Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Gynecol Endocrinol ; 39(1): 2231085, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37395213

RESUMO

Objectives: Talin1 is a cytoskeletal protein and is localized between cells and the extracellular matrix. This study aimed to investigate the mechanism by which Talin1 affects glucose metabolism and endometrial receptivity via glucose transporter proteins-4 (GLUT-4) in patients with polycystic ovary syndrome (PCOS) and insulin resistance (IR). Methods: We examined the expression of Talin1 and GLUT4 in the receptive endometrium of PCOS-IR and control patients. GLUT4 expression was examined after silencing and overexpression of Talin1 in Ishikawa cells. We validated the interaction between Talin1 and GLUT-4 proteins using a co-immunoprecipitation (Co-IP) assay. After successfully establishing the C57BL/6j mouse model of PCOS-IR, the expression of Talin1 and GLUT-4 were examined in PCOS-IR and control mice. The effect of Talin1 on embryo implantation and the number of live births in mice were examined. Results: Our study found low expression of Talin1 and GLUT-4 in the receptive endometrium of PCOS-IR patients compared to that in control patients (p < 0.01). The level of GLUT-4 expression decreased after silencing Talin1 in Ishikawa cells and increased after overexpression of Talin1. Co-IP results showed that Talin1 interacts with GLUT-4 protein. We successfully established a PCOS-IR C57BL/6j mouse model and found that Talin1 and GLUT-4 expression in the receptive endometrium of PCOS-IR mice were lower than that in control mice (p < 0.05). In vivo experiments confirmed that the knockdown of Talin1 affects embryo implantation (p < 0.05) and live birth rate in mice (p < 0.01). Conclusions: Talin1 and GLUT-4 expression were decreased in the endometrium of PCOS-IR patients, and Talin1 may affect glucose metabolism and endometrial receptivity through GLUT4.


Assuntos
Resistência à Insulina , Síndrome do Ovário Policístico , Animais , Feminino , Humanos , Camundongos , Endométrio/metabolismo , Glucose/farmacologia , Insulina/metabolismo , Resistência à Insulina/fisiologia , Camundongos Endogâmicos C57BL , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/metabolismo
2.
J Obstet Gynaecol ; 43(1): 2142767, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36357216

RESUMO

The aim of this study was to identify the high-risk factors for caesarean scar pregnancy (CSP) and establish a nomogram to predict the risk of caesarean scar pregnancy in pregnant women with a history of caesarean section. Among 1273 pregnant women with a history of caesarean section, 70% of the patients (892 patients, training sample) were randomly selected for analysis, and a prediction model was generated. The remaining patients (381 patients, validation sample) were validated for the model. Four high-risk factors for CSP were established, including: parity, number of previous abortions, uterus position, and early vaginal bleeding. The area under the curve of the nomogram for the training set was 0.867 and that for the validation set was 0.881, indicating good performance. Calibration curves for predicting CSP showed good calibrations. Decision curve analyses showed good application prospects for the model. Our results show that our nomogram for predicting CSP risks can be a practical tool to help in the early identification of CSP.Impact StatementWhat is already known on this subject? The high-risk factors for "caesarean scar pregnancy", An simple nomogram could be constructed to predict the risk of the disease through these high-risk factors.What do the results of this study add? This study can quickly predict whether the patient is a high-risk group for uterine scar pregnancy based on the patient's previous pregnancy, early vaginal bleeding and uterine position.What are the implications of these findings for clinical practice and/or further research? Caesarean scar pregnancy was secondary Long-term complications after caesarean section that with a high risk of pregnancy. In this study, we established a nomogram based on the number of cases of CSP and a control group with a history of caesarean section delivery at term, The high-risk factors were assigned a certain risk value in the early stage, if the woman contains more high-risk factors, the higher the risk of developing CSP, it should be highly valued in the early stage, and the rate of visiting a doctor should be increased.


Assuntos
Cesárea , Gravidez Ectópica , Gravidez , Humanos , Feminino , Cesárea/efeitos adversos , Cicatriz/complicações , Nomogramas , Gravidez Ectópica/etiologia , Paridade , Hemorragia Uterina/etiologia , Estudos Retrospectivos
3.
Biol Reprod ; 106(5): 992-999, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35044439

RESUMO

In the endometrium of women with recurrent implantation failure and unexplained recurrent miscarriage, the expression levels of homeobox A10 and E-cadherin were positively correlated. To explore whether homeobox A10 regulates E-cadherin during endometrial receptivity establishment, Ishikawa and RL95-2 cells were transfected with target-specific small interfering RNA (siRNA) and overexpression plasmid of homeobox A10. The expression levels of homeobox A10 and E-cadherin were measured by western blot and quantitative Real-time Polymerase Chain Reaction (qRT-PCR). Attachment assay of JEG-3 spheroids to endometrial cells were conducted to explore the adhesive functions after homeobox A10 interfered. Chromatin immunoprecipitation assays and dual luciferase reporter were used to investigate the regulatory mechanism of homeobox A10. The CD1 mice were transfected with si-homeobox A10 to confirm these results in vivo. In Ishikawa and RL95-2 cells, the expression of E-cadherin was positively correlated with homeobox A10 when it was silenced/overexpressed. Consistently, the adhesion of endometrial epithelium cells and trophoblast cells was inhibited after homeobox A10 was silenced, and exogenous restoration of E-cadherin expression reversed this effect to some extent. Homeobox A10 regulates the expression of E-cadherin by directly binding to a conserved motif (TGTACTAAAAA) located in the E-cadherin promoter region. In addition, after knockdown of homeobox A10 in CD1 mice, both the implantation and live birth rates were decreased. In conclusion, homeobox A10 can bind to the E-cadherin promoter region and directly regulate its expression, thereby improving endometrial receptivity and subsequently increasing the embryo adhesion and implantation.


Assuntos
Antígenos CD , Caderinas , Implantação do Embrião , Endométrio , Proteínas Homeobox A10 , Animais , Antígenos CD/genética , Caderinas/genética , Linhagem Celular Tumoral , Implantação do Embrião/fisiologia , Endométrio/metabolismo , Feminino , Proteínas Homeobox A10/genética , Humanos , Camundongos , RNA Interferente Pequeno/genética
4.
J Obstet Gynaecol Res ; 48(6): 1426-1432, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35261117

RESUMO

AIM: To investigate the distribution of tubal endometriosis (EM) in the right and left sides and four parts of the fallopian tube. METHODS: A retrospective, cross-sectional study was conducted on patients with tubal EM at the Fourth Affiliated Hospital of Guangxi Medical University from October 2011 to September 2021. Chi-square and binomial tests were used for analysis. RESULTS: Thirty-four patients (53.97%) had tubal resection due to EM (EM group). Twenty-nine patients (46.03%) had tubal resection due to non-EM (non-EM group). Thirty-two patients (50.80%) had left fallopian tube EM, 21 (33.33%) had right fallopian tube EM, and 10 (15.87%) had bilateral fallopian tube EM, with significant differences among them (p = 0.000). In the EM group, 15 patients (44.12%) had left fallopian tube EM, 13 (38.23%) had right fallopian tube EM, and 6 (17.65%) had bilateral fallopian tube EM (p = 0.052). In the non-EM group, statistically different (p = 0.001) diagnoses of left fallopian tube EM, right fallopian tube EM, and bilateral fallopian tube EM were 17 (58.62%), 8 (27.59%), and 4 (13.79%), respectively. In the EM group, 18 patients (52.94%) were in the ampullary region; 16 (47.06%) were in the nonampullary region (p = 0.864). In the non-EM group, 22 cases (75.86%) were in the ampullary region and 7 (24.14%) were in the nonampullary region, with a significant difference between them (p = 0.008). CONCLUSIONS: The incidence of left fallopian tube EM was higher than that of right and bilateral fallopian tube EM. The incidence of tubal ampullary EM was higher than that of nonampullary region.


Assuntos
Endometriose , China/epidemiologia , Estudos Transversais , Endometriose/diagnóstico , Endometriose/epidemiologia , Tubas Uterinas/cirurgia , Feminino , Humanos , Estudos Retrospectivos
5.
Ginekol Pol ; 2023 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-37417378

RESUMO

This study aimed to compare the effectiveness of oral contraceptives and a levonorgestrel intrauterine system in treating intermenstrual bleeding due to uterine niche. We retrospectively analyzed 72 patients with intermenstrual bleeding due to uterine niche from January 2017 to December 2021, of whom 41 were treated with oral contraceptives and 31 with a levonorgestrel intrauterine system. Post-treatment follow-ups at 1, 3, and 6 months were conducted to compare the efficiency and adverse effects between the two groups. In the oral contraceptive group, the effectiveness rate was higher than 80% at 1- and 3-months post-treatment and higher than 90% at 6 months. In the levonorgestrel intrauterine system group, the effectiveness rates were 58.06%, 54.84%, and 61.29% at 1, 3, and 6 months of treatment, respectively. Oral contraceptives were more effective than the levonorgestrel intrauterine system in treating intermenstrual bleeding caused by uterine niche (p < 0.05).

6.
J Reprod Immunol ; 158: 103953, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37209460

RESUMO

BACKGROUND: Regenerative medicine with peripheral blood mononuclear cell (PBMC) transplantation sheds light on the issue of premature ovarian insufficiency (POI). However, the efficiency of PBMC treatment in natural ovarian aging (NOA) remains unclear. METHODS: Thirteen-month-old female Sprague-Dawley (SD) rats were used to verify the NOA model. Seventy-two NOA rats were randomly divided into three groups: the NOA control group, PBMC group, and PBMC+platelet-rich plasma (PRP) group. PBMCs and PRP were transplanted by intraovarian injection. The effects on ovarian function and fertility were measured after transplantation. RESULTS: Transplantation of PBMCs could restore the normal estrous cycle, consistent with the recovery of serum sex hormone levels, increased follicle numbers at all stages, and restoration of fertility by facilitating pregnancy and live birth. Moreover, when combined with PRP injection, these effects were more significant. The male-specific SRY gene was detected in the ovary at all four time points, suggesting that PBMCs continuously survived and functioned in NOA rats. In addition, after PBMC treatment, the expression of angiogenesis-related and glycolysis-related markers in the ovaries was upregulated, which indicated that these effects were associated with angiogenesis and glycolysis. CONCLUSIONS: PBMC transplantation restores the ovarian functions and fertility of NOA rats, and PRP could enhance the efficiency. Increased ovarian vascularization, follicle production, and glycolysis are likely the major mechanisms.


Assuntos
Plasma Rico em Plaquetas , Insuficiência Ovariana Primária , Gravidez , Humanos , Ratos , Masculino , Feminino , Animais , Leucócitos Mononucleares/metabolismo , Ratos Sprague-Dawley , Fator Estimulador de Colônias de Granulócitos , Insuficiência Ovariana Primária/terapia , Plasma Rico em Plaquetas/metabolismo
7.
Front Genet ; 13: 1020757, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36324511

RESUMO

Background: Adenomyosis (AM) is a common benign uterine disease that threatens the normal life of patients. Cells associated with microenvironmental immune ecology are crucial in AM, although they are not as well understood at the cellular level. Methods: Single-cell sequencing (scRNA-seq) data were used to construct an AM global single-cell map, to further identify relevant cell clusters and infer chromosomal copy number variation (CNV) in AM samples. The biological functions of cell clusters were explored and cellular evolutionary processes were inferred by enrichment analysis and pseudotime analysis. In addition, a gene regulatory network (GRN) analysis was constructed to explore the regulatory role of transcription factors in AM progression. Results: We obtained the expression profiles of 42260 cells and identified 10 cell clusters. By comparing the differences in cell components between AM patients and controls, we found that significant abundance of endometrial cells (EC), epithelial cells (Ep), endothelial cells (En), and smooth muscle cells (SMC) in AM patients. Cell clusters with high CNV levels possessing tumour-like features existed in the ectopic endometrium samples. Moreover, the Ep clusters were significantly involved in leukocyte transendothelial cell migration and apoptosis, suggesting an association with cell apoptosis and migration. En clusters were mainly involved in pathways in cancer and apoptosis, indicating that En has certain malignant features. Conclusion: This study identified cell clusters with immune-related features, investigated the changes in the immune ecology of the microenvironment of these cells during AM, and provided a new strategy for the treatment of AM.

8.
Front Immunol ; 13: 1053819, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36439123

RESUMO

Recurrent miscarriage (RM) is a chronic, heterogeneous autoimmune disease that has serious social and personal consequences. No valid and reliable diagnostic markers or therapeutic targets for RM have been identified. Macrophages impact the innate immune system and can be used as diagnostic and prognostic markers for many diseases. We first collected 16 decidua and villi tissue samples from 5 normal patients and 3 RM patients for single-cell RNA sequencing data analysis and identified 1293 macrophage marker genes. We then screened a recurrent miscarriage cohort (GSE165004) for 186 macrophage-associated marker genes that were significantly differentially expressed between RM patients and the normal pregnancy endometrial tissues, and performed a functional enrichment analysis of differentially expressed genes. We then identified seven core genes (ACTR2, CD2AP, MBNL2, NCSTN, PUM1, RPN2, and TBC1D12) from the above differentially expressed gene group that are closely related to RM using the LASSO, Random Forest and SVM-RFE algorithms. We also used GSE26787 and our own collection of clinical specimens to further evaluate the diagnostic value of the target genes. A nomogram was constructed of the expression levels of these seven target genes to predict RM, and the ROC and calibration curves showed that our nomogram had a high diagnostic value for RM. These results suggest that ACTR2 and NCSTN may be potential targets for preventative RM treatments.


Assuntos
Aborto Habitual , Hexosiltransferases , Gravidez , Feminino , Humanos , Aborto Habitual/diagnóstico , Aborto Habitual/genética , Aborto Habitual/metabolismo , Macrófagos/metabolismo , Biomarcadores , Análise de Sequência de RNA , RNA , Proteínas de Ligação a RNA/genética , Hexosiltransferases/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo
9.
J Exp Clin Cancer Res ; 41(1): 77, 2022 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-35209949

RESUMO

BACKGROUND: Metabolic reprogramming is a hallmark of cancer. However, the roles of long noncoding RNAs (lncRNAs) in cancer metabolism, especially glucose metabolism remain largely unknown. RESULTS: In this study, we identified and functionally characterized a novel metabolism-related lncRNA, LINC00930, which was upregulated and associated with tumorigenesis, lymphatic invasion, metastasis, and poor prognosis in nasopharyngeal carcinoma (NPC). Functionally, LINC00930 was required for increased glycolysis activity and cell proliferation in multiple NPC models in vitro and in vivo. Mechanistically, LINC00930 served as a scaffold to recruit the RBBP5 and GCN5 complex to the PFKFB3 promoter and increased H3K4 trimethylation and H3K9 acetylation levels in the PFKFB3 promoter region, which epigenetically transactivating PFKFB3, and thus promoting glycolytic flux and cell cycle progression. Clinically, targeting LINC00930 and PFKFB3 in combination with radiotherapy induced tumor regression. CONCLUSIONS: Collectively, LINC00930 is mechanistically, functionally and clinically oncogenic in NPC. Targeting LINC00930 and its pathway may be meaningful for treating patients with NPC.


Assuntos
Glicólise/genética , Neoplasias Nasofaríngeas/genética , Oncogenes/genética , Fosfofrutoquinase-2/metabolismo , RNA Longo não Codificante/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Neoplasias Nasofaríngeas/patologia , Transfecção
10.
Life Sci ; 274: 119332, 2021 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-33711384

RESUMO

AIMS: Blastocyst implantation is mainly depended on the adhesion between cells and cell matrix. Endometrial adhesion plays an important role in establishing embryo implantation, but the underlying mechanisms are remains unclear. Talin1 is a local adhesion complex protein that is necessary for cell adhesion and movement. However, the role and mechanisms of Talin1 in embryo implantation are still unclear. MAIN METHODS: The expression of Talin1 and Integrin αvß3 was measured in the receptive endometrium from the RIF (Recurrent implantation failure) cohort and NC (normal fertile control group) cohort. A JEG-3 trophoblast and endometrial epithelial cell adhesion model and pregnant mouse model were established. The molecular mechanism of Talin1-mediated cell adhesion was explored by RNA sequencing, RT-qPCR, as well as western blotting assays. KEY FINDINGS: Talin1 enhances endometrial cell adhesion by regulating the Ras signaling pathway, and ultimately facilitates embryo implantation. SIGNIFICANCE: This study revealed the molecular mechanisms of regarding the pathogenesis of RIF caused by endometrial receptivity insufficiency. Further pharmacological research on the Ras signaling pathway would be valuable and might provide new therapeutic targets for RIF patients.


Assuntos
Aborto Habitual/patologia , Adesão Celular , Implantação do Embrião , Endométrio/patologia , Talina/metabolismo , Talina/fisiologia , Proteínas ras/metabolismo , Aborto Habitual/genética , Aborto Habitual/metabolismo , Animais , Apoptose , Proliferação de Células , Células Cultivadas , Endométrio/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Camundongos Knockout , Gravidez , Prognóstico , Talina/genética , Proteínas ras/genética
11.
Biomed Pharmacother ; 124: 109831, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31972354

RESUMO

BACKGROUND: Endometrial cancer is a type of malignant tumor of the female reproductive system. Preserving fertility in endometrial cancer patients is currently a formidable challenge. Interleukin-24 (IL-24) is a unique cytokine tumor suppressor gene belonging to the IL-10 cytokine family. IL-24 has broad-spectrum antitumor activity through different signaling pathways but does not affect normal cells. IL-24 gene therapy may provide a new method for the treatment of endometrial cancer. METHODS: Transfection was used for gene transfer. The expression of IL-24 and related pathway proteins in endometrial cancer tissue and the Ishikawa cell line was detected by immunohistochemistry and Western blotting, respectively. The antitumor function of IL-24 was examined in vitro and in vivo. Cell proliferation was determined by CCK-8 assay, cell migration was shown by wound-healing assay, and cell invasion was detected by Transwell assay. Apoptosis was analyzed by TUNEL assay, and HE staining was performed to observe the morphology of the samples. RESULTS: Immunohistochemical analysis showed different expression levels of IL-24 in human endometrial cancer tissues and normal endometrial tissues. IL-24 increased protein expression of BAX and Cytochrome C, while BCL-2, MMP-3, VEGF, Caspase-9 and Caspase-3 expression was decreased. Overexpression of IL-24 inhibited cell proliferation, migration and invasion, but increased cell apoptosis in endometrial cancer. Mechanistically, we demonstrated that IL-24 inhibited endometrial cancer cell growth by inducing cell apoptosis through the mitochondrial intrinsic signaling pathway. In addition, IL-24 inhibited tumor development by inducing cell apoptosis and inhibiting angiogenesis, as shown in xenograft tumor experiments. CONCLUSIONS: Our study demonstrates the antitumor effect of IL-24 on endometrial cancer and shows that IL-24 may be a promising therapeutic gene for endometrial cancer gene therapy.


Assuntos
Apoptose/genética , Proliferação de Células/genética , Neoplasias do Endométrio/patologia , Interleucinas/genética , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Neoplasias do Endométrio/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Nus , Mitocôndrias/metabolismo , Transdução de Sinais/genética , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Reprod Biol ; 20(2): 229-236, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32113856

RESUMO

The endometrium is a highly complex tissue that is vulnerable to subtle gene expression changes and is the first point of contact for an implanting blastocyst. Talin1 has previously been identified to regulate cytoskeleton and cell motility, however it has not been investigated in association with infertility. Herein, we presented that Talin1 dysregulation in the missed abortion endometrium would negatively influence endometrial adhesive capacity. Mechanistically, intracellular Talin1 inhibited the nuclear transportation of LIM and SH3 protein 1 (LASP1) and restored the expression of adhesion-associated protein. Moreover, extracellular Talin1 enforces endometrial epithelial cell adhesive capacity by interacting with Vitronectin (VTN) and activating the FAK/Src/ERK signalling pathway. This finding provides a novel insight into the potential use of Talin1 for managing endometrial epithelia cell adhesion. This study represents the first demonstration of Talin1 function in endometrial epithelial cell adhesion and endometrial receptivity. Our findings indicate that re-expression of Talin1 might represent a useful strategy for preventing and treating early pregnancy failure and infertility.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adesão Celular/fisiologia , Proteínas do Citoesqueleto/metabolismo , Endométrio/citologia , Células Epiteliais/metabolismo , Proteínas com Domínio LIM/metabolismo , Talina/metabolismo , Vitronectina/metabolismo , Aborto Retido/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Adulto , Proteínas do Citoesqueleto/genética , Regulação para Baixo , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Proteínas com Domínio LIM/genética , Gravidez , Talina/genética , Vitronectina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA