Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Reprod Biol Endocrinol ; 22(1): 41, 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38605340

RESUMO

BACKGROUND: Premature ovarian failure (POF) caused by cisplatin is a severe and intractable sequela for young women with cancer who received chemotherapy. Cisplatin causes the dysfunction of granulosa cells and mainly leads to but is not limited to its apoptosis and autophagy. Ferroptosis has been also reported to participate, while little is known about it. Our previous experiment has demonstrated that endometrial stem cells (EnSCs) can repair cisplatin-injured granulosa cells. However, it is still unclear whether EnSCs can play a repair role by acting on ferroptosis. METHODS: Western blotting and quantitative reverse-transcription polymerase chain reaction (qRT-PCR) were applied to detect the expression levels of ferroptosis-related genes. CCK-8 and 5-Ethynyl-2'-deoxyuridine (EdU) assays were used to evaluate cell viability. Transmission electron microscopy (TEM) was performed to detect ferroptosis in morphology. And the extent of ferroptosis was assessed by ROS, GPx, GSSG and MDA indicators. In vivo, ovarian morphology was presented by HE staining and the protein expression in ovarian tissue was detected by immunohistochemistry. RESULTS: Our results showed that ferroptosis could occur in cisplatin-injured granulosa cells. Ferroptosis inhibitor ferrostatin-1 (Fer-1) and EnSCs partly restored cell viability and mitigated the damage of cisplatin to granulosa cells by inhibiting ferroptosis. Moreover, the repair potential of EnSCs can be markedly blocked by ML385. CONCLUSION: Our study demonstrated that cisplatin could induce ferroptosis in granulosa cells, while EnSCs could inhibit ferroptosis and thus exert repair effects on the cisplatin-induced injury model both in vivo and in vitro. Meanwhile, Nrf2 was validated to participate in this regulatory process and played an essential role.


Assuntos
Cisplatino , Ferroptose , Fator 2 Relacionado a NF-E2 , Feminino , Humanos , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Células da Granulosa/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Células-Tronco/metabolismo
2.
Cell Mol Biol (Noisy-le-grand) ; 70(4): 202-211, 2024 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-38678604

RESUMO

DNA replication and sister chromatid cohesion 1 (DSCC1) exerts various functions including sister chromatid cohesion. DSCC1 overexpression plays an important role in cancer development, such as in colorectal, breast, and hepatocellular cancers. The specific role of DSCC1 in tumor progression remains largely unknown, necessitating a pan-cancer investigation to understand the potential function of DSCC1 in various cancers. In this study, we obtained data on physiological conditions, transcriptional expression, survival prognosis, genomic alteration, genomic instability, enriched pathways, immune infiltration, and immunotherapy from The Cancer Genome Atlas, The Genotype-Tissue Expression, cBioPortal, and other publicly available databases to systematically characterize the oncogenic and immunological roles of DSCC1 in 33 different cancers. We found that DSCC1 expression was upregulated at both mRNA and protein levels in various cancers. Additionally, DSCC1 expression was associated with higher tumor stage and grade in specific cancers. DSCC1 was a potential pan-cancer prognostic biomarker for its close association with patient prognosis and a diagnostic biomarker for its high predictive value in distinguishing tumor tissues from normal tissues. DSCC1 was universally amplified across different cancers and tightly associated with genomic instability. Moreover, DSCC1 had a close relationship with tumor immune cell infiltration; thus, it could be used as a potential biomarker for predicting the response and survival of patients with cancer who receive immune checkpoint blockade treatment. To sum up, our study revealed that DSCC1 is a promising target for tumor therapy.


Assuntos
Biomarcadores Tumorais , Regulação Neoplásica da Expressão Gênica , Instabilidade Genômica , Neoplasias , Proteínas Nucleares , Humanos , Biomarcadores Tumorais/genética , Imunoterapia , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/diagnóstico , Prognóstico , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia
3.
Neuropsychol Rehabil ; : 1-20, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38593026

RESUMO

The weekly calendar planning activity (WCPA) is a performance-based assessment of executive function (EF) via a cognitively-based instrumental activity of daily life (C-IADL). This study aimed to examine the validity of the Chinese version of the WCPA in adults with stroke and to explore the characteristics of cognitive strategy use among the population. Fifty-eight hospitalized patients with stroke aged 26-82 years and 53 controls completed the WCPA, two neuropsychological tests and instrumental activity of daily life (IADL) scale. Participants with stroke were subdivided into a stroke cognitive impaired group (Stroke-CI) and a general stroke group (Stroke-NCI) based on the Montreal Cognitive Assessment. Results showed that the WCPA was able to discriminate between Stroke-CI with controls and the Stroke-NCI group with controls. We found significant limitations in stroke patients' ability to use strategies. Concurrent and ecological validities were demonstrated through correlations between the neuropsychological test scores, IADL and the WCPA performance. This study provides initial evidence for the validity of the Chinese version of the WCPA-10 for adults with stroke and suggests the need to use performance-based tests even in patients with normal cognitive screening test results. The WCPA could provide useful information for strategy-based interventions for adults with stroke.

4.
Support Care Cancer ; 31(2): 120, 2023 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-36648565

RESUMO

PURPOSE: This study aimed to explore Chinese breast cancer patients' quality of sexual life (QSL) and factors associated with QSL. METHODS: The questionnaires in this cross-sectional study include the general information questionnaire, cognition and assessment of sexual health questionnaire, Self-acceptance Questionnaire (SAQ), Medical Coping Modes Questionnaire (MCMQ), and Quality of Sexual Life Questionnaire (QSLQ); 201 breast cancer patients were required to complete the questionnaires assessing characteristic information, cognition and assessment of sexual health, QSL, self-acceptance, and coping style. Finally, hierarchical regression was used to analyze the factors associated with QSL in Chinese breast cancer patients. RESULTS: The mean age (at the time of the survey) of the breast cancer patients was 48.31±9.15. The mean score of the QSLQ (range 28-140) was 75.14±16.57. Hierarchical regression analysis showed that the associated factors of breast cancer patients' QSL included age (at the time of the survey), education level, type of surgery, cognition and assessment of sexual health, self-acceptance, and avoidance and acceptance-resignation coping styles, that independent variables explained 60.4% of the variance. CONCLUSION: The QSL among Chinese breast cancer patients needs to be improved. Our findings indicated that breast cancer patients with older age, lower education level, or modified radical mastectomy have poor QSL. Breast cancer patients learn correct information about sexual health, enhance self-acceptance, and reduce acceptance-resignation, and avoidance coping could be intervention strategies to improve their QSL.


Assuntos
Neoplasias da Mama , Feminino , Humanos , Adaptação Psicológica , Estudos Transversais , População do Leste Asiático , Mastectomia , Qualidade de Vida , Inquéritos e Questionários , China
5.
World J Surg Oncol ; 21(1): 309, 2023 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-37759234

RESUMO

BACKGROUND: Although WD repeat and high-mobility group box DNA binding protein 1 (WDHD1) played an essential role in DNA replication, chromosome stability, and DNA damage repair, the panoramic picture of WDHD1 in human tumors remains unclear. Hence, this study aims to comprehensively characterize WDHD1 across 33 human cancers. METHODS: Based on publicly available databases such as TCGA, GTEx, and HPA, we used a bioinformatics approach to systematically explore the genomic features and biological functions of WDHD1 in pan-cancer. RESULTS: WDHD1 mRNA levels were significantly increased in more than 20 types of tumor tissues. Elevated WDHD1 expression was associated with significantly shorter overall survival (OS) in 10 tumors. Furthermore, in uterine corpus endometrial carcinoma (UCEC) and liver hepatocellular carcinoma (LIHC), WDHD1 expression was significantly associated with higher histological grades and pathological stages. In addition, WDHD1 had a high diagnostic value among 16 tumors (area under the ROC curve [AUC] > 0.9). Functional enrichment analyses suggested that WDHD1 probably participated in many oncogenic pathways such as E2F and MYC targets (false discovery rate [FDR] < 0.05), and it was involved in the processes of DNA replication and DNA damage repair (p.adjust < 0.05). WDHD1 expression also correlated with the half-maximal inhibitory concentrations (IC50) of rapamycin (4 out of 10 cancers) and paclitaxel (10 out of 10 cancers). Overall, WDHD1 was negatively associated with immune cell infiltration and might promote tumor immune escape. Our analysis of genomic alterations suggested that WDHD1 was altered in 1.5% of pan-cancer cohorts and the "mutation" was the predominant type of alteration. Finally, through correlation analysis, we found that WDHD1 might be closely associated with tumor heterogeneity, tumor stemness, mismatch repair (MMR), and RNA methylation modification, which were all processes associated with the tumor progression. CONCLUSIONS: Our pan-cancer analysis of WDHD1 provides valuable insights into the genomic characterization and biological functions of WDHD1 in human cancers and offers some theoretical support for the future use of WDHD1-targeted therapies, immunotherapies, and chemotherapeutic combinations for the management of tumors.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Biologia Computacional , Imunoterapia , Biomarcadores , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , Prognóstico , Proteínas de Ligação a DNA
6.
Reprod Biol Endocrinol ; 20(1): 39, 2022 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-35219326

RESUMO

BACKGROUND: Premature ovarian failure (POF) is a serious problem for young women who receive chemotherapy, and its pathophysiological basis is the dysfunction of granulosa cells. According to previous reports, menstrual-derived stem cells (MenSCs) can restore ovarian function and folliculogenesis in mice with chemotherapy-induced POF. Fat mass- and obesity-associated (FTO) was reported to be associated with oocyte development and maturation. FTO was decreased in POF and may be a biomarker for the occurrence of POF. Knockdown of FTO in granulosa cells promoted cell apoptosis and inhibited proliferation. But the relationship between FTO and ovarian repair was still unclear. This study was aimed at investigating the FTO expression level and the role of FTO in the MenSCs recovering the function of injured granulosa cells. METHOD: First, cisplatin was used to establish a granulosa cell injury model. Then, the MenSCs and injured granulosa cell coculture model and POF mouse model were established in this study to explore the role of FTO. Furthermore, gain- and loss-of-function studies, small interfering RNA transfection, and meclofenamic acid (MA), a highly selective inhibitor of FTO, studies were also conducted to clarify the regulatory mechanism of FTO in granulosa cells. RESULTS: MenSCs coculture could improve the function of injured granulosa cells by increasing the expression of FTO. MenSCs transplantation restored the expression of FTO in the ovaries of POF mice. Overexpression of FTO restored the injured cell proliferation and decreased apoptosis by regulating the expression of BNIP3. Down-regulation of FTO got the opposite results. CONCLUSIONS: In the treatment of MenSCs, FTO has a protective effect, which could improve the viability of granulosa cells after cisplatin treatment by decreasing the expression of BNIP3. Meanwhile, FTO may provide new insight into therapeutic targets for the chemotherapy-induced POF.


Assuntos
Dioxigenase FTO Dependente de alfa-Cetoglutarato/fisiologia , Antineoplásicos/efeitos adversos , Citoproteção/genética , Células da Granulosa/efeitos dos fármacos , Adulto , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Células Cultivadas , Cisplatino/efeitos adversos , Modelos Animais de Doenças , Feminino , Células da Granulosa/patologia , Células da Granulosa/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Insuficiência Ovariana Primária/induzido quimicamente , Insuficiência Ovariana Primária/genética , Insuficiência Ovariana Primária/patologia
7.
Reproduction ; 160(1): 21-29, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32272451

RESUMO

A successful pregnancy crucially depends on well-regulated extravillous trophoblast migration and invasion. Maternally expressed gene 3 (MEG3) is a long noncoding RNA that plays an important role in regulating trophoblast cells cell function. As previously reported, the expression of MEG3 was reduced in preeclampsia, and downregulation of MEG3 could suppress trophoblast cells migration and promote its apoptosis. However, the downstream regulatory mechanism of MEG3 remains unknown. As reported, MEG3 could inhibit cell proliferation in endometrial carcinoma by regulating Notch signaling. Our previous studies have demonstrated that Notch1 is downregulated in preeclampsia and that inhibiting the expression of Notch1 could promote trophoblast cell apoptosis. Therefore, this study was designed to investigate the role of MEG3 and its the relationship with Notch1 in trophoblasts. In this study, the mRNA expression levels of both MEG3 and Notch1 were decreased in preeclampsia placenta (n = 15) compared to the normal samples (n = 15). Exogenous upregulation and downregulation of MEG3 in HTR8/SVneo cells were performed to investigate the role of MEG3 in cell biological behavior and its effects on Notch1 expression. The results showed that MEG3 enhancement promoted trophoblast cell migration and invasion and inhibited cell apoptosis. Downregulation of MEG3 elicited the opposite results. Associated factors, such as matrix metalloproteinases 2 (MMP2), BAX, and Bcl-2, were examined at the mRNA and protein levels. Our study demonstrated that MEG3 could regulate Notch1 expression to modulate trophoblast cell migration, invasion, and apoptosis, which may represent the molecular mechanism of poor placentation during preeclampsia.


Assuntos
Apoptose , Movimento Celular , Regulação da Expressão Gênica , Pré-Eclâmpsia/patologia , RNA Longo não Codificante/antagonistas & inibidores , Receptor Notch1/antagonistas & inibidores , Trofoblastos/patologia , Adulto , Estudos de Casos e Controles , Proliferação de Células , Feminino , Humanos , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/metabolismo , Gravidez , Trofoblastos/metabolismo
8.
J Cell Biochem ; 120(9): 15997-16007, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31099432

RESUMO

Successful pregnancy depends on correct spiral artery (SpA) remodeling, and thus, on normal patterns of the vascular smooth muscle cell (VSMC) apoptosis and migration. Uterine natural killer (uNK) cells-derived transforming growth factor ß1 (TGF-ß1) is known to mediate the separation of VSMC layers via as yet unknown mechanisms. Likewise, the long noncoding RNA maternally expressed gene 3 (MEG3) is a tumor suppressor that has been shown to regulate cancer cell apoptosis and migration; however, its role in VSMC loss is unclear. Thus, the aim of the present study was to assess the effects of uNK-derived TGF-ß1 and MEG3 on VSMC function during SpA. Analyses were conducted to assess the effects of downregulating MEG3 expression, and/or administering treatments to increase or block TGF-ß1 signaling on VSMC survival and behavior. The results of these analyses showed that treating the VSMC with uNK cell-derived supernatant or recombinant human TGF-ß1 promoted MEG3 and matrix metalloprotease 2 expression and VSMC apoptosis and migration, and suppressed VSMC proliferation. Conversely, MEG3 silencing promoted VSMC proliferation and inhibited VSMC apoptosis and migration. Notably, TGF-ß1 signaling induction had no significant effect on the proliferation, apoptosis, nor migration of the MEG3-silenced VSMC. Together, these findings suggest that MEG3 is regulated by uNK-derived TGF-ß1, and itself mediates VSMC apoptosis and migration; thus, it may be an important positive regulator of VSMCs separation during maternal SpA remodeling.


Assuntos
Células Matadoras Naturais/imunologia , Músculo Liso Vascular/citologia , RNA Longo não Codificante/genética , Fator de Crescimento Transformador beta1/genética , Adulto , Apoptose , Movimento Celular , Proliferação de Células , Células Cultivadas , Feminino , Humanos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Transdução de Sinais , Adulto Jovem
9.
Mol Reprod Dev ; 86(6): 686-695, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31066488

RESUMO

Extravillous trophoblasts (EVTs) migrate into uterine decidua and induce vascular smooth muscle cell (VSMC) loss through mechanisms thought to involve migration and apoptosis, achieving complete spiral artery remodeling. Long noncoding RNA maternally expressed gene 3 (MEG3) can regulate diverse cellular processes, such as proliferation and migration, and has been discovered highly expressed in human placenta tissues. However, little is known about the role of MEG3 in modulating EVT functions and EVT-induced VSMC loss. In this study, we first examined the location of MEG3 in human first-trimester placenta by in situ hybridization. Then, exogenous upregulation of MEG3 in HTR-8/SVneo cells was performed to investigate the effects of MEG3 on EVT motility and EVT capacity to displace VSMCs. Meanwhile, the molecules mediating EVT-induced VSMC loss, such as tumor necrosis factor-α (TNF-α), Fas ligand (FasL), and tumor necrosis factor-α-related apoptosis-inducing ligand (TRAIL) were detected at transcriptional and translational levels. Finally, VSMCs were cocultured with MEG3-upregulated HTR-8/SVneo to explore the role of MEG3 on EVT-mediated VSMC migration and apoptosis. Results showed that MEG3 was expressed in trophoblasts in placental villi and decidua, and MEG3 enhancement inhibited HTR-8/SVneo migration and invasion. Meanwhile, the displacement of VSMCs by HTR-8/SVneo and the expression of TNF-α, FasL and TRAIL in HTR-8/SVneo were reduced following MEG3 overexpression in HTR-8/SVneo. Furthermore, HTR-8/SVneo with MEG3 upregulation impaired VSMC migration and apoptosis. The PI3K/Akt pathway, which is possibly downstream, was inactivated in MEG3-upregulated HTR-8/SVneo. These findings suggest that MEG3 might be a negative regulator of spiral artery remodeling via suppressing EVT invasion and EVT-mediated VSMC loss.


Assuntos
Movimento Celular , Decídua/metabolismo , Músculo Liso Vascular/metabolismo , RNA Longo não Codificante/metabolismo , Trofoblastos/metabolismo , Artéria Uterina/metabolismo , Remodelação Vascular , Linhagem Celular , Decídua/citologia , Feminino , Humanos , Músculo Liso Vascular/citologia , Gravidez , Primeiro Trimestre da Gravidez , Trofoblastos/citologia , Artéria Uterina/citologia
10.
Mol Reprod Dev ; 86(7): 896-907, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31115130

RESUMO

Successful pregnancy depends on the precise regulation of extravillous trophoblast cell invasion ability. MicroRNA-210-3p (miR-210), which is increased in the placenta of pre-eclampsia. Furthermore, miR-210 could inhibit trophoblasts invasion and might act as a serum biomarker for pre-eclampsia. Previous studies have demonstrated that miR-210 regulates HUVEC (human umbilical vein endothelial cell)-mediated angiogenesis by regulating the NOTCH1 signaling pathway. Studies by our group have previously identified that NOTCH1 plays a positive role in regulating trophoblast functions. However, the miR-210/NOTCH1 signaling pathway in the regulation of trophoblasts and pre-eclampsia has not been characterized. Therefore, this study was conducted to investigate the role of miR-210 and its relationship with NOTCH1 in trophoblasts. We first examined the expression levels of miR-210 and NOTCH1 in pre-eclamptic and normals placentas. Next, the expression and location of miR-210 and NOTCH1 in the first-trimester villi, maternal decidua, and placenta of late pregnancy were shown via in situ hybridization and immunohistochemistry. The trophoblast cell line HTR-8/SVneo was used to investigate the effects of miR-210 on the expression of NOTCH1 and cell bioactivity by upregulation and downregulation strategies. The results showed that miR-210 expression was increased, whereas NOTCH1 expression was decreased in pre-eclamptic placenta compared with controls. Upregulation of miR-210 decreased NOTCH1 expression, impaired HTR-8/SVneo proliferation, migration, invasion, and tube-like formation capabilities, and promoted apoptosis. In contrast, downregulation of miR-210 resulted in the opposite effects. These findings suggested that miR-210 might act as a contributor to trophoblast dysfunction by attenuating NOTCH1 expression.


Assuntos
MicroRNAs/metabolismo , Pré-Eclâmpsia/metabolismo , Receptor Notch1/metabolismo , Trofoblastos/metabolismo , Adulto , Apoptose/genética , Linhagem Celular , Movimento Celular/genética , Proliferação de Células/genética , Regulação para Baixo , Feminino , Humanos , Imuno-Histoquímica , Hibridização In Situ , MicroRNAs/genética , Placenta/metabolismo , Pré-Eclâmpsia/patologia , Gravidez , Primeiro Trimestre da Gravidez/metabolismo , Transfecção , Regulação para Cima
11.
J Ovarian Res ; 17(1): 62, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38491479

RESUMO

Premature ovarian failure (POF) is a devastating condition for women under 40 years old. Chemotherapy, especially the use of cisplatin, has been demonstrated to promote the apoptosis of granulosa cells in primary and secondary follicles, leading to POF. Our previous studies demonstrated that fat mass- and obesity-associated (FTO) plays an essential role in protecting granulosa cells from cisplatin-induced cytotoxicity. Various studies have suggested that the Hippo/YAP signalling pathway plays a significant role in regulating cell apoptosis and proliferation. Additionally, YAP1 is the main downstream target of the Hippo signalling pathway and is negatively regulated by the Hippo signalling pathway. However, whether the Hippo/YAP signalling pathway is involved in the protective effect of FTO on granulosa cells has not been determined. In this study, we found that after cisplatin treatment, the apoptosis of granulosa cells increased in a concentration-dependent manner, accompanied by the downregulation of FTO and YAP1. Furthermore, overexpression of FTO decreased cisplatin-induced granulosa cell apoptosis, inhibited the Hippo/YAP kinase cascade-induced phosphorylation of YAP1, and promoted the entry of YAP1 into the nucleus. The downstream targets of YAP1 (CTGF, CYR61, and ANKRD1) were also increased. Si-RNA-mediated downregulation of FTO promoted cisplatin-induced granulosa cell apoptosis, activated the Hippo/YAP kinase cascade, and inhibited the YAP1 entry into the nucleus. These effects were completely reversed by the small molecule inhibitor of YAP1-verteporfin (VP). Taken together, these data suggested that FTO-YAP1 plays a positive role in regulating the proliferation of injured granulosa cells induced by cisplatin.


Assuntos
Neoplasias , Transdução de Sinais , Feminino , Humanos , Adulto , Cisplatino/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proliferação de Células , Células da Granulosa/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo
12.
Mol Cell Endocrinol ; 589: 112248, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38663484

RESUMO

Young women undergoing anticancer treatment are at risk of premature ovarian failure (POF). Endometrial-derived stem cells (EnSCs) have demonstrated significant therapeutic potential for treating ovarian insufficiency, although the underlying mechanisms remain to be fully understood. This study aims to further investigate the therapeutic effects of EnSCs, particularly through the paracrine action of fibroblast growth factor 2 (FGF2), on POF. The findings show that exogenous FGF2 enhances the survival of ovarian granulosa cells damaged by cisplatin. FGF2 stimulates the proliferation of these damaged cells by suppressing the Hippo signaling pathway and activating YAP expression. In vivo experiments also revealed that FGF2 treatment significantly improves ovarian reserve and endocrine function in mice with POF. These results suggest that FGF2 can boost the proliferative capacity of damaged ovarian granulosa cells through the Hippo-YAP signaling pathway, providing a theoretical foundation for using EnSCs and FGF2 in clinical treatments for POF.


Assuntos
Proliferação de Células , Fator 2 de Crescimento de Fibroblastos , Células da Granulosa , Via de Sinalização Hippo , Insuficiência Ovariana Primária , Transdução de Sinais , Proteínas de Sinalização YAP , Insuficiência Ovariana Primária/metabolismo , Insuficiência Ovariana Primária/patologia , Feminino , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Células da Granulosa/metabolismo , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/patologia , Animais , Proliferação de Células/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Humanos , Camundongos , Proteínas de Sinalização YAP/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Cisplatino/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética
13.
Heliyon ; 10(10): e31639, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38831834

RESUMO

Stem cells have been documented as a new therapeutic method for ovarian injuries such as premature ovarian failure (POF). However, effects of exosomes (Exos) derived from human endometrial stem cells (EnSCs) on diminished ovarian failure remain to be carefully elucidated. Our study aims to investigate the mechanisms of EnSC-Exos in the recovery of the cisplatin-induced granulosa cell injury model in vitro or POF mouses model in vivo and whether the Hippo signaling pathway is involved in the regulation. In this study, we established successful construction of the cisplatin-induced granulosa cell injury model and evaluated Hippo signaling pathway activation in cisplatin-damaged granulosa cells (GCs). Furthermore, laser scanning confocal microscope and immunofluorescence demonstrated that EnSC-Exos can be transferred to cisplatin-damaged GCs to decrease apoptosis. In addition, the enhanced expression of YAP at the protein level as well as YAP/TEAD target genes, such as CTGF, ANKRD1, and the increase of YAP into the nucleus in immunofluorescence staining after the addition of EnSC-Exos to cisplatin-damaged GCs confirmed the suppression of Hippo signaling pathway. While in vivo, EnSC-Exos successfully remedied POF in a mouse model. Collectively, our findings suggest that chemotherapy-induced POF was associated with the activating of Hippo signaling pathway. Human EnSC-Exos significantly elevated the proliferation of ovarian GCs and the ovarian function by regulating Hippo signaling pathway. These findings provide new insights for further understanding of EnSC-Exos in the recovery of ovary function.

14.
Histol Histopathol ; 38(9): 1069-1077, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36562285

RESUMO

Qinba selenium mushroom is a mushroom belonging to the Basidiomycetes family, which is believed to have anti- oxidant, anti-tumoral and anti-mutagenic activities. However, the efficacy of Qinba selenium mushroom against multiple myeloma has not been confirmed. The present study aimed to investigate the apoptotic effect of FA-2-b-ß, the selenium mushroom extract from Qinba on multiple myeloma (MM) cells. The MM RPMI-8226 cells were treated with FA-2-b-ß at different concentrations and time points. MM RPMI-8226 cell proliferation and apoptosis were detected by the Cell Counting Kit-8 (CCK-8) assay and Annexin V/propidium iodide (PI) assay, RT-QPCR and western blotting analyses were performed to determine the proteins and pathways involved. The results of the present study demonstrated that FA-2-b-ß has high anti-proliferative activities and strong pro-apoptotic effects on MM RPMI-8226 cells, and its pharmacological effects on proliferation changes occurred in a dose- and time-dependent manner. In addition, we found that FA-2-b-ß was able to induce cell apoptosis and promote cell cycle arrest at G0/G1 phase. In summary, the results illustrate the involvement of FA-2-b-ß in mediating G0/G1 cell cycle arrest and apoptosis in MM RPMI-8226 cells, which suggested that FA-2-b-ß might have therapeutic potential against multiple myeloma as an effective compound, and may provide useful information for the development of a novel therapeutic target in this area.


Assuntos
Agaricales , Mieloma Múltiplo , Selênio , Humanos , Linhagem Celular Tumoral , Selênio/farmacologia , Selênio/uso terapêutico , Proliferação de Células , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Apoptose
15.
Cancer Res ; 83(23): 3920-3939, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37729394

RESUMO

Fatty acid metabolism reprogramming is a prominent feature of clear cell renal cell carcinoma (ccRCC). Increased lipid storage supports ccRCC progression, highlighting the importance of understanding the molecular mechanisms driving altered fatty acid synthesis in tumors. Here, we identified that malonyl-CoA decarboxylase (MLYCD), a key regulator of fatty acid anabolism, was downregulated in ccRCC, and low expression correlated with poor prognosis in patients. Restoring MLYCD expression in ccRCC cells decreased the content of malonyl CoA, which blocked de novo fatty acid synthesis and promoted fatty acid translocation into mitochondria for oxidation. Inhibition of lipid droplet accumulation induced by MLYCD-mediated fatty acid oxidation disrupted endoplasmic reticulum and mitochondrial homeostasis, increased reactive oxygen species levels, and induced ferroptosis. Moreover, overexpressing MLYCD reduced tumor growth and reversed resistance to sunitinib in vitro and in vivo. Mechanistically, HIF2α inhibited MLYCD translation by upregulating expression of eIF4G3 microexons. Together, this study demonstrates that fatty acid catabolism mediated by MLYCD disrupts lipid homeostasis to repress ccRCC progression. Activating MLYCD-mediated fatty acid metabolism could be a promising therapeutic strategy for treating ccRCC. SIGNIFICANCE: MLYCD deficiency facilitates fatty acid synthesis and lipid droplet accumulation to drive progression of renal cell carcinoma, indicating inducing MYLCD as a potential approach to reprogram fatty acid metabolism in kidney cancer.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Humanos , Carcinoma de Células Renais/patologia , Neoplasias Renais/patologia , Metabolismo dos Lipídeos , Ácidos Graxos/metabolismo
16.
Front Endocrinol (Lausanne) ; 13: 890623, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35784556

RESUMO

It is widely known that chemotherapy-induced apoptosis of granulosa was the main reason for premature ovarian failure (POF). In addition, accumulating evidence has demonstrated that autophagy was involved in it. Studies before have reported that fibroblast growth factor-2 (FGF2) could attenuate cell death via regulating autophagy. In our previous study, FGF2 could decrease granulosa cell apoptosis in cisplatin-induced POF mice. Furthermore, obesity-associated protein [fat mass and obesity-associated protein (FTO)], which decreased significantly in POF mice, could inhibit cell apoptosis via activating autophagy. Moreover, downregulation of FTO could decrease the expression of paracrine factor FGF2. However, the relationship between FTO and FGF2 in granulosa cell autophagy is still unknown. In the present study, Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2-deoxyuridine (EdU) assays showed that exogenous addition of FGF2 could promote cisplatin-induced injured granulosa cell proliferation. Western blotting indicated that FGF2 could inhibit apoptosis of injured granulosa cells via autophagy. Inhibition of autophagy by chemicals suppressed the effect of FGF2 and promoted injured cell apoptosis. In addition, the expression of FTO was decreased in injured cells, and FGF2 addition could reverse it. Overexpression of FTO reduced injured cell apoptosis via activating the autophagy process. Our findings indicated that FGF2 activates autophagy by regulating the expression of FTO, thereby reducing the apoptosis of the injured cells.


Assuntos
Cisplatino , Insuficiência Ovariana Primária , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Animais , Apoptose , Autofagia/fisiologia , Cisplatino/farmacologia , Feminino , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Camundongos , Obesidade , Insuficiência Ovariana Primária/metabolismo
17.
Front Genet ; 13: 906291, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35923695

RESUMO

Endometrial cancer (EC) kills about 76,000 women worldwide, with the highest incidence in industrialized countries. Because of the rise in disease mortality and new diagnoses, EC is now a top priority for women's health. Serine racemase (SRR) is thought to play a role in the central nervous system, but its role in cancers, particularly in EC, is largely unknown. The current study starts with a pan-cancer examination of SRR's expression and prognostic value before delving into SRR's potential cancer-suppressing effect in patients with EC. SRR may affect the endometrial tumor immune microenvironment, according to subsequent immune-related analysis. SRR expression is also linked to several genes involved in specific pathways such as ferroptosis, N6-methyladenosine methylation, and DNA damage repair. Finally, we used the expression, correlation, and survival analyses to investigate the upstream potential regulatory non-coding RNAs of SRR. Overall, our findings highlight the prognostic significance of SRR in patients with EC, and we can formulate a reasonable hypothesis that SRR influences metabolism and obstructs key carcinogenic processes in EC.

18.
J Leukoc Biol ; 111(3): 711-723, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34312907

RESUMO

Invariant natural killer T cells (iNKTs) bridge the innate immunity with the adaptive immunity and their interaction with B cells has been extensively studied. Here, we give a complete overview of these two cells, from their mechanism of interaction to clinical prospects and existing problems. In our introduction, we describe the relationship between iNKTs and B cells and explore the current research hotspots and future directions. We begin with how B cells interact and benefit from the innate and adaptive help of iNKTs. Next, we describe the multiple roles of these cells in infections, autoimmunity, and cancers. Lastly, we look into the potential immunotherapies that can be based on iNKTs and the possible treatments for infectious, autoimmune, and other diseases.


Assuntos
Células T Matadoras Naturais , Imunidade Adaptativa , Imunidade Inata , Imunoterapia , Ativação Linfocitária
19.
Front Immunol ; 13: 837991, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35359973

RESUMO

Background: Prostate cancer (PCa) is the most common malignant tumor in men. Although clinical treatments of PCa have made great progress in recent decades, once tolerance to treatments occurs, the disease progresses rapidly after recurrence. PCa exhibits a unique metabolic rewriting that changes from initial neoplasia to advanced neoplasia. However, systematic and comprehensive studies on the relationship of changes in the metabolic landscape of PCa with tumor recurrence and treatment response are lacking. We aimed to construct a metabolism-related gene landscape that predicts PCa recurrence and treatment response. Methods: In the present study, we used differentially expressed gene analysis, protein-protein interaction (PPI) networks, univariate and multivariate Cox regression, and least absolute shrinkage and selection operator (LASSO) regression to construct and verify a metabolism-related risk model (MRM) to predict the disease-free survival (DFS) and response to treatment for PCa patients. Results: The MRM predicted patient survival more accurately than the current clinical prognostic indicators. By using two independent PCa datasets (International Cancer Genome Consortium (ICGC) PCa and Taylor) and actual patients to test the model, we also confirmed that the metabolism-related risk score (MRS) was strongly related to PCa progression. Notably, patients in different MRS subgroups had significant differences in metabolic activity, mutant landscape, immune microenvironment, and drug sensitivity. Patients in the high-MRS group were more sensitive to immunotherapy and endocrine therapy, while patients in the low-MRS group were more sensitive to chemotherapy. Conclusions: We developed an MRM, which might act as a clinical feature to more accurately assess prognosis and guide the selection of appropriate treatment for PCa patients. It is promising for further application in clinical practice.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata , Humanos , Masculino , Recidiva Local de Neoplasia/genética , Prognóstico , Próstata/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/terapia , Microambiente Tumoral
20.
Front Genet ; 13: 1000339, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36199577

RESUMO

Anillin (ANLN) is a unique scaffolding, actin-binding protein, which is essential for the integrity and ingression of the cleavage furrow. It is mainly involved in the cytokinesis process, while its role in various tumors has not been fully addressed and remains largely elusive. To provide a thorough perspective of ANLN's roles among diverse malignancies, we conducted a comprehensive, pan-cancer analysis about ANLN, including but not limited to gene expression levels, prognostic value, biological functions, interacting proteins, immune-related analysis, and predictive value. As a result, when compared to normal tissues, ANLN expression is elevated in most cancers, and its expression also differs in different immune subtypes and molecular subtypes in diverse cancers. In addition, in 17 types of cancer, ANLN expression is increased in early tumor stages, and higher ANLN expression predicts worse survival outcomes in more than ten cancers. Furthermore, ANLN shows close correlations with the infiltration levels of most immune cells, and enrichment analysis using ANLN co-expressed genes reveals that ANLN plays essential roles in cell cycle, mitosis, cellular senescence, and p53 signaling pathways. In the final, ANLN exhibits high accuracy in predicting many cancers, and subsequent multivariate analysis suggests ANLN could be an independent prognostic factor in specific cancer types. Taken together, ANLN is proved to be a novel and promising biomarker for its excellent predictive utility, promising prognostic value, and potential immunological roles in pan-cancer. Targeting ANLN might be an attractive approach to tumor treatment.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA