Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Curr HIV Res ; 19(1): 61-72, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32860360

RESUMO

BACKGROUND: Vitamin D deficiency is common in HIV population and has been associated with increased comorbidity risk and poor immunologic status. OBJECTIVE: To evaluate the effect of protease inhibitor lopinavir/ritonavir monotherapy on changes in serum 25-hydroxyvitamin D [25(OH)D] over 48 weeks. METHODS: Thirty-four treatment-naïve HIV individuals initiating lopinavir/ritonavir monotherapy and receiving clinical care from private practice in Houston, Texas, were included. Serum 25-hydroxyvitamin D levels from stored plasma samples collected from IMANI-2 pilot study at both baseline and 48 weeks were analyzed using LC-MS assays. Mean 25(OH)D at baseline and 48 weeks were compared using paired t-tests. Linear regression analysis was used to evaluate factors associated with changes in 25(OH)D. Logistic regression analyses were used to determine the effect of vitamin D status and covariates on CD4 cell count recovery. RESULTS: Mean 25(OH)D was significantly higher at 48 weeks (26.3 ng/mL (SD + 14.9); p=0.0003) compared to baseline (19.8 ng/mL (SD +12.1), with fewer individuals having vitamin D deficiency (41.2%) and severe deficiency (11.8%). Both body mass index and baseline CD4 cell count were significant independent covariates associated with 25(OH)D changes over 48 weeks. Baseline vitamin D status did not affect CD4 cell count recovery. However, in a 24-week multivariate analysis, current tobacco use was significantly associated with a decreased odds of CD4 cell count recovery (AOR 0.106, 95% CI 0.018-0.606; p=0.012). CONCLUSION: Individuals treated with lopinavir/ritonavir monotherapy had significantly higher 25(OH)D after 48 weeks. Current tobacco users had significantly diminished CD4 cell count recovery after starting treatment, warranting further clinical investigation.


Assuntos
Fármacos Anti-HIV/uso terapêutico , Infecções por HIV/tratamento farmacológico , Lopinavir/uso terapêutico , Inibidores de Proteases/efeitos adversos , Ritonavir/uso terapêutico , Deficiência de Vitamina D/sangue , Deficiência de Vitamina D/induzido quimicamente , Adulto , Fármacos Anti-HIV/efeitos adversos , Terapia Antirretroviral de Alta Atividade/efeitos adversos , Feminino , Humanos , Lopinavir/efeitos adversos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Inibidores de Proteases/uso terapêutico , Ritonavir/efeitos adversos , Texas , Fatores de Tempo
4.
Sci Rep ; 6: 34710, 2016 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-27725717

RESUMO

Preterm labor caused by uterine contractions is a major contributor to neonatal morbidity and mortality. Treatment intended to reduce uterine contractions include tocolytic agents, such as indomethacin. Unfortunately, clinically used tocolytics are frequently inefficient and cross the placenta causing fetal side effects. Here we show for the first time in obstetrics the use of a targeted nanoparticle directed to the pregnant uterus and loaded with a tocolytic for reducing its placental passage and sustaining its efficacy. Nanoliposomes encapsulating indomethacin and decorated with clinically used oxytocin receptor antagonist were designed and evaluated in-vitro, ex-vivo and in-vivo. The proposed approach resulted in targeting uterine cells in-vitro, inhibiting uterine contractions ex-vivo, while doubling uterine drug concentration, decreasing fetal levels, and maintaining the preterm birth rate in vivo in a pregnant mouse model. This promising approach opens new horizons for drug development in obstetrics that could greatly impact preterm birth, which currently has no successful treatments.


Assuntos
Indometacina/farmacologia , Lipossomos/administração & dosagem , Terapia de Alvo Molecular/métodos , Nanoestruturas/administração & dosagem , Trabalho de Parto Prematuro/prevenção & controle , Nascimento Prematuro/prevenção & controle , Tocolíticos/farmacologia , Útero/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Composição de Medicamentos , Feminino , Expressão Gênica , Antagonistas de Hormônios/química , Antagonistas de Hormônios/metabolismo , Humanos , Indometacina/farmacocinética , Lipossomos/química , Camundongos , Nanoestruturas/química , Placenta/metabolismo , Gravidez , Ligação Proteica , Receptores de Ocitocina/metabolismo , Tocolíticos/farmacocinética , Contração Uterina/efeitos dos fármacos , Útero/metabolismo , Vasotocina/análogos & derivados , Vasotocina/química , Vasotocina/metabolismo
5.
JCI Insight ; 1(17): e87754, 2016 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-27777972

RESUMO

Current antiangiogenesis therapy relies on inhibiting newly developed immature tumor blood vessels and starving tumor cells. This strategy has shown transient and modest efficacy. Here, we report a better approach to target cancer-associated endothelial cells (ECs), reverse permeability and leakiness of tumor blood vessels, and improve delivery of chemotherapeutic agents to the tumor. First, we identified deregulated microRNAs (miRs) from patient-derived cancer-associated ECs. Silencing these miRs led to decreased vascular permeability and increased maturation of blood vessels. Next, we screened a thioaptamer (TA) library to identify TAs selective for tumor-associated ECs. An annexin A2-targeted TA was identified and used for delivery of miR106b-5p and miR30c-5p inhibitors, resulting in vascular maturation and antitumor effects without inducing hypoxia. These findings could have implications for improving vascular-targeted therapy.


Assuntos
Antineoplásicos/farmacologia , Aptâmeros de Nucleotídeos , Células Endoteliais/citologia , MicroRNAs/administração & dosagem , Neovascularização Patológica/prevenção & controle , Linhagem Celular Tumoral , Humanos , Nanopartículas , Neoplasias/irrigação sanguínea , Neoplasias/terapia , Transfecção
6.
Methods Mol Biol ; 1204: 123-33, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25182766

RESUMO

We developed fixed-cell multi-well plate immunoassays that increase the throughput and ease of quantification for questions formerly assessed by immunoblot scanning. The assays make use of the now abundant antibodies designed to recognize receptor subtypes and posttranslationally modified signaling proteins. By optimizing permeabilization and fixation conditions, mainly based on specific cell types, the assay can be adapted to the study of many different antigens of importance to hormonal and neurotransmitter signaling scenarios.


Assuntos
Imunoensaio/métodos , Sistema de Sinalização das MAP Quinases , Receptores de Estrogênio/análise , Animais , Linhagem Celular , Células Cultivadas , Ativação Enzimática , Ensaios Enzimáticos/métodos , Humanos
7.
Am J Obstet Gynecol ; 208(5): 388.e1-388.e10, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23497796

RESUMO

OBJECTIVE: Preeclampsia is associated with long-term adverse maternal health, such as cardiovascular and metabolic diseases. The objective of this study was to determine whether preeclampsia in a well-characterized animal model that was induced by overexpression of soluble fms-like tyrosine kinase-1 (sFlt1) results in alterations in the maternal circulating proteome that persist long after delivery. STUDY DESIGN: CD-1 mice at day 8 of gestation were injected with adenovirus that carried sFlt1 or the murine immunoglobulin G2α Fc fragment as control. Depleted maternal plasma was analyzed 6 months after delivery by label-free liquid chromatography-mass spectrometry assay. The tandem mass spectrometry data were searched against a mouse database, and the resultant intensity data were used to compare abundance of proteins across disease/control plasma pool. Results were analyzed with ingenuity pathways analysis. Right-tailed Fisher exact test was used to calculate a probability value. RESULTS: Of 150 proteins that are common for both groups, ingenuity pathways analysis determined 105 proteins that were ready for analysis. Diseases and disorders analysis showed significant enrichment of proteins that are associated with cardiovascular disease. Within this cluster, the most abundant proteins were associated with vascular disease, atherosclerosis, and atherosclerotic lesions. Other top disease clusters were inflammatory response, organismal injury and abnormalities, and hematologic and metabolic disease. CONCLUSION: Exposure to sFlt1-induced preeclampsia alters multiple biologic functions in mothers that persist later in life. Our results suggest that some of the long-term adverse outcomes that are associated with preeclampsia actually may be a consequence rather than a mere unmasking of an underlying predisposition. If similar results are found in humans, the development of preventive strategies for preeclampsia should also improve long-term maternal health.


Assuntos
Biomarcadores/sangue , Doenças Cardiovasculares/etiologia , Pré-Eclâmpsia/sangue , Proteoma/metabolismo , Animais , Doenças Cardiovasculares/sangue , Cromatografia Líquida , Modelos Animais de Doenças , Feminino , Espectrometria de Massas , Camundongos , Pré-Eclâmpsia/etiologia , Gravidez , Distribuição Aleatória , Receptor 1 de Fatores de Crescimento do Endotélio Vascular
8.
Steroids ; 77(5): 424-32, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22230296

RESUMO

UNLABELLED: Multiple physiologic estrogens (estradiol, estriol, and estrone), as well as xenoestrogenic compounds (including alkylphenols and bisphenol A), can act via nongenomic signaling initiated by liganding of the plasma membrane estrogen receptor-α (mERα). We examined heterotrimeric G protein involvement leading to extracellular-regulated kinase (ERK) activation in GH3/B6/F10 rat anterior pituitary tumor cells that express abundant mERα, and smaller amounts of mERß and GPR30. A combination of microarrays, immunoblots, and quantitative immunoassays demonstrated the expression of members of all α, ß, and γ G protein classes in these cells. Use of selective inhibitors showed that the G(αi) subtype was the primary initiator of downstream ERK signaling. Using antibodies against the GTP-bound form of G(α) protein subtypes i and s, we showed that xenoestrogens (bisphenol A, nonylphenol) activated G(αi) at 15-30s; all alkylphenols examined subsequently suppressed activation by 5min. GTP-activation of G(αi) for all estrogens was enhanced by irreversible cumulative binding to GTPγS. In contrast, G(αs) was neither activated nor deactivated by these treatments with estrogens. ERα and G(αi) co-localized outside nuclei and could be immuno-captured together. Interactions of ERα with G(αi) and caveolin I were demonstrated by epitope proximity ligation assays. An ERα/ß antagonist (ICI182780) and a selective disruptor of caveolar structures (nystatin) blocked estrogen-induced ERK activation. CONCLUSIONS: Xenoestrogens, like physiologic estrogens, can evoke downstream kinase signaling involving selective interactions of ERα with G(αi) and caveolin I, but with some different characteristics, which could explain their disruptive actions.


Assuntos
Caveolina 1/metabolismo , Receptor alfa de Estrogênio/metabolismo , Estrogênios/farmacologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fenóis/farmacologia , Animais , Compostos Benzidrílicos , Cavéolas/metabolismo , Caveolina 1/genética , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Receptor alfa de Estrogênio/genética , Estrogênios não Esteroides/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Guanosina Trifosfato/metabolismo , Immunoblotting , Microscopia de Fluorescência , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/metabolismo , Neoplasias Hipofisárias/patologia , Ligação Proteica/efeitos dos fármacos , Ratos
9.
Steroids ; 72(2): 124-34, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17174995

RESUMO

Studies of the nuclear transcriptional regulatory activities of non-physiological estrogens have not explained their actions in mediating endocrine disruption in animals and humans at the low concentrations widespread in the environment. However, xenoestrogens have rarely been tested for their ability to participate in the plethora of nongenomic steroid signaling pathways elucidated over the last several years. Here we review what is known about such responses in comparison to our recent evidence that xenoestrogens can rapidly and potently elicit signaling through nongenomic pathways culminating in functional endpoints. Both estradiol (E(2)) and compounds representing various classes of xenoestrogens (diethylstilbestrol, coumestrol, bisphenol A, DDE, nonylphenol, endosulfan, and dieldrin) act via a membrane version of the estrogen receptor-alpha on pituitary cells, and can provoke Ca(2+) influx via L-type channels, leading to prolactin (PRL) secretion. These hormones and mimetics can also cause the oscillating activation of extracellular regulated kinases (ERKs). However, individual estrogen mimetics differ in their potency and temporal phasing of these activations compared to each other and to E(2). It is perhaps in these ways that they disrupt some endocrine functions when acting in combination with physiological estrogens. Our quantitative assays allow comparison of these outcomes for each mimetic, and let us build a detailed picture of alternative signaling pathway usage. Such an understanding should allow us to determine the estrogenic or antiestrogenic potential of different types of xenoestrogens, and help us to develop strategies for preventing xenoestrogenic disruption of estrogen action in many tissues.


Assuntos
Estrogênios não Esteroides/farmacologia , Genoma/fisiologia , Animais , Cálcio/metabolismo , Cálcio/fisiologia , Linhagem Celular Tumoral , Mimetismo Molecular , Prolactina/biossíntese , Prolactina/metabolismo , Prolactina/fisiologia , Ratos , Transdução de Sinais/fisiologia
10.
Steroids ; 70(5-7): 364-71, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15862819

RESUMO

Estrogen mimetics in the environment and in foods can have important consequences for endocrine functions. When previously examined for action via genomic steroid signaling mechanisms, most of these compounds were found to be very weak agonists. We have instead tested their actions via several membrane-initiated signaling mechanisms in GH3/B6 pituitary tumor cells extensively selected for high (responsive) or low (nonresponsive) expression of the membrane version of estrogen receptor-alpha (mERalpha). We found many estrogen mimetic compounds to be potently active in our quantitative extracellular-regulated kinase (ERK) activation assays, to increase cellular Ca++ levels, and to cause rapid prolactin release. However, these compounds may activate one or both mechanisms with different potencies. For instance, some compounds activate ERKs in both pM and nM concentration ranges, while others are only active at nM and higher concentrations. Compounds also show great differences in their temporal activation patterns. While estradiol causes a bimodal time-dependent ERK activation (peaking at both 3 and 30 min), most estrogen mimetics cause either an early phase activation, a late phase activation, or an early sustained activation. One xenoestrogen known to be a relatively potent activator of estrogen response element-mediated actions (bisphenol A) is inactive as an ERK activator, and only a modest inducer of Ca++ levels and prolactin release. Many different signaling machineries culminate in ERK activation, and xenoestrogens differentially affect various pathways. Clearly individual xenoestrogens must be individually investigated for their differing abilities to activate distinct membrane-initiated signal cascades that lead to a variety of cellular functions.


Assuntos
Membrana Celular/metabolismo , Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Fitoestrógenos/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Violeta Genciana/farmacologia , Microscopia Confocal , Prolactina/metabolismo , Radioimunoensaio , Ratos , Receptores de Estrogênio/metabolismo , Transdução de Sinais , Fatores de Tempo
11.
Environ Health Perspect ; 113(4): 431-9, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15811834

RESUMO

Xenoestrogens (XEs) are widespread in our environment and are known to have deleterious effects in animal (and perhaps human) populations. Acting as inappropriate estrogens, XEs are thought to interfere with endogenous estrogens such as estradiol (E2) to disrupt normal estrogenic signaling. We investigated the effects of E2 versus several XEs representing organochlorine pesticides (dieldrin, endosulfan, o',p'-dichlorodiphenylethylene), plastics manufacturing by-products/detergents (nonylphenol, bisphenol A), a phytoestrogen (coumestrol), and a synthetic estrogen (diethylstilbestrol) on the pituitary tumor cell subline GH3/B6/F10, previously selected for expression of high levels of membrane estrogen receptor-alpha. Picomolar to nanomolar concentrations of both E2 and XEs caused intracellular Ca2+ changes within 30 sec of administration. Each XE produced a unique temporal pattern of Ca2+ elevation. Removing Ca2+ from the extracellular solution abolished both spontaneous and XE-induced intracellular Ca2+ changes, as did 10 microM nifedipine. This suggests that XEs mediate their actions via voltage-dependent L-type Ca2+ channels in the plasma membrane. None of the Ca2+ fluxes came from intracellular Ca2+ stores. E2 and each XE also caused unique time- and concentration-dependent patterns of prolactin (PRL) secretion that were largely complete within 3 min of administration. PRL secretion was also blocked by nifedipine, demonstrating a correlation between Ca2+ influx and PRL secretion. These data indicate that at very low concentrations, XEs mediate membrane-initiated intracellular CCa2+ increases resulting in PRL secretion via a mechanism similar to that for E2, but with distinct patterns and potencies that could explain their abilities to disrupt endocrine functions.


Assuntos
Cálcio/metabolismo , Receptor alfa de Estrogênio/metabolismo , Estrogênios/toxicidade , Prolactina/metabolismo , Animais , Compostos Benzidrílicos , Linhagem Celular Tumoral , Cumestrol/toxicidade , Diclorodifenil Dicloroetileno/toxicidade , Dieldrin , Dietilestilbestrol/toxicidade , Endossulfano/toxicidade , Estradiol/toxicidade , Fenóis/toxicidade , Ratos , Transdução de Sinais/efeitos dos fármacos , Xenobióticos/toxicidade
12.
Am J Physiol Endocrinol Metab ; 288(2): E388-97, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15494610

RESUMO

The role of membrane estrogen receptor-alpha (mERalpha) in rapid nongenomic responses to 17beta-estradiol (E(2)) was tested in sublines of GH3/B6 rat prolactinoma cells selected for high (GH3/B6/F10) and low (GH3/B6/D9) mERalpha expression. E(2) elicited rapid, concentration-dependent intracellular Ca(2+) concentration ([Ca(2+)](i)) increases in the F10 subline. Lack of inhibition by thapsigargin depletion of intracellular Ca(2+) pools, together with abrogation of the response in Ca(2+)-free medium, suggested an extracellular source of Ca(2+) for this response. The participation of voltage-dependent channels in the E(2)-induced [Ca(2+)](i) increase was confirmed by the specific L-type Ca(2+) channel inhibitor nifedipine. For comparison, the D9 mERalpha-depleted subline was insensitive to steroid action via this signaling mechanism. [Ca(2+)](i) elevation was correlated with prolactin (PRL) release in the F10 cell line in as little as 3 min. E(2) caused a much higher PRL release than KCl treatment (which caused maximal Ca(2+) elevation), suggesting that secretion was also controlled by additional mechanisms. Participation of mERalpha in these effects was confirmed by the ability of E(2)-peroxidase (a cell-impermeable analog of E(2)) to cause these responses, blockage of the responses with the ER antagonist ICI 182 780, and the inability of the E(2) stereoisomer 17alpha-E(2) to elicit a response. Thus rapid exocytosis of PRL is regulated in these cells by mERalpha signaling to specific Ca(2+) channels utilizing extracellular Ca(2+) sources and additional signaling mechanisms.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Membrana Celular/metabolismo , Receptor alfa de Estrogênio/metabolismo , Neoplasias Hipofisárias/metabolismo , Prolactina/farmacocinética , Prolactinoma/metabolismo , Animais , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Estradiol/farmacologia , Nifedipino/farmacologia , Ratos
13.
Environ Health Perspect ; 112(15): 1481-7, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15531431

RESUMO

Xenoestrogens can mimic or antagonize the activity of physiological estrogens, and the suggested mechanism of xenoestrogen action involves binding to estrogen receptors (ERs). However, the failure of various in vitro or in vivo assays to show strong genomic activity of xenoestrogens compared with estradiol (E2) makes it difficult to explain their ability to cause abnormalities in animal (and perhaps human) reproductive functions via this pathway of steroid action. E2 has also been shown to initiate rapid intracellular signaling, such as changes in levels of intracellular calcium, cAMP, and nitric oxide, and activations of a variety of kinases, via action at the membrane. In this study, we demonstrate that several xenoestrogens can rapidly activate extracellular-regulated kinases (ERKs) in the pituitary tumor cell line GH3/B6/F10, which expresses high levels of the membrane receptor for ER-alpha (mER). We tested a phytoestrogen (coumestrol), organochlorine pesticides or their metabolites (endosulfan, dieldrin, and DDE), and detergent by-products of plastics manufacturing (p-nonylphenol and bisphenol A). These xenoestrogens (except bisphenolA) produced rapid (3-30 min after application), concentration (10(-14)-10(-8) M)-dependent ERK-1/2 phosphorylation but with distinctly different activation patterns. To identify signaling pathways involved in ERK activation, we used specific inhibitors of ERs, epidermal growth factor receptors, Ca2+ signaling, Src and phosphoinositide-3 kinases, and a membrane structure disruption agent. Multiple inhibitors blocked ERK activation, suggesting simultaneous use of multiple pathways and complex signaling web interactions. However, inhibitors differentially affected each xenoestrogen response examined. These actions may help to explain the distinct abilities of xenoestrogens to disrupt reproductive functions at low concentrations.


Assuntos
Estrogênios/toxicidade , MAP Quinase Quinase 2/farmacologia , Proteína Quinase 3 Ativada por Mitógeno/farmacologia , Transdução de Sinais , Xenobióticos/toxicidade , Animais , Técnicas de Cultura de Células , Relação Dose-Resposta a Droga , Estradiol/farmacologia , MAP Quinase Quinase 2/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Praguicidas/toxicidade , Fosforilação , Neoplasias Hipofisárias/patologia , Prolactinoma/patologia , Ratos , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/fisiologia
14.
Steroids ; 69(3): 181-92, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15072920

RESUMO

Estradiol (E2) and other steroids have recently been shown to initiate various intracellular signaling cascades from the plasma membrane, including those stimulating mitogen-activated protein kinases (MAPKs), and particularly extracellular-regulated kinases (ERKs). In this study we demonstrated the ability of E2 to activate ERKs in the GH3/B6/F10 pituitary tumor cell line, originally selected for its enhanced expression of membrane estrogen receptor-alpha (mERalpha). We compared E2 to its cell-impermeable analog (E2 conjugated to peroxidase, E2-P), and to the synthetic estrogen diethylstilbestrol (DES). Time-dependent ERK activation was quantified with a novel fixed cell-based immunoassay developed to efficiently determine activation by multiple compounds over multiple parameters. Both E2 and DES produced bimodal responses, but with distinctly different time courses of enzyme phosphorylation (activation) and inactivation; E2-P induced a monophasic ERK activation. E2 also phosphorylated ERKs in concentration-dependent manner with two concentration optima (10(-14) and 10(-8)M). Inhibitors were employed to determine pathway (ER, EGFR, membrane organization, PI3 kinase, Src kinase, Ca2+) involvement and timing of pathway activations; all affected ERK activation as early as 3-6 min, suggesting simultaneous, not sequential, activation. Therefore, E2 and other estrogenic compounds can produce rapid ERK phosphorylations via nongenomic pathways, using more than one pathway for signal generation.


Assuntos
Membrana Celular/efeitos dos fármacos , Ácido Egtázico/análogos & derivados , Estrogênios/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Animais , Antifúngicos/farmacologia , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Quelantes/farmacologia , Dietilestilbestrol/farmacologia , Ácido Egtázico/farmacologia , Ativação Enzimática/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Fator de Crescimento Epidérmico/farmacologia , Antagonistas de Estrogênios/farmacologia , Estrogênios/química , Flavonoides/farmacologia , Nistatina/farmacologia , Peroxidase/química , Peroxidase/farmacologia , Fosforilação/efeitos dos fármacos , Pirimidinas/farmacologia , Ratos
15.
Clin Sci (Lond) ; 105(1): 21-8, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12593669

RESUMO

Disruption of cellular acid-base status alters the host defence functions of alveolar macrophages (m phi). These pH effects might be mediated by pH-sensitive changes in the signalling pathways of the effector functions of m phi. The present study examined the effects of intracellular pH (pH(i)) on the free cytosolic calcium concentration ([Ca(2+)](i)), an important second messenger for cell functions. [Ca(2+)](i) and pH(i) of rabbit resident alveolar m phi were measured using fluorescent dyes. With extracellular pH (pH(o)) of 7.4, the steady-state pH(i) and [Ca(2+)](i) were approx. 7.14 and 123 nM respectively. Incubation at pH(o) 6.8 caused a sustained cytosolic acidosis, but did not affect [Ca(2+)](i). Likewise, [Ca(2+)](i) was unchanged when m phi at pH(o) 7.4 were acidified using bafilomycin A(1) or sodium propionate. In contrast, [Ca(2+)](i) was markedly sensitive to cytosolic alkalosis. Exposure to NH(4)Cl at pH(o) 7.4 caused transient increases in both pH(i) and [Ca(2+)](i). The Ca(2+) response was mediated by release of intracellular Ca(2+) from thapsigargin-sensitive stores and was potentiated by capacitative entry of extracellular Ca(2+). Incubation at high pH(o) values (>7.4) produced sustained increases in pH(i) and [Ca(2+)](i). The sustained elevation of [Ca(2+)](i) was consistent with pH-sensitive inhibition of plasma-membrane Ca(2+)-ATPase. The response to high pH(o) was unaffected by blockade of L-type or receptor-operated Ca(2+) channels with nifedipine or SKF-96365, and was independent of extracellular Na(+). The findings indicate that pH impacts cytosolic Ca(2+) homoeostasis at multiple levels. The data suggest that cellular acid-base status can influence Ca(2+)-dependent signalling events in resident alveolar m phi, especially during alkaline disruptions of pH(i).


Assuntos
Cálcio/metabolismo , Citosol/metabolismo , Macrolídeos , Macrófagos Alveolares/metabolismo , Sistemas do Segundo Mensageiro/fisiologia , Cloreto de Amônio/farmacologia , Animais , Antibacterianos/farmacologia , Células Cultivadas , Citosol/efeitos dos fármacos , Corantes Fluorescentes , Fura-2 , Concentração de Íons de Hidrogênio , Macrófagos Alveolares/efeitos dos fármacos , Propionatos/farmacologia , Coelhos , Espectrometria de Fluorescência , ATPases Vacuolares Próton-Translocadoras/antagonistas & inibidores
16.
FASEB J ; 16(14): 1917-27, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12468456

RESUMO

We used modified immunocytochemical conditions to quantify a membrane form of estrogen receptor-alpha (mERalpha) in a rat pituitary tumor cell line, GH3/B6/F10. We studied the regulation of mERalpha vs. levels of intracellular ERalpha (iERalpha) using our 96-well plate immunoassay. The anti-ERalpha antibody C542 was used to label the ERalpha (via conjugated alkaline phosphatase) in fixed permeabilized (for iERalpha) vs. nonpermeabilized cells (for mERalpha). Expression of mERalpha was highest at low cell densities (<1000 cells/well) and decreased significantly at densities where cellular processes touched, whereas the more abundant iERalpha increased with increasing cell density over the same range. Serum starvation for 48 h caused increases in mERalpha, whereas iERalpha levels showed no significant changes. A large decline in mERalpha and iERalpha levels with cell passage number was observed. Minutes after nM 17beta-estradiol (E2) treatment, a portion of the cells rounded up and detached from the culture plate, whereas nM cholesterol had no such effect. Although E2 treatment did not change mERalpha levels, the antigen was reorganized from a fine particulate to aggregation into asymmetric large granules of staining. That common culturing conditions favor down-regulation of mERalpha may explain the relatively few reports of this protein in other experimental systems.


Assuntos
Receptores de Superfície Celular/metabolismo , Receptores de Estrogênio/metabolismo , Animais , Adesão Celular/efeitos dos fármacos , Contagem de Células , Técnicas de Cultura de Células , Tamanho Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultura Livres de Soro , Estradiol/farmacologia , Receptor alfa de Estrogênio , Imuno-Histoquímica , Cinética , Ratos , Receptores de Superfície Celular/análise , Receptores de Estrogênio/análise , Receptores de Estrogênio/imunologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA