Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Reprod Biol ; 23(3): 100785, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37392490

RESUMO

Menstruation is a specific physiological phenomenon in female humans that is regulated by complex molecular mechanisms. However, the molecular network involved in menstruation remains incompletely understood. Previous studies have suggested that C-X-C chemokine receptor 4 (CXCR4) is involved; however, how CXCR4 participates in endometrial breakdown remains unclear, as do its regulatory mechanisms. This study aimed to clarify the role of CXCR4 in endometrial breakdown and its regulation by hypoxia-inducible factor-1 alpha (HIF1A). We first confirmed that CXCR4 and HIF1A protein levels were significantly increased during the menstrual phase compared with the late secretory phase using immunohistochemistry. In our mouse model of menstruation, real-time PCR, western blotting, and immunohistochemistry showed that CXCR4 mRNA and protein expression levels gradually increased from 0 to 24 h after progesterone withdrawal during endometrial breakdown. HIF1A mRNA and HIF1A nuclear protein levels significantly increased and peaked at 12 h after progesterone withdrawal. Endometrial breakdown was significantly suppressed by the CXCR4 inhibitor AMD3100 and the HIF1A inhibitor 2-methoxyestradiol in our mouse model, and HIF1A inhibition also suppressed CXCR4 mRNA and protein expression. In vitro studies using human decidual stromal cells showed that CXCR4 and HIF1A mRNA expression levels were increased by progesterone withdrawal and that HIF1A knockdown significantly suppressed the elevation in CXCR4 mRNA expression. CD45+ leukocyte recruitment during endometrial breakdown was suppressed by both AMD3100 and 2-methoxyestradiol in our mouse model. Taken together, our preliminary findings suggest that endometrial CXCR4 expression is regulated by HIF1A during menstruation and may promote endometrial breakdown, potentially via leukocyte recruitment.


Assuntos
Menstruação , Progesterona , Animais , Feminino , Humanos , Camundongos , 2-Metoxiestradiol/metabolismo , Endométrio/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Leucócitos/metabolismo , Progesterona/metabolismo , Receptores de Quimiocinas/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , RNA Mensageiro/metabolismo
2.
Reproduction ; 165(4): 383-393, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36762768

RESUMO

In brief: Mating shuts down the 2-methoxyestradiol (2ME) nongenomic pathway that accelerates oviductal egg transport in the rat. This study shows that sperm cells, but not vaginocervical stimulation, utilize TNF-α to shut down this 2ME nongenomic pathway. Abstract: The transport of oocytes or embryos throughout the oviduct to the implantation site in the uterus is defined as egg transport. In the rat, 2-methoxyestradiol (2ME) accelerates egg transport through the oviduct via a nongenomic pathway. Mating is known to shut down this 2ME pathway and then trigger an estradiol genomic pathway that accelerates egg transport. Here, we tested whether intrauterine insemination (IUI) or vaginocervical stimulation (VCS) shuts down the 2ME nongenomic pathway that accelerates egg transport, and if these mating components require tumor necrosis factor alpha (TNF-α). Levels of TNF-α and the mRNA for TNF-α receptors were measured in the oviduct of IUI or VCS rats. The tissue distribution of TNF-α receptor proteins and the concentration of the mRNA for catechol-O-methyl transferase (Comt) and 2ME were also analyzed in the oviduct. Finally, we assessed whether 2ME accelerates egg transport in IUI or VCS rats previously treated with the TNF-α antagonist W9P9QY. Results show that IUI, but not VCS, increased TNF-α and their receptors in the oviduct. IUI and VCS did not change the tissue distribution of TNF-α receptors; however, both decreased the oviductal concentration of Comt and 2ME. IUI and VCS each blocked the 2ME-induced egg transport acceleration; however, only the IUI was antagonized by the TNF-α antagonist. We concluded that IUI and VCS inhibit the 2ME nongenomic pathway that accelerates egg transport; however, the vias of action are distinct, with a TNF-α increase on spermatozoa presence being required for the shutdown of the 2ME pathway.


Assuntos
Catecol O-Metiltransferase , Fator de Necrose Tumoral alfa , Feminino , Humanos , Ratos , Masculino , Animais , 2-Metoxiestradiol/farmacologia , 2-Metoxiestradiol/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Catecol O-Metiltransferase/metabolismo , Ratos Sprague-Dawley , Sêmen/metabolismo , Oviductos/metabolismo , Estradiol/farmacologia , Estradiol/metabolismo , Espermatozoides/metabolismo , RNA Mensageiro/metabolismo
3.
Chem Biol Interact ; 369: 110277, 2023 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-36414027

RESUMO

2-Methoxyestradiol (2-ME2) is a metabolite of 17ß-estradiol and is currently in clinical trials as an antitumor agent. Here we found 2-ME2 level remains stable in the local environment of ovaries but declines in serum in aging mice, and exogenous 2-ME2 impacts the meiotic maturation of mouse oocytes in dose-dependent manner. In vitro 2-ME2 application arrested oocytes at metaphase I (MI), with abnormal spindle structure and chromosome alignment. 2-ME2 exposure induced excessive production of reactive oxygen species (ROS) and malondialdehyde, as well as accelerated apoptosis progression. 2-ME2 unbalanced mitochondrial dynamics by increasing DRP1 and MFN1 while decreasing Opa1. Similar phenotypes were also observed in oocytes from mice injected intraperitoneally with 2-ME2. Taken together, this study indicates 2-ME2 exposure impairs oocyte meiotic maturation through inducing mitochondrial imbalance, oxidative stress and apoptosis. The gradual decline in oocyte quality and quantity may be associated with the stable 2-ME2 in ovaries during female reproductive aging.


Assuntos
Meiose , Oócitos , Feminino , Camundongos , Animais , 2-Metoxiestradiol/farmacologia , 2-Metoxiestradiol/metabolismo , Reprodução , Envelhecimento
4.
Biosens Bioelectron ; 218: 114747, 2022 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-36198238

RESUMO

Thoracic aortic aneurysm (TAA), in which arteries enlarge asymptomatically over time until dissection or rupture occurs, is a serious health risk. The mainstay of TAA treatment remains surgical repair due to the lack of effective drugs. The complex etiology and pathogenesis of TAA, including hemodynamic alterations and genetic factors, lead to inaccuracies in preclinical models for drug screening. Previously, our group designed an aorta smooth muscle-on-a-chip to emulate human aorta physiology and pathophysiology and screened three promising therapeutic drugs targeting mitochondrial dynamics in TAA. On this foundation, we updated the one-channel chip to an eighteen-well chip platform with four polydimethylsiloxane layers. Benefiting from this high-throughput chip, we rapidly screened multiple drugs simultaneously using distinct cell lines in vitro. In addition, we observed the abnormal activation of hypoxia-inducible factor 1-alpha (HIF-1alpha) in aortas from TAA patients by Western blot and bioinformatics analyses. Intriguingly, this phenomenon was replicated only when smooth muscle cells (SMCs) were strained on the chip. We then screened seven specific HIF-1alpha inhibitors and selected the two most effective drugs (2-methoxyestradiol and digoxin) by quantitative PCR and colorimetric methods. The results demonstrated that these two drugs can improve respiratory chain function and rescue the SMC contractile phenotype, showing applicability for the clinical treatment of TAA. This high-throughput aorta smooth muscle-on-a-chip will become a potential preclinical model for TAA drug screening.


Assuntos
Aneurisma da Aorta Torácica , Técnicas Biossensoriais , Humanos , Aneurisma da Aorta Torácica/tratamento farmacológico , Aneurisma da Aorta Torácica/genética , Aneurisma da Aorta Torácica/metabolismo , 2-Metoxiestradiol/metabolismo , Avaliação Pré-Clínica de Medicamentos , Dispositivos Lab-On-A-Chip , Aorta/metabolismo , Aorta/patologia , Digoxina , Dimetilpolisiloxanos , Fator 1 Induzível por Hipóxia/metabolismo , Músculo Liso/metabolismo , Músculo Liso/patologia
5.
Environ Sci Pollut Res Int ; 29(39): 59524-59533, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35384535

RESUMO

Intimal hyperplasia (IH) is a common complication of vascular interventional procedures that leads to narrowing of the vessel lumen. 2-Methoxyestradiol (2ME), an estrogen metabolite, has numerous pharmacological actions, including vasoprotective and antiproliferative activities. The present study aimed to evaluate the potential of 2ME, prepared as a self-nanoemulsifying drug delivery system (SNEDDS), to inhibit IH induced by balloon injury (BI) in the rat carotid artery. The prepared 2ME SNEDDS had a particle size of 119 ± 2.3 nm and a zeta potential of -7.1 ± 1.4 mV. Animals were divided into 5 groups, namely control, sham, BI, BI + 2ME (100 µg/kg), and BI + 2ME (250 µg/kg). The obtained data indicated that 2ME significantly inhibited IH as indicated by the histological and morphometric assessment of the intima, media and lumen areas. This was associated with enhanced expression of Bax and inhibited expression of Bcl2 mRNA. Furthermore, 2ME exhibited significant antioxidant properties as evidenced by prevention of malondialdehyde accumulation as well as superoxide dismutase and catalase enzymatic exhaustion. In addition, 2ME showed significant anti-inflammatory actions as it significantly inhibited vascular content of interleukin-6, tumor necrosis factor-alpha, and nuclear factor-κB. The observed vasoprotective activities of 2ME were accompanied by inhibition of Janus kinase/signal transducers and activators of transcription (JAK/STAT) protein expression. In conclusion, this study revealed that 2ME ameliorates balloon injury-induced IH in rats via suppressing JAK/STAT axis. This may help to develop new strategies to combat IH.


Assuntos
Artérias Carótidas , Janus Quinases , 2-Metoxiestradiol/metabolismo , Animais , Hiperplasia/metabolismo , Hiperplasia/patologia , Janus Quinases/metabolismo , NF-kappa B/metabolismo , Ratos
6.
Acta Pharmacol Sin ; 43(10): 2636-2650, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35292769

RESUMO

Diabetes mellitus is associated with series of macrovascular and microvascular pathological changes that cause a wide range of complications. Diabetic patients are highly susceptible to hindlimb ischemia (HLI), which remains incurable. Evidence shows that skeletal muscle cells secrete a number of angiogenic factors to promote neovascularization and restore blood perfusion, this paracrine function is crucial for therapeutic angiogenesis in diabetic HLI. In this study we investigated whether sotagliflozin, an anti-hyperglycemia SGLT2 inhibitor, exerted therapeutic angiogenesis effects in diabetic HLI in vitro and in vivo. In C2C12 skeletal muscle cells, we showed that high glucose (HG, 25 mM) under hypoxia markedly inhibited cell viability, proliferation and migration potentials, which were dose-dependently reversed by pretreatment with sotagliflozin (5-20 µM). Sotagliflozin pretreatment enhanced expression levels of angiogenic factors HIF-1α, VEGF-A and PDGF-BB in HG-treated C2C12 cells under hypoxia as well as secreted amounts of VEGF-A and PDGF-BB in the medium; pretreatment with the HIF-1α inhibitor 2-methoxyestradiol (2-ME2, 10 µM) or HIF-1α knockdown abrogated sotagliflozin-induced increases in VEGF-A and PDGF-BB expression, as well as sotagliflozin-stimulated cell proliferation and migration potentials. Furthermore, the conditioned media from sotagliflozin-treated C2C12 cells in HG medium enhanced the migration and proliferation capabilities of vascular endothelial and smooth muscle cells, two types of cells necessary for forming functional blood vessels. In vivo study was conducted in diabetic mice subjected to excising the femoral artery of the left limb. After the surgery, sotagliflozin (10 mg/kg) was directly injected into gastrocnemius muscle of the left hindlimb once every 3 days for 3 weeks. We showed that intramuscular injection of sotagliflozin effectively promoted the formation of functional blood vessels, leading to significant recovery of blood perfusion in diabetic HLI mice. Together, our results highlight a new indication of SGLT2 inhibitor sotagliflozin as a potential therapeutic angiogenesis agent for diabetic HLI.


Assuntos
Diabetes Mellitus Experimental , Inibidores do Transportador 2 de Sódio-Glicose , 2-Metoxiestradiol/metabolismo , 2-Metoxiestradiol/farmacologia , 2-Metoxiestradiol/uso terapêutico , Indutores da Angiogênese/farmacologia , Animais , Becaplermina/farmacologia , Meios de Cultivo Condicionados/farmacologia , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Glucose/metabolismo , Glicosídeos , Membro Posterior , Hipóxia/tratamento farmacológico , Injeções Intramusculares , Isquemia/tratamento farmacológico , Isquemia/patologia , Camundongos , Músculo Esquelético/metabolismo , Neovascularização Patológica/tratamento farmacológico , Neovascularização Fisiológica , Inibidores do Transportador 2 de Sódio-Glicose/metabolismo , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Inibidores do Transportador 2 de Sódio-Glicose/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Life Sci ; 296: 120441, 2022 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-35240160

RESUMO

AIMS: Investigating the impact of 17ß estradiol (E2) and its endogenous non-hormonal metabolite 2-methoxyestradiol (2ME) on renal ischemia-reperfusion (RIR) induced kidney injury in ovariectomized (OVX) rats and the role of catechol-O-methyltransferase (COMT) in their effects. MAIN METHODS: Eighty female rats were allocated into eight groups. Control group, Sham group, OVX group, OVX and RIR group, OVX + RIR + E2 group, OVX + RIR + 2ME group, OVX + RIR + E2 + Entacapone group and OVX + RIR + 2ME + Entacapone group, respectively. Twenty-four hours post RIR, creatinine (Cr) and blood urea nitrogen (BUN) were determined in serum, while malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), Glutathione (GSH), myeloperoxidase (MPO), as well as the expressions of COMT, hypoxia inducible factor-1α (HIF-1α) and tyrosine hydroxylase (TH) were assessed in the kidney tissues. KEY FINDINGS: Serum Cr, BUN, MPO, as well as HIF-1α and TH expressions were significantly higher with concomitant decrease in COMT expression, SOD and CAT activities and GSH content observed in OVX and RIR group compared to sham group. E2 and 2ME treatment significantly ameliorated all parameters measured in OVX and RIR rats. On the other hand, Entacapone significantly decreased the effect of E2, with no effect on 2ME treatment. SIGNIFICANCE: E2 ameliorates RIR-induced kidney injury and this effect is mediated, at least in part, via its COMT-mediated conversion to 2ME. Thus, 2ME by the virtue of its pleiotropic pharmacological effects can be used as a safe and effective treatment of RIR injury.


Assuntos
2-Metoxiestradiol/farmacologia , Estradiol/farmacologia , Rim/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Traumatismo por Reperfusão/prevenção & controle , 2-Metoxiestradiol/metabolismo , Animais , Nitrogênio da Ureia Sanguínea , Peso Corporal/efeitos dos fármacos , Catecol O-Metiltransferase/metabolismo , Catecóis/farmacologia , Enzimas/metabolismo , Estradiol/farmacocinética , Feminino , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Rim/irrigação sanguínea , Rim/patologia , Nitrilas/farmacologia , Ovariectomia , Ratos Sprague-Dawley
8.
Cell Mol Life Sci ; 78(17-18): 6161-6200, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34333711

RESUMO

Despite advances in the understanding of the molecular mechanisms underlying the basic biology and pathogenesis of pediatric central nervous system (CNS) malignancies, patients still have an extremely unfavorable prognosis. Over the years, a plethora of natural and synthetic compounds has emerged for the pharmacologic intervention of the NF-kB pathway, one of the most frequently dysregulated signaling cascades in human cancer with key roles in cell growth, survival, and therapy resistance. Here, we provide a review about the state-of-the-art concerning the dysregulation of this hub transcription factor in the most prevalent pediatric CNS tumors: glioma, medulloblastoma, and ependymoma. Moreover, we compile the available literature on the anti-proliferative effects of varied NF-kB inhibitors acting alone or in combination with other therapies in vitro, in vivo, and clinical trials. As the wealth of basic research data continues to accumulate, recognizing NF-kB as a therapeutic target may provide important insights to treat these diseases, hopefully contributing to increase cure rates and lower side effects related to therapy.


Assuntos
Neoplasias do Sistema Nervoso Central/patologia , NF-kappa B/metabolismo , 2-Metoxiestradiol/química , 2-Metoxiestradiol/metabolismo , 2-Metoxiestradiol/uso terapêutico , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Neoplasias do Sistema Nervoso Central/tratamento farmacológico , Neoplasias do Sistema Nervoso Central/metabolismo , Criança , Glioma/metabolismo , Glioma/patologia , Proteínas Hedgehog/antagonistas & inibidores , Proteínas Hedgehog/metabolismo , Humanos , Meduloblastoma/metabolismo , Meduloblastoma/patologia , NF-kappa B/antagonistas & inibidores , Proteínas Wnt/antagonistas & inibidores , Proteínas Wnt/metabolismo
11.
Artigo em Chinês | MEDLINE | ID: mdl-31623040

RESUMO

Objective:To explore the signal pathway that mediates the effect of 2-methoxyestradiol(2ME2) on human laryngeal papilloma cell line, in terms of cell proliferation and neovascularization. Method:HIF-1α expression of human laryngeal papilloma cell line(Hs840. T) was interfered using siRNA, and the cells were then processed by 2ME2 in two concentrations. RT-PCR and ELISA were performed to detect the difference of HIF-1α in cells with normal or lower HIF-1α mRNA level, with ELISA test of excretory VEGF level and CCK8 test of cell viability. Result:The IC50of 2ME2 in Hs840. T was 0.309 µmol/L in terms of the inhibition effect of cell proliferation(P<0.01). Baseline level of intracellular HIF-1α was detectable, and procession of Hs840. T cells by 2ME2 of 0.4 µmol/L inhibited the transcription and expression of HIF-1α by(76.8±2.0)% and(68.6±3.5)% [vs blank group(100.0±2.7)% and(100.0±6.9)%, P<0.01]. VEGF excretion decreased to(50.8±2.1) and(28.1±4.0)% [vs blank group(100.0±3.1)%, P<0.01]after procession by 2ME2 of 0.2 µmol/L and 0.4 µmol/L. After the successful interference of HIF-1α by siRNA, the inhibition effect on cell proliferation by 2ME2 of 0.4 µmol/L decreased to(51.5±3.8)% [vs control group(65.7±1.7)%, P<0.01]. siRNA interference of HIF-1α lead to a decrease of HIF-α mRNA and protein level to(16.3±0.9)% and(7.4±0.8)% [vs cells not interfered(76.8±2.0)% vs(68.6±3.5)%, P<0.01]. Secretory VEGF dropped to(41.0±2.9)% [vs cells not interfered(28.1±4.0)%, P<0.05]. Conclusion:2ME2 has a significant inhibitory effect on human laryngeal cell line. The inhibition of cell proliferation was mediated by a lower level of HIF-1α and therefore lower VEGF. 2ME2 might serve as a novel potential therapy for patients of recurrent respiratory papillomatosis.


Assuntos
2-Metoxiestradiol/metabolismo , Linhagem Celular Tumoral , Estradiol , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Laríngeas , Papiloma , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Steroids ; 144: 21-29, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30738071

RESUMO

An estradiol metabolite, 2-methoxyestradiol (2ME), has emerged as an important regulator of ovarian physiology. 2ME is recognized as a potent anti-angiogenic agent in clinical trials and laboratory studies. However, little is known about its molecular actions and its endogenous targets. 2ME is produced by human ovarian cells during the normal menstrual cycle, being higher during regression of the corpus luteum, and is postulated to be involved in the anti-angiogenic process that plays out during luteolysis. We utilized cell biology techniques to understand the molecular mechanism of 2ME anti-angiogenic effects on human granulosa luteal cells. The principal effect of 2ME was to alter Hypoxia Inducible Factor 1A (HIF1A) sub-cellular localization. Molecular modelling and multiple bioinformatics tools indicated that 2ME impairs Hypoxia Inducible Factor complex (HIF) nuclear translocation by binding to a buried pocket in the HIF1A Per Arnt Sim (PAS)-B domain. Binding of 2ME to HIF1A protein is predicted to perturb HIF1A-Hypoxia Inducible Factor B (HIFB) interaction, a key step in HIF nuclear translocation, preventing the transcriptional actions of HIF, including Vascular Endotelial Growth Factor (VEGF) gene activation. To our knowledge, 2ME is the first putative HIF endogenous ligand characterized with anti-angiogenic activity. This postulate has important implications for reproduction, because angiogenic processes are critical for ovarian follicular development, ovulation and corpus luteum regression. The present research could contribute to the development of novel pharmacological approaches for controlling HIF activity in human reproductive diseases.


Assuntos
2-Metoxiestradiol/metabolismo , 2-Metoxiestradiol/farmacologia , Biologia Computacional , Subunidade alfa do Fator 1 Induzível por Hipóxia/química , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Simulação de Dinâmica Molecular , Linhagem Celular , Feminino , Humanos , Células Lúteas/efeitos dos fármacos , Células Lúteas/metabolismo , Ligação Proteica , Domínios Proteicos , Multimerização Proteica/efeitos dos fármacos , Estrutura Quaternária de Proteína
13.
Steroids ; 136: 47-55, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29772242

RESUMO

The endogenous steroid 2-methoxyestradiol (1) has attracted a great interest as a lead compound towards the development of new anti-cancer drugs. Herein, the synthesis, molecular modeling, anti-proliferative and anti-angiogenic effects of ten 2-ethyl and four 2-methoxy analogs of estradiol are reported. The ethyl group was introduced to the steroid A-ring using a novel Friedel-Crafts alkylation protocol. Several analogs displayed potent anti-proliferative activity with IC50-values in the submicromolar range towards the CEM human leukemia cancer cell line. As such, all of these compounds proved to be more active than the lead compound 2-methoxyestradiol (1) in these cells. The six most cytostatic analogs were also tested as anti-angiogenic agents using an in vitro tube formation assay. The IC50-values were determined to be in the range of 0.1 µM ±â€¯0.03 and 1.1 µM ±â€¯0.2. These six compounds were also modest inhibitors against tubulin polymerization with the most potent inhibitor was 14b (IC50 = 2.1 ±â€¯0.1 µM). Binding studies using N,N'-ethylene-bis(iodoacetamide) revealed that neither14a or 14b binds to the colchicine binding site in the tubulin protein, in contrast to 2-methoxyestradiol (1). These observations were supported by molecular modeling studies. Results from a MDA-MB-231 cell cycle assay showed that both 10e and 14b gave accumulation in the G2/M phase resulting in induction of apoptosis. The results presented herein shows that the novel analogs reported exhibit their anticancer effects via several modes of action.


Assuntos
2-Metoxiestradiol/síntese química , 2-Metoxiestradiol/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Simulação de Acoplamento Molecular , 2-Metoxiestradiol/química , 2-Metoxiestradiol/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Técnicas de Química Sintética , Humanos , Conformação Proteica , Relação Estrutura-Atividade , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo
14.
Otolaryngol Head Neck Surg ; 158(3): 566-570, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29110580

RESUMO

Objective We aim to explore the correlation between serum and tissue 2-methoxyestradiol (2-ME-2) levels and recurrence of juvenile-onset respiratory papillomatosis (JORRP). Study Design Retrospective cohort studies. Settings Laboratory of Otolaryngology, Department of Head and Neck Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University. Subjects and Methods Sixty-four patients diagnosed with JORRP in our department from January 2007 to December 2012 were enrolled. Patients were divided into recurrence and nonrecurrence groups, with 32 patients in each group. ELISA detected the concentration of 2-ME-2 in serum and tissue samples collected during the first surgical procedure. Mann-Whitney analysis, receiver operating characteristic curves, logistic regression model, and Kaplan-Meier method were used for data processing. Results There was no difference in the serum 2-ME-2 concentration between the groups ( P = .237), while the tissue 2-ME-2 concentration of the recurrent group was significantly lower than that of the nonrecurrence group ( P = .0001). When the area under the curve was 0.752, the cutoff value of tissue 2-ME-2 at 670.02 pg/mL yielded the highest predictive sensitivity (71.9%) and specificity (71.9%). Regrouped by this cutoff point, patients with a lower tissue 2-ME-2 level (n = 26) had shorter disease-free survival and a higher recurrence odds ratio than patients with a higher tissue 2-ME-2 level (n = 38; P = .0408, odds ratio = 7.667). Conclusion A low tissue 2-ME-2 level is associated with a higher recurrence rate of JORRP. Tissue 2-ME-2 may be an effective target for JORRP treatment and a convenient measure for recurrence monitoring.


Assuntos
2-Metoxiestradiol/metabolismo , Neoplasias Laríngeas/metabolismo , Neoplasias Laríngeas/patologia , Papiloma/metabolismo , Papiloma/patologia , Biomarcadores/metabolismo , Criança , Pré-Escolar , China , Ensaio de Imunoadsorção Enzimática , Humanos , Lactente , Neoplasias Laríngeas/cirurgia , Laringoscopia , Papiloma/cirurgia , Recidiva , Estudos Retrospectivos
15.
Sci Rep ; 7(1): 7927, 2017 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-28801594

RESUMO

2-methoxyestradiol (2-ME), an estrogen metabolite generated via catechol-o-methyltransferase (COMT), is multifunctional methoxy-catechol. Here, we report that COMT deficiency leads to glucose intolerance and 2-ME rescues COMT-deficient-associated metabolic defects. Liver COMT protein was suppressed in high fat diet (HFD)-fed or in pregnant mice. COMT suppression, by Ro41-0960 or siRNA, in HFD fed mice or in pregnant mice exacerbated glucose intolerance; 2-ME intervention ameliorated these defects. 2-ME effects on glucose tolerance were associated with AMPK phosphorylation in the liver and in islet cells. Metformin restored liver COMT protein levels, and metformin-induced liver AMPK phosphorylation was abolished by COMT inhibition. The amelioration in glucose tolerance by 2-ME was associated with biphasic insulin secretion in an environment-dependent manner. 2-ME-induced insulin secretion was associated with the AMPK phosphorylation, PDX-1 phosphorylation, and MST-1 suppression in MIN-6 cells. Furthermore 2-ME displayed PPARγ agonist-like activity. These results suggest that COMT is an enzyme to maintain glucose homeostasis and 2-ME is a potential endogenous multi-target anti-diabetic candidate.


Assuntos
2-Metoxiestradiol/metabolismo , Catecol O-Metiltransferase/deficiência , Glucose/metabolismo , Homeostase , Animais , Dieta Hiperlipídica , Feminino , Intolerância à Glucose , Camundongos , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...