Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Cancer Immunol Res ; 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38752667

RESUMO

An immunosuppressive microenvironment promotes the occurrence and development of tumors. Low apolipoprotein A1 (ApoA1) is closely related to tumor development, but the underlying mechanisms are unclear. This study investigated the association between serum ApoA1 levels and the immune microenvironment in endometrial, ovarian, and lung cancers. The serum ApoA1 level was decreased significantly in patients with endometrial and ovarian cancers compared with healthy controls. In endometrial cancer tissues, the low serum ApoA1 group showed increased CD163+ macrophage infiltration and decreased CD8+ T-cell infiltration compared with the normal serum ApoA1 group. Compromised tumor-infiltrating CD8+ T-cell functions and decreased CD8+ T-cell infiltration also were found in tumor-bearing ApoA1-knockout mice. CD8+ T-cell depletion experiments confirmed that ApoA1 exerted its antitumor activity in a CD8+ T cell-dependent manner. In vitro experiments showed that the ApoA1 mimetic peptide L-4F directly potentiated the antitumor activity of CD8+ T cells via a HIF-1α-mediated glycolysis pathway. Mechanistically, ApoA1 suppressed ubiquitin-mediated degradation of HIF-1α protein by downregulating HIF-1α subunit α inhibitor. This regulatory process maintained the stability of HIF-1α protein and activated the HIF-1α signaling pathway. Tumor-bearing ApoA1 transgenic mice showed an increased response to anti-PD-1 therapy, leading to reduced tumor growth along with increased infiltration of activated CD8+ T cells and enhanced tumor necrosis. The data reported herein demonstrate critical roles for ApoA1 in enhancing CD8+ T-cell immune functions via HIF-1α-mediated glycolysis and support clinical investigation of combining ApoA1 supplementation with anti-PD-1 therapy for treating cancer.

2.
Nat Genet ; 56(4): 637-651, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38565644

RESUMO

Endometrial carcinoma remains a public health concern with a growing incidence, particularly in younger women. Preserving fertility is a crucial consideration in the management of early-onset endometrioid endometrial carcinoma (EEEC), particularly in patients under 40 who maintain both reproductive desire and capacity. To illuminate the molecular characteristics of EEEC, we undertook a large-scale multi-omics study of 215 patients with endometrial carcinoma, including 81 with EEEC. We reveal an unexpected association between exposome-related mutational signature and EEEC, characterized by specific CTNNB1 and SIGLEC10 hotspot mutations and disruption of downstream pathways. Interestingly, SIGLEC10Q144K mutation in EEECs resulted in aberrant SIGLEC-10 protein expression and promoted progestin resistance by interacting with estrogen receptor alpha. We also identified potential protein biomarkers for progestin response in fertility-sparing treatment for EEEC. Collectively, our study establishes a proteogenomic resource of EEECs, uncovering the interactions between exposome and genomic susceptibilities that contribute to the development of primary prevention and early detection strategies for EEECs.


Assuntos
Carcinoma Endometrioide , Hiperplasia Endometrial , Neoplasias do Endométrio , Preservação da Fertilidade , Proteogenômica , Humanos , Feminino , Progestinas/uso terapêutico , Antineoplásicos Hormonais , Hiperplasia Endometrial/tratamento farmacológico , Preservação da Fertilidade/métodos , Estudos Retrospectivos , Carcinoma Endometrioide/tratamento farmacológico , Carcinoma Endometrioide/genética , Carcinoma Endometrioide/patologia , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia
3.
Cell Tissue Res ; 394(2): 379-392, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37759141

RESUMO

Despite the high complete response rate of fertility-sparing treatment in early-stage endometrial cancer (EC), the low pregnancy rate is a clinical challenge. Whether endometrium-derived mesenchymal stem cells (eMSCs) can repair damaged endometrium after EC reversal remains unclear. This study explored the potential therapeutic effects of eMSCs with suitable scaffold materials on endometrial damage caused by EC. Here, appropriate engineering scaffold materials were compared to identify the most suitable materials to carry eMSCs. Then, safety and efficacy evaluations of eMSCs with a suitable hyaluronic acid hydrogel (eMSCs/HA-GEL) were investigated in in vivo experiments with subcutaneous xenotransplantation in Balb/C nude mice and a model of endometrial mechanical injury in rats. HA-GEL has minimal cytotoxicity to eMSCs compared to other materials. Then, in vitro experiments demonstrate that eMSCs/HA-GEL enhance the inhibitory effects of progestins on EC cell biological behaviors. eMSCs/HA-GEL significantly inhibit EC cell growth and have no potential safety hazards of spontaneous tumorigenesis in Balb/C nude mouse subcutaneous xenotransplantation assays. eMSCs/HA-GEL intrauterine transplantation effectively increases endometrial thickness and glandular number, improves endometrial blood supply, reduces fibrotic areas, and improves pregnancy rates in a rat endometrial mechanical injury model. GFP-eMSCs/HA-GEL intrauterine transplantation in rats shows more GFP-eMSCs in the endometrium than GFP-eMSCs transplantation alone, and no tumor formation or suspicious cell nodules are found in the liver, kidney, or lung tissues. Our results reveal the safety and efficacy of eMSCs/HA-GEL in animal models and provide preliminary evidence for the use of eMSCs/HA-GEL as a treatment for EC-related endometrial damage.


Assuntos
Neoplasias do Endométrio , Células-Tronco Mesenquimais , Camundongos , Humanos , Feminino , Ratos , Animais , Camundongos Nus , Endométrio/patologia , Células-Tronco Mesenquimais/fisiologia , Neoplasias do Endométrio/terapia , Neoplasias do Endométrio/patologia , Transplante Heterólogo
4.
Stem Cell Res Ther ; 14(1): 159, 2023 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-37287079

RESUMO

BACKGROUND: Mesenchymal stem cell (MSC) therapy is an attractive treatment option for various cancers. Whether MSCs can be used to treat well-differentiated endometrial cancer (EC) remains unclear. The aim of this study is to explore the potential therapeutic effects of MSCs on EC and the underlying mechanisms. METHODS: The effects of adipose-derived MSCs (AD-MSCs), umbilical-cord-derived MSCs (UC-MSCs), and endometrium-derived MSCs (eMSCs) on the malignant behaviors of EC cells were explored via in vitro and in vivo experiments. Three EC models, including patient-derived EC organoid lines, EC cell lines, and EC xenograft model in female BALB/C nude mice, were used for this study. The effects of MSCs on EC cell proliferation, apoptosis, migration, and the growth of xenograft tumors were evaluated. The potential mechanisms by which eMSCs inhibit EC cell proliferation and stemness were explored by regulating DKK1 expression in eMSCs or Wnt signaling in EC cells. RESULTS: Our results showed that eMSCs had the highest inhibitory effect on EC cell viability, and EC xenograft tumor growth in mice compared to AD-MSCs and UC-MSCs. Conditioned medium (CM) obtained from eMSCs significantly suppressed the sphere-forming ability and stemness-related gene expression of EC cells. In comparison to AD-MSCs and UC-MSCs, eMSCs had the highest level of Dickkopf-related protein 1 (DKK1) secretion. Mechanistically, eMSCs inhibited Wnt/ß-catenin signaling in EC cells via secretion of DKK1, and eMSCs suppressed EC cell viability and stemness through DKK1-Wnt/ß-catenin signaling. Additionally, the combination of eMSCs and medroxyprogesterone acetate (MPA) significantly inhibited the viability of EC organoids and EC cells compared with eMSCs or MPA alone. CONCLUSIONS: The eMSCs, but not AD-MSCs or UC-MSCs, could suppress the malignant behaviors of EC both in vivo and in vitro via inhibiting the Wnt/ß-catenin signaling pathway by secreting DKK1. The combination of eMSCs and MPA effectively inhibited EC growth, indicating that eMSCs may potentially be a new therapeutic strategy for young EC patients desiring for fertility preservation.


Assuntos
Neoplasias do Endométrio , Células-Tronco Mesenquimais , Humanos , Camundongos , Feminino , Animais , Via de Sinalização Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo , Camundongos Nus , Camundongos Endogâmicos BALB C , Neoplasias do Endométrio/terapia , Neoplasias do Endométrio/metabolismo , Endométrio/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proliferação de Células , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo
5.
J Gynecol Oncol ; 34(4): e53, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36929579

RESUMO

OBJECTIVE: This study aimed to investigate the impact of molecular classification and PTEN, KRAS and PIK3CA gene mutation on the outcome of fertility-preserving treatment in the patients with endometrioid endometrial cancer (EEC) and endometrial atypical hyperplasia (EAH). METHODS: This is a single-center retrospective study. A total of 135 patients with EEC and EAH receiving fertility-preserving treatment and molecular classification were reviewed. The distribution of the four types of molecular classification was described. The impact of non-specific molecular profile (NSMP), mismatch repair-deficiency (MMRd), and PTEN, KRAS and PIK3CA gene mutation on the outcome of fertility-preserving treatment was analyzed. RESULTS: Of the patients analyzed, 86.7% (117/136) were classified as having NSMP; 14 (10.4%), MMRd; 1 (0.7%), POLEmut EAH; and 3 (2.2%), p53abn EEC. The patients having NSMP and MMRd achieved similar 16-, 32-, and 48-week complete response rates. The patients harboring tier I and tier II PTEN mutations (PTENmut-Clin) achieved lower cumulative 32-week CR rates than those with PTEN-others (without PTENmut-Clin) (22/47, 46.8% vs. 50/74, 67.6%; p=0.023; odds ratio=0.422; 95% confidence interval [CI]=0.199-0.896). Insulin-resistance (hazard ratio [HR]=0.435; 95% CI=0.269-0.702; p=0.001) and PTENmut-Clin (HR=0.535; 95% CI=0.324-0.885; p=0.015) were independent negative predictors for lower 32-week CR rates. CONCLUSION: PTENmut-Clin is an independent risk factor for unfavorable fertility-preserving treatment outcomes in the patients with EEC and EAH. The patients with MMRd receiving fertility-preserving treatment achieved outcomes similar to those of the patients with NSMP. The molecular profiles might guide fertility-preserving treatment in the prognosis and clinical decisions.


Assuntos
Carcinoma Endometrioide , Hiperplasia Endometrial , Neoplasias do Endométrio , Feminino , Humanos , Neoplasias do Endométrio/terapia , Neoplasias do Endométrio/tratamento farmacológico , Hiperplasia , Estudos Retrospectivos , Proteínas Proto-Oncogênicas p21(ras)/genética , Carcinoma Endometrioide/genética , Carcinoma Endometrioide/terapia , Resultado do Tratamento , Hiperplasia Endometrial/terapia , Hiperplasia Endometrial/tratamento farmacológico , Classe I de Fosfatidilinositol 3-Quinases/genética , Classe I de Fosfatidilinositol 3-Quinases/uso terapêutico , Fertilidade/genética , Mutação , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/uso terapêutico
6.
Cancer Med ; 12(5): 5964-5978, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36373483

RESUMO

BACKGROUND: Progestins are used as fertility-sparing regimens for young patients with stage 1A endometrioid endometrial cancer (EEC) and atypical endometrial hyperplasia (AEH). CD163+ macrophages promote estrogen-dependent EEC development, but whether they induce progestin insensitivity remains unclear. This study aimed to investigate the possible effects of CD163+ macrophages on progestin response in AEH/EEC patients. METHODS: The number of infiltrating CD163+ macrophages in progestin-insensitive and -sensitive endometrial lesions was compared. The effects of CD163+ macrophages on progestin responses and progesterone receptor (PR) expression in EC cells were evaluated in vitro. ATAC-seq and RNA-seq were combined to identify molecular/biological changes induced by CD163+ macrophages in progestin-insensitive EC cells. RESULTS: Increased CD163+ macrophage infiltration was significantly associated with progestin insensitivity and longer treatment durations in AEH/EEC patients. Additionally, the number of CD163+ macrophages was negatively correlated with PR expression in AEH/EEC tissues. Furthermore, the CD163+ macrophage-mediated microenvironment and secreted cytokines downregulated PR expression and impaired the response of EC cells to medroxyprogesterone acetate (MPA). RNA-seq analysis demonstrated that CD163+ macrophages antagonized PR signaling by blocking or even reversing MPA-regulated differential gene expression. Based on RNA-seq and ATAC-seq analyses, extracellular matrix (ECM) signaling and ECM-related transcription factors, FOXF2, POU1F1, and RUNX1were identified to potentially be involved in CD163+ macrophage-induced progestin insensitivity in endometrial cancer patients. CONCLUSIONS: We identified CD163+ macrophages as an important mediator of progestin desensitization and an unfavorable factor for the efficacy of fertility-preserving treatment in AEH/EEC patients.


Assuntos
Carcinoma Endometrioide , Hiperplasia Endometrial , Neoplasias do Endométrio , Feminino , Humanos , Progestinas/farmacologia , Sequenciamento de Cromatina por Imunoprecipitação , RNA-Seq , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Acetato de Medroxiprogesterona/farmacologia , Progesterona , Macrófagos , Microambiente Tumoral , Fatores de Transcrição Forkhead
7.
Int J Mol Sci ; 23(20)2022 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-36293372

RESUMO

Progestin resistance is a major obstacle to conservative therapy in patients with endometrial cancer (EC) and endometrial atypical hyperplasia (EAH). However, the related inducing factor is yet unclear. In this study, thyroid hormone and its receptor α (TRα) and ß (TRß) of patients were assayed. THRB-silenced RL95-2 and KLE EC cells were cultured to investigate the response of progestins. Transcriptomics and Western blotting were performed to investigate the changes in signaling pathways. We found that THRB, rather than THRA, knockdown promoted the viability and motilities of RL95-2 cells but not KLE cells. The suppressive effect of progestins on cell growth and motility significantly decreased in THRB-silenced RL95-2 cells. Multiple proliferation-related signaling pathways were enriched, and the activities of mammalian targets of rapamycin (mTOR)/4e-binding protein 1 (4EBP1)/eukaryotic translation initiation factor 4G (eIF4G) rather than phosphorylated protein kinase B (Akt) were remarkably boosted. Progestin treatment enhanced the effects, and the augmentation was partially abated on supplementation with T3. In THRB-knockdown KLE cells, the progestins-activated partial signaling pathway expression (either mTOR or eIF4G), and supplementation with T3 did not induce noticeable alterations. The serum levels of triiodothyronine (T3) were significantly lower in patients with EC compared with healthy women. A strong expression of TRß was observed in most patients with EC and EAH sensitive to progestin treatment. In contrast, TRα positive expression was detected in less than half of the patients sensitive to progestin therapy. In conclusion, THRB knockdown enhanced the viability and motility of type I EC cells and attenuated the suppressive effects of progestins by activating the mTOR-4EBP1/eIF4G pathway. Lower expression of THRB is likely correlated with progesterone resistance.


Assuntos
Neoplasias do Endométrio , Progestinas , Animais , Humanos , Feminino , Progestinas/farmacologia , Proteínas Proto-Oncogênicas c-akt , Receptores beta dos Hormônios Tireóideos , Fator de Iniciação Eucariótico 4G , Tri-Iodotironina/farmacologia , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Serina-Treonina Quinases TOR , Sirolimo , Mamíferos
8.
Laryngoscope Investig Otolaryngol ; 7(5): 1634-1642, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36258845

RESUMO

Background: Pain management, as a key component of enhanced recovery after surgery (ERAS), can effectively relieve perioperative pain and anxiety. However, there are few studies on the application of pain management based on ERAS in pediatric surgery patients. We aimed to examine the effect of ERAS-based perioperative pain management in children with obstructive sleep apnea (OSA) undergoing adenotonsillectomy. Methods: From March 2021 to July 2021, a randomized controlled single-blind study was conducted on children with OSA and scheduled to undergo adenotonsillectomy. The children were randomly assigned to either control group (n = 60) or ERAS group (n = 60). Traditional analgesia measures were provided to children in the control group, whereas ERAS-based optimized analgesia measures were provided to children in the ERAS group. The pain scores, anxiety scores and diet quality scores were compared between the two groups. Results: The pain scores after surgery in the ERAS group were significantly lower than those in the control group at 6 h, 1 day, 3 days, and 5 days after surgery. Furthermore, the diet quality scores in the ERAS group were significantly higher than those in the control group at 6 h, 1 day, 3 days, and 5 days after surgery. The anxiety scores after surgery in the ERAS group were significantly lower than those in the control group. Conclusions: Perioperative pain management based on ERAS can significantly alleviate postoperative pain, improve quality of life, and promote the accelerated rehabilitation of children with OSA undergoing adenotonsillectomy. Level of evidence: 1.

9.
Front Genet ; 13: 952083, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36092919

RESUMO

Objective: The aim of this study was to establish predictive models based on the molecular profiles of endometrial lesions, which might help identify progestin-insensitive endometrial atypical hyperplasia (EAH) or endometrioid endometrial cancer (EEC) patients before progestin-based fertility-preserving treatment initiation. Methods: Endometrial lesions from progestin-sensitive (PS, n = 7) and progestin-insensitive (PIS, n = 7) patients were prospectively collected before progestin treatment and then analyzed by ATAC-Seq and RNA-Seq. Potential chromatin accessibility and expression profiles were compared between the PS and PIS groups. Candidate genes were identified by bioinformatics analyses and literature review. Then expanded samples (n = 35) were used for validating bioinformatics data and conducting model establishment. Results: ATAC-Seq and RNA-Seq data were separately analyzed and then integrated for the subsequent research. A total of 230 overlapping differentially expressed genes were acquired from ATAC-Seq and RNA-Seq integrated analysis. Further, based on GO analysis, REACTOME pathways, transcription factor prediction, motif enrichment, Cytoscape analysis and literature review, 25 candidate genes potentially associated with progestin insensitivity were identified. Finally, expanded samples were used for data verification, and based on these data, three predictive models comprising 9 genes (FOXO1, IRS2, PDGFC, DIO2, SOX9, BCL11A, APOE, FYN, and KLF4) were established with an overall predictive accuracy above 90%. Conclusion: This study provided potential predictive models that might help identify progestin-insensitive EAH and EEC patients before fertility-preserving treatment.

10.
Cancer Gene Ther ; 29(10): 1452-1462, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35388173

RESUMO

Progestin is one of the main hormone treatment regimens for early-stage estrogen receptor- and progesterone receptor (PR)-positive endometrial cancer (EC). However, the response rate of EC to progestins is unsatisfactory. Investigating the mechanisms related to progestin treatment could help improve treatment efficacy. Studies have demonstrated that normal endometrial stromal cells (ESCs) increase the inhibitory effect of progestin on EC cell proliferation via paracrine signaling, but the mechanisms involved remain unclear. In this study, we found that ESCs had different morphological features between progestin-sensitive and -insensitive EC tissues. ESCs presented typical decidualization changes in progestin-sensitive cases, while they remained slim in progestin-insensitive EC lesions, indicating no response. Furthermore, ESCs enhanced the inhibitory effect of medroxyprogesterone acetate (MPA) on EC cell proliferation by secreting neuron cell adhesion molecule (NrCAM). MPA treatment enhanced NrCAM secretion by ESCs. EC xenografts in BALB/C nude mice demonstrated that MPA combined with NrCAM had an increased tumor inhibitory effect compared with MPA or NrCAM alone. Mechanistically, MPA upregulated NrCAM expression in ESCs through PR. Specifically, NrCAM increased PR expression in EC cells through TET1-induced hydroxymethylation of the PRB gene promoter region. These findings indicate that NrCAM or NrCAM combined with progestins could be a new EC treatment.


Assuntos
Neoplasias do Endométrio , Progestinas , Animais , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Moléculas de Adesão Celular/farmacologia , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Endométrio , Epigênese Genética , Feminino , Humanos , Acetato de Medroxiprogesterona/metabolismo , Acetato de Medroxiprogesterona/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Oxigenases de Função Mista/genética , Progestinas/metabolismo , Progestinas/farmacologia , Proteínas Proto-Oncogênicas/genética , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Células Estromais/metabolismo , Células Estromais/patologia
11.
Arch Gynecol Obstet ; 306(4): 1135-1146, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35246715

RESUMO

OBJECTIVE: Morular metaplasia (MM) is a benign epithelial metaplasia that sometimes appears in atypical endometrial hyperplasia (AEH) and endometrioid endometrial carcinoma (EEC). However, the clinical implications of MM for fertility-preserving treatment in AEH and EEC patients are unclear. This study investigated the clinical features and impact of MM on the efficacy of fertility-preserving treatment. METHODS: We retrospectively studied 427 AEH and EEC patients who received fertility-preserving treatment. Clinical features, treatment efficacy, and onco-fertility results were compared between patients with and without MM. RESULTS: MM appeared in 147 of 427 (34.4%) patients. Among them, 49 (33.3%) had MM only before treatment (BEF group), 32 (21.8%) had sustained MM before and during treatment (SUS group), and 66 (44.9%) had MM only during treatment (DUR group). The BEF group had a higher 12-month CR rate (98.0% vs 85.7%, p = 0.017) and shorter therapeutic duration to achieve CR (4.0 vs 5.7 months, p = 0.013) than the non-MM group had. In comparison with the non-MM group, the SUS and DUR groups had a lower CR rate after 7 months of treatment (SUS vs non-MM, 37.5% vs 61.1%, p = 0.010; DUR vs non-MM 33.3% vs. 61.1%, p < 0.001), and a longer median therapeutic duration to achieve CR (SUS vs non-MM, 7.6 vs. 4.0 months, p = 0.037; DUR vs non-MM, 7.9 vs. 4.0 months, p < 0.001). CONCLUSION: Appearance of MM only before treatment was positively correlated with outcome of fertility-preserving treatment, while sustained MM or appearance of MM only during treatment implied poorer outcome of fertility-preserving treatment in AEH and EEC patients.


Assuntos
Carcinoma Endometrioide , Hiperplasia Endometrial , Neoplasias do Endométrio , Preservação da Fertilidade , Hiperplasia Endometrial/tratamento farmacológico , Hiperplasia Endometrial/patologia , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/patologia , Feminino , Fertilidade , Preservação da Fertilidade/métodos , Humanos , Metaplasia , Estudos Retrospectivos
12.
J Gynecol Oncol ; 32(5): e70, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34132069

RESUMO

OBJECTIVE: This study aimed to investigate the impact of polycystic ovary syndrome (PCOS) on fertility-sparing treatment in young patients with atypical endometrial hyperplasia (AEH) or endometrioid endometrial cancer (EEC). METHODS: A total of 285 patients with EEC (n=76, FIGO stage IA, without myometrium invasion) or AEH (n=209) who received progestin-based fertility-sparing treatment were evaluated retrospectively. Among the 285 patients, 103 (36.1%), including 70 AEH cases and 33 EEC cases, were diagnosed with PCOS. General characteristics, cumulative 16- and 32-week complete response (CR) rate, pregnancy outcome and recurrence were compared between patients with or without PCOS. RESULTS: The cumulative 16-week CR rate was lower in the PCOS group than in the non-PCOS group (18.4% vs. 33.8%, p=0.006). Patients with PCOS took longer treatment duration to achieve CR (7.0 months vs. 5.4 months, p=0.006) and shorter time to relapse after CR (9.6 months vs. 17.6 months, p=0.040) compared with non-PCOS group. After adjusting for patient age, body mass index, PCOS, homeostasis model assessment-insulin resistance index, and serum testosterone levels, we found that body mass index ≥25 kg/m² (HR=0.583; 95% CI=0.365-0.932; p=0.024) and PCOS (HR=0.545; 95% CI=0.324-0.917; p=0.022) were significantly correlated with lower 16-week CR rate. CONCLUSION: PCOS was associated with lower 16-week CR rate, longer treatment duration and shorter recurrence interval in patients with AEH or EEC receiving fertility-preserving treatment.


Assuntos
Hiperplasia Endometrial , Neoplasias do Endométrio , Preservação da Fertilidade , Síndrome do Ovário Policístico , Hiperplasia Endometrial/tratamento farmacológico , Neoplasias do Endométrio/tratamento farmacológico , Feminino , Humanos , Hiperplasia , Recidiva Local de Neoplasia , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/tratamento farmacológico , Gravidez , Estudos Retrospectivos , Resultado do Tratamento
13.
Mol Reprod Dev ; 88(6): 379-394, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34014590

RESUMO

Endometrial damage is an important cause of female reproductive problems, manifested as menstrual abnormalities, infertility, recurrent pregnancy loss, and other complications. These conditions are collectively termed "Asherman syndrome" (AS) and are typically associated with recurrent induced pregnancy terminations, repeated diagnostic curettage and intrauterine infections. Cancer treatment also has unexpected detrimental side effects on endometrial function in survivors independently of ovarian effects. Endometrial stem cells act in the regeneration of the endometrium and in repair through direct differentiation or paracrine effects. Nonendometrial adult stem cells, such as bone marrow-derived mesenchymal stem cells and umbilical cord-derived mesenchymal stem cells, with autologous and allogenic applications, can also repair injured endometrial tissue in animal models of AS and in human studies. However, there remains a lack of research on the repair of the damaged endometrium after the reversal of tumors, especially endometrial cancers. Here, we review the biological mechanisms of endometrial regeneration, and research progress and challenges for adult stem cell therapy for damaged endometrium, and discuss the potential applications of their use for endometrial repair after cancer remission, especially in endometrial cancers. Successful application of such cells will improve reproductive parameters in patients with AS or cancer. Significance: The endometrium is the fertile ground for embryos, but damage to the endometrium will greatly impair female fertility. Adult stem cells combined with tissue engineering scaffold materials or not have made great progress in repairing the injured endometrium due to benign lesions. However, due to the lack of research on the repair of the damaged endometrium caused by malignant tumors or tumor therapies, the safety and effectiveness of such stem cell-based therapies need to be further explored. This review focuses on the molecular insights and clinical application potential of adult stem cells in endometrial regeneration and discusses the possible challenges or difficulties that need to be overcome in stem cell-based therapies for tumor survivors. The development of adult stem cell-related new programs will help repair damaged endometrium safely and effectively and meet fertility needs in tumor survivors.


Assuntos
Células-Tronco Adultas/fisiologia , Endométrio/fisiologia , Ginatresia/fisiopatologia , Regeneração/fisiologia , Aborto Habitual/etiologia , Aborto Habitual/prevenção & controle , Células-Tronco Adultas/transplante , Âmnio/citologia , Animais , Antígenos de Diferenciação/análise , Células da Medula Óssea , Senescência Celular , Modelos Animais de Doenças , Neoplasias do Endométrio/fisiopatologia , Neoplasias do Endométrio/terapia , Endométrio/irrigação sanguínea , Endométrio/citologia , Endométrio/lesões , Feminino , Sangue Fetal/citologia , Ginatresia/complicações , Ginatresia/terapia , Humanos , Hidrogéis , Células-Tronco Pluripotentes Induzidas/transplante , Infertilidade Feminina/etiologia , Infertilidade Feminina/terapia , Menstruação , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Mucosa Bucal/citologia , Células da Side Population/citologia , Nicho de Células-Tronco , Engenharia Tecidual/métodos , Alicerces Teciduais
14.
J Obstet Gynaecol Res ; 47(3): 984-991, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33403812

RESUMO

PURPOSE: To explore the clinical outcomes of megestrol acetate alone or plus metformin in young women with grade 2 stage IA endometrial carcinoma who ask for preserved fertility. METHODS: Patients with stage IA grade 2 endometrial carcinoma who asked for fertility-sparing treatment in the Obstetrics and Gynecology Hospital of Fudan University between 2015 and 2017 were enrolled and retrospectively reviewed. RESULTS: Four patients were included and treated with oral megestrol acetate (160 mg per day), while metformin (500 mg, thrice daily) was added for patients with metabolic syndrome. Regular hysteroscopic examination was performed every 3 months during the conservative treatment. Overall, 75% (3/4) of the patients had a complete response, one relapsed and achieved a complete response after changing the therapy plan, and one patient had an indication of myometrial invasion during fertility-sparing treatment and chose to remove uterus. CONCLUSIONS: Fertility-sparing treatment for stage IA grade 2 endometrial carcinoma patients is worth exploration. Megestrol acetate with or without metformin combined with hysteroscopic lesion ablation may be an effective therapy.


Assuntos
Neoplasias do Endométrio , Preservação da Fertilidade , Antineoplásicos Hormonais/uso terapêutico , Tratamento Conservador , Neoplasias do Endométrio/tratamento farmacológico , Feminino , Humanos , Acetato de Megestrol/uso terapêutico , Gravidez , Estudos Retrospectivos , Resultado do Tratamento
15.
J BUON ; 24(4): 1706-1711, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31646829

RESUMO

PURPOSE: To evaluate the inhibitory effect of 5-fluorouracil (5-FU) in combination with cisplatin on the proliferation of human osteosarcoma cells in vitro. METHODS: Three groups of human osteosarcoma U2OS cells were cultured in DMEM medium supplemented with the following drugs: 20 µg/mL 5-FU, 50 µg/mL 5-FU and 50 µg/mL 5-FU in combination with 0.5 mg/L cisplatin. After culture at 24, 48 and 72 h, the inhibition of proliferation rate of U2OS cells was calculated by CCK-8 cell kit. Cell invasiveness was assessed by Transwell assay. Flow cytometry was used for monitoring the cell apoptosis. RESULTS: 5-FU inhibited the growth of osteosarcoma cells, and the results of different concentrations of 5-FU were significantly different. The growth of U2OS cells decreased significantly within 24-72 h, and the concentration of 5-FU increased with time. The inhibition of the shift was more obvious, and the combined drug inhibition was significantly higher than the 20µg/ml 5-FU Group, 0.5mg/L Cisplatin Group and 50µg/ml 5-FU Group. After 72 h, the mean inhibitory rates of 20 µg/mL 5-FU, 50 µg/mL 5-FU, 50 µg/ml 5-FU in combination with 0.5 mg/L cisplatin, and 0.5 mg/L cisplatin were 12.54±1.26%, 22.17±0.59%, 32.54±1.25%, 20.84±0.83% respectively, and the difference was significant (p<0.05). Results of cell invasion assay showed that after culturing for 48 h, the mean number of cells penetrating the membrane was 22.84±5.27 in the culture group of 0.5 mg/L cisplatin, 30.57±5.68 in the culture group of 20 µg/mL 5-FU, 18.68±4.88 in the culture group of 50 µg/mL 5-FU, and 9.84±3.64 in the culture group of 50 µg/mL 5-FU in combination with 0.5 mg/L cisplatin, respectively, and 72.00±7.52 in the control group, showing statistical differences in each group (p<0.05). The apoptosis of the control group was significantly lower than that of the other groups (p<0.05). Apoptosis rate of the 20µg/mL 5-FU group was significantly lower than that of the 0.5mg/L cisplatin group, the 50µg/mL 5-FU group and the 50 µg/ml 5-FU group in combination with 0.5mg/L cisplatin (p<0.05). There was no difference in apoptosis between 0.5mg/L cisplatin and 50µg/mL 5-FU group (p>0.05). CONCLUSION: 5-FU in combination with cisplatin exerts an inhibitory effect on the proliferation and invasion of human osteosarcoma cells in vitro, and can promote cell apoptosis.


Assuntos
Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Fluoruracila/farmacologia , Osteossarcoma/tratamento farmacológico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Humanos , Osteossarcoma/patologia
16.
Hum Cell ; 32(3): 306-315, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31104300

RESUMO

Allergic rhinitis (AR) is a common hypersensitive disease that troubles patients a lot. Nasal epithelial cells (NECs), as the outmost protection of inhalation, play an important role in AR allergic response. Adrenoceptor beta 2 (ADRB2) is an important gene in inflammatory response, which has become the hot spot for AR development and treatment in recent years. MiR-15a-5p has been proved to be involved in AR immune response as the upstream regulator of ADRB2. Human primary NECs were isolated and stimulated by IL-13. qRT-PCR assay was used to detect the RNA level of target genes. ELISA and Western blotting were applied to detect target protein levels. Luciferase reporter assay and biotin pull-down assay were performed to test molecules interaction. ADRB2 was highly expressed in nasal mucosa of AR patients and was positively correlated with IL-13 stimulation, and knockdown of ADRB2 inhibited IL-13-induced expression of GM-CSF, eotaxin, and MUC5AC in NECs. ADRB2 was directly targeted by miR-15a-5p, and miR-15a-5p inhibited IL-13-induced expression of GM-CSF, eotaxin, and MUC5AC in NECs. ADRB2 mediated the effect of miR-15a-5p on the regulation of nasal epithelial immune responses. ADRB2 is negatively regulated by miR-15a-5p, which inhibits IL-13-induced nasal epithelial inflammatory responses.


Assuntos
Células Epiteliais/imunologia , Interleucina-13/imunologia , MicroRNAs/imunologia , Mucosa Nasal/citologia , Receptores Adrenérgicos beta 2/fisiologia , Rinite Alérgica/genética , Rinite Alérgica/imunologia , Células Cultivadas , Expressão Gênica , Regulação da Expressão Gênica , Humanos , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo
17.
J Gynecol Oncol ; 30(2): e39, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30740961

RESUMO

The Asian Society of Gynecologic Oncology International Workshop 2018 on gynecologic oncology was held in the Ajou University Hospital, Suwon, Korea on the 24th to 25th August 2018. The workshop was an opportunity for Asian doctors to discuss the latest findings of gynecologic cancer, including cervical, ovarian, and endometrial cancers, as well as the future of fertility-sparing treatments, minimally invasive/radical/debulking surgery, radiotherapy, chemotherapy, targeted therapy, and immunotherapy. Clinical guidelines and position statement of Asian countries were presented by experts. Asian clinical trials for gynecologic cancers were reviewed and experts emphasized the point that original Asian study is beneficial for Asian patients. In Junior session, young gynecologic oncologists presented their latest research on gynecologic cancers.


Assuntos
Neoplasias dos Genitais Femininos/terapia , Antineoplásicos/uso terapêutico , Ásia , Ensaios Clínicos como Assunto , Feminino , Neoplasias dos Genitais Femininos/diagnóstico , Neoplasias dos Genitais Femininos/genética , Humanos , Hipertermia Induzida , Imunoterapia , Laparoscopia/métodos , Vacinas contra Papillomavirus , Guias de Prática Clínica como Assunto , Sociedades Médicas
18.
Cancer Lett ; 442: 137-147, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30420335

RESUMO

Continuous estrogen signaling is thought to be the main mechanism causing endometrial cancer (EC). Studies have demonstrated that CD163+ macrophages could promote the development of estrogen-dependent EC, but the mechanisms involved remain unclear. We found that CD163+ macrophages were the dominant macrophages in atypical endometrial hyperplasia and cancer, and their infiltration was positively associated with ERα expression. CD163+ macrophages mainly increased ERα protein levels but with little upregulatory effect on ESR1 (ERα coding gene) transcripts. The ubiquitin-editing enzyme A20, screened from the endometrial microarray obtained from mice receiving a high-fat diet and sustained estrogen-intervened, was highly expressed in endometrial lesions rich with CD163+ macrophages, and positively correlated with ERα expression. Similarly, A20 and ERα were both upregulated by CD163+ macrophages via cytokines such as IL1α, IL17A and TNFα. Mechanistically, A20 overexpression in EC cells prolonged ERα protein half-life without affecting ESR1 transcripts. A20 increased functional ERα protein levels and enhanced estrogen-driven EC cell proliferation through preventing ERα protein degradation by its deubiquitinase activity. Our study revealed that A20-mediated deubiquitination of ERα might be an important mechanism by which CD163+ macrophages sensitize EC cells to estrogen.


Assuntos
Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias do Endométrio/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Macrófagos/metabolismo , Receptores de Superfície Celular/metabolismo , Microambiente Tumoral , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/metabolismo , Animais , Comunicação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Receptor alfa de Estrogênio/genética , Feminino , Células HEK293 , Meia-Vida , Humanos , Macrófagos/patologia , Camundongos Endogâmicos C57BL , Fenótipo , Estabilidade Proteica , Transdução de Sinais/efeitos dos fármacos , Ubiquitinação
19.
Arch Gynecol Obstet ; 299(1): 185-190, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30317387

RESUMO

OBJECTIVES: Perivascular epithelioid cell tumor (PEComa) is a rare condition and the recognition of this condition is limited. Here we report five cases of uterine PEComa to add to the limited understanding of this rare condition. METHODS: Five cases from Obstetrics and Gynecology Hospital of Fudan University were diagnosed as uterine PEComas. We collected the patients' clinical and pathological data as well as their outcomes. RESULTS: All the five cases were diagnosed post-operationally. Fertility-sparing surgery was done for the first case and had a mass resection only. She delivered a healthy boy through the cesarean section in November 2016 and neither recurrence nor metastasis was found for 71 months. Hysterectomy was done for the other four cases. Adjuvant chemotherapy was also given for case 2 and case 4. Case 2 had combined endometrial cancer, which could be associated with tuberous sclerosis complex (TSC). She was followed up for 22 months and neither recurrence nor metastasis was detected. Neither recurrence nor metastasis was found in case 3 for 33 months. However, the patient in case 4 died of multiple dissemination and multiple organs failures, 10 months after the second surgery. The patient in case 5 had the hysterectomy and left adnexal resection and in this case we had no data about her long-term outcomes. CONCLUSION: It is still challenging to detect and diagnose uterine PEComa clinically and no consensus or guidelines have been established regarding the treatment of this condition. More case studies are needed to enlighten the underlying mechanism and help optimize the therapies for this condition.


Assuntos
Neoplasias de Células Epitelioides Perivasculares/patologia , Neoplasias Uterinas/patologia , Adulto , Cesárea , Neoplasias do Endométrio/complicações , Neoplasias do Endométrio/patologia , Neoplasias do Endométrio/cirurgia , Feminino , Humanos , Histerectomia , Leiomioma/complicações , Leiomioma/patologia , Pessoa de Meia-Idade , Recidiva Local de Neoplasia , Neoplasias de Células Epitelioides Perivasculares/mortalidade , Neoplasias de Células Epitelioides Perivasculares/cirurgia , Gravidez , Esclerose Tuberosa/complicações , Esclerose Tuberosa/patologia , Esclerose Tuberosa/cirurgia
20.
Biochem Biophys Res Commun ; 503(4): 2918-2923, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30139517

RESUMO

Endometrial cancer (EC) has recently become a major gynecological cancer and endometrial hyperplasia increases the risk for developing EC. Previous studies have reported that human high temperature requirement factor A3 (HtrA3), a member of ATP independent serine proteases family, is involved in endometrial carcinogenesis. However, the underlying mechanism of HtrA3 function is unclear in endometrial hyperplasia and cancer. In this study, we investigated that HtrA3 expression was reduced in endometrial hyperplasia as well as EC. The circulating levels of HtrA3 were also significantly reduced in both atypical hyperplasia and EC. Whether hypoxia is involved in the reduction of HtrA3 in EC was further investigated. Immunohistochemistry (IHC) scores of Glut1 and HtrA3 in type 1 and type 2 EC tissues showed the inverse correlation. And hypoxic condition reduced the expression of HtrA3. Furthermore, silencing HtrA3 promoted EC cell migration. Our study demonstrated the reduced levels of HtrA3 in endometrial hyperplasia including atypical hyperplasia which is a premalignant condition; and as the degree of hypoxia increases in EC, HtrA3 eventually loses its expression. Hypoxia is responsible for the reduction of HtrA3 which in turn promotes EC progression. These findings suggested that HtrA3 is an important adaptor in hypoxic regions that drives endometrial cancer development.


Assuntos
Hiperplasia Endometrial/metabolismo , Neoplasias do Endométrio/metabolismo , Serina Endopeptidases/metabolismo , Carcinogênese , Progressão da Doença , Hiperplasia Endometrial/etiologia , Feminino , Transportador de Glucose Tipo 1/análise , Humanos , Hipóxia , Imuno-Histoquímica , Oxirredução , Serina Endopeptidases/análise , Serina Endopeptidases/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...