Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
1.
Br J Cancer ; 130(4): 585-596, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38172534

RESUMO

BACKGROUND: The enriched proteins within in vitro fertilisation (IVF)-generated human embryonic microenvironment could reverse progestin resistance in endometrial cancer (EC). METHODS: The expression of thymic stromal lymphopoietin (TSLP) in EC was evaluated by immunoblot and IHC analysis. Transcriptome sequencing screened out the downstream pathway regulated by TSLP. The role of TSLP, androgen receptor (AR) and KANK1 in regulating the sensitivity of EC to progestin was verified through a series of in vitro and in vivo experiments. RESULTS: TSLP facilitates the formation of a BMP4/BMP7 heterodimer, resulting in activation of Smad5, augmenting AR signalling. AR in turn sensitises EC cells to progestin via KANK1. Downregulation of TSLP, loss of AR and KANK1 in EC patients are associated with tumour malignant progress. Moreover, exogenous TSLP could rescue the anti-tumour effect of progestin on mouse in vivo xenograft tumour. CONCLUSIONS: Our findings suggest that TSLP enhances the sensitivity of EC to progestin through the BMP4/Smad5/AR/KANK1 axis, and provide a link between embryo development and cancer progress, paving the way for the establishment of novel strategy overcoming progestin resistance using embryo original factors.


Assuntos
Neoplasias do Endométrio , Linfopoietina do Estroma do Timo , Animais , Feminino , Humanos , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Citocinas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Progestinas/farmacologia , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Transdução de Sinais , Microambiente Tumoral
2.
J Med Virol ; 96(3): e29468, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38415499

RESUMO

Cervical human papillomavirus (HPV) infection is believed to increase the risks of pregnancy failure and abortion, however, whether the uterine cavity HPV infection reduces pregnancy rate or increases miscarriage rate remains unclarified in infertile women undergoing assisted reproductive technology (ART) treatment. Therefore, we aimed to assess ART outcomes in the presence of intrauterine HPV. This was a hospital-based multicenter (five reproductive medicine centers) matched cohort study. This study involved 4153 infertile women undergoing in vitro fertilization (IVF) or intracytoplasmic sperm injection treatment in five reproductive medicine centers between October 2018 and 2020. The spent embryo transfer media sample with endometrium tissue were collected and performed with flow-through hybridization and gene chips to detect HPV DNA. According to basic characteristics, HPV-positive and negative patients were matched in a ratio of 1:4 by age, body mass index transfer timing, transfer type, and number of embryos transferred. The primary outcome was pregnancy and clinical miscarriage rates in the transfer cycle underwent HPV detection. 92 HPV-positive and 368 HPV-negative patients were screened and analyzed statistically. Univariate analysis showed uterine cavity HPV infection resulted in lower rates of ongoing pregnancy (31.5% vs. 44.6%; p = 0.023), implantation (32.3% vs. 43.1%; p = 0.026), biochemical pregnancy (47.8% vs. 62.5%; p = 0.010), and clinical pregnancy (40.2% vs. 54.3%; p = 0.015) compared with HPV negative group. The infertile female with positive HPV also had a slightly higher frequency of biochemical miscarriage (15.9% vs. 13.0%; p = 0.610) and clinical miscarriage (24.3% vs. 15.5%; p = 0.188). These findings suggest that HPV infection in the uterine cavity is a high risk for ART failure. HPV screening is recommended before ART treatment, which may be benefit to improving pregnancy outcome.


Assuntos
Aborto Espontâneo , Infertilidade Feminina , Infecções por Papillomavirus , Gravidez , Humanos , Masculino , Feminino , Infecções por Papillomavirus/diagnóstico , Infertilidade Feminina/terapia , Papillomavirus Humano , Estudos de Coortes , Sêmen , Transferência Embrionária/métodos , Técnicas de Reprodução Assistida , Fertilização in vitro , Falha de Tratamento
3.
Lab Invest ; 102(12): 1335-1345, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36038734

RESUMO

Progestin resistance is the main obstacle for the conservative therapy to maintain fertility in women with endometrial cancer. Brusatol was identified as an inhibitor of the NRF2 pathway; however, its impact on progestin resistance and the underlying mechanism remains unclear. Here, we found that brusatol sensitized endometrial cancer to progestin by suppressing NRF2-TET1-AKR1C1-mediated progestin metabolism. Brusatol transcriptionally suppressed AKR1C1 via modifying the hydroxymethylation status in its promoter region through TET1 inhibition. Suppression of AKR1C1 by brusatol resulted in decreased progesterone catabolism and maintained potent progesterone to inhibit endometrial cancer growth. This inhibition pattern has also been found in the established xenograft mouse and organoid models. Aberrant overexpression of AKR1C1 was found in paired endometrial hyperplasia and cancer samples from the same individuals with progestin resistance, whereas attenuated or loss of AKR1C1 was observed in post-treatment samples with well progestin response as compared with paired pre-treatment tissues. Our findings suggest that AKR1C1 expression pattern may serve as an important biomarker of progestin resistance in endometrial cancer.


Assuntos
Hiperplasia Endometrial , Neoplasias do Endométrio , Humanos , Feminino , Camundongos , Animais , Hiperplasia Endometrial/tratamento farmacológico , Hiperplasia Endometrial/genética , Progestinas/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Progesterona , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Oxigenases de Função Mista/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas de Ligação a DNA
4.
Small ; 18(29): e2202782, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35754168

RESUMO

The precise and comprehensive manipulation of the component, size, and geometric nano-architecture of platinum-based electrocatalysts into porous and hollow structure can effectively impart the catalysts with substantially improved electrochemical performance, yet remain formidably challenging. Herein, a straightforward fabrication of porous platinum-copper alloyed nanobowls (abbreviated as Pt3 Cu NBs hereafter) assembled by ultrafine nanoparticles (≈2.9 nm) via a one-pot hydrothermal approach with the assistance of a structure-directing agent of N,N'-methylenebisacrylamide (MBAA) is reported. The involvement of MBAA plays a decisive role in the formation of Pt-MBAA complex solid nanospheres, which serve as the self-sacrificial reactive template for the deposition/growth of Pt3 Cu nanoparticles and the eventual formation of the asymmetric open-shelled nanobowls. Benefitting from the 3D sufficient accessibility of exterior/interior surfaces, high atom-utilization efficiency, and PtCu bimetallic alloy synergy, the self-supported Pt3 Cu NBs demonstrate remarkably enhanced activity, better anti-poisoning capability, and reinforced robustness for the methanol oxidation reaction (MOR) as compared with the commercial Pt black benchmark, exhibiting great application promises in practical fuel cell systems. It is envisaged that the innovative self-templated synthetic strategy outlined here may provide a perspective to design a range of porous bowl-shaped high-performance nanocatalysts.

5.
Reprod Biomed Online ; 45(6): 1182-1187, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36085270

RESUMO

RESEARCH QUESTION: What is the effect of letrozole use in patients undergoing frozen embryo transfer (FET) with normal ovulation? Although the number of FETs is increasing, an optimal protocol for FET (particularly vitrified-warmed embryo transfer) is yet to be determined. The aim of this study was to evaluate letrozole use on patients with normal menstrual cycles compared with hormone replacement therapy (HRT) cycles and natural cycles. DESIGN: The study involved 2849 patients. Patients were divided into three groups: HRT cycle (n = 2115), letrozole cycle (n = 532) and natural cycle (n = 202). Inverse probability of treatment weighting aimed to equate each group according to measured baseline covariates to achieve a comparison with reduced selection bias and live birth rate as main pregnancy outcome was analysed. RESULTS: In the crude analysis, the letrozole group had a higher live birth rate compared with the HRT cycle (OR 1.18, 95% CI 1.06 to 1.33) and natural cycle (OR 1.24, 95% CI 1.11 to 1.41); after adjusting for confounding factors, live birth rate was consistently higher in the letrozole group. Moreover, the biochemical pregnancy, clinical pregnancy, ongoing pregnancy and full-term delivery rates were higher in the letrozole group. CONCLUSION: For infertile women with normal menstrual cycle undergoing FET, mildly stimulated cycles with letrozole present a relatively large advantage compared with HRT cycle and natural cycle, with higher live birth pregnancy, indicating that letrozole administration could improve pregnancy outcomes in this population.


Assuntos
Infertilidade Feminina , Resultado da Gravidez , Feminino , Gravidez , Humanos , Letrozol , Infertilidade Feminina/terapia , Infertilidade Feminina/epidemiologia , Taxa de Gravidez , Indução da Ovulação/métodos , Criopreservação/métodos , Transferência Embrionária/métodos , Nascido Vivo , Ovulação , Estudos Retrospectivos
6.
J Assist Reprod Genet ; 37(9): 2053-2079, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32562095

RESUMO

PURPOSE: Patients with Klinefelter syndrome (KS) who receive assisted reproductive technology (ART) treatment often experience poor pregnancy rates due to decreased fertilization, cleavage, and implantation rates and even an increased miscarriage rate. Mounting evidence from recent studies has shown that various technological advances and approaches could facilitate the success of ART treatment for KS patients. In this review, we summarize the methods for guiding KS patients during ART and for developing optimal strategies for preserving fertility, improving pregnancy rate and live birth rate, and avoiding the birth of KS infants. METHODS: We searched PubMed and Google Scholar publications related to KS patients on topics of controlled ovarian stimulation protocols, sperm extraction, fertility preservation, gamete artificial activation, round spermatid injection (ROSI), and non-invasive prenatal screening (PGD) methods. RESULTS: This review outlines the different ovulation-inducing treatments for female partners according to the individual sperm status in the KS patient. We further summarize the methods of retrieving sperm, storing, and freezing rare sperm. We reviewed different methods of gamete artificial activation and discussed the feasibility of ROSI for sterile KS patients who absolutely lack sperm. The activation of eggs in the process of intracytoplasmic sperm injection and non-invasive PGD are urgently needed to prevent the birth of KS infants. CONCLUSION: The integrated strategies will pave the way for the establishment of ART treatment approaches and improve the clinical outcome for KS patients.


Assuntos
Implantação do Embrião/genética , Síndrome de Klinefelter/terapia , Técnicas de Reprodução Assistida/tendências , Coeficiente de Natalidade , Feminino , Preservação da Fertilidade/tendências , Humanos , Síndrome de Klinefelter/genética , Síndrome de Klinefelter/patologia , Masculino , Gravidez , Taxa de Gravidez , Injeções de Esperma Intracitoplásmicas/tendências
7.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 51(3): 371-375, 2020 May.
Artigo em Chinês | MEDLINE | ID: mdl-32543145

RESUMO

OBJECTIVE: To observe the effect of ginsenoside Rg1 on the acute lung injury of sepsis in combination with the antibiotic imipenem in a mouse model of sepsis that induced by cecal puncture. METHODS: C57BL/6 mice were used to establish the model of sepsis. The model mice were randomly divided into model group, imipenem group, ginsenoside Rg1 group, and ginsenoside Rg1+imipenem group, 10 mice in each group. Another 10 mice were set as control group. Sham operation was performed in the mice of control group. Each mice was intraperitoneally injected the corresponding drug in 2 h, 26 h and 50 h after surgery for three times. They were 2 h after surgery, 26 h after surgery and 50 h after surgery. 2 h after the last administration, the oxygenation index of the arterial blood was measured, the lung tissue was taken to measure lung wet/dry ratio (W/D), and HE staining was used to observe the lung inflammation. The ELISA kits were used to detect the levels of interleukin (IL)- 1ß, IL-6, tumor necrosis factor (TNF)-α and nuclear factor-kappa B (NF-κB) inalveolar lavage fluid. Western blot and immunohistochemical staining were used to detect NF-κB p65 expression in lung tissues. RESULTS: The drug-administered groups significantly reduced the W/D of the lungs in the septic mice and increased the oxygenation index in the blood ( P<0.01), and decreased the inflammation in lung and the levels of IL-1ß, IL-6, TNF-α and NF-κB in the alveolar lavage fluid ( P<0.01), and decreased the expression of NF-κB p65 in lung tissue ( P<0.01). When ginsenoside Rg1 was combined with imipenem, the above indicators were closer to the control group than that in the ginsenoside Rg1 and imipenem groups. CONCLUSION: Rg1 can significantly inhibit lung inflammation in septic mice. It has a better therapeutic effect when combined with antibiotics.


Assuntos
Lesão Pulmonar Aguda , Ginsenosídeos , Sepse , Lesão Pulmonar Aguda/complicações , Lesão Pulmonar Aguda/tratamento farmacológico , Animais , Antibacterianos/uso terapêutico , Pulmão , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B , Sepse/complicações , Sepse/tratamento farmacológico , Fator de Necrose Tumoral alfa
8.
Glycobiology ; 28(2): 61-68, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29206917

RESUMO

GRP78 (an Mr 78 kDa calcium dependent glucose binding protein) is located in ER lumen. It functions as ER chaperone and translocates proteins for glycosylation at the asparagine residue present in the sequon Asn-X-Ser/Thr. Paraffin sections from N-glycosylation inhibitor tunicamycin treated ER-/PR-/HER2+ (double negative) breast tumor in athymic nude mice exhibited reduced N-glycan but increased GRP78 expression. We have evaluated the effect of tunicamycin on cellular localization of GRP78 in metastatic human breast cancer cells MDA-MB-231 (ER-/PR-/HER2-). Tunicamycin inhibited cell proliferation in a time and dose-dependent manner. Nonmetastatic estrogen receptor positive (ER+) MCF-7 breast cancer cells were also equally effective. GRP78 expression (protein and mRNA) was higher in tunicamycin (1.0 µg/mL) treated MCF-7 and MDA-MB-231 cells. GRP78 is an ER stress marker, so we have followed its intracellular localization using immunofluorescence microscopy after subjecting the cancer cells to various stress conditions. Unfixed cells stained with either FITC-conjugated Concanavalin A (Con A) or Texas-red conjugated wheat germ agglutinin (WGA) exhibited surface expression of N-glycans but not GRP78. GRP78 became detectable only after a brief exposure of cells to ice-cold methanol. Western blotting did not detect GRP78 in conditioned media of cancer cells whereas it did for MMP-1. The conclusion, GRP78 is expressed neither on the outer-leaflet of the (ER-/PR-/HER2-) human breast cancer cells nor it is secreted into the culture media during tunicamycin-induced ER stress. Our study therefore suggests strongly that anti-tumorigenic action of tunicamycin can be modeled to develop next generation cancer therapy, i.e., glycotherapy for treating breast and other sold tumors.


Assuntos
Estresse do Retículo Endoplasmático , Proteínas de Choque Térmico/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Chaperona BiP do Retículo Endoplasmático , Feminino , Glicosilação , Proteínas de Choque Térmico/genética , Humanos , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Tunicamicina/toxicidade
9.
Adv Exp Med Biol ; 1112: 223-244, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30637701

RESUMO

Dolichol phosphate mannose synthase (DPMS) is an inverting GT-A-folded enzyme and classified as GT2 by CAZy. DPMS sequence carries a metal-binding DXD motif, a PKA motif, and a variable number of hydrophobic domains. Human and bovine DPMS possess a single transmembrane domain, whereas that from S. cerevisiae and A. thaliana carry multiple transmembrane domains and are superimposable. The catalytic activity of DPMS is documented in all spheres of life, and the 32kDa protein is uniquely regulated by protein phosphorylation. Intracellular activation of DPMS by cAMP signaling is truly due to the activation of the enzyme and not due to increased Dol-P level. The sequence of DPMS in some species also carries a protein N-glycosylation motif (Asn-X-Ser/Thr). Apart from participating in N-glycan biosynthesis, DPMS is essential for the synthesis of GPI anchor as well as for O- and C-mannosylation of proteins. Because of the dynamic nature, DPMS actively participates in cellular proliferation enhancing angiogenesis and breast tumor progression. In fact, overexpression of DPMS in capillary endothelial cells supports increased N-glycosylation, cellular proliferation, and enhanced chemotactic activity. These are expected to be completely absent in congenital disorders of glycosylation (CDGs) due to the silence of DPMS catalytic activity. DPMS has also been found to be involved in the cross talk with N-acetylglucosaminyl 1-phosphate transferase (GPT). Inhibition of GPT with tunicamycin downregulates the DPMS catalytic activity quantitatively. The result is impairment of surface N-glycan expression, inhibition of angiogenesis, proliferation of human breast cancer cells, and induction of apoptosis. Interestingly, nano-formulated tunicamycin is three times more potent in inhibiting the cell cycle progression than the native tunicamycin and is supported by downregulation of the ratio of phospho-p53 to total-p53 as well as phospho-Rb to total Rb. DPMS expression is also reduced significantly. However, nano-formulated tunicamycin does not induce apoptosis. We, therefore, conclude that DPMS could become a novel target for developing glycotherapy treating breast tumor in the clinic.


Assuntos
Neoplasias da Mama/enzimologia , Manosiltransferases/metabolismo , Neovascularização Patológica/enzimologia , Neoplasias da Mama/patologia , Ciclo Celular , Glicosilação , Humanos , Neovascularização Patológica/patologia , Tunicamicina/farmacologia
10.
Cell Physiol Biochem ; 42(1): 185-197, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28535511

RESUMO

BACKGROUND AND AIM: Increasing evidence shows that the calpain regulatory subunit Capn4 can modulate the proliferation and metastasis of cancer cells, and plays an important role in the development of malignant tumors. However, there is no information on the clinical significance of Capn4 in epithelial ovarian carcinoma (EOC) or the molecular mechanisms by which Capn4 promotes the growth and metastasis of EOC. Therefore, the aim of this study was to clarify the role of Capn4 in EOC. METHODS: We evaluated Capn4 and osteopontin (OPN) expression in EOC cell lines and tissues from patients with ovarian cancer by western blotting and immunohistochemical analysis. We then created cell lines with downregulated and upregulated Capn4 expression, using Capn4-targeting small interfering RNA and a pcDNA3.1-Capn4 overexpression vector, respectively, to investigate its function in EOC in vitro. In addition, we investigated the potential mechanism underlying the function of Capn4 by examining the effect of modifying Capn4 expression on Wnt/ß-catenin signaling pathway-related genes by western blotting. RESULTS: Capn4 was overexpressed in clinical EOC tissues compared with that in normal ovarian epithelial tissue, and was associated with poor clinical outcomes. Upon silencing or overexpressing Capn4 in EOC cells, we concluded that Capn4 promotes cell proliferation and migration in vitro. Furthermore, Capn4 promoted EOC metastasis by interacting with the Wnt/ß-catenin signaling pathway to upregulate OPN expression. CONCLUSION: Our study indicates that Capn4 plays a critical role in the progression and metastasis of EOC, and could be a potential therapeutic target for EOC management.


Assuntos
Calpaína/metabolismo , Neoplasias Epiteliais e Glandulares/patologia , Osteopontina/metabolismo , Neoplasias Ovarianas/patologia , Via de Sinalização Wnt , beta Catenina/metabolismo , Calpaína/antagonistas & inibidores , Calpaína/genética , Carcinoma Epitelial do Ovário , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação para Baixo , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Pessoa de Meia-Idade , Metástase Neoplásica , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Epiteliais e Glandulares/mortalidade , Osteopontina/antagonistas & inibidores , Osteopontina/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/mortalidade , Plasmídeos/genética , Plasmídeos/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Regulação para Cima , Proteínas Wnt/metabolismo , beta Catenina/antagonistas & inibidores , beta Catenina/genética
11.
Biochem Biophys Res Commun ; 482(4): 857-862, 2017 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-27889612

RESUMO

Large amount of clinical evidence has demonstrated that insulin resistance is closely related to oncogenesis of endometrial cancer (EC). Despite recent studies showed the up-regulatory role of insulin in G protein-coupled estrogen receptor (GPER/GPR30) expression, GPER expression was not decreased compared to control when insulin receptor was blocked even in insulin treatment. The purpose of this study was to explore the possible mechanism by which insulin up-regulates GPER that drives EC cell proliferation. For this purpose, we first investigated the GPER expression in tissues of endometrial lesions, further explored the effect of GPER on EC cell proliferation in insulin resistance context. Then we analyzed the role of Ten-Eleven Translocation 1 (TET1) in insulin-induced GEPR expression and EC cell proliferation. The results showed that GPER was highly expressed in endometrial atypical hyperplasia and EC tissues. Mechanistically, insulin up-regulated TET1 expression and the latter played an important role in up-regulating GPER expression and activating PI3K/AKT signaling pathway. TET1 mediated GPER up-regulation was another mechanism that insulin promotes EC cell proliferation.


Assuntos
Proliferação de Células , Neoplasias do Endométrio/patologia , Endométrio/patologia , Insulina/metabolismo , Transdução de Sinais , Linhagem Celular Tumoral , Neoplasias do Endométrio/metabolismo , Endométrio/metabolismo , Feminino , Humanos , Resistência à Insulina , Oxigenases de Função Mista/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
12.
Mol Carcinog ; 56(6): 1543-1553, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28112439

RESUMO

Previously, we have demonstrated that NRF2 plays a key role in mediating cisplatin resistance in ovarian cancer. To further explore the mechanism underlying NRF2-dependent cisplatin resistance, we stably overexpressed or knocked down NRF2 in parental and cisplatin-resistant human ovarian cancer cells, respectively. These two pairs of stable cell lines were then subjected to microarray analysis, where we identified 18 putative NRF2 target genes. Among these genes, ABCF2, a cytosolic member of the ABC superfamily of transporters, has previously been reported to contribute to chemoresistance in clear cell ovarian cancer. A detailed analysis on ABCF2 revealed a functional antioxidant response element (ARE) in its promoter region, establishing ABCF2 as an NRF2 target gene. Next, we investigated the contribution of ABCF2 in NRF2-mediated cisplatin resistance using our stable ovarian cancer cell lines. The NRF2-overexpressing cell line, containing high levels of ABCF2, was more resistant to cisplatin-induced apoptosis compared to its control cell line; whereas the NRF2 knockdown cell line with low levels of ABCF2, was more sensitive to cisplatin treatment than its control cell line. Furthermore, transient overexpression of ABCF2 in the parental cells decreased apoptosis and increased cell viability following cisplatin treatment. Conversely, knockdown of ABCF2 using specific siRNA notably increased apoptosis and decreased cell viability in cisplatin-resistant cells treated with cisplatin. This data indicate that the novel NRF2 target gene, ABCF2, plays a critical role in cisplatin resistance in ovarian cancer, and that targeting ABCF2 may be a new strategy to improve chemotherapeutic efficiency.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Antineoplásicos/farmacologia , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Fator 2 Relacionado a NF-E2/genética , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Elementos de Resposta Antioxidante , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Humanos , Ovário/efeitos dos fármacos , Ovário/metabolismo , Regiões Promotoras Genéticas , Regulação para Cima
13.
Glycoconj J ; 34(4): 467-479, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28616799

RESUMO

N-glycans provide structural and functional stability to asparagine-linked (N-linked) glycoproteins, and add flexibility. Glycan biosynthesis is elaborative, multi-compartmental and involves many glycosyltransferases. Failure to assemble N-glycans leads to phenotypic changes developing infection, cancer, congenital disorders of glycosylation (CDGs) among others. Biosynthesis of N-glycans begins at the endoplasmic reticulum (ER) with the assembly of dolichol-linked tetra-decasaccharide (Glc3Man9GlcNAc2-PP-Dol) where dolichol phosphate mannose synthase (DPMS) plays a central role. DPMS is also essential for GPI anchor biosynthesis as well as for O- and C-mannosylation of proteins in yeast and in mammalian cells. DPMS has been purified from several sources and its gene has been cloned from 39 species (e.g., from protozoan parasite to human). It is an inverting GT-A folded enzyme and classified as GT2 by CAZy (carbohydrate active enZyme; http://www.cazy.org ). The sequence alignment detects the presence of a metal binding DAD signature in DPMS from all 39 species but finds cAMP-dependent protein phosphorylation motif (PKA motif) in only 38 species. DPMS also has hydrophobic region(s). Hydropathy analysis of amino acid sequences from bovine, human, S. crevisiae and A. thaliana DPMS show PKA motif is present between the hydrophobic domains. The location of PKA motif as well as the hydrophobic domain(s) in the DPMS sequence vary from species to species. For example, the domain(s) could be located at the center or more towards the C-terminus. Irrespective of their catalytic similarity, the DNA sequence, the amino acid identity, and the lack of a stretch of hydrophobic amino acid residues at the C-terminus, DPMS is still classified as Type I and Type II enzyme. Because of an apparent bio-sensing ability, extracellular signaling and microenvironment regulate DPMS catalytic activity. In this review, we highlight some important features and the molecular diversities of DPMS.


Assuntos
Manosiltransferases/química , Manosiltransferases/metabolismo , Sequência de Aminoácidos , Animais , Biocatálise , Glicosilação , Saúde , Humanos
14.
Arch Gynecol Obstet ; 295(4): 987-995, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28239742

RESUMO

PURPOSE: Stroma-derived factor-1 (SDF-1) and its receptor C-X-C chemokine receptor-4 (CXCR4) are involved in human endometrial carcinoma (EC) progression. CXCR7 is another important receptor of SDF-1 and has a higher affinity with SDF-1 compared with that of CXCR4. This paper aims to study the effects of the SDF-1/CXCR7 axis on the growth and invasion ability of EC cells. METHODS: CXCR7 expression was evaluated by quantitative RT-PCR, immunohistochemistry, immunocytochemistry and Western blotting in EC cell lines and 30 cases of primary EC tissue from patients. EC cell line proliferation and migration were assessed following knockdown of CXCR7 by MTT and transwell assays. RESULTS: The results showed that CXCR7 was highly expressed at both mRNA and protein levels in the EC cells and tissue. siCXCR7 effectively silenced CXCR7 in Ishikawa and AN3CA cells. Treatment with 17ß-oestradiol (17ß-E2) significantly increased the levels of CXCR7 and SDF-1 in Con, siCon and siCXCR7 treated Ishikawa. siCXCR7 persistently inhibited CXCR7 expression, even in cells treated with 17ß-E2. Moreover, in vitro functional analyses, silencing CXCR7 resulted in decreased proliferation in Ishikawa and AN3CA cells. Treatment with 17ß-E2 and SDF-1 significantly promoted the growth and migration in siCon treated Ishikawa and AN3CA. Interestingly, in response to 17ß-E2 and SDF-1 stimulation, siCXCR7 continuously inhibited the growth and invasion of Ishikawa and AN3CA cells. CONCLUSION: Our results indicate that SDF-1/CXCR7 plays a positive role in the proliferation and invasion of EC cells. CXCR7 inhibition treatment may provide a promising strategy for anti-tumour therapy for EC.


Assuntos
Proliferação de Células/genética , Quimiocina CXCL12/fisiologia , Neoplasias do Endométrio/patologia , Células Endoteliais/patologia , Invasividade Neoplásica/genética , Receptores CXCR/fisiologia , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quimiocina CXCL12/genética , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Estradiol/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imuno-Histoquímica , RNA Mensageiro/metabolismo , Receptores CXCR/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
15.
Ann Rheum Dis ; 75(12): 2124-2132, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27165176

RESUMO

OBJECTIVE: Ankylosing spondylitis (AS) is a chronic inflammatory disease of unknown origin in which interleukin (IL) 17 has been genetically and therapeutically recognised as a key player. Identification of the cellular sources and inducers of IL-17 is crucial in our understanding of the drivers of inflammation in AS. Recently, mucosal-associated invariant T (MAIT) cells have been implicated in autoimmune diseases. Their gut origin, effector phenotype and expression of multiple AS-associated genes, such as IL7R and IL23R, makes them potential contributors to the pathogenesis of AS. METHODS: Mononuclear cells from patients with AS, healthy controls (HCs) and patients with rheumatoid arthritis were isolated from blood and synovial fluid (SF). Flow cytometry was used to identify MAIT cells. Phenotype was assessed by intracellular staining for cytokines and granzyme. Function was assessed by antigen-specific stimulation using Salmonella, or antigen non-specific activation via priming with IL-7 or IL-23. RESULTS: MAIT cells were reduced in frequency in the blood of patients with AS compared with HCs, yet patients with AS had an elevated frequency IL-17A+ MAIT cells. There was an enrichment of MAIT cells in SF, which had an exaggerated IL-17 phenotype. IL-17 elevation in AS MAIT cells was dependent on priming with IL-7 but not IL-23 or antigen stimulation. The AS-associated IL7R single nucleotide polymorphism (SNP), rs11742270, had no effect on IL-7R expression or function in the experiments performed. CONCLUSIONS: This study reveals a potential role for MAIT cells in patients with AS and is the first linking IL-7 to the elevated IL-17 profile in patients through the AS-associated risk gene IL7R.


Assuntos
Interleucina-17/metabolismo , Interleucina-7/metabolismo , Ativação Linfocitária , Células T Invariantes Associadas à Mucosa/imunologia , Espondilite Anquilosante/imunologia , Adulto , Idoso , Artrite Reumatoide/sangue , Artrite Reumatoide/imunologia , Estudos de Casos e Controles , Feminino , Citometria de Fluxo , Humanos , Interleucina-23/metabolismo , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Receptores de Interleucina-7/genética , Receptores de Interleucina-7/metabolismo , Espondilite Anquilosante/sangue , Espondilite Anquilosante/genética , Líquido Sinovial/citologia , Células Th17/imunologia
16.
Tumour Biol ; 37(7): 9671-80, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26801672

RESUMO

NANOG is a key transcription factor that is overexpressed and plays an important role in various cancers. Its overexpression is associated with highly tumorigenic, drug-resistant, and poor prognosis. However, the underlying mechanism of action of NANOG in ovarian cancer remains unclear. Epithelial-mesenchymal transition (EMT), which is a critical process in cancer invasion and metastasis, is also associated with drug resistance. We determined whether NANOG is associated with EMT and chemoresistance in epithelial ovarian cancer cells. NANOG expression was increased in epithelial ovarian cancer cells (HEY and SKOV3) compared with normal epithelial ovarian cells (Moody). Low expression of NANOG increased the expression of E-cadherin and decreased the expression of vimentin, ß-catenin, and Snail. Furthermore, the cell migration and invasion abilities were decreased. The multidrug resistance genes MDR-1 and GST-π were also downregulated when NANOG was lowly expressed. The cells that were transfected with the si-NANOG plasmid were more sensitive to cisplatin compared with the cells that were transfected with empty vector. The data demonstrated that Stat3 was correlated with NANOG-mediated EMT and drug resistance. The silencing of Stat3 expression abrogated NANOG-mediated EMT changes and increased the sensitivity of the cells to chemotherapy. These results suggest that NANOG mediates EMT and drug resistance through activation of the Stat3 pathway in epithelial ovarian cancer.


Assuntos
Resistência a Múltiplos Medicamentos/genética , Resistencia a Medicamentos Antineoplásicos/genética , Transição Epitelial-Mesenquimal/genética , Proteína Homeobox Nanog/genética , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Ovarianas/genética , Fator de Transcrição STAT3/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Cisplatino/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Neoplasias Epiteliais e Glandulares/etiologia , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/etiologia , Neoplasias Ovarianas/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fatores de Transcrição da Família Snail/genética , Transfecção/métodos , Vimentina/genética , beta Catenina/genética
17.
Biochem Biophys Res Commun ; 461(3): 525-32, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-25911324

RESUMO

Our previous study showed that Octamer-binding transcription factor 4 (OCT4) expression was upregulated and significantly associated with histological grade through the analysis of OCT4 expression in 159 ovarian cancer tissue samples, and OCT4 mediated follicle-stimulating hormone (FSH)-induced anti-apoptosis in epithelial ovarian cancer. Nevertheless, whether OCT4 participates in FSH-induced invasion in ovarian cancer is still unknown. Therefore, the present study aimed to define whether FSH-induced ovarian cancer invasion is mediated by OCT4. In present study, we showed that FSH induced not only the epithelial-mesenchymal transition (EMT) and invasive phenotype but also the upregulation of OCT4 expression in a dose- and time-dependent manner in epithelial ovarian cancer cells. In addition, the expression of FSH receptor (FSHR) was upregulated by FSH induction, and knockdown of FSHR inhibited FSH-stimulated OCT4 expression. ERK1/2 signaling pathway participated in the enhanced expression of OCT4 and Snail induced by FSH. We further showed that the activated expression of Snail and N-cadherin, the suppressed expression of E-cadherin and the morphological change of the cells stimulated by FSH were blocked by OCT4-specific small interfering RNA. Moreover, our results showed that OCT4 mediated the increase in invasive capacity induced by FSH in ovarian cancer cells. Taken together, our work reveals that OCT4 is an essential mediator in FSH-induced EMT and invasion in epithelial ovarian cancer and may act as a potential therapeutic target.


Assuntos
Hormônio Foliculoestimulante/fisiologia , Sistema de Sinalização das MAP Quinases , Invasividade Neoplásica , Neoplasias Epiteliais e Glandulares/patologia , Fator 3 de Transcrição de Octâmero/fisiologia , Neoplasias Ovarianas/patologia , Sequência de Bases , Carcinoma Epitelial do Ovário , Humanos , Neoplasias Epiteliais e Glandulares/enzimologia , Neoplasias Epiteliais e Glandulares/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/metabolismo , Reação em Cadeia da Polimerase , RNA Interferente Pequeno , Receptores do FSH/fisiologia
18.
Tumour Biol ; 36(3): 1529-37, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25608836

RESUMO

The purpose of the study was to explore the role and mechanism of ataxia-telangiectasia mutated (ATM) protein in endometrial carcinogenesis. A reverse-phase protein array (RPPA) was used to analyze the expression of ATM signal pathway proteins in Ishikawa and progesterone-insensitive Ishikawa. ATM expression was detected in endometrium specimens by immunohistochemistry, including 8 cases with proliferative endometrium, 6 cases with secretory endometrium, 10 cases with simple hyperplasia (SH), 13 cases of complex hyperplasia (CH), 11 cases of endometrial atypical hyperplasia (EAH), and 83 cases with type I endometrial cancer. The relationship between ATM expression and other clinicopathological indicators was also examined in type I endometrial cancer patients. The mechanisms of ATM were explored in vitro with the endometrial cell lines Ishikawa and RL95-2. A cell counting kit-8 (CCK-8) test and Western blot analysis were performed to test proliferation and protein expression. Statistical analysis was performed with SPSS19.0. The significance level was set at 0.05. ATM was increased with medroxyprogesterone acetate (MPA) stimulation in Ishikawa in RPPA. ATM expression gradually decreased in endometrial hyperplasic lesions compared with the normal proliferative and secretory endometrium and was the lowest in type I endometrial cancer. ATM expression was negatively correlated with pathological grades in type I endometrial cancer. In vitro, ATM silencing retarded proliferation inhibition in Ishikawa and RL95-2 treated with MPA. ATM silencing could down-regulate the MPA-stimulated signal proteins, including Chk2, P53, and caspase-3 in vitro. MPA might exert its role through activating the ATM-associated pathway, ATM-Chk2-P53-caspase-3 (active), preserving normal endometrium and protecting it from malignancies. ATM might be a promising indicator for endometrial hyperplasia and cancer.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Carcinogênese/metabolismo , Neoplasias do Endométrio/metabolismo , Progesterona/genética , Proteínas Mutadas de Ataxia Telangiectasia/genética , Carcinogênese/genética , Carcinogênese/patologia , Caspase 3/genética , Caspase 3/metabolismo , Proliferação de Células/genética , Quinase do Ponto de Checagem 2/genética , Quinase do Ponto de Checagem 2/metabolismo , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Endométrio/metabolismo , Endométrio/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Progesterona/metabolismo , Fatores de Proteção , Transdução de Sinais , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
20.
Gynecol Oncol ; 132(3): 606-10, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24418201

RESUMO

OBJECTIVE: To explore the relationship between metabolic abnormalities and DPE (disordered proliferative endometrium), EH (endometrial hyperplasia) and type I EC (endometrial cancer). METHODS: We conducted a prospective cross-sectional study that lasted from September 2011 to September 2012 at the Obstetrics and Gynecology Hospital of Fudan University. 314 cases were enrolled, including 56 cases of DPE, 194 cases of EH and 25 cases of type I EC. 39 healthy female cases were collected as a control group. General information was collected, and blood tests, including blood lipids, OGTT (75-g oral glucose tolerance test) and insulin release tests were examined. Statistical analysis was performed using SPSS 19.0 (Chicago, USA), and 0.05 was chosen as the significance test level. RESULTS: The median (inter-quartile) age of the 314 study subjects was 44 (12) years, with the ages ranging from 21 to 75 years. Elevated insulin level was correlated with DPE, EH without/with atypia and EC. The risk increased when HOMA-IR (homeostasis model assessment-insulin resistance)≥ 2.95 (the lower limit of the top quartile of HOMA-IR distribution in non-diabetic patients); the OR (odds ratio) for DPE was 9.973 (95% CI (coefficient interval): 2.038-48.789, P=0.005), that for EH without atypia was 8.481 (95% CI:1.860-38.672, P=0.006), that for EH with atypia was 18.716 (95% CI: 3.091-113.335, P=0.001) and that for type I EC was 45.199 (95% CI: 5.886-347.065, P<0.001). Opposite trends were observed for the QUICKI (Quantitative Insulin Sensitivity Check Index). CONCLUSIONS: Hyperinsulinemia is associated with DPE and EH without/with atypia, not only with type I EC, and it might be a key factor that initiates and promotes endometrial hyperplastic lesions.


Assuntos
Hiperplasia Endometrial/sangue , Endométrio/patologia , Hiperinsulinismo/patologia , Estudos Transversais , Neoplasias do Endométrio/sangue , Neoplasias do Endométrio/patologia , Feminino , Humanos , Hiperinsulinismo/sangue , Pessoa de Meia-Idade , Estudos Prospectivos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa