Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Apoptosis ; 29(5-6): 757-767, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38358580

ABSTRACT

Autophagy has emerged as an important process of cell metabolism. With continuous in-depth research on autophagy, TFEB has been a key transcription factor regulating autophagy levels in recent years. Studies have established that TFEB regulates autophagy and apoptosis in various diseases. However, the relationship between TFEB and the pathogenesis of endometriosis remains unclear. This study aimed to investigate the effect of TFEB on the mechanism of endometriosis progression. The results showed that TFEB and autophagy-related protein LC3 are highly expressed in ectopic endometrium of patients with endometriosis, overexpression of TFEB in cultured human endometrial stromal cells (HESCs) by lentivirus not only promoted autophagy but also inhibited apoptosis. In addition, the migration and invasion ability of HESCs were enhanced by TFEB overexpression. Furthermore, inhibiting autophagy with specific inhibitors can attenuate migration and invasion of HESCs induced by TFEB. The rat models of endometriosis show that TFEB knockdown can suppress lesion growth in vivo. Our results suggest that autophagy may be involved in the progression mechanism of endometriosis, and the mechanism of autophagy disorder in endometriosis is probably related to TFEB. TFEB may be a key molecule in promoting endometriosis.


Subject(s)
Apoptosis , Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cell Movement , Endometriosis , Endometrium , Adult , Animals , Female , Humans , Rats , Apoptosis/genetics , Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Movement/genetics , Endometriosis/metabolism , Endometriosis/pathology , Endometriosis/genetics , Endometrium/metabolism , Endometrium/pathology , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Rats, Sprague-Dawley , Stromal Cells/metabolism , Stromal Cells/pathology
2.
Gynecol Obstet Invest ; : 1-9, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39321785

ABSTRACT

OBJECTIVE: Laparoendoscopic single-site myomectomy (LESS-M) is widely applied for the treatment of uterine leiomyoma. The purposes of this study were to investigate differences in hidden blood loss between LESS-M and conventional laparoscopic myomectomy (CLM) during treatment of uterine leiomyoma and to identify the associated risk factors. DESIGN: This is a retrospective study. PARTICIPANTS: The participants of this study were patients who underwent laparoscopic myomectomy (114 and 156 for LESS-M and CLM, respectively) between July 1, 2019, and October 10, 2020, at the Second Affiliated Hospital of Wenzhou Medical University. SETTING: The study was conducted at the Second Affiliated Hospital of Wenzhou Medical University. METHODS: We enrolled a total of 114 and 156 patients who were treated with LESS-M and CLM, respectively, between July 1, 2019, and October 10, 2020. We collected clinical data, then applied the Nadler and Gross formula and multiple linear regression analysis to calculate the HBL and identify the associated risk factors, respectively. RESULTS: Patients in the LESS-M group had a VBL of 115.4 ± 180.6 mL and an HBL of 364.3 ± 252.6 mL, accounting for 74.4 ± 22.4% of true TBL. On the other hand, patients in the CLM group had VBL of 187.9 ± 198.5 mL, and HBL of 306.8 ± 304.7 mL, accounting for 58.9 ± 30.2% of true TBL. HBL was significantly higher in the LESS-M than the CLM group (p = 0.000). LIMITATIONS: This study was the small sample size used. CONCLUSIONS: HBL accounted for a significant percentage of TBL in laparoscopic myomectomy, especially in patients treated with LESS-M. Paying attention to perioperative blood changes coupled with fully understanding HBL might promote postoperative recovery of patients.

3.
Int Immunopharmacol ; 128: 111531, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38281338

ABSTRACT

OBJECTIVE: To investigate the influence and significance of cGAS-STING signaling pathway and autophagy on the occurrence and development of preeclampsia. DESIGN: A case-control experimental study, in vitro cell culture study, and in vivo animal research. METHODS: Human placenta tissue was collected and the differences in HE staining were observed. Immunohistochemistry and Western blot were used to verify differences in cGAS, STING and autophagy associated proteins. The PE rat model was established, the pathological changes of placenta and kidney were observed by HE staining, and the expression levels of related proteins were detected. In the lv-STING transfected HTR-8/SVneo trophoblast cell model, the expressions of autophagy indexes such as P62 and LC3 were verified by RT-PCR, Western blot and cell fluorescence experiments, and then the invasion and migration ability of cells were detected by Transwell and scrape tests. As an effective STING antagonist, C176 was administered to PE rats to observe whether it was effective in the treatment of PE disease. RESULTS: The expression levels of cGAS, STING and autophagy related proteins were increased in human and rat placental tissues. In the HTR-8/SVneo cell model which transfected by lv-STING, the expression levels of autophagy related indicators such as P62 and LC3 were increased. The invasion and migration ability of HTR-8/SVneo cells were significantly inhibited, which was improved by the autophagy inhibitor chloroquine. Acting as an effective STING antagonist in vivo, C176 significantly reversed the outcome of PE, alleviated and prevented the occurrence and development of PE. CONCLUSION: Our study proved that the cGAS-STING signaling pathway and autophagy levels are elevated in preeclampsia disease, and the cGAS-STING signaling pathway promotes the occurrence and development of preeclampsia through up-regulation of autophagy. This finding provides new insights into the pathogenesis of preeclampsia. Targeting this pathway may provide a potential therapeutic strategy for the treatment of preeclampsia.


Subject(s)
Placenta , Pre-Eclampsia , Pregnancy , Humans , Female , Animals , Rats , Placenta/metabolism , Cell Line , Pre-Eclampsia/metabolism , Nucleotidyltransferases/metabolism , Autophagy , Cell Movement
4.
Int Immunopharmacol ; 117: 109644, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36878046

ABSTRACT

OBJECTIVE: To investigate the roles of the cGAS-STING signal pathway and autophagy in the disease progression of endometriosis and to explore the regulatory mechanism of the cGAS-STING signal pathway on autophagy. DESIGN: A case-control experimental study, in vitro primary cell culture study, and in vivo animal research. MAIN OUTCOME MEASURES: Immunohistochemistry, RT-PCR and Western Blot were used to detect cGAS-STING signal pathway and autophagy expression differences in human and rat models. The lentivirus was used to overexpress STING in cells. The expression level of autophagy in human endometrial stromal cells (HESCs) transfected with lv-STING was detected by Western Blot, RT-PCR, and immunofluorescence. Transwell migration and invasion assays were conducted to assess cellular motility. The STING antagonist was applicated in vivo to investigate the therapeutic effects. RESULTS: The expression levels of the cGAS-STING signal pathway and autophagy in Human and Rat ectopic endometrium were increased. STING overexpression promotes the expression of autophagy in human endometrial stromal cells (HESCs). STING overexpression enhances the migration and invasion of the human endometrial stromal cells (HESCs), but the addition of autophagy antagonists could significantly reverse this. STING antagonists inhibited the expression of autophagy in vivo and reduced the volume of ectopic lesions. CONCLUSION: The expression levels of the cGAS-STING signal pathway and autophagy were increased in endometriosis. cGAS-STING signal pathway promotes the development of endometriosis by upregulating autophagy.


Subject(s)
Autophagy , Endometriosis , Animals , Female , Humans , Rats , Autophagy/physiology , Cell Movement , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Signal Transduction/physiology
5.
Int J Gynaecol Obstet ; 157(2): 424-430, 2022 May.
Article in English | MEDLINE | ID: mdl-34328652

ABSTRACT

OBJECTIVE: To quantify the volume of hidden blood loss (HBL) between conventional laparoscopic surgery (CLS) and laparoendoscopic single-site surgery (LESS) for ovarian cyst and to explore its risk factors. METHODS: A total of 310 patients who underwent CLS or LESS were enrolled in this study. The Nadler formula and Gross formula were used to calculate each patient's estimated blood volume and total blood loss, multiple linear regression analysis was applied to identify the risk factors. RESULTS: The HBL in LESS was more than in CLS (P = 0.000). Operative time (p = 0.015), pre-hematocrit (P = 0.002), pre-hemoglobin (P = 0.015), and pelvic adhesions (P = 0.037) were positively correlated with HBL in CLS. Intraoperative bleeding (P = 0.026), operative time (P = 0.000), pre-hematocrit (P = 0.042), CA125 (P = 0.047), and cyst volume (P = 0.012) were independent risk factors for HBL in LESS. CONCLUSION: A large amount of HBL occurs in ovarian cystectomy surgery and cannot be ignored in clinical work; fully and correctly understanding HBL and exploring its causes can ensure the safety and improve the prognosis of patients.


Subject(s)
Cystectomy , Laparoscopy , Blood Loss, Surgical , Cystectomy/adverse effects , Female , Humans , Laparoscopy/adverse effects , Retrospective Studies , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL