Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Proc Natl Acad Sci U S A ; 114(16): E3344-E3353, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28373577

RESUMO

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.


Assuntos
Cálcio/metabolismo , Claudinas/fisiologia , Extremidades/fisiologia , Regulação da Expressão Gênica , Hormônio Paratireóideo/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Junções Íntimas/fisiologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas/genética , Transdução de Sinais
2.
Am J Physiol Renal Physiol ; 315(5): F1261-F1270, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29993278

RESUMO

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.


Assuntos
Fatores de Crescimento de Fibroblastos/sangue , Glucuronidase/metabolismo , Hiperfosfatemia/sangue , Túbulos Renais Proximais/metabolismo , Hormônio Paratireóideo/sangue , Fosfatos/sangue , Reabsorção Renal , Animais , Calcitriol/sangue , Cálcio/sangue , Células Cultivadas , Fator de Crescimento de Fibroblastos 23 , Predisposição Genética para Doença , Glucuronidase/deficiência , Glucuronidase/genética , Hiperfosfatemia/genética , Hiperfosfatemia/fisiopatologia , Túbulos Renais Proximais/fisiopatologia , Proteínas Klotho , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Receptor Tipo 1 de Hormônio Paratireóideo/deficiência , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/metabolismo , Regulação para Cima
3.
Int J Mol Sci ; 19(11)2018 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-30463190

RESUMO

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.


Assuntos
Transição Epitelial-Mesenquimal , Palato/embriologia , Palato/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Transição Epitelial-Mesenquimal/genética , Humanos , Fenótipo , Transdução de Sinais/genética
4.
J Cell Physiol ; 232(1): 192-201, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27137755

RESUMO

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.


Assuntos
Receptores ErbB/metabolismo , Farnesiltranstransferase/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Receptores de Estrogênio/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Caderinas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Feminino , Humanos , Quinolonas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/genética
5.
Kidney Int ; 92(3): 599-611, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28396120

RESUMO

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.


Assuntos
Envelhecimento/metabolismo , Distúrbio Mineral e Ósseo na Doença Renal Crônica/patologia , Glucuronidase/metabolismo , Osteócitos/metabolismo , Osteogênese/fisiologia , Animais , Densidade Óssea , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Diferenciação Celular , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/metabolismo , Glucuronidase/genética , Humanos , Imuno-Histoquímica , Proteínas Klotho , Camundongos , Camundongos Knockout , Osteoblastos/fisiologia , Osteoclastos/fisiologia , Cultura Primária de Células , Transdução de Sinais
6.
Bioorg Med Chem Lett ; 27(4): 929-935, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28129980

RESUMO

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.


Assuntos
ATP Citrato (pro-S)-Liase/antagonistas & inibidores , Ácidos Carboxílicos/farmacologia , Inibidores Enzimáticos/farmacologia , Furanos/química , Ácidos Carboxílicos/química , Linhagem Celular , Descoberta de Drogas , Inibidores Enzimáticos/química , Ensaios de Triagem em Larga Escala , Humanos , Simulação de Acoplamento Molecular , Estrutura Molecular
7.
Kidney Int ; 90(2): 348-362, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27292223

RESUMO

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.


Assuntos
25-Hidroxivitamina D3 1-alfa-Hidroxilase/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Glucuronidase/metabolismo , Túbulos Renais/fisiologia , Fosfatos/metabolismo , Eliminação Renal , Senilidade Prematura/metabolismo , Animais , Calcitriol/metabolismo , Feminino , Fator de Crescimento de Fibroblastos 23 , Glucuronidase/genética , Humanos , Hiperfosfatemia/sangue , Hiperfosfatemia/genética , Imuno-Histoquímica , Túbulos Renais/citologia , Proteínas Klotho , Camundongos , Camundongos Endogâmicos C57BL , Fosfatos/urina , Cultura Primária de Células , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIc/metabolismo
8.
Nat Med ; 12(6): 642-9, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16751767

RESUMO

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.


Assuntos
Antígenos CD/metabolismo , Pré-Eclâmpsia/metabolismo , Prenhez , Receptores de Superfície Celular/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Adulto , Sequência de Aminoácidos , Animais , Antígenos CD/genética , Endoglina , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feminino , Idade Gestacional , Hemodinâmica , Humanos , Rim/metabolismo , Rim/patologia , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Dados de Sequência Molecular , Óxido Nítrico Sintase Tipo III/metabolismo , Placenta/metabolismo , Placenta/patologia , Pré-Eclâmpsia/etiologia , Pré-Eclâmpsia/fisiopatologia , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores de Superfície Celular/genética , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta1 , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética
9.
J Cell Physiol ; 227(4): 1709-20, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21688263

RESUMO

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.


Assuntos
ATP Citrato (pro-S)-Liase/antagonistas & inibidores , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/terapia , ATP Citrato (pro-S)-Liase/genética , Animais , Apoptose , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Terapia Combinada , Transição Epitelial-Mesenquimal , Receptores ErbB/genética , Feminino , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Mutação , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Nat Med ; 9(7): 964-8, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12808448

RESUMO

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.


Assuntos
Proteínas Morfogenéticas Ósseas/farmacologia , Túbulos Renais/citologia , Mesoderma/citologia , Nefrite/tratamento farmacológico , Fator de Crescimento Transformador beta/farmacologia , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/metabolismo , Animais , Proteína Morfogenética Óssea 7 , Proteínas Morfogenéticas Ósseas/metabolismo , Caderinas/efeitos dos fármacos , Caderinas/genética , Caderinas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Doença Crônica , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/embriologia , Mesoderma/efeitos dos fármacos , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos , Nefrite/patologia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Proteína Smad3 , Proteína Smad5 , Transativadores/genética , Transativadores/metabolismo , Transfecção , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1
11.
J Clin Invest ; 117(12): 3940-51, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17992259

RESUMO

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.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Lovastatina/efeitos adversos , Proteínas Musculares/metabolismo , Transtornos Musculares Atróficos/enzimologia , Proteínas Ligases SKP Culina F-Box/metabolismo , Transativadores/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Colesterol/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Hidroximetilglutaril-CoA Redutases/genética , Hidroximetilglutaril-CoA Redutases/metabolismo , Camundongos , Fibras Musculares Esqueléticas/enzimologia , Fibras Musculares Esqueléticas/patologia , Proteínas Musculares/genética , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Transtornos Musculares Atróficos/induzido quimicamente , Transtornos Musculares Atróficos/genética , Transtornos Musculares Atróficos/patologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Proteínas Ligases SKP Culina F-Box/genética , Transativadores/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
12.
FASEB J ; 23(9): 2844-54, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19406843

RESUMO

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.


Assuntos
Proteínas F-Box/genética , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Fibras Musculares Esqueléticas/patologia , Proteínas Musculares/genética , Atrofia Muscular/induzido quimicamente , Prenilação/genética , Proteínas Ligases SKP Culina F-Box/genética , Proteínas de Peixe-Zebra/genética , Animais , Células Cultivadas , Proteínas de Ligação ao GTP , Lovastatina/efeitos adversos , Camundongos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Atrofia Muscular/etiologia , Ativação Transcricional , Peixe-Zebra
13.
J Cell Biol ; 158(3): 529-39, 2002 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-12147676

RESUMO

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.


Assuntos
Inibidores da Angiogênese/metabolismo , Colágeno/metabolismo , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neovascularização Patológica/metabolismo , Oócitos/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/genética , Xenopus laevis/anormalidades , Proteínas de Peixe-Zebra , Inibidores da Angiogênese/genética , Animais , Padronização Corporal/genética , Movimento Celular/genética , Colágeno/genética , Colágeno Tipo XVIII , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Endostatinas , Endotélio Vascular/citologia , Feminino , Substâncias de Crescimento/farmacologia , Proteoglicanas de Heparan Sulfato/genética , Proteoglicanas de Heparan Sulfato/metabolismo , Humanos , Mutação/fisiologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oócitos/citologia , Fragmentos de Peptídeos/genética , Estrutura Terciária de Proteína/genética , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fase S/efeitos dos fármacos , Fase S/fisiologia , Transativadores/genética , Transativadores/metabolismo , Células Tumorais Cultivadas , Ubiquitina-Proteína Ligases , Proteínas Wnt , Proteínas de Xenopus , Xenopus laevis/genética , Xenopus laevis/metabolismo , beta Catenina
15.
Mol Cancer Res ; 4(11): 821-9, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17114340

RESUMO

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.


Assuntos
Proteínas de Fase Aguda/fisiologia , Neovascularização Patológica/genética , Proteínas Proto-Oncogênicas/fisiologia , Trombospondina 1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas ras/antagonistas & inibidores , Proteínas de Fase Aguda/genética , Animais , Caveolina 1/genética , Caveolina 1/metabolismo , Linhagem Celular Transformada , Regulação para Baixo , Humanos , Ferro/metabolismo , Ferro/farmacologia , Lipocalina-2 , Lipocalinas , Camundongos , Camundongos Endogâmicos BALB C , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neovascularização Patológica/enzimologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Transdução de Sinais , Trombospondina 1/genética , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética , Proteínas ras/genética
16.
Clin Calcium ; 17(5): 704-10, 2007 May.
Artigo em Japonês | MEDLINE | ID: mdl-17470999

RESUMO

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.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Rim/fisiologia , Animais , Células Endoteliais/fisiologia , Feto/fisiologia , Nefropatias/fisiopatologia , Camundongos , Transdução de Sinais/fisiologia , Proteínas Smad Reguladas por Receptor/fisiologia , Fatores de Transcrição/fisiologia , Fator de Crescimento Transformador beta/fisiologia
17.
Sci Rep ; 7(1): 4537, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28674429

RESUMO

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.


Assuntos
Ciclo do Ácido Cítrico , Ácido Cítrico/metabolismo , Modelos Biológicos , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Ácido Cítrico/farmacologia , Ciclo do Ácido Cítrico/efeitos dos fármacos , Citocinas/biossíntese , Modelos Animais de Doenças , Feminino , Glicólise/efeitos dos fármacos , Humanos , Mediadores da Inflamação/metabolismo , Leucócitos/metabolismo , Leucócitos/patologia , Camundongos , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas ras/genética , Proteínas ras/metabolismo
18.
Eur J Med Chem ; 126: 920-928, 2017 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-27997879

RESUMO

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.


Assuntos
ATP Citrato (pro-S)-Liase/antagonistas & inibidores , Desenho de Fármacos , Emodina/síntese química , Emodina/farmacologia , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , ATP Citrato (pro-S)-Liase/química , ATP Citrato (pro-S)-Liase/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Técnicas de Química Sintética , Emodina/química , Emodina/metabolismo , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Humanos , Simulação de Acoplamento Molecular , Células-Tronco Neoplásicas/efeitos dos fármacos , Domínios Proteicos , Relação Estrutura-Atividade
19.
Cell Metab ; 25(3): 661-672, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28162969

RESUMO

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.


Assuntos
Células da Medula Óssea/citologia , Linhagem da Célula/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Hormônio Paratireóideo/farmacologia , Adipócitos/metabolismo , Adipogenia , Tecido Adiposo/metabolismo , Animais , Biomarcadores/metabolismo , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Osso e Ossos , Contagem de Células , Humanos , Integrases/metabolismo , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Osteoblastos/metabolismo , Osteoporose/patologia , Fenótipo , Ligante RANK/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Transdução de Sinais , Crânio/citologia
20.
FASEB J ; 19(2): 270-1, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15579670

RESUMO

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.


Assuntos
Diferenciação Celular/genética , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/genética , Análise em Microsséries/métodos , Neovascularização Fisiológica/genética , Pericitos/citologia , Animais , Linhagem Celular , Técnicas de Cocultura , Embrião de Mamíferos/citologia , Genes/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C3H , Células-Tronco Multipotentes/química , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Pericitos/química , Pericitos/metabolismo , Veias Umbilicais/química , Veias Umbilicais/citologia , Veias Umbilicais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA