Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Cell Mol Life Sci ; 81(1): 221, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38763964

RESUMO

In females, the pathophysiological mechanism of poor ovarian response (POR) is not fully understood. Considering the expression level of p62 was significantly reduced in the granulosa cells (GCs) of POR patients, this study focused on identifying the role of the selective autophagy receptor p62 in conducting the effect of follicle-stimulating hormone (FSH) on antral follicles (AFs) formation in female mice. The results showed that p62 in GCs was FSH responsive and that its level increased to a peak and then decreased time-dependently either in ovaries or in GCs after gonadotropin induction in vivo. GC-specific deletion of p62 resulted in subfertility, a significantly reduced number of AFs and irregular estrous cycles, which were same as pathophysiological symptom of POR. By conducting mass spectrum analysis, we found the ubiquitination of proteins was decreased, and autophagic flux was blocked in GCs. Specifically, the level of nonubiquitinated Wilms tumor 1 homolog (WT1), a transcription factor and negative controller of GC differentiation, increased steadily. Co-IP results showed that p62 deletion increased the level of ubiquitin-specific peptidase 5 (USP5), which blocked the ubiquitination of WT1. Furthermore, a joint analysis of RNA-seq and the spatial transcriptome sequencing data showed the expression of steroid metabolic genes and FSH receptors pivotal for GCs differentiation decreased unanimously. Accordingly, the accumulation of WT1 in GCs deficient of p62 decreased steroid hormone levels and reduced FSH responsiveness, while the availability of p62 in GCs simultaneously ensured the degradation of WT1 through the ubiquitin‒proteasome system and autophagolysosomal system. Therefore, p62 in GCs participates in GC differentiation and AF formation in FSH induction by dynamically controlling the degradation of WT1. The findings of the study contributes to further study the pathology of POR.


Assuntos
Hormônio Foliculoestimulante , Células da Granulosa , Folículo Ovariano , Proteína Sequestossoma-1 , Ubiquitinação , Proteínas WT1 , Animais , Hormônio Foliculoestimulante/metabolismo , Hormônio Foliculoestimulante/farmacologia , Feminino , Proteínas WT1/metabolismo , Proteínas WT1/genética , Camundongos , Folículo Ovariano/metabolismo , Folículo Ovariano/efeitos dos fármacos , Células da Granulosa/metabolismo , Células da Granulosa/efeitos dos fármacos , Proteína Sequestossoma-1/metabolismo , Proteína Sequestossoma-1/genética , Camundongos Endogâmicos C57BL , Autofagia/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Humanos , Camundongos Knockout
2.
Sci Bull (Beijing) ; 69(8): 1122-1136, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38302330

RESUMO

In a growing follicle, the survival and maturation of the oocyte largely depend on support from somatic cells to facilitate FSH-induced mutual signaling and chemical communication. Although apoptosis and autophagy in somatic cells are involved in the process of FSH-induced follicular development, the underlying mechanisms require substantial study. According to our study, along with FSH-induced antral follicles (AFs) formation, both lysine-specific demethylase 1 (LSD1) protein levels and autophagy increased simultaneously in granulosa cells (GCs) in a time-dependent manner, we therefore evaluated the importance of LSD1 upon facilitating the formation of AFs correlated to autophagy in GCs. Conditional knockout of Lsd1 in GCs resulted in significantly decreased AF number and subfertility in females, accompanied by marked suppression of the autophagy in GCs. On the one hand, depletion of Lsd1 resulted in accumulation of Wilms tumor 1 homolog (WT1), at both the protein and mRNA levels. WT1 prevented the expression of FSH receptor (Fshr) in GCs and thus reduced the responsiveness of the secondary follicles to FSH induction. On the other hand, depletion of LSD1 resulted in suppressed level of autophagy by upregulation of ATG16L2 in GCs. We finally approved that LSD1 contributed to these sequential activities in GCs through its H3K4me2 demethylase activity. Therefore, the importance of LSD1 in GCs is attributable to its roles in both accelerating autophagy and suppressing WT1 expression to ensure the responsiveness of GCs to FSH during AFs formation.


Assuntos
Células da Granulosa , Folículo Ovariano , Feminino , Autofagia/genética , Hormônio Foliculoestimulante/farmacologia , Células da Granulosa/metabolismo , Folículo Ovariano/metabolismo , Transdução de Sinais
3.
Toxicol Appl Pharmacol ; 476: 116671, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37633598

RESUMO

Impaired fertility is the major side effect of chemotherapy for female cancer patients, accumulated evidence indicates this is associated with damage on oocyte quality, but the underlying mechanisms remain unclear. Previously we reported that doxorubicin (DXR) exposure, one of the most widely used chemotherapy drugs, disrupted mouse oocyte meiotic maturation in vitro. In the current study, we identified that SIRT1 expression was remarkably reduced in DXR exposure oocytes. Next, we found that increasing SIRT1 expression by resveratrol partially alleviated the effects of DXR exposure on oocyte maturation, which was counteracted by SIRT1 inhibition. Furthermore, we revealed that increasing SIRT1 expression mitigated DXR induced oocyte damage through reducing ROS levels, increasing antioxidant enzyme MnSOD expression, and preventing spindle and chromosome disorganization, lowering the incidence of aneuploidy. Importantly, by performing in vitro fertilization and embryo transfer assays, we demonstrated that increasing SIRT1 expression significantly improved the fertilization ability, developmental competence of oocytes and early embryos. In summary, our data uncover that SIRT1 reduction represents one mechanism that mediates the effects of DXR exposure on oocyte quality.


Assuntos
Oócitos , Sirtuína 1 , Feminino , Animais , Camundongos , Sirtuína 1/genética , Estresse Oxidativo , Antioxidantes , Doxorrubicina/toxicidade
4.
Fertil Steril ; 120(4): 899-910, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37247688

RESUMO

OBJECTIVE: To explore the dynamic transcriptional regulatory network of primordial follicle fate in obese mice to elucidate the potential mechanism of primordial follicle depletion. DESIGN: Experimental study and transcriptomic analysis. ANIMALS: Healthy (n=15) and obese (n=15) female mice. INTERVENTIONS: Six-week-old CD-1 mice were divided into healthy and high-fat diet groups and fed continuously for 12 weeks. The diet of healthy mice contained 10% fat. The diet of high-fat mice contained 60% fat. MAIN OUTCOME MEASURES: Primordial to primary follicle transition rate, gene expression changes, enriched Kyoto Encyclopedia of Genes and Genomes pathways, and ferroptosis. RESULTS: Primordial follicle depletion was increased in the ovaries of obese mice. We found that deposited fat around primordial and primary follicles of obese mice was higher than that for healthy mice. The proliferation of granulosa cells around primary follicles was increased in obese mice. In addition, we uncovered specific gene signatures associated with the primordial to primary follicle transition (PPT) in obese mice using laser capture microdissection RNA sequencing analysis. Gene set enrichment analysis indicated that ferroptosis, cell oxidation, vascular endothelial growth factor, and mammalian target of rapamycin signaling were increased significantly in the primordial follicles of obese mice. Notably, the ferritin, acyl CoA synthetase long-chain family member 4, and solid carrier family 7 member 11 associated proteins of the ferroptosis signaling pathway were significantly increased in the PPT phase of obese mice. CONCLUSION: Our work suggests that ferroptosis is a key pathway activated within immature ovarian follicles in the context of obesity and that the process may be involved in the physiological regulation of the PPT as well.


Assuntos
Transcriptoma , Fator A de Crescimento do Endotélio Vascular , Feminino , Animais , Camundongos , Camundongos Obesos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Folículo Ovariano/fisiologia , Células da Granulosa , Mamíferos
5.
Molecules ; 28(5)2023 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-36903358

RESUMO

Premature ovarian insufficiency (POI) is a complex disease which causes amenorrhea, hypergonadotropism and infertility in patients no more than 40 years old. Recently, several studies have reported that exosomes have the potential to protect ovarian function using a POI-like mouse model induced by chemotherapy drugs. In this study, the therapeutic potential of exosomes derived from human pluripotent stem cell-mesenchymal stem cells (hiMSC exosomes) was evaluated through a cyclophosphamide (CTX)-induced POI-like mouse model. POI-like pathological changes in mice were determined by serum sex-hormones levels and the available number of ovarian follicles. The expression levels of cellular proliferation proteins and apoptosis-related proteins in mouse ovarian granulosa cells were measured using immunofluorescence, immunohistochemistry and Western blotting. Notably, a positive effect on the preservation of ovarian function was evidenced, since the loss of follicles in the POI-like mouse ovaries was slowed. Additionally, hiMSC exosomes not only restored the levels of serum sex hormones, but also significantly promoted the proliferation of granulosa cells and inhibited cell apoptosis. The current study suggests that the administration of hiMSC exosomes in the ovaries can preserve female-mouse fertility.


Assuntos
Exossomos , Células-Tronco Mesenquimais , Insuficiência Ovariana Primária , Humanos , Feminino , Camundongos , Animais , Adulto , Exossomos/metabolismo , Insuficiência Ovariana Primária/induzido quimicamente , Insuficiência Ovariana Primária/patologia , Insuficiência Ovariana Primária/terapia , Ciclofosfamida/farmacologia , Células da Granulosa/metabolismo , Apoptose , Proliferação de Células , Células-Tronco Mesenquimais/metabolismo
6.
J Proteomics ; 272: 104772, 2023 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-36414229

RESUMO

Early ovarian follicular development is regulated by multiple proteins and signaling pathways, including the Wnt gene. To explore the regulatory mechanism of Wnt signaling on early ovarian follicular development, ovaries from 17.5 days post coitum (17.5 dpc) mice were collected and cultured in vitro for four days in the presence of IWP2 as a Wnt activity inhibitor and KN93 as a CaMKII inhibitor. LFQ proteomics technique was then used to analyze the significant differentially abundant (P-SDA) 93 and 262 proteins in the IWP2 and KN93 groups, respectively. Of these, 63 up-regulated proteins and 30 down-regulated proteins were identified for IWP2, along with 3 significant KEGG pathways (P < 0.05). For the KN93 group, 168 up-regulated proteins and 94 down-regulated ones were P-SDA, with 9 significant KEGG pathways also noted (P < 0.05). In both IWP2 and KN93 groups, key pathways (Wnt signaling pathway, Notch signaling pathway, P53 signaling pathway, TGF-ß signaling pathway, ovarian steroid production) and metabolic regulation (energy metabolism, metal ion metabolism) were found to be related to early ovarian follicular development. Finally, western blotting demonstrated the regulatory role of Wnt/P53/Caspase3 signaling pathway in mouse ovarian development. These results contribute new knowledge to the understanding of regulatory factors of early ovarian follicular development. SIGNIFICANCE: In this study, label-free quantification (LFQ) was used in combination with liquid chromatography-mass spectrometer (LC-MS/MS) to study potential changes in the proteomic profiles of embryonic mice subjected to Wnt inhibitor IWP2 and CaMKIIinhibitor KN93. In addition, bioinformatics and comparative analyses were performed using publicly available proteomics databases to further explore the underlying mechanisms associated with early mouse ovarian growth and development.


Assuntos
Proteômica , Proteína Supressora de Tumor p53 , Camundongos , Animais , Cromatografia Líquida , Proteômica/métodos , Espectrometria de Massas em Tandem , Via de Sinalização Wnt
7.
J Mol Cell Biol ; 13(9): 646-661, 2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34097060

RESUMO

Embryo implantation in both humans and rodents is initiated by the attachment of a blastocyst to the uterine epithelium. For blastocyst attachment, the uterine epithelium needs to transform at both the structural and molecular levels first, and then initiate the interaction with trophectoderm. Any perturbation during this process will result in implantation failure or long-term adverse pregnancy outcomes. Endocrine steroid hormones, which function through nuclear receptors, combine with the local molecules produced by the uteri or embryo to facilitate implantation. The insulin-like growth factor (IGF) signaling has been reported to play a vital role during pregnancy. However, its physiological function during implantation remains elusive. This study revealed that mice with conditional deletion of Igf1r gene in uteri suffered from subfertility, mainly due to the disturbed uterine receptivity and abnormal embryo implantation. Mechanistically, we uncovered that in response to the nidatory estrogen on D4 of pregnancy, the epithelial IGF1R, stimulated by the stromal cell-produced IGF1, facilitated epithelial STAT3 activation to modulate the epithelial depolarity. Furthermore, embryonic derived IGF2 could activate both the epithelial ERK1/2 and STAT3 signaling through IGF1R, which was critical for the transcription of Cox2 and normal attachment reaction. In brief, our data revealed that epithelial IGF1R was sequentially activated by the uterine stromal IGF1 and embryonic IGF2 to guarantee normal epithelium differentiation during the implantation process.


Assuntos
Implantação do Embrião , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Receptor IGF Tipo 1/metabolismo , Animais , Blastocisto/citologia , Blastocisto/fisiologia , Diferenciação Celular , Células Epiteliais/metabolismo , Estrogênios/metabolismo , Feminino , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Receptor IGF Tipo 1/genética , Fator de Transcrição STAT3/metabolismo , Células Estromais/metabolismo , Útero/metabolismo
8.
Aging (Albany NY) ; 12(9): 7717-7728, 2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32352929

RESUMO

Doxorubicin (DXR), a widely used chemotherapeutic drug, has adverse effects on female fertility in young cancer patients. However, the underlying mechanisms of doxorubicin exposure on female fertility and how to prevent it have not been well studied yet. Here, mouse oocytes were employed to investigate the issues mentioned above. The results showed that doxorubicin treatment impaired oocyte meiotic maturation by destroying spindle assembly and chromosome arrangement. In addition, doxorubicin caused oxidative stress by increasing reactive oxygen species (ROS) levels. Furthermore, doxorubicin led to severe DNA damage in oocytes, which eventually induced apoptosis through DNA damage-P63-Caspase3 pathway. Conversely, resveratrol (RES) effectively improved oocyte quality by restoring spindle and chromosome configuration, reducing ROS levels and inhibiting apoptosis. In summary, our results indicate that RES can protect oocytes against doxorubicin-induced damage.


Assuntos
Oócitos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Prenhez , Espécies Reativas de Oxigênio/metabolismo , Resveratrol/farmacologia , Animais , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Dano ao DNA , Feminino , Camundongos , Modelos Animais , Oócitos/citologia , Gravidez
9.
Biotech Histochem ; 95(8): 605-612, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32292080

RESUMO

As much as 90% of cancer associated mortality follows metastasis of a primary tumor. Circulating tumor cells (CTCs) and CTC clusters are important for metastasis. Compared to CTCs, CTC clusters formed by collective invasion exhibit a 23-50 fold increase in metastatic potential, but the factors that influence collective invasion are largely unknown. Using well defined three-dimensional matrices and different extracellular matrix (ECM) concentrations, we found that cancer cells were more prone to collective invasion at low ECM concentration. Moreover, despite variation of biological factors, changes in ECM microarchitecture, especially the pore size of the matrix, was correlated with the probability of collective invasion, which indicates that the physical microarchitecture of ECM plays an important role in collective invasion of cancer cells.


Assuntos
Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Matriz Extracelular , Alicerces Teciduais , Humanos , Células MCF-7 , Mastodinia
10.
Cancer Sci ; 111(2): 467-476, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31845453

RESUMO

Collective invasion of cancer cells is the key process of circulating tumor cell (CTC) cluster formation, and greatly contributes to metastasis. Cancer stem-like cells (CSC) have a distinct advantage of motility for metastatic dissemination. To verify the role of CSC in the collective invasion, we performed 3D assays to investigate the collective invasion from cancer cell spheroids. The results demonstrated that CSC can significantly promote both collective and single-cell invasion. Further study showed that CSC prefer to move outside and lead the collective invasion. More interestingly, approximately 60% of the leader CSC in collective invasion co-expressed both epithelial and mesenchymal genes, while only 4% co-expressed in single invasive CSC, indicating that CSC with hybrid epithelial/mesenchymal phenotype play a key role in cancer cell collective invasion.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/patologia , Invasividade Neoplásica/patologia , Células-Tronco Neoplásicas/patologia , Esferoides Celulares/patologia , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Camundongos , Metástase Neoplásica , Transplante de Neoplasias , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo
11.
Nat Commun ; 10(1): 5719, 2019 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-31844300

RESUMO

It is known that granulosa cells (GCs) mediate gonadotropin-induced oocyte meiosis resumption by releasing EGF-like factors in mammals, however, the detailed molecular mechanisms remain unclear. Here, we demonstrate that luteinizing hormone (LH) surge-induced histone deacetylase 3 (HDAC3) downregulation in GCs is essential for oocyte maturation. Before the LH surge, HDAC3 is highly expressed in GCs. Transcription factors, such as FOXO1, mediate recruitment of HDAC3 to the amphiregulin (Areg) promoter, which suppresses AREG expression. With the LH surge, decreased HDAC3 in GCs enables histone H3K14 acetylation and binding of the SP1 transcription factor to the Areg promoter to initiate AREG transcription and oocyte maturation. Conditional knockout of Hdac3 in granulosa cells in vivo or inhibition of HDAC3 activity in vitro promotes the maturation of oocytes independent of LH. Taking together, HDAC3 in GCs within ovarian follicles acts as a negative regulator of EGF-like growth factor expression before the LH surge.


Assuntos
Anfirregulina/genética , Regulação da Expressão Gênica no Desenvolvimento , Histona Desacetilases/metabolismo , Meiose/genética , Oócitos/crescimento & desenvolvimento , Oogênese/genética , Acetilação , Animais , Células Cultivadas , Feminino , Proteína Forkhead Box O1/metabolismo , Técnicas de Inativação de Genes , Células da Granulosa/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/genética , Histonas/metabolismo , Hormônio Luteinizante/metabolismo , Camundongos , Oogênese/efeitos dos fármacos , Cultura Primária de Células , Regiões Promotoras Genéticas/genética , Fator de Transcrição Sp1/metabolismo
12.
FASEB J ; 33(12): 14703-14716, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31693862

RESUMO

In female mammals, the majority of primordial follicles (PFs) are physiologically quiescent, and only a few of them are activated and enter the growing follicle pool. Specific molecules, such as mammalian target of rapamycin (mTOR) and the serine/threonine kinase Akt (AKT), have been proven to be important for PF activation. However, how the transcription of these genes is regulated is not clear. Although activators of mTOR or AKT have been successfully used to rescue the fertility of patients with premature ovarian insufficiency, the low efficacy and unclear safety profile of these drugs hinder their clinical use in the in vitro activation (IVA) of PFs. Here, sirtuin 1 (SIRT1), an NAD-dependent deacetylase, was demonstrated to activate mouse PFs independent of its deacetylase activity. SIRT1 was prominently expressed in pregranulosa cells (pGCs) and oocytes, and its expression was increased during PF activation. PF activation was achieved by either up-regulating SIRT1 with a specific activator or overexpressing SIRT1. Moreover, SIRT1 knockdown in oocytes or pGCs could significantly suppress PF activation. Further studies demonstrated that SIRT1 enhanced both Akt1 and mTOR expression by acting more as a transcription cofactor, directly binding to the respective gene promoters, than as a deacetylase. Importantly, we explored the potential clinical applications of targeting SIRT1 in IVA via short-term treatment of cultured ovaries from mice and human ovarian tissues to activate PFs by applying the SIRT1 activator resveratrol. RSV-induced IVA could be a candidate strategy to develop more efficient procedures for future clinical treatment of infertility.-Zhang, T., Du, X., Zhao, L., He, M., Lin, L., Guo, C., Zhang, X., Han, J., Yan, H., Huang, K., Sun, G., Yan, L., Zhou, B., Xia, G., Qin, Y., Wang, C. SIRT1 facilitates primordial follicle recruitment independent of deacetylase activity through directly modulating Akt1 and mTOR transcription.


Assuntos
Folículo Ovariano/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Sirtuína 1/metabolismo , Serina-Treonina Quinases TOR/genética , Ativação Transcricional , Animais , Células Cultivadas , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Endogâmicos NOD , Camundongos SCID , Folículo Ovariano/citologia , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sirtuína 1/genética , Serina-Treonina Quinases TOR/metabolismo
13.
Hum Reprod ; 33(11): 2087-2095, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30304446

RESUMO

STUDY QUESTION: What is the function of CAV1 in folliculogenesis and female reproduction? SUMMARY ANSWER: CAV1 regulates germline cyst breakdown and primordial follicle (PF) formation in mice, and CAV1 mutation may be related to premature ovarian insufficiency (POI). WHAT IS KNOWN ALREADY: Pre-granulosa cells are essential for the establishment of the PF pool, which determines female fertility and reproductive lifespan. Cav1 participates in vascularization in fetal mouse ovaries. However, the role of CAV1 in early folliculogenesis and POI pathogenesis remains unclear. STUDY DESIGN, SIZE, DURATION: Cav1 function was investigated in mice and Human Embryonic Kidney 293 cells. Ovaries (six per group) were randomly assigned to Cav1-vivo-morpholino, control and control-morpholino groups, and all experiments were repeated at least three times. To investigate CAV1 mutations in women, 200 Chinese women with POI and 200 control individuals with regular menstrual cycles and normal endocrine profiles were recruited from the Center for Reproductive Medicine of Shandong University between September 2012 and December 2013. PARTICIPANTS/MATERIALS, SETTING, METHODS: Wild-type CD1 mice, Lgr5-EGFP-ires-CreERT2 (Lgr5-KI) reporter mice and Human Embryonic Kidney 293 cells were used for these experiments. Protein expression was detected by Western blot, and quantitative RT-PCR was used to detect gene expression. The expression pattern of CAV1 in mouse ovaries and the phenotype of Cav1 deficiency in mice were detected by immunofluorescence. Pre-granulosa cell proliferation in ovaries was detected by bromodeoxyuridine (BrdU) assay and immunofluorescence. The coding region of the CAV1 gene was sequenced in 200 women with POI and 200 controls. The functional effect of the novel mutation c.142 G > C (p.Glu48Gln) was investigated by Cell Counting Kit-8 (CCK8) assays and Western blot. MAIN RESULTS AND THE ROLE OF CHANCE: We confirmed that Cav1 deficiency in mouse ovary induced by CAV1-vivo-morpholino resulted in more multi-oocyte follicles than in the control and control-morpholino groups (P < 0.01). Suppression of Cav1 decreased Leucine rich repeat containing G protein coupled receptor 5 (Lgr5)-positive cell proliferation (P < 0.01) and reduced the number of Lgr5 and Forkhead box L2 (Foxl2) double-positive cells (P < 0.01). Furthermore, suppression of Cav1 inhibited ovarian epithelial Lgr5-positive cell proliferation and differentiation through the Notch2 signalling pathway. Two of the POI women carried novel CAV1 mutations (c.45 C > G synonymous and c.142 G > C [Glu48Gln]). The deleterious effect of p.Glu48Gln was corroborated by showing that it adversely affected the function of CAV1 in cell proliferation and NOTCH2 expression in HEK293FT cells. LARGE-SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: The novel Glu48Gln mutation was only detected in one of 200 POI patients and we were unable to investigate its effects in the ovary. WIDER IMPLICATIONS OF THE FINDINGS: The identification of CAV1 as a potentially causative gene for POI provides a theoretical basis to devise treatments for POI in women. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by the National Basic Research Program of China (973 Programs: 2012CB944700; 2013CB945501; 2013CB911400; 2014CB943202), the National Key Research and Development Program of China (2016YFC1000604, 2017YFC1001301), the State Key Program of National Natural Science Foundation of China (81430029), and the National Natural Science Foundation of China (31571540, 81522018, 81471509, 81601245, 81701406, 81571406). The authors declare no competing financial interests.


Assuntos
Caveolina 1/metabolismo , Menopausa Precoce/genética , Folículo Ovariano/metabolismo , Receptor Notch2/genética , Adulto , Animais , Feminino , Humanos , Camundongos , Mutação , Ovário , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais
14.
iScience ; 9: 14-26, 2018 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-30368078

RESUMO

Therapeutic effects of photodynamic therapy (PDT) remain largely limited because of tumor hypoxia. Herein, we report safe and versatile nanocatalysts (NCs) for endogenous oxygen generation and imaging-guided enhanced PDT. The NCs (named as PSP) are prepared by coating Prussian blue (PB) with mesoporous silica to load photosensitizer (zinc phthalocyanine, ZnPc), followed by the modification of polyethylene glycol chains. The inner PB not only acts like a catalase for hydrogen peroxide decomposition but also serves as a photothermal agent to increase the local temperature and then speed up the oxygen supply under near-infrared irradiation. The loaded ZnPc can immediately transform the formed oxygen to generate cytotoxic singlet oxygen upon the same laser irradiation due to the overlapped absorption between PB and ZnPc. Results indicate that the PSP-ZnPc (PSPZP) NCs could realize the photothermally controlled improvement of hypoxic condition in cancer cells and tumor tissues, therefore demonstrating enhanced cancer therapy by the incorporation of PDT and photothermal therapy.

15.
BMC Biol ; 16(1): 73, 2018 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-29976179

RESUMO

BACKGROUND: In mammalian females, progressive activation of dormant primordial follicles in adulthood is crucial for the maintenance of the reproductive lifespan. Misregulated activation of primordial follicles leads to various ovarian diseases, such as premature ovarian insufficiency (POI). Although recent studies have revealed that several functional genes and pathways, such as phosphoinositide 3-kinase (PI3K) signaling, play roles in controlling the activation of primordial follicles, our understanding of the molecular networks regulating the activation progress is still incomplete. RESULTS: Here, we identify a new role for cell division cycle 42 (CDC42) in regulating the activation of primordial follicles in mice. Our results show that CDC42 expression increases in oocytes during the activation of primordial follicles in the ovary. Disruption of CDC42 activity with specific inhibitors or knockdown of Cdc42 expression significantly suppresses primordial follicle activation in cultured mouse ovaries. Conversely, the follicle activation ratio is remarkably increased by overexpression of CDC42 in ovaries. We further demonstrate that CDC42 governs the process of primordial follicle activation by binding to phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit beta (p110ß) and regulating the expression levels of PTEN in oocytes. Finally, we extend our study to potential clinical applications and show that a short-term in vitro treatment with CDC42 activators could significantly increase the activation rates of primordial follicles in both neonatal and adult mouse ovaries. CONCLUSION: Our results reveal that CDC42 controls the activation of primordial follicles in the mammalian ovary and that increasing the activity of CDC42 with specific activators might improve the efficiency of in vitro activation approaches, opening avenues for infertility treatments.


Assuntos
Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Oócitos/metabolismo , Folículo Ovariano/crescimento & desenvolvimento , Ovário/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Proteína cdc42 de Ligação ao GTP/fisiologia , Animais , Feminino , Camundongos , Proteína cdc42 de Ligação ao GTP/genética
16.
Clin Sci (Lond) ; 132(7): 759-776, 2018 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-29535265

RESUMO

Polycystic ovary syndrome (PCOS), which is characterized by hyperandrogenism, is a complex endocrinopathy that affects the fertility of 9-18% of reproductive-aged women. However, the exact mechanism of PCOS, especially hyperandrogen-induced anovulation, is largely unknown to date. Physiologically, the natriuretic peptide type C/natriuretic peptide receptor 2 (CNP/NPR2) system is essential for sustaining oocyte meiotic arrest until the preovulatory luteinizing hormone (LH) surge. We therefore hypothesized that the CNP/NPR2 system is also involved in PCOS and contributes to arresting oocyte meiosis and ovulation. Here, based on a dehydroepiandrosterone (DHEA)-induced PCOS-like mouse model, persistent high levels of CNP/NPR2 were detected in anovulation ovaries. Meanwhile, oocytes arrested at the germinal vesicle stage correlated with persistent high levels of androgen and estrogen. We further showed that ovulation failure in these mice could be a result of elevated Nppc/Npr2 gene transcription that was directly increased by androgen (AR) and estrogen (ER) receptor signaling. Consistent with this, anovulation was alleviated by administration of either exogenous human chorionic gonadotropin (hCG) or inhibitors of AR or ER to reduce the level of CNP/NPR2. Additionally, the CNP/NPR2 expression pattern in the anovulated follicles was, to some extent, consistent with the clinical expression in PCOS patients. Therefore, our study highlights the important role an overactive CNP/NPR2 system caused by hyperandrogenism in preventing oocytes from maturation and ovulation in PCOS mice. Our findings provide insight into potential mechanisms responsible for infertility in women with PCOS.


Assuntos
Anovulação/etiologia , Hiperandrogenismo/metabolismo , Peptídeo Natriurético Tipo C/metabolismo , Síndrome do Ovário Policístico/metabolismo , Receptores do Fator Natriurético Atrial/metabolismo , Adulto , Antagonistas de Receptores de Andrógenos , Animais , Estudos de Casos e Controles , Gonadotropina Coriônica , Modelos Animais de Doenças , Antagonistas do Receptor de Estrogênio , Feminino , Células HEK293 , Humanos , Camundongos Endogâmicos BALB C , Ovário/metabolismo , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/metabolismo , Adulto Jovem
17.
ACS Biomater Sci Eng ; 4(8): 3047-3054, 2018 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-33435024

RESUMO

Biocompatible core-shell Fe3O4@C@MnO2 nanoparticles (named as FOCMO NPs) with an average size at 130 nm prepared through an effortless yet efficient strategy were employed as pH-activatable T1-T2* dual-modality magnetic resonance imaging (MRI) contrast agents (CAs). The release rate of Mn ions in acidic PBS (pH = 5.0) was approximately 10 times to that under condition with pH value of 7.4. Benefiting from excellent acid responsiveness, which facilitates the release of ions from FOCMO NPs at tumor region with acidic microenvironment and organelles, the diagnosis accuracy was commendably improved. After intravenous injection of FOCMO NPs, an efficiently intensive contrast in tumors are realized with a distinct enhancement of 127% in T1 MRI signal 24 h after the administration. Moreover, a significant decreasement of 71% is witnessed in T2 MRI signal. Those demonstrated that FOCMO NPs can achieve the purpose of positive/negative MRI simultaneously. Furthermore, obtained FOCMO NPs showed great hemocompatibility and negligible toxicity.

18.
J Cell Physiol ; 233(1): 226-237, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28218391

RESUMO

The majority of ovarian primordial follicles are preserved in a dormant state to maintain the female reproductive lifespan, and only a few primordial follicles are activated to enter the growing follicle pool in each wave. Recent studies have shown that primordial follicular activation depends on mammalian target of rapamycin complex 1 (mTORC1)-KIT ligand (KITL) signaling in pre-granulosa cells and its receptor (KIT)-phosphoinositol 3 kinase (PI3K) signaling in oocytes. However, the upstream regulator of mTORC1 signaling is unclear. The results of the present study showed that the phosphorylated mitogen-activated protein kinase3/1 (MAPK3/1) protein is expressed in some primordial follicles and all growing follicles. Culture of 3 days post-parturition (dpp) ovaries with the MAPK3/1 signaling inhibitor U0126 significantly reduced the number of activated follicles and was accompanied by dramatically reduced granulosa cell proliferation and increased oocyte apoptosis. Western blot and immunofluorescence analyses showed that U0126 significantly decreased the phosphorylation levels of Tsc2, S6K1, and rpS6 and the expression of KITL, indicating that U0126 inhibits mTORC1-KITL signaling. Furthermore, U0126 decreased the phosphorylation levels of Akt, resulting in a decreased number of oocytes with Foxo3 nuclear export. To further investigate MAPK3/1 signaling in primordial follicle activation, we used phosphatase and tensin homolog deleted on chromosome 10 (PTEN) inhibitor bpV(HOpic) to promote primordial follicle activation. In this model, U0126 also inhibited the activation of primordial follicles and mTORC1 signaling. Thus, these results suggest that MAPK3/1 participates in primordial follicle activation through mTORC1-KITL signaling.


Assuntos
Células da Granulosa/enzimologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Complexos Multiproteicos/metabolismo , Oócitos/enzimologia , Transdução de Sinais , Fator de Células-Tronco/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Proliferação de Células , Ativação Enzimática , Feminino , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos Endogâmicos ICR , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Oócitos/efeitos dos fármacos , Oócitos/patologia , PTEN Fosfo-Hidrolase/antagonistas & inibidores , PTEN Fosfo-Hidrolase/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Técnicas de Cultura de Tecidos
19.
Biol Open ; 7(1)2018 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-29242197

RESUMO

In female mammals, primordial follicles consist of two types of cells, namely, oocytes and pregranulosa cells that surround the oocytes. The size of the primordial follicle pool determines the reproductive ability of female mammals. However, the underlying mechanisms controlling primordial follicle assembly remain unclear. In this study, we show that oocyte-derived Janus kinase (JAK) signaling is vital for germline cyst breakdown and primordial follicle formation in vitro JAK2 and JAK3 activity is increased while germline cysts are breaking down. Inhibition of either JAK2 or JAK3 prevents germline cyst breakdown and primordial follicle formation. We further show that specific suppression of JAK2 delays germ cell loss through the downregulation of p53, but has no influence on pregranulosa cell proliferation. Alternatively, specific inhibition of JAK3 decreases pregranulosa cell proliferation by downregulating Notch2 signaling, implying that JAK3 acts on pregranulosa cells by controlling the extracellular secretion of oocyte-derived factors. In summary, our results indicate that JAK signaling contributes to germline cyst breakdown and primordial follicle formation by regulating oocyte loss and pregranulosa cell proliferation in the fetal mouse ovary. Our findings contribute to a better understanding of the molecular mechanism of mammalian folliculogenesis.

20.
J Clin Invest ; 128(1): 175-189, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29202468

RESUMO

Natural and synthetic progestogens have been commonly used to prevent recurrent pregnancy loss in women with inadequate progesterone secretion or reduced progesterone sensitivity. However, the clinical efficacy of progesterone and its analogs for maintaining pregnancy is variable. Additionally, the underlying cause of impaired endometrial progesterone responsiveness during early pregnancy remains unknown. Here, we demonstrated that uterine-selective depletion of BMI1, a key component of the polycomb repressive complex-1 (PRC1), hampers uterine progesterone responsiveness and derails normal uterine receptivity, resulting in implantation failure in mice. We further uncovered genetic and biochemical evidence that BMI1 interacts with the progesterone receptor (PR) and the E3 ligase E6AP in a polycomb complex-independent manner and regulates the PR ubiquitination that is essential for normal progesterone responsiveness. A close association of aberrantly low endometrial BMI1 expression with restrained PR responsiveness in women who had previously had a miscarriage indicated that the role of BMI1 in endometrial PR function is conserved in mice and in humans. In addition to uncovering a potential regulatory mechanism of BMI1 that ensures normal endometrial progesterone responsiveness during early pregnancy, our findings have the potential to help clarify the underlying causes of spontaneous pregnancy loss in women.


Assuntos
Implantação do Embrião , Endométrio/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Progesterona/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Progesterona/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Aborto Espontâneo/genética , Aborto Espontâneo/metabolismo , Aborto Espontâneo/patologia , Animais , Endométrio/patologia , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Complexo Repressor Polycomb 1/genética , Gravidez , Proteínas Proto-Oncogênicas/genética , Receptores de Progesterona/genética , Ubiquitina-Proteína Ligases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA