Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Basic Res Cardiol ; 115(5): 51, 2020 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-32699940

RESUMEN

The concentration of fibroblast growth factor 23 (FGF23) rises progressively in renal failure (RF). High FGF23 concentrations have been consistently associated with adverse cardiovascular outcomes or death, in chronic kidney disease (CKD), heart failure or liver cirrhosis. We identified the mechanisms whereby high concentrations of FGF23 can increase the risk of death of cardiovascular origin. We studied the effects of FGF23 and Klotho in adult rat ventricular cardiomyocytes (ARVMs) and on the heart of mice with CKD. We show that FGF23 increases the frequency of spontaneous calcium waves (SCWs), a marker of cardiomyocyte arrhythmogenicity, in ARVMs. FGF23 increased sarcoplasmic reticulum Ca2+ leakage, basal phosphorylation of Ca2+-cycling proteins including phospholamban and ryanodine receptor type 2. These effects are secondary to a decrease in phosphodiesterase 4B (PDE4B) in ARVMs and in heart of mice with RF. Soluble Klotho, a circulating form of the FGF23 receptor, prevents FGF23 effects on ARVMs by increasing PDE3A and PDE3B expression. Our results suggest that the combination of high FGF23 and low sKlotho concentrations decreases PDE activity in ARVMs, which favors the occurrence of ventricular arrhythmias and may participate in the high death rate observed in patients with CKD.


Asunto(s)
Arritmias Cardíacas/etiología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Glucuronidasa/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Arritmias Cardíacas/metabolismo , Señalización del Calcio , Cardiomegalia/etiología , AMP Cíclico/metabolismo , Acoplamiento Excitación-Contracción , Factor-23 de Crecimiento de Fibroblastos , Proteínas Klotho , Masculino , Ratones , Nefrectomía , Cultivo Primario de Células , Ratas Wistar
2.
Blood ; 126(15): 1802-12, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26320102

RESUMEN

CD9, a member of the tetraspanin family, has been implicated in hematopoietic and leukemic stem cell homing. We investigated the role of CD9 in the dissemination of B acute lymphoblastic leukemia (B-ALL) cells, by stably downregulating CD9 in REH and NALM6 cells. CD9 expression was associated with higher levels of REH cell adhesion to fibronectin and C-X-C motif chemokine receptor 4 (CXCR4)-mediated migration. Death occurred later in NOD/SCID mice receiving REH cells depleted of CD9 for transplantation than in mice receiving control cells. After C-X-C motif chemokine ligand 12 (CXCL12) stimulation, CD9 promoted the formation of long cytoplasmic actin-rich protrusions. We demonstrated that CD9 enhanced RAC1 activation, in both REH cells and blasts from patients. Conversely, the overexpression of a competing CD9 C-terminal tail peptide in REH cytoplasm decreased RAC1 activation and cytoplasmic extension formation in response to CXCL12. Finally, the inhibition of RAC1 activation decreased migration in vitro, and the depletion of RAC1 protein from transplanted REH cells increased mouse survival. Furthermore, a testis-conditioned medium induced the migration of REH and NALM6 cells, and this migration was impeded by an anti-CD9 antibody. The level of CD9 expression also influenced the homing of these cells in mouse testes. These findings demonstrate, for the first time, that CD9 plays a key role in the CXCR4-mediated migration and engraftment of B-ALL cells in the bone marrow or testis, through RAC1 activation.


Asunto(s)
Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Neuropéptidos/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Receptores CXCR4/metabolismo , Tetraspanina 29/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Apoptosis , Western Blotting , Médula Ósea/metabolismo , Médula Ósea/patología , Adhesión Celular , Proliferación Celular , Quimiocina CXCL12/metabolismo , Humanos , Inmunoprecipitación , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Testículo/metabolismo , Testículo/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
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
4.
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
5.
Am J Case Rep ; 24: e941096, 2023 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-37798877

RESUMEN

BACKGROUND Profound transaminitis (>1000 international units per liter [IU/L]) is typically associated with ischemic and viral or toxic hepatitis. Pancreaticobiliary causes are less likely to be included in the workup, especially in patients who have undergone cholecystectomy. We present a case of recurrent choledocholithiasis in a 52-year-old woman 7 years after cholecystectomy, presenting with severe transaminitis, illustrating the diagnostic challenges of this presentation. CASE REPORT A 52-year-old woman presented to the Emergency Department (ED) with acute upper abdominal pain. Computed tomography (CT) of the abdomen without contrast showed no abnormalities and mild common bile duct (CBD) dilation was noted on ultrasound (US) abdomen. Laboratory studies were significant for elevated transaminases greater than 1000 units/L and alkaline phosphatase (ALP) greater than 200 units/L. She was diagnosed with acute hepatitis of unknown etiology without undergoing further investigation of the biliary tract and was discharged after improvement with supportive therapy. She returned 4 months later with similar symptoms and laboratory findings, but with more CBD dilation and intrahepatic biliary dilation on CT and US. Endoscopic retrograde cholangiopancreatography (ERCP) was performed, and multiple stones and sludge were removed from the CBD. CONCLUSIONS This report has shown that pancreaticobiliary causes should be included in the workup of severe transaminitis, even in patients with a remote history of cholecystectomy. Failure to do so may subject patients to extensive, unnecessary workup and delay correct management.


Asunto(s)
Colecistectomía Laparoscópica , Coledocolitiasis , Femenino , Humanos , Persona de Mediana Edad , Coledocolitiasis/diagnóstico por imagen , Coledocolitiasis/cirugía , Colecistectomía/efectos adversos , Colangiopancreatografia Retrógrada Endoscópica/efectos adversos , Dolor Abdominal/etiología , Ultrasonografía , Colecistectomía Laparoscópica/efectos adversos
6.
Sci Rep ; 13(1): 85, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36596813

RESUMEN

Biallelic pathogenic variants in the SLC34A3 gene, encoding for the NPT2c cotransporter, cause Hereditary Hypophosphatemic Rickets with Hypercalciuria (HHRH). However, the associated phenotype is highly variable. In addition, mice deleted for Slc34a3 exhibit a different phenotype compared to humans, without urinary phosphate leakage. The mechanisms by which SLC34A3 variants disrupt phosphate/calcium metabolism are un-completely understood. In this study we explored these mechanisms in vitro using SLC34A3 variants identified in patients with urinary phosphate leakage. We analyzed the consequences of these variants on NPT2c function and the link with the phenotype of the patients. We studied 20 patients with recurrent nephrolithiasis and low serum phosphate concentration harboring variants in the SLC34A3 gene. Half of the patients carried homozygous or composite heterozygous variants. Three patients had in addition variants in SLC34A1 and SLC9A3R1 genes. All these patients benefited from a precise analysis of their phenotype. We generated 13 of these mutants by site-directed mutagenesis. Then we carried out transient transfections of these mutants in HEK cells and measured their phosphate uptake capacity under different conditions. Among the 20 patients included, 3 had not only mutations in NPT2c but also in NPT2a or NHERF1 genes. Phosphate uptake was decreased in 8 NPT2c mutants studied and normal for 5. Four variants were initially categorized as variants of uncertain significance. Expression of the corresponding mutants showed that one did not modify phosphate transport, two reduced it moderately and one abolished it. Co-transfection of the NPT2c mutants with the wild-type plasmid of NPT2c or NPT2a did not reveal dominant negative effect of the mutants on NPT2c-mediated phosphate transport. A detailed analysis of patient phenotypes did not find a link between the severity of the disorder and the level of phosphate transport impairment. NPT2c mutations classified as ACMG3 identified in patients with renal phosphate leak should be characterized by in vitro study to check if they alter NPT2c-mediated phosphate transport since phosphate uptake capacity may not be affected. In addition, research for mutations in NHERF1 and NPT2a genes should always be associated to NPT2c sequencing.


Asunto(s)
Raquitismo Hipofosfatémico Familiar , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIc , Animales , Humanos , Ratones , Raquitismo Hipofosfatémico Familiar/genética , Raquitismo Hipofosfatémico Familiar/patología , Riñón/metabolismo , Mutación , Fenotipo , Fosfatos/metabolismo
7.
Aging (Albany NY) ; 14(9): 3728-3756, 2022 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-35507806

RESUMEN

Naked mole-rats (NMR) are subterranean rodents characterized by an unusual longevity coupled with an unexplained resistance to aging. In the present study, we performed extensive in situ analysis and single-cell RNA-sequencing comparing young and older animals. At variance with other species, NMR exhibited a striking stability of skin compartments and cell types, which remained stable over time without aging-associated changes. Remarkably, the number of stem cells was constant throughout aging. We found three classical cellular states defining a unique keratinocyte differentiation trajectory that were not altered after pseudo-temporal reconstruction. Epidermal gene expression did not change with aging either. Langerhans cell clusters were conserved, and only a higher basal stem cell expression of Igfbp3 was found in aged animals. In accordance, NMR skin healing closure was similar in young and older animals. Altogether, these results indicate that NMR skin is characterized by peculiar genetic and cellular features, different from those previously demonstrated for mice and humans. The remarkable stability of the aging NMR skin transcriptome likely reflects unaltered homeostasis and resilience.


Asunto(s)
Ratas Topo , Transcriptoma , Envejecimiento/genética , Animales , Longevidad/genética , Ratones , Ratas Topo/genética , Células Madre
8.
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
9.
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
10.
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
11.
Nat Cell Biol ; 22(9): 1091-1102, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32868900

RESUMEN

Organs and cells must adapt to shear stress induced by biological fluids, but how fluid flow contributes to the execution of specific cell programs is poorly understood. Here we show that shear stress favours mitochondrial biogenesis and metabolic reprogramming to ensure energy production and cellular adaptation in kidney epithelial cells. Shear stress stimulates lipophagy, contributing to the production of fatty acids that provide mitochondrial substrates to generate ATP through ß-oxidation. This flow-induced process is dependent on the primary cilia located on the apical side of epithelial cells. The interplay between fluid flow and lipid metabolism was confirmed in vivo using a unilateral ureteral obstruction mouse model. Finally, primary cilium-dependent lipophagy and mitochondrial biogenesis are required to support energy-consuming cellular processes such as glucose reabsorption, gluconeogenesis and cytoskeletal remodelling. Our findings demonstrate how primary cilia and autophagy are involved in the translation of mechanical forces into metabolic adaptation.


Asunto(s)
Autofagia/fisiología , Cilios/metabolismo , Cilios/fisiología , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Riñón/metabolismo , Riñón/fisiología , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Gluconeogénesis/fisiología , Glucosa/metabolismo , Metabolismo de los Lípidos/fisiología , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Estrés Mecánico
12.
Biochim Biophys Acta ; 1781(11-12): 685-93, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18755291

RESUMEN

Long chain polyunsaturated fatty acids (PUFAs) play an important role in cardioprotection. These effects have been largely attributed to membrane docosahexaenoic acid. Conversely, saturated fatty acids trigger apoptosis in cardiomyocytes, with modifications of mitochondrial properties including cardiolipin loss, cytochrome c release and caspase-3 activation. The purpose of this study was to investigate the chronic effect of eicosapentaenoic acid (EPA) on mitochondrial apoptosis induced by palmitate treatment and the associated signalling pathways. Confluent cultures of rat neonatal cardiomyocytes were treated for 2 days in media enriched with either EPA or arachidonic acid (AA) and then exposed to palmitate (0.5 mM) to induce apoptosis, in the absence of PUFA supplements. The EPA treatment resulted in significant membrane enrichment in n-3 PUFAs, especially in docosapentaenoic acid (DPA), and a large decrease in AA. Both AA and EPA treatments prevented caspase-3 activation, translocation of Bax to the mitochondria and release of cytochrome c induced by palmitate treatment. Furthermore, EPA, but not AA prevented the loss of mitochondrial cardiolipin due to apoptosis. These results suggest that EPA supplementation is able to protect cardiomyocytes against palmitate-induced apoptosis via an implication of different mitochondrial elements, possibly through its elongation to DPA, which is very efficient in cardiomyocytes.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácido Eicosapentaenoico/farmacología , Miocitos Cardíacos/efectos de los fármacos , Palmitatos/toxicidad , Animales , Animales Recién Nacidos , Apoptosis/fisiología , Ácido Araquidónico/farmacología , Western Blotting , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Células Cultivadas , Citocromos c/metabolismo , Activación Enzimática/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Wistar , Proteína X Asociada a bcl-2/metabolismo
13.
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
14.
Oxid Med Cell Longev ; 2019: 4502819, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30881592

RESUMEN

The oxidative stress theory of aging, linking reactive oxygen species (ROS) to aging, has been accepted for more than 60 years, and numerous studies have associated ROS with various age-related diseases. A more precise version of the theory specifies that mitochondrial oxidative stress is a direct cause of aging. The naked mole rat, a unique animal with exceptional longevity (32 years in captivity), appears to be an ideal model to study successful aging and the role of ROS in this process. Several studies in the naked mole rat have shown that these animals exhibit a remarkable resistance to oxidative stress. At low concentrations, ROS serve as second messengers, and these important intracellular signalling functions are crucial for the regulation of cellular processes. In this review, we examine the literature on ROS and their functions as signal transducers. We focus specifically on the longest-lived rodent, the naked mole rat, which is a perfect example of the paradox of living an exceptionally long life with slow aging despite high levels of oxidative damage from a young age.


Asunto(s)
Estrés Oxidativo/fisiología , Animales , Ratas Topo , Ratas
15.
Aging (Albany NY) ; 11(14): 4783-4800, 2019 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-31346149

RESUMEN

The naked mole-rat (Heterocephalus glaber) is characterized by a more than tenfold higher life expectancy compared to another rodent species of the same size, namely, the laboratory mouse (Mus musculus). We used mass spectrometric metabolomics to analyze circulating plasma metabolites in both species at different ages. Interspecies differences were much more pronounced than age-associated alterations in the metabolome. Such interspecies divergences affected multiple metabolic pathways involving amino, bile and fatty acids as well as monosaccharides and nucleotides. The most intriguing metabolites were those that had previously been linked to pro-health and antiaging effects in mice and that were significantly increased in the long-lived rodent compared to its short-lived counterpart. This pattern applies to α-tocopherol (also known as vitamin E) and polyamines (in particular cadaverine, N8-acetylspermidine and N1,N8-diacetylspermidine), all of which were more abundant in naked mole-rats than in mice. Moreover, the age-associated decline in spermidine and N1-acetylspermidine levels observed in mice did not occur, or is even reversed (in the case of N1-acetylspermidine) in naked mole-rats. In short, the present metabolomics analysis provides a series of testable hypotheses to explain the exceptional longevity of naked mole-rats.


Asunto(s)
Envejecimiento/metabolismo , Longevidad/fisiología , Ratas Topo/metabolismo , Animales , Metabolómica , Ratones , Ratas , Especificidad de la Especie
16.
Sci Rep ; 9(1): 1808, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30755642

RESUMEN

PiT1/SLC20A1 is an inorganic phosphate transporter with additional functions including the regulation of TNFα-induced apoptosis, erythropoiesis, cell proliferation and insulin signaling. Recent data suggest a relationship between PiT1 and NF-κB-dependent inflammation: (i) Pit1 mRNA is up-regulated in the context of NF-κB pathway activation; (ii) NF-κB target gene transcription is decreased in PiT1-deficient conditions. This led us to investigate the role of PiT1 in lipopolysaccharide (LPS)-induced inflammation. MCP-1 and IL-6 concentrations were impaired in PiT1-deficient bone marrow derived macrophages (BMDMs) upon LPS stimulation. Lower MCP-1 and IL-6 serum levels were observed in Mx1-Cre; Pit1lox/lox mice dosed intraperitoneally with LPS. Lower PiT1 expression correlated with decreased in vitro wound healing and lower reactive oxygen species levels. Reduced IκB degradation and lower p65 nuclear translocation were observed in PiT1-deficient cells stimulated with LPS. Conversely, PiT1 expression was induced in vitro upon LPS stimulation. Addition of an NF-κB inhibitor abolished LPS-induced PiT1 expression. Furthermore, we showed that p65 expression activated Pit1 promoter activity. Finally, ChIP assays demonstrated that p65 directly binds to the mPit1 promoter in response to LPS. These data demonstrate a completely novel function of PiT1 in the response to LPS and provide mechanistic insights into the regulation of PiT1 expression by NF-κB.


Asunto(s)
Inflamación/inducido químicamente , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo , Factor de Transcripción Pit-1/metabolismo , Animales , Apoptosis/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , NADPH Oxidasa 2/metabolismo , FN-kappa B/metabolismo , Peritonitis/inducido químicamente , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Tioglicolatos/toxicidad , Factor de Transcripción Pit-1/genética , Factor de Necrosis Tumoral alfa/metabolismo , Cicatrización de Heridas/efectos de los fármacos
17.
J Clin Med ; 8(4)2019 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-31013726

RESUMEN

Hyperphosphatemia is commonly present in end-stage renal disease. Klotho (KL) is implicated in phosphate homeostasis since it acts as obligate co-receptor for the fibroblast growth factor 23 (FGF23), a major phosphaturic hormone. We hypothesized that genetic variation in the KL gene might be associated with alterations in phosphate homeostasis resulting in hyperphosphatemia. We performed sequencing for determining KL gene variants in a group of resistant hyperphosphatemic dialysis patients. In a 67-year-old female, blood DNA sequencing revealed a heterozygous deletion of a T at position 1041 (c.1041delT) in exon 2. This variation caused a frameshift with substitution of isoleucine for phenylalanine and introduction of a premature termination codon (p.Ile348Phefs*28). cDNA sequencing showed absence of deletion-carrier transcripts in peripheral blood mononuclear cells suggesting degradation of these through a nonsense-mediated RNA decay pathway. Experiments in vitro showed that p.Ile348Phefs*28 variant impaired FGF23 signaling pathway, indicating a functional inactivation of the gene. In the patient, serum levels of KL were 2.9-fold lower than the mean level of a group of matched dialysis subjects, suggesting a compromise in the circulating protein concentration due to haploinsufficiency. These findings provide a new loss-of-function variant in the human KL gene, suggesting that genetic determinants might be associated to clinical resistant hyperphosphatemia.

19.
Nat Chem ; 9(10): 1025-1033, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28937680

RESUMEN

Cancer stem cells (CSCs) represent a subset of cells within tumours that exhibit self-renewal properties and the capacity to seed tumours. CSCs are typically refractory to conventional treatments and have been associated to metastasis and relapse. Salinomycin operates as a selective agent against CSCs through mechanisms that remain elusive. Here, we provide evidence that a synthetic derivative of salinomycin, which we named ironomycin (AM5), exhibits a more potent and selective activity against breast CSCs in vitro and in vivo, by accumulating and sequestering iron in lysosomes. In response to the ensuing cytoplasmic depletion of iron, cells triggered the degradation of ferritin in lysosomes, leading to further iron loading in this organelle. Iron-mediated production of reactive oxygen species promoted lysosomal membrane permeabilization, activating a cell death pathway consistent with ferroptosis. These findings reveal the prevalence of iron homeostasis in breast CSCs, pointing towards iron and iron-mediated processes as potential targets against these cells.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Hierro/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Piranos/farmacología , Antineoplásicos/química , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Homeostasis/efectos de los fármacos , Humanos , Lisosomas/química , Conformación Molecular , Células Madre Neoplásicas/metabolismo , Piranos/química , Especies Reactivas de Oxígeno/análisis , Especies Reactivas de Oxígeno/metabolismo
20.
Cell Rep ; 16(10): 2736-2748, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27568561

RESUMEN

The liver plays a central role in whole-body lipid and glucose homeostasis. Increasing dietary fat intake results in increased hepatic fat deposition, which is associated with a risk for development of insulin resistance and type 2 diabetes. In this study, we demonstrate a role for the phosphate inorganic transporter 1 (PiT1/SLC20A1) in regulating metabolism. Specific knockout of Pit1 in hepatocytes significantly improved glucose tolerance and insulin sensitivity, enhanced insulin signaling, and decreased hepatic lipogenesis. We identified USP7 as a PiT1 binding partner and demonstrated that Pit1 deletion inhibited USP7/IRS1 dissociation upon insulin stimulation. This prevented IRS1 ubiquitination and its subsequent proteasomal degradation. As a consequence, delayed insulin negative feedback loop and sustained insulin signaling were observed. Moreover, PiT1-deficient mice were protected against high-fat-diet-induced obesity and diabetes. Our findings indicate that PiT1 has potential as a therapeutic target in the context of metabolic syndrome, obesity, and diabetes.


Asunto(s)
Glucosa/metabolismo , Hepatocitos/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Insulina/metabolismo , Transducción de Señal , Factor de Transcripción Pit-1/metabolismo , Peptidasa Específica de Ubiquitina 7/metabolismo , Tejido Adiposo/patología , Envejecimiento/patología , Animales , Dieta Alta en Grasa , Hígado Graso/complicaciones , Hígado Graso/patología , Fibroblastos/metabolismo , Gluconeogénesis , Prueba de Tolerancia a la Glucosa , Inflamación/complicaciones , Inflamación/patología , Resistencia a la Insulina , Ratones Noqueados , Obesidad/patología , Especificidad de Órganos , Fenotipo , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Insulina/metabolismo , Ubiquitinación , Aumento de Peso
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA