Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Hum Reprod ; 36(12): 3049-3061, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34647126

RESUMO

STUDY QUESTION: Is the protein l-arginine methyltransferase 3 (PRMT3)/asymmetrical dimethylarginine (ADMA)/nitric oxide (NO) pathway involved in the development of recurrent miscarriage (RM), and what is the potential mechanism? SUMMARY ANSWER: Elevated levels of PRMT3 and ADMA inhibit NO formation in the decidua, thereby impairing the functions of trophoblast cells at the maternal-foetal interface. WHAT IS KNOWN ALREADY: Decreased NO bioavailability is associated with RM. ADMA, an endogenous inhibitor of nitric oxide synthase (NOS), is derived from the methylation of protein arginine residues by PRMTs and serves as a predictor of mortality in critical illness. STUDY DESIGN, SIZE, DURATION: A total of 145 women with RM and 149 healthy women undergoing elective termination of an early normal pregnancy were enrolled. Ninety-six female CBA/J, 24 male DBA/2 and 24 male BALB/c mice were included. CBA/J × DBA/2 matings represent the abortion group, while CBA/J × BALB/c matings represent the normal control group. The CBA/J pregnant mice were then categorised into four groups: (i) normal + vehicle group (n = 28), (ii) abortion + vehicle group (n = 28), (iii) normal + SGC707 (a PRMT3 inhibitor) group (n = 20) and (iv) abortion + SGC707 group (n = 20). All injections were made intraperitoneally on Days 0.5, 3.5 and 6.5 of pregnancy. Decidual tissues were collected on Days 8.5, 9.5 and 10.5 of gestation. The embryo resorption rates were calculated on Day 9.5 and Day 10.5 of gestation. PARTICIPANTS/MATERIALS, SETTING, METHODS: NO concentration, ADMA content, NOS activity, expression levels of NOS and PRMTs in decidual tissues were determined using conventional assay kits or western blotting. PRMT3 expression was further analysed in decidual stromal cells, macrophages and natural killer cells. A co-culture system between decidual macrophages (DMs) and HTR-8/SVneo trophoblasts was constructed to study the roles of the PRMT3/ADMA/NO signalling pathway. Trophoblast apoptosis was analysed via Annexin V-fluorescein isothiocyanate/propidium iodide staining. CBA/J × DBA/2 mouse models were used to investigate the effects of SGC707 on embryo resorption rates. MAIN RESULTS AND THE ROLE OF CHANCE: Our results show that NO concentration and NOS activity were decreased, but ADMA content and PRMT3 expression were increased in the decidua of RM patients. Moreover, compared with the normal control subjects, PRMT3 expression was significantly up-regulated in the macrophages but not in the natural killer cells or stromal cells of the decidua from RM patients. The inhibition of PRMT3 results in a significant decrease in ADMA accumulation and an increase in NO concentration in macrophages. When co-cultured with DMs, which were treated with SGC707 and ADMA, trophoblast apoptosis was suppressed and induced, respectively. In vivo experiments revealed that the administration of SGC707 reduced the embryo resorption rate of CBA/J × DBA/2 mice. LIMITATIONS, REASONS FOR CAUTION: All sets of experiments were not performed with the same samples. The main reason is that each tissue needs to be reserved for clinical diagnosis and only a small piece of each tissue can be cut and collected for this study. WIDER IMPLICATIONS OF THE FINDINGS: Our results indicate that the PRMT3/ADMA/NO pathway is a potential marker and target for the clinical diagnosis and therapy of RM. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by the National Key Research and Development Program of China (2017YFC1001401), National Natural Science Foundation of China (81730039, 82071653, 81671460, 81971384 and 82171657) and Shanghai Municipal Medical and Health Discipline Construction Projects (2017ZZ02015). The authors have declared no conflict of interest. TRIAL REGISTRATION NUMBER: N/A.


Assuntos
Aborto Habitual , Arginina , Macrófagos , Óxido Nítrico , Proteína-Arginina N-Metiltransferases/metabolismo , Trofoblastos , Aborto Habitual/metabolismo , Animais , Apoptose , Arginina/análogos & derivados , Arginina/metabolismo , China , Decídua/metabolismo , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Óxido Nítrico/metabolismo , Gravidez , Trofoblastos/metabolismo
2.
Nat Commun ; 12(1): 3428, 2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34103526

RESUMO

Dysregulated extravillous trophoblast invasion and proliferation are known to increase the risk of recurrent spontaneous abortion (RSA); however, the underlying mechanism remains unclear. Herein, in our retrospective observational case-control study we show that villous samples from RSA patients, compared to healthy controls, display reduced succinate dehydrogenase complex iron sulfur subunit (SDHB) DNA methylation, elevated SDHB expression, and reduced succinate levels, indicating that low succinate levels correlate with RSA. Moreover, we find high succinate levels in early pregnant women are correlated with successful embryo implantation. SDHB promoter methylation recruited MBD1 and excluded c-Fos, inactivating SDHB expression and causing intracellular succinate accumulation which mimicked hypoxia in extravillous trophoblasts cell lines JEG3 and HTR8 via the PHD2-VHL-HIF-1α pathway; however, low succinate levels reversed this effect and increased the risk of abortion in mouse model. This study reveals that abnormal metabolite levels inhibit extravillous trophoblast function and highlights an approach for RSA intervention.


Assuntos
Aborto Habitual/metabolismo , Vilosidades Coriônicas/metabolismo , Ácido Succínico/metabolismo , Aborto Habitual/enzimologia , Aborto Habitual/genética , Animais , Estudos de Casos e Controles , Hipóxia Celular , Linhagem Celular Tumoral , Ilhas de CpG/genética , Metilação de DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Regulação da Expressão Gênica , Glicólise , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Metaboloma , Camundongos Endogâmicos C57BL , Gravidez , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fatores de Risco , Succinato Desidrogenase/genética , Succinato Desidrogenase/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Trofoblastos/metabolismo , Trofoblastos/patologia
3.
Bioorg Chem ; 95: 103566, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31935604

RESUMO

A docking study of a novel series of benzofuran derivatives with ERα was conducted. In this study, we report the synthesis of a novel series of benzofuran derivatives and evaluation of their anticancer activity in vitro against MCF-7 human breast cancer cells, as well as their potential toxicity to ER-independent MDA-MB-231 breast cancer cells, human renal epithelial HEK-293 cells, and human immortal keratinocytes (HaCaT cells) by using the MTT colorimetric assay. The screening results indicated that the target compounds exhibited anti-breast cancer activity. The target compound 2-benzoyl-3-methyl-6-[2-(morpholin-4-yl)ethoxy]benzofuran hydrochloride (4e) exhibited excellent activity against anti-oestrogen receptor-dependent breast cancer cells and low toxicity. The preliminary structure-activity relationships of the target benzofuran derivatives have been summarised. In conclusion, the novel benzofuran scaffold may be a promising lead for the development of potential oestrogen receptor inhibitors.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Benzofuranos/química , Benzofuranos/farmacologia , Neoplasias da Mama/patologia , Desenho de Fármacos , Receptores de Estrogênio/metabolismo , Antineoplásicos/síntese química , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Simulação de Acoplamento Molecular , Análise Espectral/métodos , Relação Estrutura-Atividade
4.
Autophagy ; 14(8): 1376-1397, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29962266

RESUMO

Impaired NK cell cytotoxic activity contributes to the local dysfunctional immune environment in endometriosis (EMS), which is an estrogen-dependent gynecological disease that affects the function of ectopic endometrial tissue clearance. The reason for the impaired cytotoxic activity of NK cells in an ectopic lesion microenvironment (ELM) is largely unknown. In this study, we show that the macroautophagy/autophagy level of endometrial stromal cells (ESCs) from EMS decreased under negative regulation of estrogen. The ratio of peritoneal FCGR3- NK to FCGR3+ NK cells increases as EMS progresses. Moreover, the autophagy suppression results in the downregulation of HCK (hematopoietic cellular kinase) by inactivating STAT3 (signal transducer and activator of transcription 3), as well as the increased secretion of the downstream molecules CXCL8/IL8 and IL23A by ESCs, and this increase induced the upregulation of FCGR3- NK cells and decline of cytotoxic activity in ELM. This process is mediated through the depression of microRNA MIR1185-1-3p, which is associated with the activation of the target gene PTGS2 in NK cells. FCGR3- NK with a phenotype of PTGS2/COX2high IFNGlow PRF1low GZMBlow induced by hck knockout (hck-/-) or 3-methyladenine (3-MA, an autophagy inhibitor)-stimulated ESCs accelerates ESC's growth both in vitro and in vivo. These results suggest that the estrogen-autophagy-STAT3-HCK axis participates in the differentiation of PTGS2high IFNGlow PRF1low GZMBlow FCGR3- NK cells in ELM and contributes to the development of EMS. This result provides a scientific basis for potential therapeutic strategies to treat diseases related to impaired NK cell cytotoxic activity. ABBREVIATIONS: anti-FCGR3: anti-FCGR3 with neutralizing antibody; Ctrl-ESC: untreated ESCs; CXCL8: C-X-C motif chemokine ligand 8; ectoESC: ESCs from ectopic lesion; ELM: ectopic lesion microenvironment; EMS: endometriosis; ESCs: endometrial stromal cells; eutoESC:eutopic ESCs; HCK: hematopoietic cellular kinase; HCK(OE): overexpression of HCK; IFNG: interferon gamma; IL23A (OE): overexpression of IL23A; KLRK1: Killer cell lectin like receptor K1; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; 3 -MA: 3-methyladenine; 3-MA-ESC: 3-MA-treated ESCs; MIR1185-1-3p+: overexpression of HsMIR1185-1-3p; NK: natural killer; normESCs: normal ESCs; Rap-ESC:rapamycin-treated ESCs; PCNA: proliferating cell nuclear antigen; PF: peritoneal fluid; SFKs: SRC family of cytoplasmic tyrosine kinases; si-HCK: silencing of HCK; siIL23A: silencing of IL23A; USCs: uterus stromal cells.


Assuntos
Autofagia , Diferenciação Celular , Ciclo-Oxigenase 2/metabolismo , Endométrio/patologia , Células Matadoras Naturais/citologia , Proteínas Proto-Oncogênicas c-hck/metabolismo , Receptores de IgG/metabolismo , Transdução de Sinais , Adulto , Animais , Microambiente Celular , Coristoma/patologia , Progressão da Doença , Regulação para Baixo , Células-Tronco Embrionárias/metabolismo , Feminino , Granzimas/metabolismo , Humanos , Interferon gama/metabolismo , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Fator de Transcrição STAT3/metabolismo , Células Estromais/patologia
5.
Cell Death Dis ; 9(5): 574, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29760378

RESUMO

Endometriosis (EMS) is an estrogen-dependent gynecological disease with a low autophagy level of ectopic endometrial stromal cells (eESCs). Impaired NK cell cytotoxic activity is involved in the clearance obstruction of the ectopic endometrial tissue in the abdominopelvic cavity. Protopanaxadiol (PPD) and protopanaxatriol (PPT) are two metabolites of ginsenosides, which have profound biological functions, such as anti-cancer activities. However, the role and mechanism of ginsenosides and metabolites in endometriosis are completely unknown. Here, we found that the compounds PPD, PPT, ginsenoside-Rg3 (G-Rg3), ginsenoside-Rh2 (G-Rh2), and esculentoside A (EsA) led to significant decreases in the viability of eESCs, particularly PPD (IC50 = 30.64 µM). In vitro and in vivo experiments showed that PPD promoted the expression of progesterone receptor (PR) and downregulated the expression of estrogen receptor α (ERα) in eESCs. Treatment with PPD obviously induced the autophagy of eESCs and reversed the inhibitory effect of estrogen on eESC autophagy. In addition, eESCs pretreated with PPD enhanced the cytotoxic activity of NK cells in response to eESCs. PPD decreased the numbers and suppressed the growth of ectopic lesions in a mouse EMS model. These results suggest that PPD plays a role in anti-EMS activation, possibly by restricting estrogen-mediated autophagy regulation and enhancing the cytotoxicity of NK cells. This result provides a scientific basis for potential therapeutic strategies to treat EMS by PPD or further structural modification.


Assuntos
Autofagia/efeitos dos fármacos , Endometriose/imunologia , Endométrio/imunologia , Ginsenosídeos/farmacologia , Células Matadoras Naturais/imunologia , Receptores de Estrogênio/imunologia , Sapogeninas/farmacologia , Autofagia/imunologia , Endometriose/tratamento farmacológico , Endometriose/patologia , Endométrio/patologia , Feminino , Humanos , Células Matadoras Naturais/patologia , Células Estromais/imunologia , Células Estromais/patologia
6.
Mol Med Rep ; 17(2): 2869-2878, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29257317

RESUMO

Embryo implantation is essential for a successful pregnancy, and leads to the decidualization of endometrial stromal cells (ESCs) in the secretory phase of the menstrual cycle. It has previously been demonstrated that decidual stromal cells (DSCs) co­express interleukin (IL)­25/IL­17RB and that IL­25 further promotes the proliferation of DSCs via activating c­Jun n­terminal kinase and protein kinase B signals, therefore the present study primarily focused on the role of IL­25 in the process of decidualization in vitro. It was demonstrated that the expression of IL­25/IL­17RB in ESCs was decreased compared with DSCs. In addition, following decidualization, the expression levels of IL­25/IL­17RB in ESCs were significantly elevated. Recombinant human (rh) IL­25 promoted the decidualization of ESCs in the presence of 8­bromoadenosine 3',5'­cyclic monophosphate sodium salt and 6α­methyl17α­acetoxyprogesterone, which was partially inhibited by anti­human IL­25 neutralizing antibody (anti­IL­25) or anti­IL­17RB. In addition, decidual natural killer (dNK) cells not only secreted IL­25, however also further accelerated the decidualization in vitro. Therefore, these findings indicated that ESCs differentiate into DSCs in the presence of ovarian hormones, resulting in the upregulation of IL­25/IL­17RB expression in ESCs. Furthermore, IL­25 secreted by ESCs and dNK cells further facilitates the decidualization of ESCs, which may form a positive feedback mechanism at the maternal­fetal interface and thus contribute to the establishment and maintenance of normal pregnancy.


Assuntos
Decídua/citologia , Endométrio/citologia , Interleucina-17/metabolismo , Células Matadoras Naturais/citologia , Células Estromais/citologia , Adulto , Diferenciação Celular , Células Cultivadas , Técnicas de Cocultura , Decídua/metabolismo , Implantação do Embrião , Endométrio/metabolismo , Feminino , Humanos , Interleucina-17/análise , Células Matadoras Naturais/metabolismo , Pessoa de Meia-Idade , Gravidez , Células Estromais/metabolismo
7.
Cell Death Dis ; 8(10): e3105, 2017 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-29022922

RESUMO

Decidual macrophages (dMϕ) contribute to maternal-fetal tolerance. However, the mechanism of dMϕ differentiation during pregnancy is still largely unknown. Here, we report that receptor activator for nuclear factor-κ B ligand (RANKL), secreted by human embryonic trophoblasts and maternal decidual stromal cells (DSCs), polarizes dMϕ toward a M2 phenotype. This polarization is mediated through activation of Akt/signal transducer and activator of transcription 6 (STAT6) signaling, which is associated with the upregulation of histone H3 lysine-27 demethylase Jmjd3 and IRF4 in dMϕ. Such differentiated dMϕ can induce a Th2 bias that promotes maternal-fetal tolerance. Impaired expression of RANKL leads to dysfunction of dMϕ in vivo and increased rates of fetal loss in mice. Transfer of RANK+Mϕ reverses mouse fetal loss induced by Mϕ depletion. Compared with normal pregnancy, there are abnormally low levels of RANKL/RANK in villi and decidua from miscarriage patients. These results suggest that RANKL is a pivotal regulator of maternal-fetal tolerance by licensing dMϕ to ensure a successful pregnancy outcome. This observation provides a scientific basis on which a potential therapeutic strategy can be targeted to prevent pregnancy loss.


Assuntos
Aborto Espontâneo/patologia , Decídua/imunologia , Tolerância Imunológica/imunologia , Macrófagos/imunologia , Troca Materno-Fetal/imunologia , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Animais , Decídua/citologia , Ativação Enzimática/fisiologia , Feminino , Humanos , Fatores Reguladores de Interferon/biossíntese , Histona Desmetilases com o Domínio Jumonji/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Gravidez , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição STAT6/metabolismo , Transdução de Sinais/imunologia , Células Estromais/metabolismo , Células Th2/imunologia , Trofoblastos/metabolismo
8.
J Cancer ; 8(15): 2915-2923, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28928882

RESUMO

Ovarian cancer, the third most common with highest mortality rates gynecological malignancy among women in China, is characterized by a unique tumor immune microenvironment. Immune-cell population infiltrated into the tumor tissue among patients with ovarian cancer are associated positively or negatively with antitumor activity. The imbalance between immune activation and immune suppression can result in oncogenesis and cancer progression. Therefore, intense investigation of the immunologic mechanism of ovarian cancer is urgently needed, and a comprehensive understanding of the network in which immune cells interact with the microenvironment, tumor cells and each other will greatly promote the development of more effective immunotherapies for ovarian cancer. In this review, we will focus on the main immune-cell population in ovarian tumor microenvironment, discuss their role in tumor progression and try to give the readers a new perspective in finding more promising therapeutic targets for cancers.

9.
Mol Med Rep ; 16(4): 3887-3893, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28765940

RESUMO

Interleukin-33 (IL-33) promotes migration of cancer cells through downregulating the expression of E-cadherin. Previous studies have demonstrated that IL­33 stimulates the proliferation of trophoblasts. However, the effect of IL­33 on the adhesion and invasion of trophoblasts has not been investigated in detail. In the present study, the expression of IL­33 and its receptor, IL­1 receptor­like 1 (ST2), was examined in villi from women during early pregnancy using immunohistochemistry. ST2 expression on human trophoblast and choriocarcinoma cell lines JAR, BeWo, JEG3 and HTR8 was confirmed by flow cytometry (FCM) assay. The effect of recombinant human IL­33 (rhIL­33) on adhesion, invasion and associated molecules was analyzed by cell adhesion, Matrigel invasion and FCM assays. The current study identified that human trophoblasts expressed IL­33 and ST2. RhIL­33 inhibited trophoblast invasion and adhesion, and decreased adhesion and invasion­associated molecules such as integrin α4ß1 and CD62L. Therefore, these results suggest that IL­33 may serve an important role in limiting invasion and implantation of trophoblasts by adhesion and invasion­associated molecules, contributing to the formation of the placenta and maintenance of normal pregnancy during early pregnancy.


Assuntos
Regulação para Baixo , Integrina alfa4beta1/metabolismo , Interleucina-33/farmacologia , Selectina L/metabolismo , Trofoblastos/metabolismo , Trofoblastos/patologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Humanos , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Invasividade Neoplásica , Trofoblastos/efeitos dos fármacos
10.
Cell Death Dis ; 8(3): e2666, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28300844

RESUMO

Endometriosis is an estrogen-dependent inflammatory disease. The anti-inflammatory cytokine IL-10 is also increased in endometriosis. IL-10 production by Th17 cells is critical for limiting autoimmunity and inflammatory responses. However, the mechanism of inducing IL-10-producing Th17 cells is still largely unknown. The present study investigated the differentiation mechanism and role of IL-10-producing Th17 cells in endometriosis. Here, we report that IL-10+Th17 cells are significantly increased in the peritoneal fluid of women with endometriosis, along with an elevation of IL-27, IL-6 and TGF-ß. Compared with peripheral CD4+ T cells, endometrial CD4+ T cells highly expressed IL-27 receptors, especially the ectopic endometrium. Under external (2,3,7,8-tetrachlorodibenzo-p-dioxin, TCDD) and local (estrogen, IL-6 and TGF-ß) environmental regulation, IL-27 from macrophages and endometrial stromal cells (ESCs) induces IL-10 production in Th17 cells in vitro and in vivo. This process may be mediated through the interaction between c-musculoaponeurotic fibrosarconna (c-Maf) and retinoic acid-related orphan receptor gamma t (RORγt), and associated with the upregulation of downstream B lymphocyte-induced maturation protein-1 (Blimp-1). IL-10+Th17 cells, in turn, stimulate the proliferation and implantation of ectopic lesions and accelerate the progression of endometriosis. These results suggest that IL-27 is a pivotal regulator in endometriotic immune tolerance by triggering Th17 cells to produce IL-10 and promoting the rapid growth and implantation of ectopic lesions. This finding provides a scientific basis for potential therapeutic strategies aimed at preventing the development of endometriosis, especially for patients with high levels of IL-10+Th17 cells.


Assuntos
Endometriose/metabolismo , Interleucina-10/metabolismo , Interleucinas/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-maf/metabolismo , Proteínas Repressoras/metabolismo , Células Th17/metabolismo , Adulto , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Diferenciação Celular/fisiologia , Progressão da Doença , Endometriose/patologia , Endométrio/metabolismo , Endométrio/patologia , Feminino , Humanos , Interleucina-6/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Fator 1 de Ligação ao Domínio I Regulador Positivo , Células Estromais/metabolismo , Células Estromais/patologia , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima/fisiologia
11.
Am J Reprod Immunol ; 77(1)2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27976427

RESUMO

Thymic stromal lymphopoietin (TSLP) is produced mainly by epithelial cells and was originally found to stimulate the growth and activation of B cells, but it is now shown to function on both hematopoietic and non-hematopoietic cell lineages. Despite a number of studies focusing on the role of TSLP in Th2 response for allergic disorders, it is becoming increasingly evident that TSLP may also be involved in multiple other disease states. This review will focus on recent advances on the effects of TSLP on obstetrical and gynecological diseases, including pregnancy failure, endometriosis, and cervical cancer.


Assuntos
Citocinas/metabolismo , Endometriose/metabolismo , Doenças do Recém-Nascido/metabolismo , Infertilidade Feminina/metabolismo , Neoplasias do Colo do Útero/metabolismo , Animais , Feminino , Hematopoese , Humanos , Recém-Nascido , Linfopoietina do Estroma do Timo
12.
Reproduction ; 152(6): 673-682, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27624484

RESUMO

Macrophages play an important role in the origin and development of endometriosis. Estrogen promoted the growth of decidual stromal cells (DSCs) by downregulating the level of interleukin (IL)-24. The aim of this study was to clarify the role and mechanism of IL-24 and its receptors in the regulation of biological functions of endometrial stromal cells (ESCs) during endometriosis. The level of IL-24 and its receptors in endometrium was measured by immunohistochemistry. In vitro analysis was used to measure the level of IL-24 and receptors and the biological behaviors of ESCs. Here, we found that the expression of IL-24 and its receptors (IL-20R1 and IL-20R2) in control endometrium was significantly higher than that in eutopic and ectopic endometrium of women with endometriosis. Recombinant human IL-24 (rhIL-24) significantly inhibited the viability of ESCs in a dosage-dependent manner. Conversely, blocking IL-24 with anti-IL-24 neutralizing antibody promoted ESCs viability. In addition, rhIL-24 could downregulate the invasiveness of ESCs in vitro After co-culture, macrophages markedly reduced the expression of IL-24 and IL-20R1 in ESCs, but not IL-22R1. Moreover, macrophages significantly restricted the inhibitory effect of IL-24 on the viability, invasion, the proliferation relative gene Ki-67, proliferating cell nuclear antigen (PCNA) and cyclooxygenase2 (COX-2), and the stimulatory effect on the tumor metastasis suppressor gene CD82 in ESCs. These results indicate that the abnormally low level of IL-24 in ESCs possibly induced by macrophages may lead to the enhancement of ESCs' proliferation and invasiveness and contribute to the development of endometriosis.


Assuntos
Movimento Celular , Endometriose/patologia , Endométrio/patologia , Interleucinas/antagonistas & inibidores , Macrófagos/patologia , Células Estromais/patologia , Adulto , Proliferação de Células , Técnicas de Cocultura , Endometriose/metabolismo , Endométrio/metabolismo , Feminino , Humanos , Interleucinas/metabolismo , Macrófagos/metabolismo , Transdução de Sinais , Células Estromais/metabolismo , Adulto Jovem
13.
Am J Reprod Immunol ; 75(6): 609-18, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26833725

RESUMO

Discovered 30 years ago, γδT cells remain an enigmatic T-cell subset. Although they account for a small portion of the total human circulating T-cell pool, their associations with other immune cells and their potential regulatory roles in related diseases have been explored but still require further investigation. γδT cells which are MHC-unrestricted innate-like lymphocytes with more unique antigen receptors than αßT cells and B cells are considered to bridge innate and adaptive immunity. They have APC functions and initiate adaptive immunity. Due to their distribution in specific tissues, secretion of Th1-, Th2-, and Th17-type cytokines, and other characteristics, they are involved in a variety of physiology and pathology processes. They are barometers in HIV infection. However, different γδT cell subsets play opposing roles in HBV infections, autoimmune diseases, and several types of tumors. Moreover, decidual γδT cells have protective roles during pregnancies by synthesizing several cytokines. This emerging evidence provides an improved understanding of the immune mechanism of infection, autoimmunity, cancer, and other related disorders and better insights regarding the potential roles of γδT cells in immunological therapeutic strategies.


Assuntos
Doenças Autoimunes/imunologia , Infecções por HIV/imunologia , Imunidade Inata , Imunoterapia Adotiva , Neoplasias/imunologia , Linfócitos T/imunologia , Imunidade Adaptativa , Animais , Apresentação de Antígeno , Doenças Autoimunes/terapia , Infecções por HIV/terapia , Humanos , Imunomodulação , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Linfócitos T/transplante
14.
Am J Cancer Res ; 5(10): 3072-84, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26693060

RESUMO

Cervical cancer is often associated with hypoxia and many kinds of chemokines. But the relationship and role of hypoxia and Chemokine (C-C motif) ligand 17 (CCL17) in cervical cancer are still unknown. Here, we found that CCL17 was high expressed in cervical cancer. HeLa and SiHa cells could secrete CCL17 in a time-dependent manner. Hypoxia increased expression of CCL17 receptor (CCR4) on HeLa and SiHa cells. Treatment with recombination human CCL17 (rhCCL17) led to an elevation of cell proliferation in HeLa and SiHa cells in a dose-dependent manner. In contrast, blocking CCL17 with anti-human CCL17 neutralizing antibody (α-CCL17) played an oppose effect. However, rhCCL17 had no effect on apoptosis in cervical cancer cells. Further analysis showed that hypoxia promoted the proliferation of HeLa and SiHa cells, and these effects could be reversed by α-CCL17. Stimulation with the inhibitor for c-Jun N-terminal kinase (JNK) or signal transducers and activator of transcription 5 (STAT5) signal pathway not only directly decreased the proliferation of HeLa and SiHa cells, but also abrogated the stimulatory effect of rhCCL17 on the proliferation of HeLa and SiHa cells. These results suggest that a high level of CCL17 in cervical cancer lesions is an important regulator in the proliferation of cervical cancer cells through JNK and STAT5 signaling pathways. In this process, hypoxia magnifies this effect by up-regulating CCR4 expression and strengthening the interaction of CCL17/CCR4.

15.
Reproduction ; 150(5): 417-27, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26316550

RESUMO

Chemokine CCL24, acting through receptor CCR3, is a potent chemoattractant for eosinophil in allergic diseases and parasitic infections. We recently reported that CCL24 and CCR3 are co-expressed by trophoblasts in human early pregnant uterus. Here we prove with evidence that steroid hormones estradiol (E), progesterone (P), and human chorionic gonadotropin (hCG), as well as decidual stromal cells (DSCs) could regulate the expression of CCL24 and CCR3 of trophoblasts. We further investigate how trophoblast-derived CCL24 mediates the function of trophoblasts in vitro, and conclude that CCL24/CCR3 promotes the proliferation, viability and invasiveness of trophoblasts. In addition, analysis of the downstream signaling pathways of CCL24/CCR3 show that extracellular signal-regulated kinases (ERK1/2) and phosphoinositide 3-kinase (PI3K) pathways may contribute to the proliferation, viability and invasiveness of trophoblasts by activating intracellular molecules Ki67 and matrix metallopeptidase 9 (MMP9). However, we did not observe any inhibitory effect on trophoblasts when blocking c-Jun N-terminal kinase (JNK) or p38 pathways. In conclusion, our data suggests that trophoblast-derived CCL24 at the maternal-fetal interface promotes trophoblasts cell growth and invasiveness by ERK1/2 and PI3K pathways. Meanwhile, pregnancy-related hormones (P and hCG), as well as DSCs could up-regulate CCL24/CCR3 expression in trophoblasts, which may indirectly influence the biological functions of trophoblasts. Thus, our results provide a possible explanation for the growth and invasion of trophoblasts in human embryo implantation.


Assuntos
Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quimiocina CCL24/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Trofoblastos/patologia , Adulto , Apoptose/efeitos dos fármacos , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Técnicas Imunoenzimáticas , Gravidez , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo , Adulto Jovem
16.
PLoS One ; 10(3): e0122036, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25825911

RESUMO

Stearoyl-acyl carrier protein desaturase (SAD), locating in the plastid stroma, is an important fatty acid biosynthetic enzyme in higher plants. SAD catalyzes desaturation of stearoyl-ACP to oleyl-ACP and plays a key role in determining the homeostasis between saturated fatty acids and unsaturated fatty acids, which is an important player in cold acclimation in plants. Here, four new full-length cDNA of SADs (ScoSAD, SaSAD, ScaSAD and StSAD) were cloned from four Solanum species, Solanum commersonii, S. acaule, S. cardiophyllum and S. tuberosum, respectively. The ORF of the four SADs were 1182 bp in length, encoding 393 amino acids. A sequence alignment indicated 13 amino acids varied among the SADs of three wild species. Further analysis showed that the freezing tolerance and cold acclimation capacity of S. commersonii are similar to S. acaule and their SAD amino acid sequences were identical but differed from that of S. cardiophyllum, which is sensitive to freezing. Furthermore, the sequence alignments between StSAD and ScoSAD indicated that only 7 different amino acids at residues were found in SAD of S. tuberosum (Zhongshu8) against the protein sequence of ScoSAD. A phylogenetic analysis showed the three wild potato species had the closest genetic relationship with the SAD of S. lycopersicum and Nicotiana tomentosiformis but not S. tuberosum. The SAD gene from S. commersonii (ScoSAD) was cloned into multiple sites of the pBI121 plant binary vector and transformed into the cultivated potato variety Zhongshu 8. A freeze tolerance analysis showed overexpression of the ScoSAD gene in transgenic plants significantly enhanced freeze tolerance in cv. Zhongshu 8 and increased their linoleic acid content, suggesting that linoleic acid likely plays a key role in improving freeze tolerance in potato plants. This study provided some new insights into how SAD regulates in the freezing tolerance and cold acclimation in potato.


Assuntos
Genes de Plantas , Oxigenases de Função Mista/genética , Solanum tuberosum/genética , Adaptação Fisiológica/genética , Sequência de Aminoácidos , Clonagem Molecular , DNA Complementar/genética , Evolução Molecular , Congelamento , Oxigenases de Função Mista/química , Dados de Sequência Molecular , Plantas Geneticamente Modificadas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Solanum tuberosum/enzimologia , Solanum tuberosum/fisiologia
17.
Int J Clin Exp Pathol ; 7(9): 5762-71, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25337217

RESUMO

It has reported that interleukin-22 (IL-22) promotes the invasion of tumor cells. IL-22 in the endometriotic milieu stimulates the proliferation of human endometrial stromal cells (ESCs). The present study aimed to elucidate whether and how IL-22 regulates the invasion of ESCs from adenomyosis. The expression of IL-22 and its receptors in normal endometrium, eutopic endometrium and ectopic lesion was analyzed by immunohistochemistry; the invasiveness of ESCs in vitro was verified by Matrigel invasion assay; and the effects of IL-22 on the correspondent functional molecules were investigated by ELISA and flow cytometry. Here we found that IL-22 and its receptors IL-22R1 and IL-10R2 in eutopic endometrium and ectopic lesion of adenomyosis were significantly higher than that of normal endometrium. Recombinant human IL-22 (rhIL-22) increased IL-22R1 and IL-10R2 levels on ESCs. Moreover, rhIL-22 promoted the invasiveness of ESCs, and inhibited the expression of metastasis suppressor gene CD82, stimulated the secretion of IL-8, RANTES, IL-6 and VEGF of ESCs. On the contrary, the neutralizing antibody for IL-22 reversed these effects. Our current study has demonstrated that IL-22 has a positive feedback on the expression of its receptors IL-22R1 and IL-10R2 on ESCs. This autocrine effect of IL-22 promotes the invasion of ESCs possibly through regulating invasion-related molecules, suggesting that the abnormal high expression of IL-22 may play an important role in ESCs invasion and finally contribute to the origin and development of adenomyosis.


Assuntos
Adenomiose/metabolismo , Comunicação Autócrina , Movimento Celular , Endométrio/metabolismo , Interleucinas/metabolismo , Células Estromais/metabolismo , Adenomiose/patologia , Estudos de Casos e Controles , Células Cultivadas , Quimiocina CCL5/metabolismo , Endométrio/patologia , Feminino , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Proteína Kangai-1/metabolismo , Receptores de Interleucina/metabolismo , Transdução de Sinais , Células Estromais/patologia , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/metabolismo , Interleucina 22
18.
Int J Clin Exp Pathol ; 7(6): 2743-57, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25031694

RESUMO

In the peritoneal fluid, macrophages and their secretory cytokines are essential for endometriosis, but the factors that favor their involvement in the endometriosis-associated inflammatory response are still elusive. Given the anomalous expression of indoleamine 2,3-dioxygenase-1 (IDO1) in endometrial stromal cells (ESCs) and its potentially important roles in immune modulation, we aimed to determine the effects of IDO1 in ESCs on macrophages and the mechanism of those effects. Normal ESCs and ectopic ESCs transfected with the SD11-IDO1 shRNA (short hairpin RNA) or vector-only plasmid SD11 were subsequently co-cultured with peripheral blood (PB)-derived monocytes (PBMC)-driven macrophages directly and indirectly. Cytokine production was determined by analyzing the supernatant of the co-culture unit by enzyme-linked immunosorbent assay (ELISA). The phenotypes and the phagocytic ability of the macrophages were determined by flow cytometry. Compared to normal ESCs, the PBMC-driven macrophages co-cultured with ectopic ESCs displayed lower phagocytic ability. Additionally, macrophages co-cultured with ectopic ESCs exhibited higher levels of CD163 and CD209 and lower levels of HLA-DR and CD11c. Moreover, both the intracellular expression and extracellular secretion of interleukin-10 (IL-10) and transforming growth factor-ß1 (TGF-ß1) were significantly increased, while that of IL-12p70 was decreased in macrophages after being co-cultured with ectopic ESCs. However, there was no significant difference in macrophage phagocytic ability, immunophenotype or cytokine secretion between the direct and indirect co-culture units. Reversely, SD11-IDO1 shRNA transfection of ectopic ESCs could abrogate the decreased phagocytic ability and alternative activation of macrophages in ectopic ESC-macrophage co-culture unit, suggesting that higher IDO1 in ectopic ESCs was indispensable for the induction of macrophage tolerance. Furthermore, the decrease in phagocytic macrophages and alternatively activated macrophages induced by IDO1 in ectopic ESCs was reversed by the addition of an IL-33 inhibitor, that is, soluble ST2 (sST2). Therefore, through the activation of IL-33, the increased expression of IDO1 in ectopic ESCs contributed to the truncated phagocytic ability of macrophages in endometriosis.


Assuntos
Endometriose/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Interleucinas/metabolismo , Macrófagos/metabolismo , Células Estromais/metabolismo , Adulto , Técnicas de Cocultura , Progressão da Doença , Endometriose/imunologia , Endometriose/patologia , Endométrio/imunologia , Endométrio/metabolismo , Endométrio/patologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Interleucina-33 , Macrófagos/patologia , Fagocitose/imunologia , Células Estromais/imunologia , Células Estromais/patologia , Transfecção , Adulto Jovem
19.
J Endocrinol ; 222(1): 151-60, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24860148

RESUMO

Mammalian proprotein convertases (PCs) play an important role in folliculogenesis, as they proteolytically activate a variety of substrates such as the transforming growth factor beta (TGFß) superfamily. PC subtilism/kexin 6 (PCSK6) is a member of the PC family and is ubiquitously expressed and implicated in many physiological and pathological processes. However, in human granulosa cells, the expression of the PC family members, their hormonal regulation, and the function of PCs are not clear. In this study, we found that PCSK6 is the most highly expressed PC family member in granulosa cells. LH increased PCSK6 mRNA level and PCSK6 played an anti-apoptosis function in KGN cells. Knockdown of PCSK6 not only increased the secretion of activin A and TGFß2 but also decreased the secretion of follistatin, estrogen, and the mRNA levels of FSH receptor (FSHR) and P450AROM (CYP19A1). We also found that, in the KGN human granulosa cell line, TGFß2 and activin A could promote the apoptosis of KGN cells and LH could regulate the follistatin level. These data indicate that PCSK6, which is regulated by LH, is highly expressed in human primary granulosa cells of pre-ovulatory follicles and plays important roles in regulating a series of downstream molecules and apoptosis of KGN cells.


Assuntos
Ativinas/metabolismo , Apoptose/fisiologia , Células da Granulosa/metabolismo , Hormônio Luteinizante/farmacologia , Pró-Proteína Convertases/efeitos dos fármacos , Pró-Proteína Convertases/metabolismo , Serina Endopeptidases/efeitos dos fármacos , Serina Endopeptidases/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Apoptose/efeitos dos fármacos , Aromatase/metabolismo , Linhagem Celular , Células Cultivadas , Estrogênios/metabolismo , Feminino , Folistatina/metabolismo , Células da Granulosa/citologia , Células da Granulosa/efeitos dos fármacos , Humanos , Técnicas In Vitro , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Receptores do FSH/metabolismo , Regulação para Cima/efeitos dos fármacos
20.
Fertil Steril ; 102(1): 257-63, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24746746

RESUMO

OBJECTIVE: To clarify the role and mechanism of interleukin (IL)-25 in regulating the biological functions of decidual stromal cells (DSCs) in human early pregnancy. DESIGN: Laboratory study of the effect of IL-25 induced by hCG on the proliferation of DSCs. SETTING: Research laboratories. PATIENT(S): Women aged 23-47 years with normal pregnancy and abortion. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Signal transduction from IL-25. RESULT(S): Here we show that DSCs coexpress IL-25/IL-17RB. Human chorionic gonadotropin promotes the expression of IL-25/IL-17RB. In contrast to anti-human IL-25 neutralizing antibody, recombinant human IL-25 (rhIL-25) significantly stimulates the proliferation of DSCs in dosage- and time-dependent manners. RhIL-25 promotes the phosphorylation of AKT, extracellular-signal-regulated kinases 1/2 (ERK1/2), c-Jun N-terminal kinases (JNK), and nuclear factor κB in DSCs. Interestingly, blocking JNK or AKT signal with inhibitors down-regulates the stimulatory effect on DSC proliferation induced by rhIL-25. In addition, the results of Western blot show that the expression of IL-25/IL-17RB in DSCs from normal pregnancy was higher than that from abortion. CONCLUSION(S): Our results have revealed that hCG derived of trophoblasts up-regulates the expression of IL-25/IL-17RB in DSCs and that IL-25 further stimulates the proliferation of DSCs through activating JNK and AKT signals, which finally contributes to the establishment and maintenance of physiological pregnancy.


Assuntos
Proliferação de Células , Gonadotropina Coriônica/metabolismo , Decídua/enzimologia , Interleucina-17/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Células Estromais/enzimologia , Adulto , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Decídua/efeitos dos fármacos , Ativação Enzimática , Feminino , Humanos , Interleucina-17/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Pessoa de Meia-Idade , Fosforilação , Gravidez , Primeiro Trimestre da Gravidez , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Receptores de Interleucina/metabolismo , Receptores de Interleucina-17 , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Células Estromais/efeitos dos fármacos , Fatores de Tempo , Regulação para Cima , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA