Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 114(16): E3344-E3353, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28373577

RESUMEN

Renal Ca2+ reabsorption is essential for maintaining systemic Ca2+ homeostasis and is tightly regulated through the parathyroid hormone (PTH)/PTHrP receptor (PTH1R) signaling pathway. We investigated the role of PTH1R in the kidney by generating a mouse model with targeted deletion of PTH1R in the thick ascending limb of Henle (TAL) and in distal convoluted tubules (DCTs): Ksp-cre;Pth1rfl/fl Mutant mice exhibited hypercalciuria and had lower serum calcium and markedly increased serum PTH levels. Unexpectedly, proteins involved in transcellular Ca2+ reabsorption in DCTs were not decreased. However, claudin14 (Cldn14), an inhibitory factor of the paracellular Ca2+ transport in the TAL, was significantly increased. Analyses by flow cytometry as well as the use of Cldn14-lacZ knock-in reporter mice confirmed increased Cldn14 expression and promoter activity in the TAL of Ksp-cre;Pth1rfl/fl mice. Moreover, PTH treatment of HEK293 cells stably transfected with CLDN14-GFP, together with PTH1R, induced cytosolic translocation of CLDN14 from the tight junction. Furthermore, mice with high serum PTH levels, regardless of high or low serum calcium, demonstrated that PTH/PTH1R signaling exerts a suppressive effect on Cldn14. We therefore conclude that PTH1R signaling directly and indirectly regulates the paracellular Ca2+ transport pathway by modulating Cldn14 expression in the TAL. Finally, systemic deletion of Cldn14 completely rescued the hypercalciuric and lower serum calcium phenotype in Ksp-cre;Pth1rfl/fl mice, emphasizing the importance of PTH in inhibiting Cldn14. Consequently, suppressing CLDN14 could provide a potential treatment to correct urinary Ca2+ loss, particularly in patients with hypoparathyroidism.


Asunto(s)
Calcio/metabolismo , Claudinas/fisiología , Extremidades/fisiología , Regulación de la Expresión Génica , Hormona Paratiroidea/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Uniones Estrechas/fisiología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Transducción de Señal
2.
Am J Physiol Renal Physiol ; 315(5): F1261-F1270, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29993278

RESUMEN

Phosphate homeostasis is primarily maintained in the renal proximal tubules, where the expression of sodium/phosphate cotransporters (Npt2a and Npt2c) is modified by the endocrine actions of both fibroblast growth factor 23 (FGF23) and parathyroid hormone (PTH). However, the specific contribution of each regulatory pathway in the proximal tubules has not been fully elucidated in vivo. We have previously demonstrated that proximal tubule-specific deletion of the FGF23 coreceptor Klotho results in mild hyperphosphatemia with little to no change in serum levels of FGF23, 1,25(OH)2D3, and PTH. In the present study, we characterized mice in which the PTH receptor PTH1R was specifically deleted from the proximal tubules, either alone or in combination with Klotho ( PT-PTH1R-/- and PT-PTH1R/KL-/-, respectively). PT-PTH1R-/- mice showed significant increases in serum FGF23 and PTH levels, whereas serum phosphate levels were maintained in the normal range, and Npt2a and Npt2c expression in brush border membrane (BBM) did not change compared with control mice. In contrast, PT-PTH1R/KL-/- mice displayed hyperphosphatemia and an increased abundance of Npt2a and Npt2c in the renal BBM, along with increased circulating FGF23 levels. While serum calcium was normal, 1,25(OH)2D3 levels were significantly decreased, leading to extremely high levels of PTH. Collectively, mice with a deletion of PTH1R alone in proximal tubules results in only minor changes in phosphate regulation, whereas deletion of both PTH1R and Klotho leads to a severe disturbance, including hyperphosphatemia with increased sodium/phosphate cotransporter expression in BBM. These results suggest an important interplay between the PTH/PTH1R and FGF23/Klotho pathways to affect renal phosphate handling in the proximal tubules.


Asunto(s)
Factores de Crecimiento de Fibroblastos/sangre , Glucuronidasa/metabolismo , Hiperfosfatemia/sangre , Túbulos Renales Proximales/metabolismo , Hormona Paratiroidea/sangre , Fosfatos/sangre , Reabsorción Renal , Animales , Calcitriol/sangre , Calcio/sangre , Células Cultivadas , Factor-23 de Crecimiento de Fibroblastos , Predisposición Genética a la Enfermedad , Glucuronidasa/deficiencia , Glucuronidasa/genética , Hiperfosfatemia/genética , Hiperfosfatemia/fisiopatología , Túbulos Renales Proximales/fisiopatología , Proteínas Klotho , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Receptor de Hormona Paratiroídea Tipo 1/deficiencia , Receptor de Hormona Paratiroídea Tipo 1/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIc/metabolismo , Regulación hacia Arriba
3.
Int J Mol Sci ; 19(11)2018 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-30463190

RESUMEN

Signaling by transforming growth factor (TGF)-ß plays an important role in development, including in palatogenesis. The dynamic morphological process of palatal fusion occurs to achieve separation of the nasal and oral cavities. Critically and specifically important in palatal fusion are the medial edge epithelial (MEE) cells, which are initially present at the palatal midline seam and over the course of the palate fusion process are lost from the seam, due to cell migration, epithelial-mesenchymal transition (EMT), and/or programed cell death. In order to define the role of TGF-ß signaling during this process, several approaches have been utilized, including a small interfering RNA (siRNA) strategy targeting TGF-ß receptors in an organ culture context, the use of genetically engineered mice, such as Wnt1-cre/R26R double transgenic mice, and a cell fate tracing through utilization of cell lineage markers. These approaches have permitted investigators to distinguish some specific traits of well-defined cell populations throughout the palatogenic events. In this paper, we summarize the current understanding on the role of TGF-ß signaling, and specifically its association with MEE cell fate during palatal fusion. TGF-ß is highly regulated both temporally and spatially, with TGF-ß3 and Smad2 being the preferentially expressed signaling molecules in the critical cells of the fusion processes. Interestingly, the accessory receptor, TGF-ß type 3 receptor, is also critical for palatal fusion, with evidence for its significance provided by Cre-lox systems and siRNA approaches. This suggests the high demand of ligand for this fine-tuned signaling process. We discuss the new insights in the fate of MEE cells in the midline epithelial seam (MES) during the palate fusion process, with a particular focus on the role of TGF-ß signaling.


Asunto(s)
Transición Epitelial-Mesenquimal , Hueso Paladar/embriología , Hueso Paladar/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Transición Epitelial-Mesenquimal/genética , Humanos , Fenotipo , Transducción de Señal/genética
4.
J Cell Physiol ; 232(1): 192-201, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27137755

RESUMEN

The aggressiveness of triple-negative breast cancer (TNBC), which lacks estrogen receptor, progesterone receptor and epidermal growth factor receptor 2 (HER2), represents a major challenge in breast cancer. Migratory and self-renewal capabilities are integral components of invasion, metastasis and recurrence of TNBC. Elevated hypoxia-inducible factor-1α (HIF-1α) expression is associated with aggressiveness of cancer. Nonetheless, how HIF-1α expression is regulated and how HIF-1α induces aggressive phenotype are not completely understood in TNBC. The cytotoxic effects of farnesyltransferase (FTase) inhibitors (FTIs) have been studied in cancer and leukemia cells. In contrast, the effect of FTIs on HIF-1α expression has not yet been studied. Here, we show that clinically relevant low-dose FTI, tipifarnib (300 nM), decreased HIF-1α expression, migration and tumorsphere formation in human MDA-MB-231 TNBC cells under a normoxic condition. In contrast, the low-dose FTIs did not inhibit cell growth and activity of the Ras pathway in MDA-MB 231 cells. Tipifarnib-induced decrease in HIF-1α expression was associated with amelioration of the Warburg effect, hypermetabolic state, increases in Snail expression and ATP release, and suppressed E-cadherin expression, major contributors to invasion, metastasis and recurrence of TBNC. These data suggest that FTIs may be capable of ameliorating the aggressive phenotype of TNBC by suppressing the HIF-1α-Snail pathway. J. Cell. Physiol. 232: 192-201, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Receptores ErbB/metabolismo , Farnesiltransferasa/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Receptores de Estrógenos/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Cadherinas/metabolismo , Línea Celular Tumoral , Proliferación Celular/fisiología , Femenino , Humanos , Quinolonas/farmacología , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/genética
5.
Kidney Int ; 92(3): 599-611, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28396120

RESUMEN

Osteocytes within the mineralized bone matrix control bone remodeling by regulating osteoblast and osteoclast activity. Osteocytes express the aging suppressor Klotho, but the functional role of this protein in skeletal homeostasis is unknown. Here we identify Klotho expression in osteocytes as a potent regulator of bone formation and bone mass. Targeted deletion of Klotho from osteocytes led to a striking increase in bone formation and bone volume coupled with enhanced osteoblast activity, in sharp contrast to what is observed in Klotho hypomorphic (kl/kl) mice. Conversely, overexpression of Klotho in cultured osteoblastic cells inhibited mineralization and osteogenic activity during osteocyte differentiation. Further, the induction of chronic kidney disease with high-turnover renal osteodystrophy led to downregulation of Klotho in bone cells. This appeared to offset the skeletal impact of osteocyte-targeted Klotho deletion. Thus, our findings establish a key role of osteocyte-expressed Klotho in regulating bone metabolism and indicate a new mechanism by which osteocytes control bone formation.


Asunto(s)
Envejecimiento/metabolismo , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/patología , Glucuronidasa/metabolismo , Osteocitos/metabolismo , Osteogénesis/fisiología , Animales , Densidad Ósea , Huesos/metabolismo , Huesos/patología , Diferenciación Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Glucuronidasa/genética , Humanos , Inmunohistoquímica , Proteínas Klotho , Ratones , Ratones Noqueados , Osteoblastos/fisiología , Osteoclastos/fisiología , Cultivo Primario de Células , Transducción de Señal
6.
Bioorg Med Chem Lett ; 27(4): 929-935, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28129980

RESUMEN

The enzyme ATP citrate lyase (ACL) catalyzes the formation of cytosolic acetyl CoA, the starting material for de novo lipid and cholesterol biosynthesis. The dysfunction and upregulation of ACL in numerous cancers makes it an attractive target for developing anticancer therapies. ACL inhibition by shRNA knockdown limits cancer cell proliferation and reduces cancer stemness. We designed and implemented a dual docking protocol to select virtual ACL inhibitors that were scored among the top 10 percentiles by both the Autodock Vina and the Glamdock algorithms. Via this in silico screens of a focused furoic acid library, we discovered four subtypes of furans and benzofurans as novel ACL inhibitors. The hit rate of our in silico protocol was 45.8% with 11 of 24 virtual hits confirmed as active in an in vitro ACL enzymatic assay. The IC50 of the most potent ACL inhibitor A1 is 4.1µM. Our results demonstrated remarkable hit rate by the dual docking approach and provided novel chemical scaffolds for the development of ACL inhibitors for the treatment of cancer.


Asunto(s)
ATP Citrato (pro-S)-Liasa/antagonistas & inhibidores , Ácidos Carboxílicos/farmacología , Inhibidores Enzimáticos/farmacología , Furanos/química , Ácidos Carboxílicos/química , Línea Celular , Descubrimiento de Drogas , Inhibidores Enzimáticos/química , Ensayos Analíticos de Alto Rendimiento , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular
7.
Kidney Int ; 90(2): 348-362, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27292223

RESUMEN

Klotho is a transmembrane protein expressed in the renal tubules where it acts as a permissive coreceptor for fibroblast growth factor 23 (FGF23). FGF23 signaling reduces the abundance of CYP27b1 and phosphate cotransporters NPT2a and NPT2c, leading to a decrease in 1,25(OH)2D3 synthesis and a rise in urinary phosphate excretion, respectively. Systemic or whole-nephron deletion of Klotho in mice results in renal FGF23 resistance characterized by high 1,25(OH)2D3 and phosphate levels and premature aging. Expression of Klotho is highest in the distal tubules, whereas 25OH vitamin D 1α hydroxylation and phosphate reabsorption predominantly occur in the proximal tubules. Currently, the segment-specific roles of Klotho in renal tubules are not fully understood. Here we have generated mice with Klotho specifically ablated from the proximal tubules using 3 different Cre mouse strains. All 3 models displayed impaired urinary phosphate excretion and increased abundance of NPT2a in the brush border membrane. Notably, hyperphosphatemia in knockout mice was mild or nonexistent under basal conditions but occurred upon high phosphate loading, indicating the presence of compensatory mechanisms. Effects on 1,25(OH)2D3 varied between mouse strains but were modest overall. Thus, Klotho expressed in the proximal tubules has a defined but limited role in renal phosphate handling in vivo.


Asunto(s)
25-Hidroxivitamina D3 1-alfa-Hidroxilasa/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Glucuronidasa/metabolismo , Túbulos Renales/fisiología , Fosfatos/metabolismo , Eliminación Renal , Envejecimiento Prematuro/metabolismo , Animales , Calcitriol/metabolismo , Femenino , Factor-23 de Crecimiento de Fibroblastos , Glucuronidasa/genética , Humanos , Hiperfosfatemia/sangre , Hiperfosfatemia/genética , Inmunohistoquímica , Túbulos Renales/citología , Proteínas Klotho , Ratones , Ratones Endogámicos C57BL , Fosfatos/orina , Cultivo Primario de Células , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIc/metabolismo
8.
Nat Med ; 12(6): 642-9, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16751767

RESUMEN

Preeclampsia is a pregnancy-specific hypertensive syndrome that causes substantial maternal and fetal morbidity and mortality. Maternal endothelial dysfunction mediated by excess placenta-derived soluble VEGF receptor 1 (sVEGFR1 or sFlt1) is emerging as a prominent component in disease pathogenesis. We report a novel placenta-derived soluble TGF-beta coreceptor, endoglin (sEng), which is elevated in the sera of preeclamptic individuals, correlates with disease severity and falls after delivery. sEng inhibits formation of capillary tubes in vitro and induces vascular permeability and hypertension in vivo. Its effects in pregnant rats are amplified by coadministration of sFlt1, leading to severe preeclampsia including the HELLP (hemolysis, elevated liver enzymes, low platelets) syndrome and restriction of fetal growth. sEng impairs binding of TGF-beta1 to its receptors and downstream signaling including effects on activation of eNOS and vasodilation, suggesting that sEng leads to dysregulated TGF-beta signaling in the vasculature. Our results suggest that sEng may act in concert with sFlt1 to induce severe preeclampsia.


Asunto(s)
Antígenos CD/metabolismo , Preeclampsia/metabolismo , Preñez , Receptores de Superficie Celular/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Adulto , Secuencia de Aminoácidos , Animales , Antígenos CD/genética , Endoglina , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Edad Gestacional , Hemodinámica , Humanos , Riñón/metabolismo , Riñón/patología , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Noqueados , Persona de Mediana Edad , Datos de Secuencia Molecular , Óxido Nítrico Sintasa de Tipo III/metabolismo , Placenta/metabolismo , Placenta/patología , Preeclampsia/etiología , Preeclampsia/fisiopatología , Embarazo , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/genética , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta1 , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
9.
J Cell Physiol ; 227(4): 1709-20, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21688263

RESUMEN

ATP citrate lyase (ACL) catalyzes the conversion of cytosolic citrate to acetyl-CoA and oxaloacetate. A definitive role for ACL in tumorigenesis has emerged from ACL RNAi and chemical inhibitor studies, showing that ACL inhibition limits tumor cell proliferation and survival and induces differentiation in vitro. In vivo, it reduces tumor growth leading to a cytostatic effect and induces differentiation. However, the underlying molecular mechanisms are poorly understood and agents that could enhance the efficacy of ACL inhibition have not been identified. Our studies focus on non-small cell lung cancer (NSCLC) lines, which show phosphatidylinositol 3-kinase (PI3K)/AKT activation secondary to a mutation in the K-Ras gene or the EGFR gene. Here we show that ACL knockdown promotes apoptosis and differentiation, leading to the inhibition of tumor growth in vivo. Moreover, in contrast to most studies, which elucidate how activation/suppression of signaling pathways can modify metabolism, we show that inhibition of a metabolic pathway "reverse signals" and attenuates PI3K/AKT signaling. Additionally, we find that statins, inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, which act downstream of ACL in the cholesterol synthesis pathway, dramatically enhance the anti-tumor effects of ACL inhibition, even regressing established tumors. With statin treatment, both PI3K/AKT and the MAPK pathways are affected. Moreover, this combined treatment is able to reduce the growth of EGF receptor resistant tumor cell types. Given the essential role of lipid synthesis in numerous cancers, this work may impact therapy in a broad range of tumors.


Asunto(s)
ATP Citrato (pro-S)-Liasa/antagonistas & inhibidores , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/terapia , ATP Citrato (pro-S)-Liasa/genética , Animales , Apoptosis , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Terapia Combinada , Transición Epitelial-Mesenquimal , Receptores ErbB/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Mutación , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Nat Med ; 9(7): 964-8, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12808448

RESUMEN

Bone morphogenic protein (BMP)-7 is a 35-kDa homodimeric protein and a member of the transforming growth factor (TGF)-beta superfamily. BMP-7 expression is highest in the kidney, and its genetic deletion in mice leads to severe impairment of eye, skeletal and kidney development. Here we report that BMP-7 reverses TGF-beta1-induced epithelial-to-mesenchymal transition (EMT) by reinduction of E-cadherin, a key epithelial cell adhesion molecule. Additionally, we provide molecular evidence for Smad-dependent reversal of TGF-beta1-induced EMT by BMP-7 in renal tubular epithelial cells and mammary ductal epithelial cells. In the kidney, EMT-induced accumulation of myofibroblasts and subsequent tubular atrophy are considered key determinants of renal fibrosis during chronic renal injury. We therefore tested the potential of BMP-7 to reverse TGF-beta1-induced de novo EMT in a mouse model of chronic renal injury. Our results show that systemic administration of recombinant human BMP-7 leads to repair of severely damaged renal tubular epithelial cells, in association with reversal of chronic renal injury. Collectively, these results provide evidence of cross talk between BMP-7 and TGF-beta1 in the regulation of EMT in health and disease.


Asunto(s)
Proteínas Morfogenéticas Óseas/farmacología , Túbulos Renales/citología , Mesodermo/citología , Nefritis/tratamiento farmacológico , Factor de Crecimiento Transformador beta/farmacología , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Animales , Proteína Morfogenética Ósea 7 , Proteínas Morfogenéticas Óseas/metabolismo , Cadherinas/efectos de los fármacos , Cadherinas/genética , Cadherinas/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Enfermedad Crónica , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/embriología , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Ratones , Ratones Endogámicos , Nefritis/patología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal , Proteína smad3 , Proteína Smad5 , Transactivadores/genética , Transactivadores/metabolismo , Transfección , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1
11.
J Clin Invest ; 117(12): 3940-51, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17992259

RESUMEN

Statins inhibit HMG-CoA reductase, a key enzyme in cholesterol synthesis, and are widely used to treat hypercholesterolemia. These drugs can lead to a number of side effects in muscle, including muscle fiber breakdown; however, the mechanisms of muscle injury by statins are poorly understood. We report that lovastatin induced the expression of atrogin-1, a key gene involved in skeletal muscle atrophy, in humans with statin myopathy, in zebrafish embryos, and in vitro in murine skeletal muscle cells. In cultured mouse myotubes, atrogin-1 induction following lovastatin treatment was accompanied by distinct morphological changes, largely absent in atrogin-1 null cells. In zebrafish embryos, lovastatin promoted muscle fiber damage, an effect that was closely mimicked by knockdown of zebrafish HMG-CoA reductase. Moreover, atrogin-1 knockdown in zebrafish embryos prevented lovastatin-induced muscle injury. Finally, overexpression of PGC-1alpha, a transcriptional coactivator that induces mitochondrial biogenesis and protects against the development of muscle atrophy, dramatically prevented lovastatin-induced muscle damage and abrogated atrogin-1 induction both in fish and in cultured mouse myotubes. Collectively, our human, animal, and in vitro findings shed light on the molecular mechanism of statin-induced myopathy and suggest that atrogin-1 may be a critical mediator of the muscle damage induced by statins.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Lovastatina/efectos adversos , Proteínas Musculares/metabolismo , Trastornos Musculares Atróficos/enzimología , Proteínas Ligasas SKP Cullina F-box/metabolismo , Transactivadores/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Colesterol/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Hidroximetilglutaril-CoA Reductasas/genética , Hidroximetilglutaril-CoA Reductasas/metabolismo , Ratones , Fibras Musculares Esqueléticas/enzimología , Fibras Musculares Esqueléticas/patología , Proteínas Musculares/genética , Músculo Esquelético/enzimología , Músculo Esquelético/patología , Trastornos Musculares Atróficos/inducido químicamente , Trastornos Musculares Atróficos/genética , Trastornos Musculares Atróficos/patología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Proteínas Ligasas SKP Cullina F-box/genética , Transactivadores/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/genética
12.
FASEB J ; 23(9): 2844-54, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19406843

RESUMEN

Statins are widely used to treat hypercholesterolemia but can lead to a number of side effects in muscle, including rhabdomyolysis. Our recent findings implicated the induction of atrogin-1, a gene required for the development of muscle atrophy, in statin-induced muscle damage. Since statins inhibit many biochemical reactions besides cholesterol synthesis, we sought to define the statin-inhibited pathways responsible for atrogin-1 expression and muscle damage. We report here that lovastatin-induced atrogin-1 expression and muscle damage in cultured mouse myotubes and zebrafish can be prevented in the presence of geranylgeranol but not farnesol. Further, inhibitors of the transfer of geranylgeranyl isoprene units to protein targets cause statin muscle damage and atrogin-1 induction in cultured cells and in fish. These findings support the concept that dysfunction of small GTP-binding proteins lead to statin-induced muscle damage since these molecules require modification by geranylgeranyl moieties for their cellular localization and activity. Collectively, our animal and in vitro findings shed light on the molecular mechanism of statin-induced myopathy and suggest that atrogin-1 may be regulated by novel signaling pathways.


Asunto(s)
Proteínas F-Box/genética , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Fibras Musculares Esqueléticas/patología , Proteínas Musculares/genética , Atrofia Muscular/inducido químicamente , Prenilación/genética , Proteínas Ligasas SKP Cullina F-box/genética , Proteínas de Pez Cebra/genética , Animales , Células Cultivadas , Proteínas de Unión al GTP , Lovastatina/efectos adversos , Ratones , Fibras Musculares Esqueléticas/efectos de los fármacos , Atrofia Muscular/etiología , Activación Transcripcional , Pez Cebra
13.
J Cell Biol ; 158(3): 529-39, 2002 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-12147676

RESUMEN

Endostatin (ES) is a fragment of collagen XVIII that possesses antiangiogenic activity. To gain insight into ES-mediated signaling, we studied the effects of ES RNA on Xenopus embryogenesis and observed developmental abnormalities consistent with impaired Wnt signaling. ES RNA blocked the axis duplication induced by beta-catenin, partially suppressed Wnt-dependent transcription, and stimulated degradation of both wild-type and "stabilized" forms of beta-catenin, the latter suggesting that ES signaling does not involve glycogen synthase kinase 3. Moreover, ES uses a pathway independent of the Siah1 protein in targeting beta-catenin for proteasome-mediated degradation. ES failed to suppress the effects of T cell-specific factor (TCF)-VP16 (TVP), a constitutive downstream transcriptional activator that acts independently of beta-catenin. Importantly, these data were replicated in endothelial cells and also in the DLD-1 colon carcinoma cells with the mutated adenomatous polyposis coli protein. Finally, suppression of endothelial cell migration and inhibition of cell cycle by ES were reversed by TVP. Though high levels of ES were used in both the Xenopus and endothelial cell studies and the effects on beta-catenin signaling were modest, these data argue that at pharmacological concentrations ES may impinge on Wnt signaling and promote beta-catenin degradation.


Asunto(s)
Inhibidores de la Angiogénesis/metabolismo , Colágeno/metabolismo , Endotelio Vascular/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Neovascularización Patológica/metabolismo , Oocitos/metabolismo , Fragmentos de Péptidos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/genética , Xenopus laevis/anomalías , Proteínas de Pez Cebra , Inhibidores de la Angiogénesis/genética , Animales , Tipificación del Cuerpo/genética , Movimiento Celular/genética , Colágeno/genética , Colágeno Tipo XVIII , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Endostatinas , Endotelio Vascular/citología , Femenino , Sustancias de Crecimiento/farmacología , Proteoglicanos de Heparán Sulfato/genética , Proteoglicanos de Heparán Sulfato/metabolismo , Humanos , Mutación/fisiología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oocitos/citología , Fragmentos de Péptidos/genética , Estructura Terciaria de Proteína/genética , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Fase S/efectos de los fármacos , Fase S/fisiología , Transactivadores/genética , Transactivadores/metabolismo , Células Tumorales Cultivadas , Ubiquitina-Proteína Ligasas , Proteínas Wnt , Proteínas de Xenopus , Xenopus laevis/genética , Xenopus laevis/metabolismo , beta Catenina
15.
Mol Cancer Res ; 4(11): 821-9, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17114340

RESUMEN

Lipocalin 2 is an iron-binding secreted protein that converts embryonic kidney mesenchyme to epithelia. Previously, we reported that lipocalin 2 could revert 4T1-ras-transformed mesenchymal tumor cells to a more epithelial phenotype, increase E-cadherin expression, and suppress cell invasiveness in vitro and in vivo, indicating that lipocalin 2 is a metastasis suppressor. Here, we show that lipocalin 2 can suppress the ras-induced expression of vascular endothelial growth factor in 4T1 cells via down-regulation of ras mitogen-activated protein kinase and ras phosphatidylinositol-3-kinase signaling. In addition, the expression of thrombospondin-1 (an antiangiogenic molecule) was increased in tumors formed by 4T1-ras cells into which lipocalin 2 was stably introduced. Tumor angiogenesis, assessed via an intradermal tumor angiogenesis assay, was also suppressed by lipocalin 2. We also show that caveolin-1 is a critical mediator of this activity. These data provide new insights into the action of lipocalin 2 and raise the possibility that the administration of lipocalin 2 may be useful for inhibiting tumor angiogenesis, in addition to suppressing tumor metastasis, in cancers which show ras activation.


Asunto(s)
Proteínas de Fase Aguda/fisiología , Neovascularización Patológica/genética , Proteínas Proto-Oncogénicas/fisiología , Trombospondina 1/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas ras/antagonistas & inhibidores , Proteínas de Fase Aguda/genética , Animales , Caveolina 1/genética , Caveolina 1/metabolismo , Línea Celular Transformada , Regulación hacia Abajo , Humanos , Hierro/metabolismo , Hierro/farmacología , Lipocalina 2 , Lipocalinas , Ratones , Ratones Endogámicos BALB C , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Neovascularización Patológica/enzimología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Transducción de Señal , Trombospondina 1/genética , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/genética , Proteínas ras/genética
16.
Clin Calcium ; 17(5): 704-10, 2007 May.
Artículo en Japonés | MEDLINE | ID: mdl-17470999

RESUMEN

Bone morphogenetic proteins (BMPs) are multipotent signaling molecules that belong to the transforming growth factor-beta (TGF-beta) superfamily. Developmentally these proteins promote endochondral bone formation and are involved in the cascade of body patterning and morphogenesis. Moreover, BMPs play an important role in the pathophysiology of several diseases, including osteoporosis, arthritis, pulmonary hypertension, cerebrovascular diseases, cancer and kidney diseases. In this review, BMP signaling and regulation, the pathophysiological role of BMP in kidney diseases and potential therapeutic applications have been discussed.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Riñón/fisiología , Animales , Células Endoteliales/fisiología , Feto/fisiología , Enfermedades Renales/fisiopatología , Ratones , Transducción de Señal/fisiología , Proteínas Smad Reguladas por Receptores/fisiología , Factores de Transcripción/fisiología , Factor de Crecimiento Transformador beta/fisiología
17.
Sci Rep ; 7(1): 4537, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28674429

RESUMEN

In this study we have tested the efficacy of citrate therapy in various cancer models. We found that citrate administration inhibited A549 lung cancer growth and additional benefit accrued in combination with cisplatin. Interestingly, citrate regressed Ras-driven lung tumors. Further studies indicated that citrate induced tumor cell differentiation. Additionally, citrate treated tumor samples showed significantly higher infiltrating T-cells and increased blood levels of numerous cytokines. Moreover, we found that citrate inhibited IGF-1R phosphorylation. In vitro studies suggested that citrate treatment inhibited AKT phosphorylation, activated PTEN and increased expression of p-eIF2a. We also found that p-eIF2a was decreased when PTEN was depleted. These data suggest that citrate acts on the IGF-1R-AKT-PTEN-eIF2a pathway. Additionally, metabolic profiling suggested that both glycolysis and the tricarboxylic acid cycle were suppressed in a similar manner in vitro in tumor cells and in vivo but only in tumor tissue. We reproduced many of these observations in an inducible Her2/Neu-driven breast cancer model and in syngeneic pancreatic tumor (Pan02) xenografts. Our data suggests that citrate can inhibit tumor growth in diverse tumor types and via multiple mechanisms. Dietary supplementation with citrate may be beneficial as a cancer therapy.


Asunto(s)
Ciclo del Ácido Cítrico , Ácido Cítrico/metabolismo , Modelos Biológicos , Receptor IGF Tipo 1/metabolismo , Transducción de Señal , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Ácido Cítrico/farmacología , Ciclo del Ácido Cítrico/efectos de los fármacos , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Femenino , Glucólisis/efectos de los fármacos , Humanos , Mediadores de Inflamación/metabolismo , Leucocitos/metabolismo , Leucocitos/patología , Ratones , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas ras/genética , Proteínas ras/metabolismo
18.
Eur J Med Chem ; 126: 920-928, 2017 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-27997879

RESUMEN

Aberrant cellular metabolism drives cancer proliferation and metastasis. ATP citrate lyase (ACL) plays a critical role in generating cytosolic acetyl CoA, a key building block for de novo fatty acid and cholesterol biosynthesis. ACL is overexpressed in cancer cells, and siRNA knockdown of ACL limits cancer cell proliferation and reduces cancer stemness. We characterized a new class of ACL inhibitors bearing the key structural feature of the natural product emodin. Structure-activity relationship (SAR) study led to the identification of 1d as a potent lead that demonstrated dose-dependent inhibition of proliferation and cancer stemness of the A549 lung cancer cell line. Computational modeling indicates this class of inhibitors occupies an allosteric binding site and blocks the entrance of the substrate citrate to its binding site.


Asunto(s)
ATP Citrato (pro-S)-Liasa/antagonistas & inhibidores , Diseño de Fármacos , Emodina/síntesis química , Emodina/farmacología , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , ATP Citrato (pro-S)-Liasa/química , ATP Citrato (pro-S)-Liasa/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Técnicas de Química Sintética , Emodina/química , Emodina/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Células Madre Neoplásicas/efectos de los fármacos , Dominios Proteicos , Relación Estructura-Actividad
19.
Cell Metab ; 25(3): 661-672, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28162969

RESUMEN

Intermittent PTH administration builds bone mass and prevents fractures, but its mechanism of action is unclear. We genetically deleted the PTH/PTHrP receptor (PTH1R) in mesenchymal stem cells using Prx1Cre and found low bone formation, increased bone resorption, and high bone marrow adipose tissue (BMAT). Bone marrow adipocytes traced to Prx1 and expressed classic adipogenic markers and high receptor activator of nuclear factor kappa B ligand (Rankl) expression. RANKL levels were also elevated in bone marrow supernatant and serum, but undetectable in other adipose depots. By cell sorting, Pref1+RANKL+ marrow progenitors were twice as great in mutant versus control marrow. Intermittent PTH administration to control mice reduced BMAT significantly. A similar finding was noted in male osteoporotic patients. Thus, marrow adipocytes exhibit osteogenic and adipogenic characteristics, are uniquely responsive to PTH, and secrete RANKL. These studies reveal an important mechanism for PTH's therapeutic action through its ability to direct mesenchymal cell fate.


Asunto(s)
Células de la Médula Ósea/citología , Linaje de la Célula/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Hormona Paratiroidea/farmacología , Adipocitos/metabolismo , Adipogénesis , Tejido Adiposo/metabolismo , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Huesos , Recuento de Células , Humanos , Integrasas/metabolismo , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Osteoblastos/metabolismo , Osteoporosis/patología , Fenotipo , Ligando RANK/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Transducción de Señal , Cráneo/citología
20.
FASEB J ; 19(2): 270-1, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15579670

RESUMEN

The vasculature consists of endothelial cells (ECs) lined by pericyte/vascular smooth muscle cells (vSMCs). Pericyte/vSMCs provide support to the mature vasculature but are also essential for normal blood vessel development. To determine how pericyte-EC communication influences vascular development, we used the well-established in vitro model of TGFbeta-stimulated differentiation of 10T1/2 cells into pericyte/vSMCs. Microarray analysis was performed to identify genes that were differentially expressed by induced vs. uninduced 10T1/2 cells. We discovered that these cells show an angiogenic program of gene expression, with up-regulation of several genes previously implicated in angiogenesis, including VEGF, IL-6, VEGF-C, HB-EGF, CTGF, tenascin C, integrin alpha5, and Eph receptor A2. Up-regulation of some genes was validated by Western blots and immunocytochemistry. We also examined the functional significance of these gene expression changes. VEGF and IL-6 alone and in combination were important in 10T1/2 cell differentiation. Furthermore, we used a coculture system of 10T1/2 and human umbilical vein ECs (HUVECs), resulting in the formation of cordlike structures by the HUVECs. This cordlike structure formation was disrupted when neutralizing antibodies to VEGF or IL-6 were added to the coculture system. The results of these studies show that factors produced by pericytes may be responsible for recruiting ECs and promoting angiogenesis. Therefore, a further understanding of the genes involved in pericyte differentiation could provide a novel approach for developing anti-angiogenic therapies.


Asunto(s)
Diferenciación Celular/genética , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Análisis por Micromatrices/métodos , Neovascularización Fisiológica/genética , Pericitos/citología , Animales , Línea Celular , Técnicas de Cocultivo , Embrión de Mamíferos/citología , Genes/fisiología , Humanos , Ratones , Ratones Endogámicos C3H , Células Madre Multipotentes/química , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Pericitos/química , Pericitos/metabolismo , Venas Umbilicales/química , Venas Umbilicales/citología , Venas Umbilicales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA