:253-260).


Asunto(s)
Canales de Cloruro/metabolismo , Animales , Células HEK293 , Humanos , Túbulos Renales/metabolismo , Técnicas de Placa-Clamp , Proteínas Recombinantes/metabolismo , Xenopus laevis
8.
Cell Death Dis ; 15(1): 20, 2024 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-38195526

RESUMEN

In recent years, primary familial brain calcification (PFBC), a rare neurological disease characterized by a wide spectrum of cognitive disorders, has been associated to mutations in the sodium (Na)-Phosphate (Pi) co-transporter SLC20A2. However, the functional roles of the Na-Pi co-transporters in the brain remain still largely elusive. Here we show that Slc20a1 (PiT-1) and Slc20a2 (PiT-2) are the most abundant Na-Pi co-transporters expressed in the brain and are involved in the control of hippocampal-dependent learning and memory. We reveal that Slc20a1 and Slc20a2 are differentially distributed in the hippocampus and associated with independent gene clusters, suggesting that they influence cognition by different mechanisms. Accordingly, using a combination of molecular, electrophysiological and behavioral analyses, we show that while PiT-2 favors hippocampal neuronal branching and survival, PiT-1 promotes synaptic plasticity. The latter relies on a likely Otoferlin-dependent regulation of synaptic vesicle trafficking, which impacts the GABAergic system. These results provide the first demonstration that Na-Pi co-transporters play key albeit distinct roles in the hippocampus pertaining to the control of neuronal plasticity and cognition. These findings could provide the foundation for the development of novel effective therapies for PFBC and cognitive disorders.


Asunto(s)
Cognición , Simportadores , Transporte Iónico , Plasticidad Neuronal/genética , Fosfatos
9.
Cell Mol Life Sci ; 68(2): 205-18, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20848155

RESUMEN

Although considerable advances in our understanding of the mechanisms of phosphate homeostasis and skeleton mineralization have recently been made, little is known about the initial events involving the detection of changes in the phosphate serum concentrations and the subsequent downstream regulation cascade. Recent data has strengthened a long-established hypothesis that a phosphate-sensing mechanism may be present in various organs. Such a phosphate sensor would detect changes in serum or local phosphate concentration and would inform the body, the local environment, or the individual cell. This suggests that phosphate in itself could represent a signal regulating multiple factors necessary for diverse biological processes such as bone or vascular calcification. This review summarizes findings supporting the possibility that phosphate represents a signaling molecule, particularly in bone and cartilage, but also in other tissues. The involvement of various signaling pathways (ERK1/2), transcription factors (Fra-1, Runx2) and phosphate transporters (PiT1, PiT2) is discussed.


Asunto(s)
Huesos/fisiología , Proteínas de Transporte de Fosfato , Fosfatos , Transducción de Señal , Animales , Huesos/química , Huesos/citología , Calcificación Fisiológica , Cartílago/metabolismo , Diferenciación Celular , Homeostasis/fisiología , Humanos , Mamíferos , Osteoblastos/citología , Osteoblastos/metabolismo , Proteínas de Transporte de Fosfato/metabolismo , Fosfatos/química , Fosfatos/fisiología , Transducción de Señal/fisiología
10.
Front Endocrinol (Lausanne) ; 13: 921073, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36465661

RESUMEN

The common cellular origin between bone marrow adipocytes (BMAds) and osteoblasts contributes to the intimate link between bone marrow adipose tissue (BMAT) and skeletal health. An imbalance between the differentiation ability of BMSCs towards one of the two lineages occurs in conditions like aging or osteoporosis, where bone mass is decreased. Recently, we showed that the sodium-phosphate co-transporter PiT2/SLC20A2 is an important determinant for bone mineralization, strength and quality. Since bone mass is reduced in homozygous mutant mice, we investigated in this study whether the BMAT was also affected in PiT2-/- mice by assessing the effect of the absence of PiT2 on BMAT volume between 3 and 16 weeks, as well as in an ovariectomy-induced bone loss model. Here we show that the absence of PiT2 in juveniles leads to an increase in the BMAT that does not originate from an increased adipogenic differentiation of bone marrow stromal cells. We show that although PiT2-/- mice have higher BMAT volume than control PiT2+/+ mice at 3 weeks of age, BMAT volume do not increase from 3 to 16 weeks of age, leading to a lower BMAT volume in 16-week-old PiT2-/- compared to PiT2+/+ mice. In contrast, the absence of PiT2 does not prevent the increase in BMAT volume in a model of ovariectomy-induced bone loss. Our data identify SLC20a2/PiT2 as a novel gene essential for the maintenance of the BMAd pool in adult mice, involving mechanisms of action that remain to be elucidated, but which appear to be independent of the balance between osteoblastic and adipogenic differentiation of BMSCs.


Asunto(s)
Enfermedades Óseas Metabólicas , Osteoporosis , Femenino , Ratones , Animales , Médula Ósea , Tejido Adiposo , Osteoporosis/genética , Densidad Ósea
11.
J Biol Chem ; 285(45): 34408-18, 2010 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-20817733

RESUMEN

PiT1/SLC20A1 is a sodium-dependent P(i) transporter expressed by most mammalian cells. Interestingly, PiT1 transcription has been shown to be up-regulated by the tumor necrosis factor α (TNF), and we have now investigated the possible involvement of PiT1 in TNF-induced apoptosis. We show that PiT1-depleted cells are more sensitive to the proapoptotic activity of TNF (i.e. when the antiapoptotic NFκB pathway is inactivated). These observations were made in the human HeLa cancer cell line either transiently or stably depleted in PiT1 by RNA interference and in immortalized mouse embryonic fibroblasts isolated from PiT1 knock-out embryos. Depletion of the closely related family member PiT2 had no effect on TNF-induced apoptosis, showing that this effect was specific to PiT1. The increased sensitivity of PiT1-depleted cells was evident regardless of the presence or absence of extracellular P(i), suggesting that a defect in P(i) uptake was not involved in the observed phenotype. Importantly, we show that the re-expression of a P(i) uptake mutant of PiT1 in PiT1(-/-) mouse embryonic fibroblasts delays apoptosis as efficiently as the WT protein, showing that this function of PiT1 is unrelated to its transport activity. Caspase-8 is more activated in PiT1-depleted cells, and our data reveal that the sustained activation of the MAPK JNK is up-regulated in response to TNF. JNK activity is actually involved in PiT1-depleted cell death because specific JNK inhibitors delay apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Fibroblastos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos , Animales , Apoptosis/fisiología , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Caspasa 8/genética , Caspasa 8/metabolismo , Línea Celular Transformada , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Fibroblastos/citología , Células HeLa , Humanos , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Noqueados , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética , Transcripción Genética/efectos de los fármacos
12.
N Engl J Med ; 359(11): 1128-35, 2008 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-18784102

RESUMEN

Impaired renal phosphate reabsorption, as measured by dividing the tubular maximal reabsorption of phosphate by the glomerular filtration rate (TmP/GFR), increases the risks of nephrolithiasis and bone demineralization. Data from animal models suggest that sodium-hydrogen exchanger regulatory factor 1 (NHERF1) controls renal phosphate transport. We sequenced the NHERF1 gene in 158 patients, 94 of whom had either nephrolithiasis or bone demineralization. We identified three distinct mutations in seven patients with a low TmP/GFR value. No patients with normal TmP/GFR values had mutations. The mutants expressed in cultured renal cells increased the generation of cyclic AMP (cAMP) by parathyroid hormone (PTH) and inhibited phosphate transport. These NHERF1 mutations suggest a previously unrecognized cause of renal phosphate loss in humans.


Asunto(s)
Desmineralización Ósea Patológica/genética , Cálculos Renales/genética , Nefrolitiasis/genética , Hormona Paratiroidea/metabolismo , Fosfatos/metabolismo , Fosfoproteínas/genética , Intercambiadores de Sodio-Hidrógeno/genética , Adulto , Animales , Transporte Biológico/genética , Desmineralización Ósea Patológica/metabolismo , Desmineralización Ósea Patológica/fisiopatología , Células Cultivadas , AMP Cíclico/biosíntesis , AMP Cíclico/orina , Análisis Mutacional de ADN , Femenino , Tasa de Filtración Glomerular/genética , Humanos , Hipercalciuria/genética , Riñón/citología , Riñón/metabolismo , Cálculos Renales/metabolismo , Cálculos Renales/fisiopatología , Masculino , Persona de Mediana Edad , Mutación , Mutación Missense , Nefrolitiasis/metabolismo , Zarigüeyas , Hormona Paratiroidea/sangre , Fosfatos/sangre
13.
J Biol Chem ; 284(45): 31363-74, 2009 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-19726692

RESUMEN

PiT1 is a Na(+)-phosphate (P(i)) cotransporter located at the plasma membrane that enables P(i) entry into the cell. Its broad tissue expression pattern has led to the idea that together with the closely related family member PiT2, PiT1 is the ubiquitous supplier of P(i) to the cell. Moreover, the role of P(i) in phosphorylation reactions, ATP production, DNA structure, and synthesis has led to the view that P(i) availability could be an important determinant of cell growth. However, these issues have not been clearly addressed to date, and the role of either P(i) or PiT proteins in cell proliferation is unknown. Using RNA interference in HeLa and HepG2 cells, we show that transient or stable PiT1 depletion markedly reduces cell proliferation, delays cell cycle, and impairs mitosis and cytokinesis. In vivo, PiT1 depletion greatly reduced tumor growth when engineered HeLa cells were injected into nude mice. We provide evidence that this effect on cell proliferation is specific to PiT1 and not shared by PiT2 and is not the consequence of impaired membrane Na(+)-P(i) transport. Moreover, we show that modulation of cell proliferation by PiT1 is independent from its transport function because the proliferation of PiT1-depleted cells can be rescued by non-transporting PiT1 mutants. PiT1 depletion leads to the phosphorylation of p38 mitogen-activated protein (MAP) kinase, whereas other MAP kinases and downstream targets of mammalian target of rapamycin (mTOR) remain unaffected. This study is the first to describe the effects of a P(i) transporter in cell proliferation, tumor growth, and cell signaling.


Asunto(s)
Proliferación Celular , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo , Transporte Biológico , Ciclo Celular , Células HeLa , Humanos , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética
14.
J Mol Endocrinol ; 65(3): R53-R63, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32755995

RESUMEN

The critical role of phosphate (Pi) in countless biological processes requires the ability to control its concentration both intracellularly and extracellularly. At the body level, this concentration is finely regulated by numerous hormones, primarily parathyroid hormone (PTH) and fibroblast growth factor 23 (FGF23). While this control of the body's Pi homeostasis is now well documented, knowledge of the mechanisms that allow the cell and the body to detect extracellular Pi variations is much less known. These systems are well described in bacteria, yeasts and plants, but as will be discussed in this review, knowledge obtained from these organisms is not entirely relevant to the requirements of Pi biology in mammals. In this review, we present the latest findings on extracellular Pi sensing in mammals, and describe the mammalian Pi sensors identified to date, such as SLC20A1 (PIT1)/SLC20A2 (PIT2) heterodimers and the calcium-sensing receptor (CaSR). While there are many questions remaining to be resolved, a clarification of the Pi sensing mechanisms in mammals is critical to understanding the deregulation of Pi balance in certain life-threatening disease states, such as end-stage renal disease and associated vascular calcifications, and to proposing relevant therapeutic approaches.


Asunto(s)
Espacio Extracelular/metabolismo , Mamíferos/metabolismo , Fosfatos/metabolismo , Animales , Huesos/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Homeostasis/fisiología , Humanos , Redes y Vías Metabólicas/fisiología , Receptores Sensibles al Calcio/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/fisiología , Vitamina D/metabolismo
15.
J Ren Nutr ; 19(1): 50-6, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19121771

RESUMEN

The discovery that two recently identified molecules, klotho and fibroblast growth factor 23 (FGF23), played an important role in calcium, phosphate, and vitamin D metabolism has transformed our traditional physiological view in which bone and mineral homeostasis was mainly regulated by parathyroid hormone, vitamin D, and calcitonin, according to mineral body needs. FGF23 is a 251-amino acid secreted protein produced by osteoblasts and osteocytes in bone following the stimulation by phosphate and vitamin D or the inhibition by dentin matrix protein 1. Originally isolated from tumoral cells of patients with tumor-induced osteomalacia and hypophosphatemia, FGF23 inhibits phosphate reabsorption in renal proximal tubular cells and 1alpha-hydroxylase activity, resulting in decreased synthesis of calcitriol. To exert these actions, FGF23 requires the conversion, by klotho, of the canonical FGF receptor 1 (IIIc) in a specific high affinity FGF23 receptor. On the other hand, klotho is a putative antiaging gene identified in 1997 when a particular mouse strain, created by random insertion mutagenesis, was found to be short-lived and displayed premature atherosclerosis, osteopenia, skin atrophy, pulmonary emphysema, hyperphosphatemia, hypercalcemia, and high serum calcitriol levels. The gene of klotho encodes a 1012-amino acid cell-surface protein with a short cytoplasmic tail and an extracellular domain that consists in tandem duplicated copies of a beta-glucuronidase-like sequence, which can be released into the circulation as soluble forms after being cleaved by metalloproteinases such as ADAM10 and ADAM17. By modulating FGF23 action, klotho regulates urinary phosphate excretion and calcitriol synthesis. By virtue of its beta-glucuronidase activity, klotho deglycosylates the calcium channel TRPV5 (transient receptor potential vallinoid-5) and regulates urinary calcium excretion. klotho also binds to Na(+),K(+)-ATPase in parathyroid cells and regulates calcium-stimulated PTH secretion. Finally, klotho extends life span via several mechanisms, including the reduction of calcitriol synthesis, serum calcium, and phosphorus levels; the induction of insulin resistance; and by increasing the resistance to oxidative stress.


Asunto(s)
Calcio/metabolismo , Factores de Crecimiento de Fibroblastos/fisiología , Glucuronidasa/fisiología , Fosfatos/metabolismo , Vitamina D/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Glucuronidasa/genética , Homeostasis , Humanos , Fallo Renal Crónico/metabolismo , Fallo Renal Crónico/mortalidad , Fallo Renal Crónico/terapia , Proteínas Klotho , Diálisis Renal/métodos , Transducción de Señal/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo
16.
J Bone Miner Res ; 34(2): 387-398, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30347511

RESUMEN

During skeletal mineralization, the sodium-phosphate co-transporter PiT1Slc20a1 is assumed to meet the phosphate requirements of bone-forming cells, although evidence is missing. Here, we used a conditional gene deletion approach to determine the role of PiT1 in growth plate chondrocytes. We show that PiT1 ablation shortly after birth generates a rapid and massive cell death in the center of the growth plate, together with an uncompensated endoplasmic reticulum (ER) stress, characterized by morphological changes and increased Chop, Atf4, and Bip expression. PiT1 expression in chondrocytes was not found at the cell membrane but co-localized with the ER marker ERp46, and was upregulated by the unfolded protein response cascade. In addition, we identified the protein disulfide isomerase (Pdi) ER chaperone as a PiT1 binding partner and showed that PiT1 ablation impaired Pdi reductase activity. The ER stress induced by PiT1 deficiency in chondrocytes was associated with intracellular retention of aggrecan and vascular endothelial growth factor A (Vegf-A), which was rescued by overexpressing a phosphate transport-deficient mutant of PiT1. Our data thus reveal a novel, Pi-transport independent function of PiT1, as a critical modulator of ER homeostasis and chondrocyte survival during endochondral ossification. © 2018 American Society for Bone and Mineral Research.


Asunto(s)
Condrocitos/metabolismo , Retículo Endoplásmico , Placa de Crecimiento/metabolismo , Homeostasis , Osteogénesis , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo , Animales , Condrocitos/citología , Regulación de la Expresión Génica , Placa de Crecimiento/citología , Ratones , Ratones Transgénicos , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética , Respuesta de Proteína Desplegada
17.
Nutrients ; 11(12)2019 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-31888255

RESUMEN

Polyphenols are widely acknowledged for their health benefits, especially for the prevention of inflammatory and age-related diseases. We previously demonstrated that hydroxytyrosol (HT) and procyanidins (PCy), alone or in combination, drive preventive anti-osteoathritic effects in vivo. However, the lack of sufficient clinical evidences on the relationship between dietary phytochemicals and osteoarthritis remains. In this light, we investigated in humans the potential osteoarticular benefit of a grapeseed and olive extract (OPCO) characterized for its hydroxytyrosol (HT) and procyanidins (PCy) content. We first validated, in vitro, the anti-inflammatory and chondroprotective properties of the extract on primary cultured human articular chondrocytes stimulated by interleukin-1 beta (IL-1 ß). The sparing effect involved a molecular mechanism dependent on the nuclear transcription factor-kappa B (NF-κB) pathway. To confirm the clinical relevance of such a nutritional strategy, we designed an innovative clinical approach taking into account the metabolites that are formed during the digestion process and that appear in circulation after the ingestion of the OPCO extract. Blood samples from volunteers were collected following ingestion, absorption, and metabolization of the extract and then were processed and applied on human primary chondrocyte cultures. This original ex vivo methodology confirmed at a clinical level the chondroprotective properties previously observed in vitro and in vivo.


Asunto(s)
Absorción Fisicoquímica/efectos de los fármacos , Antiinflamatorios/farmacología , Condrocitos/efectos de los fármacos , Extracto de Semillas de Uva/farmacología , Extractos Vegetales/farmacología , Polifenoles/farmacología , Adulto , Células Cultivadas , Voluntarios Sanos , Humanos , Interleucina-1beta/sangre , Masculino , FN-kappa B/sangre , Alcohol Feniletílico/análogos & derivados , Alcohol Feniletílico/farmacología , Proantocianidinas/farmacología , Adulto Joven
18.
J Bone Miner Res ; 34(6): 1101-1114, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30721528

RESUMEN

Osteoporosis is characterized by low bone mineral density (BMD) and fragility fracture and affects over 200 million people worldwide. Bone quality describes the material properties that contribute to strength independently of BMD, and its quantitative analysis is a major priority in osteoporosis research. Tissue mineralization is a fundamental process requiring calcium and phosphate transporters. Here we identify impaired bone quality and strength in Slc20a2-/- mice lacking the phosphate transporter SLC20A2. Juveniles had abnormal endochondral and intramembranous ossification, decreased mineral accrual, and short stature. Adults exhibited only small reductions in bone mass and mineralization but a profound impairment of bone strength. Bone quality was severely impaired in Slc20a2-/- mice: yield load (-2.3 SD), maximum load (-1.7 SD), and stiffness (-2.7 SD) were all below values predicted from their bone mineral content as determined in a cohort of 320 wild-type controls. These studies identify Slc20a2 as a physiological regulator of tissue mineralization and highlight its critical role in the determination of bone quality and strength. © 2019 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.


Asunto(s)
Huesos/fisiología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética , Animales , Animales Recién Nacidos , Desarrollo Óseo , Resorción Ósea/fisiopatología , Huesos/diagnóstico por imagen , Calcificación Fisiológica , Calcinosis/diagnóstico por imagen , Calcinosis/genética , Células Cultivadas , Condrocitos/metabolismo , Humanos , Incisivo/ultraestructura , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/metabolismo , Fenotipo , Cráneo/diagnóstico por imagen , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/deficiencia , Diente/crecimiento & desarrollo , Microtomografía por Rayos X
19.
Mol Metab ; 11: 197-204, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29551636

RESUMEN

OBJECTIVE: The canonical role of the bone-derived fibroblast growth factor 23 (Fgf23) is to regulate the serum inorganic phosphate (Pi) level. As part of a feedback loop, serum Pi levels control Fgf23 secretion through undefined mechanisms. We recently showed in vitro that the two high-affinity Na+-Pi co-transporters PiT1/Slc20a1 and PiT2/Slc20a2 were required for mediating Pi-dependent signaling. Here, we addressed the contribution of PiT1 and PiT2 to the regulation of Fgf23 secretion. METHODS: To this aim, we used PiT2 KO and DMP1Cre; PiT1lox/lox fed Pi-modified diets, as well as ex vivo isolated long bone shafts. Fgf23 secretion and expression of Pi homeostasis-related genes were assessed. RESULTS: In vivo, PiT2 KO mice responded inappropriately to low-Pi diets, displaying abnormally normal serum levels of intact Fgf23. Despite the high iFgf23 level, serum Pi levels remained unaffected, an effect that may relate to lower αKlotho expression in the kidney. Moreover, consistent with a role of PiT2 as a possible endocrine Pi sensor, the iFGF23/cFGF23 ratios were suppressed in PiT2 KO mice, irrespective of the Pi loads. While deletion of PiT1 in osteocytes using the DMP1-Cre mice was inefficient, adenovirus-mediated deletion of PiT1 in isolated long bone shafts suggested that PiT1 does not contribute to Pi-dependent regulation of Fgf23 secretion. In contrast, using isolated bone shafts from PiT2 KO mice, we showed that PiT2 was necessary for the appropriate Pi-dependent secretion of Fgf23, independently from possible endocrine regulatory loops. CONCLUSIONS: Our data provide initial mechanistic insights underlying the Pi-dependent regulation of Fgf23 secretion in identifying PiT2 as a potential player in this process, at least in high Pi conditions. Targeting PiT2, therefore, could improve excess FGF23 in hyperphosphatemic conditions such as chronic kidney disease.


Asunto(s)
Factores de Crecimiento de Fibroblastos/sangre , Fosfatos/sangre , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética , Animales , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Riñón/metabolismo , Ratones , Ratones Endogámicos C57BL , Osteocitos/metabolismo , Transducción de Señal , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo
20.
J Ren Nutr ; 16(2): 87-99, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16567265

RESUMEN

Secondary hyperparathyroidism (SHPT) is a common and serious complication of chronic kidney disease (CKD). It affects more than 300,000 end-stage renal disease patients treated by dialysis and probably more than 3 million patients with CKD worldwide. For a long time, traditional therapies for SHPT had consisted of correcting the hypocalcemia using calcium salts and vitamin D derivatives, preventing the hyperphosphatemia by calcium- or aluminum-containing intestinal phosphate binders, and recently by using no metal-containing intestinal phosphate binders; however, these therapies are limited by the occurrence of hypercalcemia, hyperphosphatemia, and the lack of specificity and long-term efficacy. Moreover, surgical parathyroidectomy (PTX), which remains the gold standard therapy, is not exempt from risk. PTX exposes patients to anesthesia risks, presurgical and postsurgical complications, and in many cases a permanent state of hypoparathyroidism. Thus, the medical treatment of SHPT became an ideal target for the development of new therapies and strategies. The purpose of this article is to provide an overview of these new therapies, including vitamin D analogs, intestinal phosphate binders, calcimimetics, parathyroidectomies, tyrosine kinase inhibitors, azydothymidine, anticalcineurins, N-terminal truncated parathyroid hormone fragments, bisphosphonates, calcitonin, osteoprotegerin, and others. The use of these new therapies alone or in combination may help to optimize the future treatment of SHPT in CKD patients.


Asunto(s)
Hiperparatiroidismo Secundario/terapia , Uremia/complicaciones , Acidosis/complicaciones , Acidosis/terapia , Anticuerpos/farmacología , División Celular/efectos de los fármacos , Humanos , Hiperparatiroidismo Secundario/tratamiento farmacológico , Hiperparatiroidismo Secundario/etiología , Hipocalcemia/complicaciones , Hipocalcemia/terapia , Glándulas Paratiroides/efectos de los fármacos , Glándulas Paratiroides/patología , Glándulas Paratiroides/fisiopatología , Hormona Paratiroidea/antagonistas & inhibidores , Hormona Paratiroidea/inmunología , Hormona Paratiroidea/metabolismo , Paratiroidectomía , Fosfatos/sangre , Receptor de Hormona Paratiroídea Tipo 1/antagonistas & inhibidores , Deficiencia de Vitamina D/complicaciones , Deficiencia de Vitamina D/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA