Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 4989, 2023 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-37591837

RESUMO

The estrogen receptor (ER) designated ERα has actions in many cell and tissue types that impact glucose homeostasis. It is unknown if these include mechanisms in endothelial cells, which have the potential to influence relative obesity, and processes in adipose tissue and skeletal muscle that impact glucose control. Here we show that independent of impact on events in adipose tissue, endothelial ERα promotes glucose tolerance by enhancing endothelial insulin transport to skeletal muscle. Endothelial ERα-deficient male mice are glucose intolerant and insulin resistant, and in females the antidiabetogenic actions of estradiol (E2) are absent. The glucose dysregulation is due to impaired skeletal muscle glucose disposal that results from attenuated muscle insulin delivery. Endothelial ERα activation stimulates insulin transcytosis by skeletal muscle microvascular endothelial cells. Mechanistically this involves nuclear ERα-dependent upregulation of vesicular trafficking regulator sorting nexin 5 (SNX5) expression, and PI3 kinase activation that drives plasma membrane recruitment of SNX5. Thus, coupled nuclear and non-nuclear actions of ERα promote endothelial insulin transport to skeletal muscle to foster normal glucose homeostasis.


Assuntos
Receptor alfa de Estrogênio , Insulina , Animais , Feminino , Masculino , Camundongos , Células Endoteliais , Glucose , Músculo Esquelético , Receptores de Estrogênio
4.
Circulation ; 140(24): 2005-2018, 2019 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-31597453

RESUMO

BACKGROUND: Obesity-related hypertension is a common disorder, and attempts to combat the underlying obesity are often unsuccessful. We previously revealed that mice globally deficient in the inhibitory immunoglobulin G (IgG) receptor FcγRIIB are protected from obesity-induced hypertension. However, how FcγRIIB participates is unknown. Studies were designed to determine if alterations in IgG contribute to the pathogenesis of obesity-induced hypertension. METHODS: Involvement of IgG was studied using IgG µ heavy chain-null mice deficient in mature B cells and by IgG transfer. Participation of FcγRIIB was interrogated in mice with global or endothelial cell-specific deletion of the receptor. Obesity was induced by high-fat diet (HFD), and blood pressure (BP) was measured by radiotelemetry or tail cuff. The relative sialylation of the Fc glycan on mouse IgG, which influences IgG activation of Fc receptors, was evaluated by Sambucus nigra lectin blotting. Effects of IgG on endothelial NO synthase were assessed in human aortic endothelial cells. IgG Fc glycan sialylation was interrogated in 3442 human participants by mass spectrometry, and the relationship between sialylation and BP was evaluated. Effects of normalizing IgG sialylation were determined in HFD-fed mice administered the sialic acid precursor N-acetyl-D-mannosamine (ManNAc). RESULTS: Mice deficient in B cells were protected from obesity-induced hypertension. Compared with IgG from control chow-fed mice, IgG from HFD-fed mice was hyposialylated, and it raised BP when transferred to recipients lacking IgG; the hypertensive response was absent if recipients were FcγRIIB-deficient. Neuraminidase-treated IgG lacking the Fc glycan terminal sialic acid also raised BP. In cultured endothelial cells, via FcγRIIB, IgG from HFD-fed mice and neuraminidase-treated IgG inhibited vascular endothelial growth factor activation of endothelial NO synthase by altering endothelial NO synthase phosphorylation. In humans, obesity was associated with lower IgG sialylation, and systolic BP was inversely related to IgG sialylation. Mice deficient in FcγRIIB in endothelium were protected from obesity-induced hypertension. Furthermore, in HFD-fed mice, ManNAc normalized IgG sialylation and prevented obesity-induced hypertension. CONCLUSIONS: Hyposialylated IgG and FcγRIIB in endothelium are critically involved in obesity-induced hypertension in mice, and supportive evidence was obtained in humans. Interventions targeting these mechanisms, such as ManNAc supplementation, may provide novel means to break the link between obesity and hypertension.


Assuntos
Hexosaminas/farmacologia , Hipertensão/tratamento farmacológico , Ácido N-Acetilneuramínico/metabolismo , Obesidade/tratamento farmacológico , Animais , Suplementos Nutricionais , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Hipertensão/metabolismo , Imunoglobulina G/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Receptores de IgG/metabolismo
5.
Nature ; 569(7757): 565-569, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31019307

RESUMO

Atherosclerosis, which underlies life-threatening cardiovascular disorders such as myocardial infarction and stroke1, is initiated by passage of low-density lipoprotein (LDL) cholesterol into the artery wall and its engulfment by macrophages, which leads to foam cell formation and lesion development2,3. It is unclear how circulating LDL enters the artery wall to instigate atherosclerosis. Here we show in mice that scavenger receptor class B type 1 (SR-B1) in endothelial cells mediates the delivery of LDL into arteries and its accumulation by artery wall macrophages, thereby promoting atherosclerosis. LDL particles are colocalized with SR-B1 in endothelial cell intracellular vesicles in vivo, and transcytosis of LDL across endothelial monolayers requires its direct binding to SR-B1 and an eight-amino-acid cytoplasmic domain of the receptor that recruits the guanine nucleotide exchange factor dedicator of cytokinesis 4 (DOCK4)4. DOCK4 promotes internalization of SR-B1 and transport of LDL by coupling the binding of LDL to SR-B1 with activation of RAC1. The expression of SR-B1 and DOCK4 is increased in atherosclerosis-prone regions of the mouse aorta before lesion formation, and in human atherosclerotic arteries when compared with normal arteries. These findings challenge the long-held concept that atherogenesis involves passive movement of LDL across a compromised endothelial barrier. Interventions that inhibit the endothelial delivery of LDL into artery walls may represent a new therapeutic category in the battle against cardiovascular disease.


Assuntos
Artérias/metabolismo , Aterosclerose/metabolismo , LDL-Colesterol/metabolismo , Células Endoteliais/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Receptores Depuradores Classe B/metabolismo , Transcitose , Animais , Aorta/citologia , Aorta/metabolismo , Aorta/patologia , Artérias/citologia , Artérias/patologia , Aterosclerose/patologia , Células Cultivadas , Feminino , Humanos , Macrófagos/metabolismo , Masculino , Camundongos , Neuropeptídeos/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
6.
Endocrinology ; 159(11): 3848-3859, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30256928

RESUMO

Estrogens provide neuroprotection in animal models of stroke, but uterotrophic effects and cancer risk limit translation. Classic estrogen receptors (ERs) serve as transcription factors, whereas nonnuclear ERs govern numerous cell processes and exert beneficial cardiometabolic effects without uterine or breast cancer growth in mice. Here, we determined how nonnuclear ER stimulation with pathway-preferential estrogen (PaPE)-1 affects stroke outcome in mice. Ovariectomized female mice received vehicle, estradiol (E2), or PaPE-1 before and after transient middle cerebral artery occlusion (tMCAo). Lesion severity was assessed with MRI, and poststroke motor function was evaluated through 2 weeks after tMCAo. Circulating, spleen, and brain leukocyte subpopulations were quantified 3 days after tMCAo by flow cytometry, and neurogenesis and angiogenesis were evaluated histologically 2 weeks after tMCAo. Compared with vehicle, E2 and PaPE-1 reduced infarct volumes at 3 days after tMCAo, though only PaPE-1 reduced leukocyte infiltration into the ischemic brain. Unlike E2, PaPE-1 had no uterotrophic effect. Both interventions had negligible effect on long-term poststroke neuronal or vascular plasticity. All mice displayed a decline in motor performance at 2 days after tMCAo, and vehicle-treated mice did not improve thereafter. In contrast, E2 and PaPE-1 treatment afforded functional recovery at 6 days after tMCAo and beyond. Thus, the selective activation of nonnuclear ER by PaPE-1 decreased stroke severity and improved functional recovery in mice without undesirable uterotrophic effects. The beneficial effects of PaPE-1 are also associated with attenuated neuroinflammation in the brain. PaPE-1 and similar molecules may warrant consideration as efficacious ER modulators providing neuroprotection without detrimental effects on the uterus or cancer risk.


Assuntos
Estradiol/farmacologia , Estrogênios/farmacologia , Infarto da Artéria Cerebral Média/fisiopatologia , Desempenho Psicomotor/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Recuperação de Função Fisiológica , Animais , Comportamento Animal/efeitos dos fármacos , Feminino , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Camundongos , Plasticidade Neuronal , Ovariectomia , Índice de Gravidade de Doença , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Útero/efeitos dos fármacos
7.
Blood ; 131(19): 2097-2110, 2018 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-29500169

RESUMO

In the antiphospholipid syndrome (APS), antiphospholipid antibody (aPL) recognition of ß2 glycoprotein I promotes thrombosis, and preclinical studies indicate that this is due to endothelial nitric oxide synthase (eNOS) antagonism via apolipoprotein E receptor 2 (apoER2)-dependent processes. How apoER2 molecularly links these events is unknown. Here, we show that, in endothelial cells, the apoER2 cytoplasmic tail serves as a scaffold for aPL-induced assembly and activation of the heterotrimeric protein phosphatase 2A (PP2A). Disabled-2 (Dab2) recruitment to the apoER2 NPXY motif promotes the activating L309 methylation of the PP2A catalytic subunit by leucine methyl transferase-1. Concurrently, Src homology domain-containing transforming protein 1 (SHC1) recruits the PP2A scaffolding subunit to the proline-rich apoER2 C terminus along with 2 distinct regulatory PP2A subunits that mediate inhibitory dephosphorylation of Akt and eNOS. In mice, the coupling of these processes in endothelium is demonstrated to underlie aPL-invoked thrombosis. By elucidating these intricacies in the pathogenesis of APS-related thrombosis, numerous potential new therapeutic targets have been identified.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Anticorpos Antifosfolipídeos/imunologia , Autoanticorpos/imunologia , Endotélio/metabolismo , Proteínas Relacionadas a Receptor de LDL/metabolismo , Proteína Fosfatase 2/metabolismo , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Células Endoteliais/metabolismo , Endotélio/imunologia , Endotélio Vascular/metabolismo , Humanos , Masculino , Camundongos , Modelos Biológicos , Complexos Multiproteicos , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/metabolismo , Trombose/etiologia , Trombose/metabolismo , Trombose/patologia
8.
J Clin Invest ; 128(1): 309-322, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29202472

RESUMO

Type 2 diabetes mellitus (T2DM) is a common complication of obesity. Here, we have shown that activation of the IgG receptor FcγRIIB in endothelium by hyposialylated IgG plays an important role in obesity-induced insulin resistance. Despite becoming obese on a high-fat diet (HFD), mice lacking FcγRIIB globally or selectively in endothelium were protected from insulin resistance as a result of the preservation of insulin delivery to skeletal muscle and resulting maintenance of muscle glucose disposal. IgG transfer in IgG-deficient mice implicated IgG as the pathogenetic ligand for endothelial FcγRIIB in obesity-induced insulin resistance. Moreover, IgG transferred from patients with T2DM but not from metabolically healthy subjects caused insulin resistance in IgG-deficient mice via FcγRIIB, indicating that similar processes may be operative in T2DM in humans. Mechanistically, the activation of FcγRIIB by IgG from obese mice impaired endothelial cell insulin transcytosis in culture and in vivo. These effects were attributed to hyposialylation of the Fc glycan, and IgG from T2DM patients was also hyposialylated. In HFD-fed mice, supplementation with the sialic acid precursor N-acetyl-D-mannosamine restored IgG sialylation and preserved insulin sensitivity without affecting weight gain. Thus, IgG sialylation and endothelial FcγRIIB may represent promising therapeutic targets to sever the link between obesity and T2DM.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Imunoglobulina G/metabolismo , Resistência à Insulina , Obesidade/metabolismo , Receptores de IgG/metabolismo , Animais , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Hexosaminas/farmacologia , Imunoglobulina G/genética , Camundongos , Camundongos Knockout , Obesidade/genética , Obesidade/patologia , Receptores de IgG/genética , Transcitose/efeitos dos fármacos
9.
J Mol Cell Cardiol ; 107: 41-51, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28457941

RESUMO

Steroid hormone receptors including estrogen receptors (ER) classically function as ligand-regulated transcription factors. However, estrogens also elicit cellular effects through binding to extra-nuclear ER (ERα, ERß, and G protein-coupled ER or GPER) that are coupled to kinases. How extra-nuclear ER actions impact cardiac ischemia-reperfusion (I/R) injury is unknown. We treated ovariectomized wild-type female mice with estradiol or an estrogen-dendrimer conjugate (EDC), which selectively activates extra-nuclear ER, or vehicle interventions for two weeks. I/R injury was then evaluated in isolated Langendorff perfused hearts. Two weeks of treatment with estradiol significantly decreased infarct size and improved post-ischemic contractile function. Similarly, EDC treatment significantly decreased infarct size and increased post-ischemic functional recovery compared to vehicle-treated hearts. EDC also caused an increase in myocardial protein S-nitrosylation, consistent with previous studies showing a role for this post-translational modification in cardioprotection. In further support of a role for S-nitrosylation, inhibition of nitric oxide synthase, but not soluble guanylyl cyclase blocked the EDC mediated protection. The administration of ICI182,780, which is an agonist of G-protein coupled estrogen receptor (GPER) and an antagonist of ERα and ERß, did not result in protection; however, ICI182,780 significantly blocked EDC-mediated cardioprotection, indicating participation of ERα and/or ERß. In studies determining the specific ER subtype and cellular target involved, EDC decreased infarct size and improved functional recovery in mice lacking ERα in cardiomyocytes. In contrast, protection was lost in mice deficient in endothelial cell ERα. Thus, extra-nuclear ERα activation in endothelium reduces cardiac I/R injury in mice, and this likely entails increased protein S-nitrosylation. Since EDC does not stimulate uterine growth, in the clinical setting EDC-like compounds may provide myocardial protection without undesired uterotrophic and cancer-promoting effects.


Assuntos
Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Isquemia/genética , Traumatismo por Reperfusão/genética , Animais , Endotélio/metabolismo , Endotélio/patologia , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor beta de Estrogênio/antagonistas & inibidores , Estrogênios/genética , Estrogênios/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Isquemia/metabolismo , Isquemia/patologia , Camundongos , Ovariectomia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Receptores de Estrogênio/antagonistas & inibidores , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Transdução de Sinais/efeitos dos fármacos
10.
Endocrinology ; 157(10): 3731-3741, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27552247

RESUMO

Estrogens have the potential to afford atheroprotection, to prevent excess adiposity and its metabolic complications including insulin resistance, and to lessen hepatic steatosis. Cellular responses to estrogens occur through gene regulation by nuclear estrogen receptors (ERs), and through signal initiation by plasma membrane-associated ER. Leveraging the potentially favorable cardiometabolic actions of estrogens has been challenging, because their reproductive tract and cancer-promoting effects adversely impact the risk to benefit ratio of the therapy. In previous works, we discovered that an estrogen dendrimer conjugate (EDC) comprised of ethinyl-estradiol (E2) molecules linked to a poly(amido)amine dendrimer selectively activates nonnuclear ER, and in mice, EDC does not invoke a uterotrophic response or support ER-positive breast cancer growth. In the present investigation, we employed EDC to determine how selective nonnuclear ER activation impacts atherosclerosis, adiposity, glucose homeostasis, and hepatic steatosis in female mice. In contrast to E2, EDC did not blunt atherosclerosis in hypercholesterolemic apoE-/- mice. Also in contrast to E2, EDC did not prevent the increase in adiposity caused by Western diet feeding in wild-type mice, and it did not affect Western diet-induced glucose intolerance. However, E2 and EDC had comparable favorable effect on diet-induced hepatic steatosis, and this was related to down-regulation of fatty acid and triglyceride synthesis genes in the liver. Predictably, only E2 caused a uterotrophic response. Thus, although nonnuclear ER activation does not prevent atherosclerosis or diet-induced obesity or glucose intolerance, it may provide a potential new strategy to combat hepatic steatosis without impacting the female reproductive tract or increasing cancer risk.


Assuntos
Aterosclerose/prevenção & controle , Dendrímeros/uso terapêutico , Estrogênios/uso terapêutico , Fígado Gorduroso/prevenção & controle , Adiposidade/efeitos dos fármacos , Animais , Aterosclerose/etiologia , Composição Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Metabolismo dos Carboidratos/efeitos dos fármacos , Dendrímeros/farmacologia , Dieta Hiperlipídica , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Estrogênios/farmacologia , Fígado Gorduroso/etiologia , Feminino , Glucose/metabolismo , Homeostase/efeitos dos fármacos , Hipercolesterolemia/complicações , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Camundongos Endogâmicos C57BL
11.
Diabetes ; 65(7): 1996-2005, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27207525

RESUMO

Modest elevations in C-reactive protein (CRP) are associated with type 2 diabetes. We previously revealed in mice that increased CRP causes insulin resistance and mice globally deficient in the CRP receptor Fcγ receptor IIB (FcγRIIB) were protected from the disorder. FcγRIIB is expressed in numerous cell types including endothelium and B lymphocytes. Here we investigated how endothelial FcγRIIB influences glucose homeostasis, using mice with elevated CRP expressing or lacking endothelial FcγRIIB. Whereas increased CRP caused insulin resistance in mice expressing endothelial FcγRIIB, mice deficient in the endothelial receptor were protected. The insulin resistance with endothelial FcγRIIB activation was due to impaired skeletal muscle glucose uptake caused by attenuated insulin delivery, and it was associated with blunted endothelial nitric oxide synthase (eNOS) activation in skeletal muscle. In culture, CRP suppressed endothelial cell insulin transcytosis via FcγRIIB activation and eNOS antagonism. Furthermore, in knock-in mice harboring constitutively active eNOS, elevated CRP did not invoke insulin resistance. Collectively these findings reveal that by inhibiting eNOS, endothelial FcγRIIB activation by CRP blunts insulin delivery to skeletal muscle to cause insulin resistance. Thus, a series of mechanisms in endothelium that impairs insulin movement has been identified that may contribute to type 2 diabetes pathogenesis.


Assuntos
Endotélio Vascular/metabolismo , Resistência à Insulina/fisiologia , Insulina/metabolismo , Músculo Esquelético/metabolismo , Receptores de IgG/metabolismo , Animais , Aorta/citologia , Aorta/metabolismo , Transporte Biológico , Proteína C-Reativa/metabolismo , Bovinos , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Glucose/metabolismo , Homeostase/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Óxido Nítrico Sintase Tipo III/metabolismo , Transdução de Sinais/fisiologia
12.
Sci Signal ; 9(429): ra53, 2016 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-27221711

RESUMO

There is great medical need for estrogens with favorable pharmacological profiles that support desirable activities for menopausal women, such as metabolic and vascular protection, but that lack stimulatory activities on the breast and uterus. We report the development of structurally novel estrogens that preferentially activate a subset of estrogen receptor (ER) signaling pathways and result in favorable target tissue-selective activity. Through a process of structural alteration of estrogenic ligands that was designed to preserve their essential chemical and physical features but greatly reduced their binding affinity for ERs, we obtained "pathway preferential estrogens" (PaPEs), which interacted with ERs to activate the extranuclear-initiated signaling pathway preferentially over the nuclear-initiated pathway. PaPEs elicited a pattern of gene regulation and cellular and biological processes that did not stimulate reproductive and mammary tissues or breast cancer cells. However, in ovariectomized mice, PaPEs triggered beneficial responses both in metabolic tissues (adipose tissue and liver) that reduced body weight gain and fat accumulation and in the vasculature that accelerated repair of endothelial damage. This process of designed ligand structure alteration represents a novel approach to develop ligands that shift the balance in ER-mediated extranuclear and nuclear pathways to obtain tissue-selective, non-nuclear PaPEs, which may be beneficial for postmenopausal hormone replacement. The approach may also have broad applicability for other members of the nuclear hormone receptor superfamily.


Assuntos
Desenho de Fármacos , Estrogênios/metabolismo , Receptores de Estrogênio/metabolismo , Tecido Adiposo/efeitos dos fármacos , Animais , Peso Corporal , Proliferação de Células , Cromatina/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Ligantes , Fígado/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases , Células MCF-7 , Glândulas Mamárias Animais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica , Conformação Proteica , Transdução de Sinais , Útero/efeitos dos fármacos
13.
Hypertension ; 65(2): 456-62, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25368023

RESUMO

There is a well-recognized association between obesity, inflammation, and hypertension. Why obesity causes hypertension is poorly understood. We previously demonstrated using a C-reactive protein (CRP) transgenic mouse that CRP induces hypertension that is related to NO deficiency. Our prior work in cultured endothelial cells identified the Fcγ receptor IIB (FcγRIIB) as the receptor for CRP whereby it antagonizes endothelial NO synthase. Recognizing known associations between CRP and obesity and hypertension in humans, in the present study we tested the hypothesis that FcγRIIB plays a role in obesity-induced hypertension in mice. Using radiotelemetry, we first demonstrated that the hypertension observed in transgenic mouse-CRP is mediated by the receptor, indicating that FcγRIIB is capable of modifying blood pressure. We then discovered in a model of diet-induced obesity yielding equal adiposity in all study groups that whereas FcγRIIB(+/+) mice developed obesity-induced hypertension, FcγRIIB(-/-) mice were fully protected. Levels of CRP, the related pentraxin serum amyloid P component which is the CRP-equivalent in mice, and total IgG were unaltered by diet-induced obesity; FcγRIIB expression in endothelium was also unchanged. However, whereas IgG isolated from chow-fed mice had no effect, IgG from high-fat diet-fed mice inhibited endothelial NO synthase in cultured endothelial cells, and this was an FcγRIIB-dependent process. Thus, we have identified a novel role for FcγRIIB in the pathogenesis of obesity-induced hypertension, independent of processes regulating adiposity, and it may entail an IgG-induced attenuation of endothelial NO synthase function. Approaches targeting FcγRIIB may potentially offer new means to treat hypertension in obese individuals.


Assuntos
Hipertensão/etiologia , Óxido Nítrico Sintase Tipo III/fisiologia , Obesidade/complicações , Receptores de IgG/fisiologia , Animais , Pressão Sanguínea/fisiologia , Proteína C-Reativa/efeitos adversos , Proteína C-Reativa/análise , Proteína C-Reativa/genética , Células Cultivadas , Gorduras na Dieta/toxicidade , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Células Endoteliais/imunologia , Hipertensão/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Imunoglobulina G/isolamento & purificação , Imunoglobulina G/farmacologia , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Óxido Nítrico/fisiologia , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Obesidade/imunologia , Receptores de IgG/deficiência , Receptores de IgG/genética , Componente Amiloide P Sérico/análise
14.
Am J Physiol Endocrinol Metab ; 307(3): E345-54, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24939737

RESUMO

Despite the capacity of estrogens to favorably regulate body composition and glucose homeostasis, their use to combat obesity and type 2 diabetes is not feasible, because they promote sex steroid-responsive cancers. The novel selective estrogen receptor modulator (SERM) bazedoxifene acetate (BZA) uniquely antagonizes both breast cancer development and estrogen-related changes in the female reproductive tract. How BZA administered with conjugated estrogen (CE) or alone impacts metabolism is unknown. The effects of BZA or CE + BZA on body composition and glucose homeostasis were determined in ovariectomized female mice fed a Western diet for 10-12 wk. In contrast to vehicle, estradiol (E2), CE, BZA, and CE + BZA equally prevented body weight gain by 50%. In parallel, all treatments caused equal attenuation of the increase in body fat mass invoked by the diet as well as the increases in subcutaneous and visceral white adipose tissue. Diet-induced hepatic steatosis was attenuated by E2 or CE, and BZA alone or with CE provided even greater steatosis prevention; all interventions improved pyruvate tolerance tests. Glucose tolerance tests and HOMA-IR were improved by E2, CE, and CE + BZA. Whereas E2 or CE alone invoked a uterotrophic response, BZA alone or CE + BZA had negligible impact on the uterus. Thus, CE + BZA affords protection from diet-induced adiposity, hepatic steatosis, and insulin resistance with minimal impact on the female reproductive tract in mice. These combined agents may provide a valuable new means to favorably regulate body composition and glucose homeostasis and combat fatty liver.


Assuntos
Diabetes Mellitus Tipo 2/prevenção & controle , Terapia de Reposição de Estrogênios , Estrogênios Conjugados (USP)/uso terapêutico , Estrogênios/uso terapêutico , Fígado Gorduroso/prevenção & controle , Obesidade/prevenção & controle , Moduladores Seletivos de Receptor Estrogênico/uso terapêutico , Gordura Abdominal/efeitos dos fármacos , Gordura Abdominal/patologia , Adiposidade/efeitos dos fármacos , Animais , Diabetes Mellitus Tipo 2/patologia , Dieta Hiperlipídica/efeitos adversos , Relação Dose-Resposta a Droga , Quimioterapia Combinada/efeitos adversos , Terapia de Reposição de Estrogênios/efeitos adversos , Estrogênios/administração & dosagem , Estrogênios/efeitos adversos , Estrogênios Conjugados (USP)/administração & dosagem , Estrogênios Conjugados (USP)/efeitos adversos , Fígado Gorduroso/etiologia , Fígado Gorduroso/patologia , Feminino , Indóis/administração & dosagem , Indóis/efeitos adversos , Indóis/uso terapêutico , Resistência à Insulina , Fígado/efeitos dos fármacos , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica , Obesidade/etiologia , Obesidade/patologia , Tamanho do Órgão/efeitos dos fármacos , Ovariectomia/efeitos adversos , Moduladores Seletivos de Receptor Estrogênico/administração & dosagem , Moduladores Seletivos de Receptor Estrogênico/efeitos adversos , Útero/efeitos dos fármacos , Útero/patologia
15.
Steroids ; 81: 64-9, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24252382

RESUMO

Estrogen receptors (ER) classically function as transcription factors regulating gene expression. More recently, evidence has continued to accumulate that ER additionally serve numerous important functions remote from the nucleus in a variety of cell types, particularly in non-reproductive tissues. The identification of post-translational modifications of ERα and protein-protein interactions with the receptor that are critical to its non-nuclear functions has afforded opportunities to gain greater insights into these novel non-genomic roles of the receptor. The development of a stable ligand that selectively activates non-nuclear ER has also been invaluable. In this review focused on ERα, recent new understanding of the processes underlying non-nuclear ER action and their in vivo consequences will be discussed. Further research into the non-nuclear capacities by which ER modulate cellular behavior is essential to ultimately harnessing these processes for therapeutic gain in numerous disease contexts.


Assuntos
Osso e Ossos/metabolismo , Receptor alfa de Estrogênio/metabolismo , Receptores Nucleares Órfãos/metabolismo , Animais , Núcleo Celular/metabolismo , Humanos , Receptores X do Fígado , Microdomínios da Membrana/metabolismo , Mutagênese , Neoplasias/metabolismo , Neoplasias/patologia , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Transdução de Sinais
16.
Mol Endocrinol ; 27(4): 649-56, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23443267

RESUMO

Extensive evidence has suggested that at least some of the effects of estrogens on bone are mediated via extranuclear estrogen receptor α signaling. However, definitive proof for this contention and the extent to which such effects may contribute to the overall protective effects of estrogens on bone maintenance have remained elusive. Here, we investigated the ability of a 17ß-estradiol (E2) dendrimer conjugate (EDC), incapable of stimulating nuclear-initiated actions of estrogen receptor α, to prevent the effects of ovariectomy (OVX) on the murine skeleton. We report that EDC was as potent as an equimolar dose of E2 in preventing bone loss in the cortical compartment that represents 80% of the entire skeleton, but was ineffective on cancellous bone. In contrast, E2 was effective in both compartments. Consistent with its effect on cortical bone mass, EDC partially prevented the loss of both vertebral and femoral strength. In addition, EDC, as did E2, prevented the OVX-induced increase in osteoclastogenesis, osteoblastogenesis, and oxidative stress. Nonetheless, the OVX-induced decrease in uterine weight was unaltered by EDC but was restored by E2. These results demonstrate that the protection of cortical bone mass by estrogens is mediated, at least in part, via a mechanism that is distinct from the classic mechanism of estrogen action on reproductive organs.


Assuntos
Osso e Ossos/metabolismo , Osso e Ossos/patologia , Núcleo Celular/metabolismo , Receptor alfa de Estrogênio/metabolismo , Animais , Atrofia , Densidade Óssea/efeitos dos fármacos , Remodelação Óssea/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Estradiol/farmacologia , Feminino , Fêmur/efeitos dos fármacos , Fêmur/patologia , Fêmur/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteogênese/efeitos dos fármacos , Ovariectomia , Estresse Oxidativo/efeitos dos fármacos , Coluna Vertebral/efeitos dos fármacos , Coluna Vertebral/patologia , Coluna Vertebral/fisiopatologia , Útero/efeitos dos fármacos , Útero/patologia
17.
Mol Endocrinol ; 27(1): 2-11, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23242705

RESUMO

A subpopulation of plasma membrane-associated estrogen receptor (ER)α interact directly with G proteins and mediate nonnuclear receptor signaling. This mechanism underlies numerous processes, including important cardiovascular protective actions of estradiol (E(2)), such as the activation of endothelial NO synthase (eNOS) and endothelial cell growth and migration. In the present work we sought a genetic approach to differentiate nonnuclear from nuclear ERα actions. We generated single alanine substitutions within the Gαi-binding domain of ERα (amino acids 251-260) and tested signaling to eNOS or ERK1,2 and activation of luciferase (Luc) reporters signifying transactivation via direct or indirect ERα-DNA binding in HeLa cells. The point mutants ERα-R256A, ERα-K257A, ERα-D258A, and ERα-R260A were all incapable of activating eNOS in response to E(2), and ERα-R256A and ERα-D258A also showed loss of ERK1,2 activation. In contrast, ERα-R256A, ERα-K257A, ERα-D258A, and ERα-R260A all displayed normal capacity to invoke E(2)-induced transactivation of estrogen response element (ERE)-Luc or Sp1-Luc. However, whereas activator protein 1-Luc activation by ERα-R256A and ERα-D258A was intact, ERα-K257A and ERα-R260A were incapable of activator protein 1-Luc activation. In in vitro pull-down assays with the two mutants that lack all nonnuclear functions tested and retain all nuclear functions tested, ERα-R256A and ERα-D258A, there was normal direct interaction between Gαi and ERα-R256A and an absence of interaction between Gαi and ERα-D258A. When expressed in endothelial cells, these two mutants prevented E(2)-induced migration and eNOS activation mediated by endogenous receptor, indicative of dominant-negative action. Thus, the point mutants ERα-R256A and ERα-D258A in the receptor GαI-binding domain provide genetic segregation of nonnuclear from nuclear ERα function.


Assuntos
Núcleo Celular/metabolismo , Receptor alfa de Estrogênio/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Mutação Puntual , Sequência de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Estradiol/fisiologia , Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Mutagênese Sítio-Dirigida , Óxido Nítrico Sintase Tipo III/metabolismo , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , Ativação Transcricional
18.
Circ Res ; 109(10): 1132-40, 2011 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-21940940

RESUMO

RATIONALE: Elevations in C-reactive protein (CRP) are associated with increased cardiovascular disease risk and endothelial dysfunction. CRP antagonizes endothelial nitric oxide synthase (eNOS) through processes mediated by the IgG receptor Fcγ receptor IIB (FcγRIIB), its immunoreceptor tyrosine-based inhibitory motif, and SH2 domain-containing inositol 5'-phosphatase 1. In mice, CRP actions on eNOS blunt carotid artery re-endothelialization. OBJECTIVE: How CRP activates FcγRIIB in endothelium is not known. We determined the role of Fcγ receptor I (FcγRI) and the basis for coupling of FcγRI to FcγRIIB in endothelium. METHODS AND RESULTS: In cultured endothelial cells, FcγRI-blocking antibodies prevented CRP antagonism of eNOS, and CRP activated Src via FcγRI. CRP-induced increases in FcγRIIB immunoreceptor tyrosine-based inhibitory motif phosphorylation and SH2 domain-containing inositol 5'-phosphatase 1 activation were Src-dependent, and Src inhibition prevented eNOS antagonism by CRP. Similar processes mediated eNOS antagonism by aggregated IgG used to mimic immune complex. Carotid artery re-endothelialization was evaluated in offspring from crosses of CRP transgenic mice (TG-CRP) with either mice lacking the γ subunit of FcγRI (FcRγ(-/-)) or FcγRIIB(-/-) mice. Whereas re-endothelialization was impaired in TG-CRP vs wild-type, it was normal in both FcRγ(-/-); TG-CRP and FcγRIIB(-/-); TG-CRP mice. CONCLUSIONS: CRP antagonism of eNOS is mediated by the coupling of FcγRI to FcγRIIB by Src kinase and resulting activation of SH2 domain-containing inositol 5'-phosphatase 1, and consistent with this mechanism, both FcγRI and FcγRIIB are required for CRP to blunt endothelial repair in vivo. Similar mechanisms underlie eNOS antagonism by immune complex. FcγRI and FcγRIIB may be novel therapeutic targets for preventing endothelial dysfunction in inflammatory or immune complex-mediated conditions.


Assuntos
Proteína C-Reativa/metabolismo , Lesões das Artérias Carótidas/enzimologia , Lesões das Artérias Carótidas/imunologia , Células Endoteliais/enzimologia , Células Endoteliais/imunologia , Receptores de IgG/metabolismo , Quinases da Família src/metabolismo , Animais , Complexo Antígeno-Anticorpo/metabolismo , Proteína C-Reativa/genética , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/patologia , Bovinos , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/patologia , Ativação Enzimática , Humanos , Inositol Polifosfato 5-Fosfatases , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Óxido Nítrico Sintase Tipo III/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação , Coelhos , Receptores de IgG/deficiência , Receptores de IgG/genética , Transdução de Sinais , Fatores de Tempo , Transfecção
19.
J Biol Chem ; 286(17): 14737-43, 2011 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-21343284

RESUMO

In addition to the classical function of estrogen receptors (ER) as transcription factors, evidence continues to accumulate that they mediate non-nuclear processes in numerous cell types, including the endothelium, in which they activate endothelial NO synthase. Non-nuclear ER signaling entails unique post-translational modifications and protein-protein interactions of the receptor with adaptor molecules, kinases, and G proteins. Recent in vitro and in vivo studies in mice using an estrogen-dendrimer conjugate that is excluded from the nucleus indicate that non-nuclear ER activation underlies the migration and growth responses of endothelial cells to estrogen but not the growth responses of endometrial or breast cancer cells to the hormone. In this minireview, the features of ERα and protein-protein interactions that enable it to invoke extranuclear signaling in the endothelium and the consequences of that signaling are discussed.


Assuntos
Endotélio/metabolismo , Receptor alfa de Estrogênio/metabolismo , Transdução de Sinais/fisiologia , Animais , Humanos , Ligação Proteica , Processamento de Proteína Pós-Traducional
20.
J Clin Invest ; 120(7): 2319-30, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20577047

RESUMO

Steroid hormone receptors function classically in the nucleus as transcription factors. However, recent data indicate that there are also non-nuclear subpopulations of steroid hormone receptors, including estrogen receptors (ERs), that mediate membrane-initiated signaling of unclear basis and significance. Here we have shown that an estrogen-dendrimer conjugate (EDC) that is excluded from the nucleus stimulates endothelial cell proliferation and migration via ERalpha, direct ERalpha-Galphai interaction, and endothelial NOS (eNOS) activation. Analysis of mice carrying an estrogen response element luciferase reporter, ER-regulated genes in the mouse uterus, and eNOS enzyme activation further indicated that EDC specifically targets non-nuclear processes in vivo. In mice, estradiol and EDC equally stimulated carotid artery reendothelialization in an ERalpha- and G protein-dependent manner, and both agents attenuated the development of neointimal hyperplasia following endothelial injury. In contrast, endometrial carcinoma cell growth in vitro and uterine enlargement and MCF-7 cell breast cancer xenograft growth in vivo were stimulated by estradiol but not EDC. Thus, EDC is a non-nuclear selective ER modulator (SERM) in vivo, and in mice, non-nuclear ER signaling promotes cardiovascular protection. These processes potentially could be harnessed to provide vascular benefit without increasing the risk of uterine or breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Receptores de Estrogênio/metabolismo , Útero/metabolismo , Animais , Antineoplásicos/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Proliferação de Células , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Ativação Enzimática/genética , Estradiol/genética , Estradiol/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Estrogênios/genética , Estrogênios/metabolismo , Feminino , Humanos , Luciferases/genética , Luciferases/metabolismo , Camundongos , Camundongos Nus , Óxido Nítrico Sintase Tipo III , Receptores de Estrogênio/genética , Elementos de Resposta , Transdução de Sinais/genética , Útero/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...