Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Arthroplasty ; 33(3): 823-828, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29217393

RESUMO

BACKGROUND: Trabecular Metal (TM) augments are one option when reconstructing bone loss during acetabular side revision surgery. METHODS: We studied 38 consecutive patients with Paprosky type 3 defects that were revised using a TM shell and one or more augments over a 6-year period. There were 29 Paprosky type 3A defects and 9 Paprosky type 3B defects. The mean age of the patients at the time of surgery was 68.2 years (range 48-84). The mean length of follow-up was 36 months (range 18-74). RESULTS: The mean preoperative short form 12 health survey improved from 27.7 before operation to 30.1 at the time of final follow-up (P = .001). The mean Western Ontario and McMaster Universities Osteoarthritis Index score improved from 53 preoperatively to a mean of 78.8 at final follow-up (P < .0001). There was evidence of radiographic loosening in 7 of the cup-augment constructs. One patient developed a deep infection requiring re-revision. Two patients required revision for aseptic loosening. CONCLUSION: The use of TM in complex acetabular reconstruction is associated with good outcome in the short to medium term.


Assuntos
Acetábulo/cirurgia , Artroplastia de Quadril/instrumentação , Artroplastia de Quadril/métodos , Prótese de Quadril , Reoperação/instrumentação , Reoperação/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Parafusos Ósseos , Osso Esponjoso , Feminino , Inquéritos Epidemiológicos , Humanos , Estimativa de Kaplan-Meier , Masculino , Metais , Pessoa de Meia-Idade , Osteoartrite/cirurgia , Período Pré-Operatório , Falha de Prótese , Estudos Retrospectivos , Índice de Gravidade de Doença , Pele
2.
Hepatology ; 62(2): 615-26, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25482505

RESUMO

UNLABELLED: Liver X receptors (LXRs) are determinants of hepatic stellate cell (HSC) activation and liver fibrosis. Freshly isolated HSCs from Lxrαß(-/-) mice have increased lipid droplet (LD) size, but the functional consequences of this are unknown. Our aim was to determine whether LXRs link cholesterol to retinoid storage in HSCs and how this impacts activation. Primary HSCs from Lxrαß(-/-) and wild-type mice were profiled by gene array during in vitro activation. Lipid content was quantified by high-performance liquid chromatography and mass spectroscopy. Primary HSCs were treated with nuclear receptor ligands, transfected with small interfering RNA and plasmid constructs, and analyzed by immunocytochemistry. Lxrαß(-/-) HSCs have increased cholesterol and retinyl esters. The retinoid increase drives intrinsic retinoic acid receptor signaling, and activation occurs more rapidly in Lxrαß(-/-) HSCs. We identify Rab18 as a novel retinoic acid-responsive, LD-associated protein that helps mediate stellate cell activation. Rab18 mRNA, protein, and membrane insertion increase during activation. Both Rab18 guanosine triphosphatase activity and isoprenylation are required for stellate cell LD loss and induction of activation markers. These phenomena are accelerated in Lxrαß(-/-) HSCs, where there is greater retinoic acid flux. Conversely, Rab18 knockdown retards LD loss in culture and blocks activation, just like the functional mutants. Rab18 is also induced with acute liver injury in vivo. CONCLUSION: Retinoid and cholesterol metabolism are linked in stellate cells by the LD-associated protein Rab18. Retinoid overload helps explain the profibrotic phenotype of Lxrαß(-/-) mice, and we establish a pivotal role for Rab18 GTPase activity and membrane insertion in wild-type stellate cell activation. Interference with Rab18 may have significant therapeutic benefit in ameliorating liver fibrosis.


Assuntos
Células Estreladas do Fígado/metabolismo , Metabolismo dos Lipídeos , Cirrose Hepática/metabolismo , Receptores Nucleares Órfãos/metabolismo , Retinoides/farmacologia , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Gotículas Lipídicas/metabolismo , Cirrose Hepática/patologia , Receptores X do Fígado , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos , Análise em Microsséries , Receptores Nucleares Órfãos/efeitos dos fármacos , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real/métodos , Transdução de Sinais
3.
Proc Natl Acad Sci U S A ; 111(28): E2866-74, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24982199

RESUMO

PET is a powerful technique for quantifying and visualizing biochemical pathways in vivo. Here, we develop and validate a novel PET probe, [(18)F]-2-deoxy-2-fluoroarabinose ([(18)F]DFA), for in vivo imaging of ribose salvage. DFA mimics ribose in vivo and accumulates in cells following phosphorylation by ribokinase and further metabolism by transketolase. We use [(18)F]DFA to show that ribose preferentially accumulates in the liver, suggesting a striking tissue specificity for ribose metabolism. We demonstrate that solute carrier family 2, member 2 (also known as GLUT2), a glucose transporter expressed in the liver, is one ribose transporter, but we do not know if others exist. [(18)F]DFA accumulation is attenuated in several mouse models of metabolic syndrome, suggesting an association between ribose salvage and glucose and lipid metabolism. These results describe a tool for studying ribose salvage and suggest that plasma ribose is preferentially metabolized in the liver.


Assuntos
Fígado , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/farmacologia , Ribose/metabolismo , Animais , Arabinose/análogos & derivados , Arabinose/farmacologia , Linhagem Celular , Modelos Animais de Doenças , Radioisótopos de Flúor/farmacologia , Glucose/genética , Glucose/metabolismo , Transportador de Glucose Tipo 2/metabolismo , Humanos , Metabolismo dos Lipídeos , Fígado/diagnóstico por imagem , Fígado/metabolismo , Síndrome Metabólica/diagnóstico por imagem , Síndrome Metabólica/metabolismo , Camundongos , Especificidade de Órgãos , Radiografia
4.
Sci Signal ; 6(276): ra36, 2013 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-23695162

RESUMO

Estrogen induces signal transduction through estrogen receptor α (ERα), which localizes to both the plasma membrane and nucleus. Using wild-type mice, ERα knockout (ERKO) mice, or transgenic mice expressing only the ligand-binding domain of ERα exclusively at the plasma membrane (MOER), we compared the transcriptional profiles of liver tissue extracts after mice were injected with the ERα agonist propyl-pyrazole-triol (PPT). The expression of many lipid synthesis-related genes was comparably decreased in livers from MOER or wild-type mice but was not suppressed in ERKO mice, indicating that only membrane-localized ERα was necessary for their suppression. Cholesterol, triglyceride, and fatty acid content was decreased only in livers from wild-type and MOER mice exposed to PPT, but not in the livers from the ERKO mice, validating the membrane-driven signaling pathway on a physiological level. PPT-triggered activation of ERα at the membrane induced adenosine monophosphate-activated protein kinase to phosphorylate sterol regulatory element-binding factor 1 (Srebf1), preventing its association with and therefore its proteolytic cleavage by site-1 protease. Consequently, Srebf1 was sequestered in the cytoplasm, preventing the expression of cholesterol synthesis-associated genes. Thus, we showed that inhibition of gene expression mediated by membrane-localized ERα caused a metabolic phenotype that did not require nuclear ERα.


Assuntos
Estrogênios/fisiologia , Lipídeos/análise , Fígado/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Adenilato Quinase/metabolismo , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Receptor alfa de Estrogênio/metabolismo , Fígado/citologia , Fígado/enzimologia , Camundongos , Camundongos Knockout , Ligação Proteica , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo
5.
J Physiol ; 591(11): 2813-31, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23529131

RESUMO

The cAMP-regulated potassium channel KCNQ1:KCNE3 plays an essential role in transepithelial Cl(-) secretion. Recycling of K(+) across the basolateral membrane provides the driving force necessary to maintain apical Cl(-) secretion. The steroid hormone oestrogen (17ß-oestradiol; E2), produces a female-specific antisecretory response in rat distal colon through the inhibition of the KCNQ1:KCNE3 channel. It has previously been shown that rapid inhibition of the channel conductance results from E2-induced uncoupling of the KCNE3 regulatory subunit from the KCNQ1 channel pore complex. The purpose of this study was to determine the mechanism required for sustained inhibition of the channel function. We found that E2 plays a role in regulation of KCNQ1 cell membrane abundance by endocytosis. Ussing chamber experiments have shown that E2 inhibits both Cl(-) secretion and KCNQ1 current in a colonic cell line, HT29cl.19A, when cultured as a confluent epithelium. Following E2 treatment, KCNQ1 was retrieved from the plasma membrane by a clathrin-mediated endocytosis, which involved the association between KCNQ1 and the clathrin adaptor, AP-2. Following endocytosis, KCNQ1 was accumulated in early endosomes. Following E2-induced endocytosis, rather than being degraded, KCNQ1 was recycled by a biphasic mechanism involving Rab4 and Rab11. Protein kinase Cδ and AMP-dependent kinase were rapidly phosphorylated in response to E2 on their activating phosphorylation sites, Ser643 and Thr172, respectively (as previously shown). Both kinases are necessary for the E2-induced endocytosis, because E2 failed to induce KCNQ1 internalization following pretreatment with specific inhibitors of both protein kinase Cδ and AMP-dependent kinase. The ubiquitin ligase Nedd4.2 binds KCNQ1 in response to E2 to induce channel internalization. This study has provided the first demonstration of hormonal regulation of KCNQ1 trafficking. In conclusion, we propose that internalization of KCNQ1 is a key event in the sustained antisecretory response to oestrogen.


Assuntos
Colo/metabolismo , Endocitose/efeitos dos fármacos , Estradiol/farmacologia , Mucosa Intestinal/metabolismo , Canal de Potássio KCNQ1/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Potenciais de Ação , Complexo 2 de Proteínas Adaptadoras/metabolismo , Animais , Colo/citologia , Colo/fisiologia , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Feminino , Células HT29 , Humanos , Mucosa Intestinal/fisiologia , Ubiquitina-Proteína Ligases Nedd4 , Ligação Proteica , Proteína Quinase C-delta/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Ubiquitina-Proteína Ligases/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab4 de Ligação ao GTP/metabolismo
6.
Mol Endocrinol ; 26(12): 2058-70, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23028062

RESUMO

Most cancers use glucose as substrate for aerobic glycolysis in preference to oxidative phosphorylation. However, variable glucose concentrations within the in-vivo tumor microenvironment may necessitate metabolic plasticity. Furthermore, little information exists on a role for estrogen receptors in modulating possible metabolic adaptations in breast cancer cells. Here we find that MCF-7 cells switch between metabolic pathways depending on glucose availability and 17ß-estradiol (E(2)) potentiates adaptation. In high glucose conditions E(2) up-regulates glycolysis via enhanced AKT kinase activity and suppresses tricarboxylic acid cycle activity. After a decrease in extracellular glucose, mitochondrial pathways are activated in preference to glycolysis. In this setting, E(2) suppresses glycolysis and rescues cell viability by stimulating the tricarboxylic acid cycle via the up-regulation of pyruvate dehydrogenase (PDH) activity. E(2) also increases ATP in low glucose-cultured cells, and the novel phosphorylation of PDH by AMP kinase is required for these metabolic compensations. Capitalizing on metabolic vulnerability, knockdown of PDH in the low-glucose state strongly potentiates ionizing radiation-induced apoptosis and reverses the cell survival effects of E(2). We propose that lowering glucose substrate and inhibiting PDH may augment adjuvant therapies for estrogen receptor-positive breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Estrogênios/metabolismo , Glucose/metabolismo , Glicólise/genética , Cetona Oxirredutases/metabolismo , Trifosfato de Adenosina , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular/genética , Ciclo do Ácido Cítrico , Estradiol/metabolismo , Feminino , Humanos , Cetona Oxirredutases/genética , Células MCF-7 , Mitocôndrias/metabolismo , Fosforilação Oxidativa , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Espécies Reativas de Oxigênio , Receptores de Estrogênio/metabolismo , Transdução de Sinais/genética , Microambiente Tumoral , Regulação para Cima
7.
J Physiol ; 589(Pt 21): 5091-107, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21911611

RESUMO

The KCNQ1 potassium channel associates with various KCNE ancillary subunits that drastically affect channel gating and pharmacology. Co-assembly with KCNE3 produces a current with nearly instantaneous activation, some time-dependent activation at very positive potentials, a linear current-voltage relationship and a 10-fold higher sensitivity to chromanol 293B. KCNQ1:KCNE3 channels are expressed in colonic crypts and mediate basolateral K(+) recycling required for Cl(-) secretion. We have previously reported the female-specific anti-secretory effects of oestrogen via KCNQ1:KCNE3 channel inhibition in colonic crypts. This study was designed to determine whether sex and oestrogen regulate the expression and function of KCNQ1 and KCNE3 in rat distal colon. Colonic crypts were isolated from Sprague-Dawley rats and used for whole-cell patch-clamp and to extract total RNA and protein. Sheets of epithelium were used for short-circuit current recordings. KCNE1 and KCNE3 mRNA and protein abundance were significantly higher in male than female crypts. No expression of KCNE2 was found and no difference was observed in KCNQ1 expression between male and female (at oestrus) colonic crypts. Male crypts showed a 2.2-fold higher level of association of KCNQ1 and KCNE3 compared to female cells. In female colonic crypts, KCNQ1 and KCNE3 protein expression fluctuated throughout the oestrous cycle and 17ß-oestradiol (E2 10 nM) produced a rapid (<15 min) dissociation of KCNQ1 and KCNE3 in female crypts only. Whole-cell K(+) currents showed a linear current-voltage relationship in male crypts, while K(+) currents in colonic crypts isolated from females displayed voltage-dependent outward rectification. Currents in isolated male crypts and epithelial sheets were 10-fold more sensitive to specific KCNQ1 inhibitors, such as chromanol 293B and HMR-1556, than in female. The effect of E2 on K(+) currents mediated by KCNQ1 with or without different ß-subunits was assayed from current-voltage relations elicited in CHO cells transfected with KCNQ1 and KCNE3 or KCNE1 cDNA. E2 (100 nM) reduced the currents mediated by the KCNQ1:KCNE3 potassium channel and had no effect on currents via KCNQ1:KCNE1 or KCNQ1 alone. Currents mediated by the complex formed by KCNQ1 and the mutant KCNE3-S82A ß-subunit (mutation of the site for PKCδ-promoted phosphorylation and modulation of the activity of KCNE3) showed rapid run-down and insensitivity to E2. Together, these data suggest that oestrogen regulates the expression of the KCNE1 and KCNE3 and with it the gating and pharmacological properties of the K(+) conductance required for Cl(-) secretion. The decreased association of the KCNQ1:KCNE3 channel complex promoted by oestrogen exposure underlies the molecular mechanism for the sexual dimorphism and oestrous cycle dependence of the anti-secretory actions of oestrogen in the intestine.


Assuntos
Colo/fisiologia , Estrogênios/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Animais , Células CHO , Cricetinae , Cricetulus , Feminino , Técnicas In Vitro , Mucosa Intestinal/metabolismo , Canal de Potássio KCNQ1/fisiologia , Masculino , Técnicas de Patch-Clamp , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Caracteres Sexuais
8.
Front Physiol ; 2: 33, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21747769

RESUMO

Berberine is a plant alkaloid with multiple pharmacological actions, including antidiarrhoeal activity and has been shown to inhibit Cl(-) secretion in distal colon. The aims of this study were to determine the molecular signaling mechanisms of action of berberine on Cl(-) secretion and the ion transporter targets. Monolayers of T84 human colonic carcinoma cells grown in permeable supports were placed in Ussing chambers and short-circuit current measured in response to secretagogues and berberine. Whole-cell current recordings were performed in T84 cells using the patch-clamp technique. Berberine decreased forskolin-induced short-circuit current in a concentration-dependent manner (IC(50) 80 ± 8 µM). In apically permeabilized monolayers and whole-cell current recordings, berberine inhibited a cAMP-dependent and chromanol 293B-sensitive basolateral membrane K(+) current by 88%, suggesting inhibition of KCNQ1 K(+) channels. Berberine did not affect either apical Cl(-) conductance or basolateral Na(+)-K(+)-ATPase activity. Berberine stimulated p38 MAPK, PKCα and PKA, but had no effect on p42/p44 MAPK and PKCδ. However, berberine pre-treatment prevented stimulation of p42/p44 MAPK by epidermal growth factor. The inhibitory effect of berberine on Cl(-) secretion was partially blocked by HBDDE (∼65%), an inhibitor of PKCα and to a smaller extent by inhibition of p38 MAPK with SB202190 (∼15%). Berberine treatment induced an increase in association between PKCα and PKA with KCNQ1 and produced phosphorylation of the channel. We conclude that berberine exerts its inhibitory effect on colonic Cl(-) secretion through inhibition of basolateral KCNQ1 channels responsible for K(+) recycling via a PKCα-dependent pathway.

9.
Steroids ; 76(9): 867-76, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21600231

RESUMO

Excessive Cl(-) secretion is the driving force for secretory diarrhea. 17ß-Estradiol has been shown to inhibit Cl(-) secretion in rat distal colon through a nongenomic pathway. We examined whether 17ß-estradiol inhibits Cl(-) secretion in an animal model of secretory diarrhea and the downstream effectors involved. The effect of 17ß-estradiol on cholera toxin and heat-stable enterotoxin induced Cl(-) secretion in rat colonic mucosal sheets was studied by current-voltage clamping. Selective permeabilization of apical or basolateral membranes with amphotericin B or nystatin was used to isolate basolateral K(+) channel and apical Cl(-) channel activity, respectively. 17ß-Estradiol dose-dependently inhibited secretory responses to both toxins with IC(50) values of approximately 1nM. This effect was female-gender specific, with no inhibition observed in male tissues. 17ß-Estradiol responses were insensitive to the pure anti-estrogen ICI 182,720. 17ß-Estradiol exerted its effects downstream of enterotoxin-induced production of second messengers (cAMP and cGMP) but was dependent on PKCδ activation. In nystatin-permeabilized tissues, apical Cl(-) currents were unaffected by 17ß-estradiol treatment while basolateral K(+) current was profoundly inhibited by the hormone. This current was sensitive to the specific KCNQ1 channel inhibitors chromanol 293B and HMR-1556. In conclusion, 17ß-estradiol inhibits enterotoxin-induced Cl(-) secretion via a PKCδ-dependent mechanism involving inhibition of basolateral KCNQ1 channels. These data elucidate mechanisms of 17ß-estradiol inhibition of Cl(-) secretion induced by enterotoxins in intestinal epithelia, which may be relevant for the treatment of diarrheal diseases.


Assuntos
Toxinas Bacterianas/farmacologia , Cloretos/metabolismo , Toxina da Cólera/farmacologia , Colo/metabolismo , Enterotoxinas/farmacologia , Escherichia coli , Estradiol/farmacologia , Estrogênios/farmacologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Androgênios/farmacologia , Androgênios/fisiologia , Animais , Canais de Cloreto/metabolismo , Colo/citologia , Colo/efeitos dos fármacos , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Ativação Enzimática , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Proteínas de Escherichia coli , Estradiol/fisiologia , Estrogênios/fisiologia , Feminino , Canal de Potássio KCNQ1/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Progesterona/farmacologia , Progesterona/fisiologia , Progestinas/farmacologia , Progestinas/fisiologia , Proteína Quinase C-delta/metabolismo , Ratos , Ratos Sprague-Dawley , Testosterona/farmacologia , Testosterona/fisiologia
10.
Mol Endocrinol ; 24(11): 2152-65, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20810711

RESUMO

Development of cardiac fibrosis portends the transition and deterioration from hypertrophy to dilation and heart failure. Here we examined how estrogen blocks this important development. Angiotensin II (AngII) and endothelin-1 induce cardiac hypertrophy and fibrosis in humans. and we find that these agents directly stimulate the transition of the cardiac fibroblast to a myofibroblast. AngII and endothelin-1 stimulated TGFß1 synthesis in the fibroblast, an inducer of fibrosis that signaled via c-jun kinase to Sma- and Mad-related protein 3 phosphorylation and nuclear translocation in myofibroblasts. As a result, mesenchymal proteins fibronectin and vimentin were produced, as were collagens I and III, the major forms found in fibrotic hearts. 17ß-Estradiol (E2) or dipropylnitrile, an estrogen receptor (ER)ß agonist, comparably blocked all these events, reversed by estrogen receptor (ER)ß small interfering RNA. E2 and dipropylnitrile signaling through cAMP and protein kinase A prevented myofibroblast formation and blocked activation of c-jun kinase and important events of fibrosis. In the hearts of ovariectomized female mice, cardiac hypertrophy and fibrosis were induced by AngII infusion and prevented by E2 administration to wild type but not ERß knockout rodents. Our results establish the cardiac fibroblast as an important target for hypertrophic/fibrosis-inducing peptides the actions of which were mitigated by E2/ERß acting in these stromal cells.


Assuntos
Receptor beta de Estrogênio/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Animais , AMP Cíclico/biossíntese , Estradiol/farmacologia , Feminino , Fibrose , Camundongos , Modelos Biológicos , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Biossíntese de Proteínas/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo
11.
Mol Endocrinol ; 23(11): 1885-99, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19846538

RESUMO

The secretion of Cl(-) across distal colonic crypt cells provides the driving force for the movement of fluid into the luminal space. 17beta-Estradiol (E2) produces a rapid and sustained reduction in secretion in females, which is dependent on the novel protein kinase C delta (PKC delta) isozyme and PKA isoform I targeting of KCNQ1 channels. This sexual dimorphism in the E2 response is associated with a higher expression level of PKC delta in female compared with the male tissue. The present study revealed the antisecretory response is regulated throughout the female reproductive (estrous) cycle and is primed by genomic regulation of the kinases. E2 (1-10 nm) decreased cAMP-dependent secretion in colonic epithelia during the estrus, metestrus, and diestrus stages. A weak inhibition of secretion was demonstrated in the proestrus stage. The expression levels of PKC delta and PKA fluctuated throughout the estrous cycle and correlated with the potency of the antisecretory effect of E2. The expression of PKC delta and PKA were up-regulated by estrogen at a transcriptional level via a PKC delta-MAPK-cAMP response element-binding protein-regulated pathway indicating a genomic priming of the antisecretory response. PK Cdelta was activated by the membrane-impermeant E2-BSA, and this response was inhibited by the estrogen receptor antagonist ICI 182,780. The 66-kDa estrogen receptor-alpha isoform was present at the plasma membrane of female colonic crypt cells with a lower abundance found in male colonic crypts. The study demonstrates estrogen regulation of intestinal secretion both at a rapid and transcriptional level, demonstrating an interdependent relationship between both nongenomic and genomic hormone responses.


Assuntos
Colo/metabolismo , Estrogênios/metabolismo , Ciclo Estral/metabolismo , Genômica , Animais , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Estradiol/metabolismo , Feminino , Sistema de Sinalização das MAP Quinases , Modelos Biológicos , Fosforilação , Isoformas de Proteínas , Proteína Quinase C-delta/metabolismo , RNA Mensageiro/metabolismo , Ratos , Transcrição Gênica
12.
J Physiol ; 587(Pt 21): 5039-44, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19723780

RESUMO

The intestine is an oestrogen responsive organ and circulatory oestrogens suppress Cl(-) secretion across the epithelium of the colon to promote fluid retention at the luteal stage of the menstrual cycle. Ion transporters in the colon which are involved in Cl(-) secretion show differential expression between males and females as do the signalling protein kinase intermediates involved in acutely regulating these transporters. Work from our laboratory has identified the KCNQ1/KCNE3 channel as one of the principal targets for oestrogen-induced signalling cascades in the distal colon. Through inhibition of the KCNQ1 channel, basolateral K(+) recycling is decreased so reducing the favourable electrochemical gradient for Cl(-) extrusion at the apical membrane. The actions of oestrogen on non-reproductive tissues such as the colon, kidney, lung and sweat gland will affect whole body electrolyte and fluid homeostasis and also have consequences for reproductive potential.


Assuntos
Cloro/metabolismo , Estrogênios/metabolismo , Intestinos/fisiologia , Canais Iônicos/fisiologia , Ciclo Menstrual/fisiologia , Potássio/metabolismo , Transdução de Sinais/fisiologia , Animais , Feminino , Humanos , Fatores Sexuais
13.
J Biol Chem ; 284(6): 3488-95, 2009 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-19054762

RESUMO

Estrogen receptors (ERs) alpha and beta exist as nuclear, cytoplasmic, and membrane cellular pools in a wide variety of organs. The relative contributions of each ERalpha pool to in vivo phenotypes resulting from estrogen signaling have not been determined. To address this, we generated a transgenic mouse expressing only a functional E domain of ERalpha at the plasma membrane (MOER). Cells isolated from many organs showed membrane only localized E domain of ERalpha and no other receptor pools. Liver cells from MOER and wild type mice responded to 17-beta-estradiol (E2) with comparable activation of ERK and phosphatidylinositol 3-kinase, not seen in cells from ERalphaKO mice. Mating the MOER female mice with proven male wild type breeders produced no pregnancies because the uterus and vagina of the MOER female mice were extremely atrophic. Ovaries of MOER and homozygous Strasbourg ERalphaKO mice showed multiple hemorrhagic cysts and no corpus luteum, and the mammary gland development in both MOER and ERalphaKO mice was rudimentary. Despite elevated serum E2 levels, serum LH was not suppressed, and prolactin levels were low in MOER mice. MOER and Strasbourg female mice showed plentiful abdominal visceral and other depots of fat and increased body weight compared to wild type mice despite comparable food consumption. These results provide strong evidence that the normal development and adult functions of important organs in female mice requires nuclear ERalpha and is not rescued by membrane ERalpha domain expression alone.


Assuntos
Membrana Celular/metabolismo , Receptor alfa de Estrogênio/metabolismo , Fenótipo , Animais , Atrofia , Peso Corporal/genética , Membrana Celular/genética , Membrana Celular/patologia , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Corpo Lúteo/metabolismo , Corpo Lúteo/patologia , Citoplasma/genética , Citoplasma/metabolismo , Citoplasma/patologia , Ingestão de Alimentos/genética , Estradiol/sangue , Receptor alfa de Estrogênio/genética , Feminino , Infertilidade Feminina/metabolismo , Infertilidade Feminina/patologia , Masculino , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Camundongos , Camundongos Knockout , Estrutura Terciária de Proteína/genética , Transdução de Sinais/genética , Útero/crescimento & desenvolvimento , Útero/metabolismo , Útero/patologia , Vagina/crescimento & desenvolvimento , Vagina/metabolismo , Vagina/patologia
14.
Steroids ; 73(9-10): 889-94, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18374379

RESUMO

Previous studies from our laboratory demonstrated that 17beta-estradiol (E2) rapidly inhibits Cl(-) secretion in rat and human distal colonic epithelium. The inhibition has been shown to occur via targeting of a basolateral K(+) channel identified as the KCNQ1 (KvLQT1) channel. E2 indirectly modulates the channel activity via a cascade of second messengers which are rapidly phosphorylated in response to E2. The anti-secretory mechanism may be the manner by which E2 induces fluid retention in the intestine during periods of high circulating plasma E2. Here we review the sex-dependent and estrous cycle regulation of this novel rapid response to E2. The inhibition of KCNQ1 channel activity and Cl(-) secretion will be of interest in the future in the investigation of the retentive effects of estrogen in female tissue and also in the study of secretory disorders and drugable targets of the intestine.


Assuntos
Colo/citologia , Estradiol/metabolismo , Transdução de Sinais , Animais , Colo/metabolismo , Estrogênios/metabolismo , Ciclo Estral , Feminino , Humanos , Mucosa Intestinal/metabolismo , Canal de Potássio KCNQ1/metabolismo , Masculino , Modelos Biológicos , Canais de Potássio/metabolismo , Ratos , Fatores Sexuais
15.
Am J Physiol Cell Physiol ; 294(6): C1362-70, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18400987

RESUMO

Alterations in EGF receptor (EGFR) signaling occur in intestinal disorders associated with dysregulated epithelial transport. In the present study, we investigated a role for the EGFR in the chronic regulation of intestinal epithelial secretory function. Epithelial Cl(-) secretion was measured as changes in short-circuit current (Isc) across voltage-clamped monolayers of T84 cells in Ussing chambers. Acute treatment of T84 cells with EGF (100 ng/ml, 15 min) chronically enhanced Isc responses to a broad range of secretagogues. This effect was apparent within 3 h, maximal by 6 h, and sustained for 24 h after treatment with EGF. The Na+/K+/2Cl(-) cotransporter (NKCC1) inhibitor bumetanide (100 microM) abolished the effect of EGF, indicating increased responses are due to potentiated Cl(-) secretion. Neither basal nor agonist-stimulated levels of intracellular Ca2+ or PKA activity were altered by EGF, implying that the effects of the growth factor are not due to chronic alterations in levels of second messengers. EGF increased the expression of NKCC1 with a time course similar to that of its effects on Cl(-) secretion. This effect of EGF was maximal after 6 h, at which time NKCC1 expression in EGF-treated cells was 199.9 +/- 21.9% of that in control cells (n = 21, P < 0.005). EGF-induced NKCC1 expression was abolished by actinomycin D, and RT-PCR analysis demonstrated EGF increased expression of NKCC1 mRNA. These data increase our understanding of mechanisms regulating intestinal fluid and electrolyte transport and reveal a novel role for the EGFR in the chronic regulation of epithelial secretory capacity through upregulation of NKCC1 expression.


Assuntos
Cloretos/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Mucosa Intestinal/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Bumetanida/farmacologia , Cálcio/metabolismo , Linhagem Celular , Colecistocinina/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dactinomicina/farmacologia , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/enzimologia , Potenciais da Membrana , Neuregulina-1/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Simportadores de Cloreto de Sódio-Potássio/genética , Membro 2 da Família 12 de Carreador de Soluto , Fatores de Tempo , Fator de Crescimento Transformador alfa/metabolismo , Regulação para Cima
16.
J Biol Chem ; 282(34): 24563-73, 2007 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-17556370

RESUMO

The estrogen sex steroid 17beta-estradiol rapidly inhibits secretagogue-stimulated cAMP-dependent Cl(-) secretion in the female rat distal colonic crypt by the inhibition of basolateral K(+) channels. In Ussing chamber studies, both the anti-secretory response and inhibition of basolateral K(+) current was shown to be attenuated by pretreatment with rottlerin, a PKCdelta-specific inhibitor. In whole cell patch-clamp analysis, 17beta-estradiol inhibited a chromanol 293B-sensitive KCNQ1 channel current in isolated female rat distal colonic crypts. Estrogen had no effect on KCNQ1 channel currents in colonic crypts isolated from male rats. Female distal colonic crypts expressed a significantly higher amount of PKCdelta in comparison to male tissue. PKCdelta and PKA were activated at 5 min in response to 17beta-estradiol in female distal colonic crypts only. Both PKCdelta- and PKA-associated with the KCNQ1 channel in response to 17beta-estradiol in female distal colonic crypts, and no associations were observed in crypts from males. PKA activation, association with KCNQ1, and phosphorylation of the channel were regulated by PKCdelta as the responses were blocked by pretreatment with rottlerin. Taken together, our experiments have identified the molecular targets underlying the anti-secretory response to estrogen involving the inhibition of KCNQ1 channel activity via PKCdelta- and PKA-dependent signaling pathways. This is a novel gender-specific mechanism of regulation of an ion channel by estrogen. The anti-secretory response described in this study provides molecular insights whereby estrogen causes fluid retention effects in the female during periods of high circulating plasma estrogen levels.


Assuntos
Colo/metabolismo , Estradiol/metabolismo , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/fisiologia , Animais , Transporte Biológico , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Estrogênios/sangue , Estrogênios/metabolismo , Feminino , Masculino , Modelos Biológicos , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores Sexuais
17.
Carcinogenesis ; 26(11): 1856-67, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15958521

RESUMO

The incidence of melanoma is increasing rapidly, with advanced lesions generally failing to respond to conventional chemotherapy. Here, we utilized DNA microarray-based gene expression profiling techniques to identify molecular determinants of melanoma progression within a unique panel of isogenic human melanoma cell lines. When a poorly tumorigenic cell line, derived from an early melanoma, was compared with two increasingly aggressive derivative cell lines, the expression of 66 genes was significantly changed. A similar pattern of differential gene expression was found with an independently derived metastatic cell line. We further examined these melanoma progression-associated genes via use of a tailored TaqMan Low Density Array (LDA), representing the majority of genes within our cohort of interest. Considerable concordance was seen between the transcriptomic profiles determined by DNA microarray and TaqMan LDA approaches. A range of novel markers were identified that correlated here with melanoma progression. Most notable was TSPY, a Y chromosome-specific gene that displayed extensive down-regulation in expression between the parental and derivative cell lines. Examination of a putative CpG island within the TSPY gene demonstrated that this region was hypermethylated in the derivative cell lines, as well as metastatic melanomas from male patients. Moreover, treatment of the derivative cell lines with the DNA methyltransferase inhibitor, 2'-deoxy-5-azacytidine (DAC), restored expression of the TSPY gene to levels comparable with that found in the parental cells. Additional DNA microarray studies uncovered a subset of 13 genes from the above-mentioned 66 gene cohort that displayed re-activation of expression following DAC treatment, including TSPY, CYBA and MT2A. DAC suppressed tumor cell growth in vitro. Moreover, systemic treatment of mice with DAC attenuated growth of melanoma xenografts, with consequent re-expression of TSPY mRNA. Overall, our data support the hypothesis that multiple genes are targeted, either directly or indirectly, by DNA hypermethylation during melanoma progression.


Assuntos
Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Melanoma Experimental/genética , Neoplasias Cutâneas/genética , Animais , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Azacitidina/uso terapêutico , Biomarcadores Tumorais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Metilases de Modificação do DNA/antagonistas & inibidores , Metilases de Modificação do DNA/genética , Metilases de Modificação do DNA/metabolismo , Decitabina , Progressão da Doença , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Epigênese Genética , Feminino , Perfilação da Expressão Gênica , Humanos , Melanoma Experimental/metabolismo , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/metabolismo , RNA Neoplásico/metabolismo , Neoplasias Cutâneas/prevenção & controle , Neoplasias Cutâneas/secundário , Transplante Heterólogo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...