Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 278
Filtrar
1.
Biol Reprod ; 110(3): 509-520, 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38123510

RESUMO

Granulosa cell tumors are relatively rare, posing challenges for comprehension and therapeutic development due to limited cases and preclinical models. Metabolic reprogramming, a hallmark of cancer, manifests in granulosa cell tumors with notable lipid accumulation and increased expression of peroxisome proliferator-activated receptor gamma (PPARγ), a key lipid metabolism regulator. The roles of these features, however, remain unclear. In our previous work, we established a granulosa cell tumor model in mice by introducing a constitutively active Pik3ca mutant in oocytes, enabling the study of predictable tumor patterns from postnatal day 50. In this study, we characterized metabolic alterations during tumorigenesis (postnatal day 8 to day 50) and tumor growth (day 50 to day 65) in this model and explored the impact of PPARγ antagonism on human granulosa cell tumor proliferation. The tumor exhibited significant lipid accumulation, with PPARγ and the proliferation marker Ki67 co-localizing at postnatal day 65. Transcriptome analysis demonstrates that pathways for lipid metabolism and mitochondrial oxidation are promoted during tumorigenesis and tumor growth, respectively. Overlappingly upregulated genes during tumorigenesis and tumor growth are associated with lipid metabolism pathways. Correspondingly, mouse granulosa cell tumor shows overexpression of peroxisome proliferator-activated receptor gamma and DGAT2 proteins at postnatal day 65. Furthermore, GW9662 reduces the proliferation of KGN human granulosa cell tumor cells and decreases the phosphorylation of AKT and SMAD3. Our findings identify metabolic abnormalities in ooPIK3CA* granulosa cell tumor model and suggest peroxisome proliferator-activated receptor gamma as a potential driver for primary granulosa cell tumor growth.


Assuntos
Tumor de Células da Granulosa , Neoplasias Ovarianas , Feminino , Humanos , Animais , Camundongos , Tumor de Células da Granulosa/genética , Tumor de Células da Granulosa/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Carcinogênese , Lipídeos
2.
Cells ; 12(23)2023 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-38067144

RESUMO

The transforming growth factor ß (TGFß) superfamily, consisting of protein ligands, receptors, and intracellular SMAD transducers, regulates fundamental biological processes and cancer development. Our previous study has shown that sustained activation of TGFß receptor 1 (TGFBR1) driven by anti-Mullerian hormone receptor type 2 (Amhr2)-Cre in the mouse testis induces the formation of testicular granulosa cell tumors (TGCTs). As Amhr2-Cre is expressed in both Sertoli cells and Leydig cells, it remains unclear whether the activation of TGFBR1 in Sertoli cells alone is sufficient to induce TGCT formation. Therefore, the objective of this study was to determine whether Sertoli cell-activation of TGFBR1 drives oncogenesis in the testis. Our hypothesis was that overactivation of TGFBR1 in Sertoli cells would promote their transdifferentiation into granulosa-like cells and the formation of TGCTs. To test this hypothesis, we generated mice harboring constitutive activation of TGFBR1 in Sertoli cells using anti-Mullerian hormone (Amh)-Cre. Disorganized seminiferous tubules and tumor nodules were found in TGFBR1CA; Amh-Cre mice. A histological analysis showed that Sertoli cell-specific activation of TGFBR1 led to the development of neoplasms resembling granulosa cell tumors, which derailed spermatogenesis. Moreover, TGCTs expressed granulosa cell markers including FOXL2, FOXO1, and INHA. Using a dual fluorescence reporter line, the membrane-targeted tdTomato (mT)/membrane-targeted EGFP (mG) mouse, we provided evidence that Sertoli cells transdifferentiated toward a granulosa cell fate during tumorigenesis. Thus, our findings indicate that Sertoli cell-specific activation of TGFBR1 leads to the formation of TGCTs, supporting a key contribution of Sertoli cell reprogramming to the development of this testicular malignancy in our model.


Assuntos
Tumor de Células da Granulosa , Neoplasias Ovarianas , Neoplasias Testiculares , Masculino , Humanos , Feminino , Camundongos , Animais , Células de Sertoli/metabolismo , Tumor de Células da Granulosa/metabolismo , Tumor de Células da Granulosa/patologia , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Neoplasias Testiculares/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Hormônio Antimülleriano/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Neoplasias Ovarianas/patologia
3.
Cancer ; 129(14): 2224-2234, 2023 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-37011030

RESUMO

BACKGROUND: Engaging with online social media consumer groups for rare cancers may help to develop collaborations between consumers and researchers. This study, a collaboration with the Granulosa Cell Tumor-Survivor Sisters (GCT-SS) Facebook group, explores the results of their survey of member's treatment and follow-up experiences. METHODS: Members of the closed multinational GCT-SS Facebook group completed a 43-item survey covering symptoms, diagnosis, treatment, recurrence, follow-up, and possible risk factors for GCT. Group members could have adult (aGCT) or juvenile (jGCT) disease. Data was collected via an online survey between 2014 and 2019. RESULTS: A total of 743 members (average 4.4 years [SD = 5.9] post-diagnosis) participated including 52 with jGCT. A total of 67% had stage I disease and 8% had stage III-IV at diagnosis, although 30% of aGCT and 25% of jGCT reported recurrent disease at survey completion. A total of 48% of aGCT had laparoscopic surgery, tumor encapsulation was reported by 49%, and tumor bagging reported by 29% overall (37% laparoscopic; 8% open). Recurrence rates were higher when the tumor was cut or ruptured (ruptured: p < .001; cut: p = .01). A total of 19% of aGCT had chemotherapy with this most common for stage II-III disease. Bleomycin, etoposide, and cisplatin protocols became less common over time (diagnosed before 2015: 47% vs. diagnosed post-2015: 21%). CONCLUSIONS: This is one of the largest surveys of GCT treatment. Members of the GCT-SS group report treatment patterns generally in line with those found from clinical audits. Using naturally forming consumer groups may assist with developing the evidence base for care and supporting those living with GCT ovarian cancer. PLAIN LANGUAGE SUMMARY: This study is a collaboration between members of Granulosa Cell Tumor-Survivor Sisters (GCT-SS) Facebook group and researchers to assess members' experiences of treatment and follow-up. A total of 743 members (52 with juvenile GCT) completed an online survey. A total of 67% had stage I disease at diagnosis. Treatment patterns were generally in line with those found from clinical audits: 95% had surgery and 19% of those with adult GCT had chemotherapy. A total of 30% reported recurrent disease, with recurrence occurring within 5 years of diagnosis for 33%. Using naturally forming consumer groups may assist with developing the evidence base for care and supporting those living with GCT ovarian cancer.


Assuntos
Tumor de Células da Granulosa , Neoplasias Ovarianas , Mídias Sociais , Adulto , Feminino , Humanos , Tumor de Células da Granulosa/terapia , Tumor de Células da Granulosa/metabolismo , Tumor de Células da Granulosa/patologia , Neoplasias Ovarianas/patologia , Cisplatino , Etoposídeo
4.
Endocrinology ; 164(6)2023 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-37075218

RESUMO

Granulosa cell tumors (GCTs) are rare ovarian tumors comprising an adult and a juvenile subtype. They have a generally good prognosis, but the survival rate drastically declines in patients with late-stage or recurring tumors. Due to the rarity of GCTs, the tumor type is largely understudied and lacks a specific treatment strategy. Estrogen receptor beta (ERß/ESR2) has been found to be highly expressed in GCTs, which could be of therapeutic importance since it can be targeted with small molecules. However, its role in GCTs is not known. In this review, we summarize the current knowledge about the action of ERß in the ovary and discuss its prospective role in GCTs.


Assuntos
Tumor de Células da Granulosa , Neoplasias Ovarianas , Feminino , Humanos , Receptor beta de Estrogênio/genética , Tumor de Células da Granulosa/metabolismo , Recidiva Local de Neoplasia , Neoplasias Ovarianas/metabolismo
5.
Reproduction ; 165(5): 521-531, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36880885

RESUMO

In brief: The role of visfatin in ovarian granulosa cell tumor (GCT) invasion and glucose metabolism reprogramming is largely unexplored. These studies imply that visfatin or its inhibitor is involved in regulating ovarian granuloma invasion by reprogramming glucose metabolism and may be a potential candidate for the diagnosis and treatment of ovarian GCT. Abstract: Visfatin is an adipokine with nicotinamide phosphoribosyltransferase (NAMPT) activity, the concentration of which is higher in ascitic fluid than in serum, and is associated with ovarian cancer peritoneal dissemination. Potentially important effects of visfatin on glucose metabolism have been previously reported. However, the mechanism underlying the effects of visfatin on ovarian cancer cell invasion, and whether this involves altered glucose metabolism, has not been elucidated. Here, we tested the hypothesis that visfatin, which can reprogram cancer metabolism, promotes invasion by ovarian cancer spheroids. Visfatin increased glucose transporter (GLUT)1 expression and glucose uptake in adult granulosa cell tumor-derived spheroid cells (KGN) and also increased the activities of hexokinase 2 and lactate dehydrogenase. We showed a visfatin-induced increase in glycolysis in KGN cells. Moreover, visfatin increased the potential invasiveness of KGN spheroid cells by upregulating MMP2 (matrix metalloproteinase 2) and downregulating CLDN3 and CLDN4 (claudin 3 and 4) gene expression. Interestingly, an inhibitor of GLUT1 and lactate dehydrogenase (LDHA) abolished the stimulatory effect of visfatin on the potential invasiveness of KGN cells. More importantly, silencing expression of the NAMPT gene in KGN cells demonstrated its important effect on glycolysis and invasiveness in adult granulosa cell tumor cells (AGCTs). In summary, visfatin appears to increase AGCT invasiveness through effects on glucose metabolism and to be an important regulator of glucose metabolism in these cells.


Assuntos
Tumor de Células da Granulosa , Neoplasias Ovarianas , Feminino , Adulto , Humanos , Tumor de Células da Granulosa/metabolismo , Nicotinamida Fosforribosiltransferase/genética , Nicotinamida Fosforribosiltransferase/metabolismo , Nicotinamida Fosforribosiltransferase/farmacologia , Metaloproteinase 2 da Matriz , Neoplasias Ovarianas/patologia , Glucose/farmacologia , Lactato Desidrogenases
6.
Cancer Res ; 83(2): 239-250, 2023 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-36409821

RESUMO

Adult-type granulosa cell tumors (AGCT) are the most common type of malignant ovarian sex cord-stromal tumors. Most AGCTs carry the somatic variant c.402C>G (p.C134W) affecting the transcription factor FOXL2. Germline dominant variants in FOXL2 are responsible for blepharophimosis syndrome, which is characterized by underdevelopment of the eyelid. In this work, we generated a mouse model harboring the C134W variant of FOXL2 to evaluate in vivo the poorly understood oncogenic role of FOXL2. The mutation was dominant regarding eyelid hypoplasia, reminiscent of blepharophimosis syndrome. Interestingly, Foxl2+/C134W female mice had reduced fertility and developed AGCTs through a progression from abnormal ovaries with aberrant granulosa cells to ovaries with stromal hyperplasia and atypia and on to tumors in adut mice. The genes dysregulated in mouse AGCTs exhibited the hallmarks of cancer and were consistent with a gain-of-function of the mutated allele affecting TGFß signaling. A comparison of these data with previous results on human AGCTs indicated similar deregulated pathways. Finally, a mutational analysis of mouse AGCT transcriptomic data suggested the absence of additional driver mutations apart from FOXL2-C134W. These results provide a clear in vivo example in which a single mutational hit triggers tumor development associated with profound transcriptomic alterations. SIGNIFICANCE: A newly generated mouse model carrying a FOXL2 mutation characteristic of adult-type granulosa cell tumors shows that FOXL2 C134W shifts the transcriptome towards a signature of granulosa cell cancer and drives tumorigenesis.


Assuntos
Tumor de Células da Granulosa , Neoplasias Ovarianas , Anormalidades da Pele , Adulto , Feminino , Humanos , Animais , Camundongos , Tumor de Células da Granulosa/genética , Tumor de Células da Granulosa/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Neoplasias Ovarianas/genética , Mutação , Proteína Forkhead Box L2/genética
7.
Int J Mol Sci ; 23(22)2022 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-36430923

RESUMO

Genetic alterations of the RUNX1 gene are associated with a variety of malignancies, including female-related cancers. The role of RUNX1 as either a tumor suppressor gene or an oncogene is tissue-dependent and varies based on the cancer type. Both the amplification and deletion of the RUNX1 gene have been associated with ovarian cancer in humans. In this study, we investigated the effects of Runx1 loss on ovarian pathogenesis in mice. A conditional loss of Runx1 in the somatic cells of the ovary led to an increased prevalence of ovarian tumors in aged mice. By the age of 15 months, 27% of Runx1 knockout (KO) females developed ovarian tumors that presented characteristics of granulosa cell tumors. While ovaries from young adult mice did not display tumors, they all contained abnormal follicle-like lesions. The granulosa cells composing these follicle-like lesions were quiescent, displayed defects in differentiation and were organized in a rosette-like pattern. The RNA-sequencing analysis further revealed differentially expressed genes in Runx1 KO ovaries, including genes involved in metaplasia, ovarian cancer, epithelial cell development, tight junctions, cell-cell adhesion, and the Wnt/beta-catenin pathway. Together, this study showed that Runx1 is required for normal granulosa cell differentiation and prevention of ovarian tumor development in mice.


Assuntos
Tumor de Células da Granulosa , Neoplasias Ovarianas , Humanos , Camundongos , Feminino , Animais , Lactente , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Células da Granulosa/metabolismo , Neoplasias Ovarianas/patologia , Tumor de Células da Granulosa/metabolismo , Carcinoma Epitelial do Ovário/patologia
8.
J Pathol ; 256(3): 335-348, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34860414

RESUMO

Granulosa cell tumor (GCT) is a form of ovarian tumor characterized by its tendency to recur years after surgical ablation. Little is known about the mechanisms involved in GCT development and progression. GCTs can produce estradiol (E2), but whether this hormone could play a role in this cancer through its nuclear receptors, i.e. ERα and ERß, remains unknown. Here, we addressed this issue by cell-based and molecular studies on human GCTs and GCT cell lines. Importantly, we observed that E2 significantly increased the growth of GCT cells by promoting cell survival. The use of selective agonists of each type of receptor, together with Esr1 (ERα) or Esr2 (ERß)-deleted GCT cells, revealed that E2 mediated its effects through ERα-dependent genomic mechanisms and ERß/ERα-dependent extra-nuclear mechanisms. Notably, the expression of Greb1, a prototypical ER target gene, was dose-dependently upregulated by E2 specifically through ERα in GCT cells. Accordingly, using GCTs from patients, we found that GREB1 mRNA abundance was positively correlated to intra-tumoral E2 concentrations. Tissue microarray analyses showed that there were various combinations of ER expression in primary and recurrent GCTs, and that ERα expression persisted only in combination with ERß in ~40% of recurrent tumors. Altogether, this study demonstrates that E2 can promote the progression of GCTs, with a clear dependence on ERα. In addition to demonstrating that GCTs can be classified as a hormone-related cancer, our results also highlight that the nature of ER forms present in recurrent GCTs could underlie the variable efficiency of endocrine therapies. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Estradiol/farmacologia , Receptor alfa de Estrogênio/agonistas , Tumor de Células da Granulosa/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Ovarianas/metabolismo , Idoso , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Tumor de Células da Granulosa/genética , Tumor de Células da Granulosa/patologia , Humanos , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Regulação para Cima
9.
Reprod Biol Endocrinol ; 19(1): 170, 2021 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-34814928

RESUMO

BACKGROUND: Polycystic ovary syndrome (PCOS) is an endocrine-related follicular developmental disorder that affects 50 %-70 % of reproductive-aged women diagnosed with ovulation-related infertility. Abnormal proliferation and apoptosis of granulosa cells (GCs) are thought to be the critical factors leading to abnormal maturation of follicles. It has been shown that microRNAs (miRNAs) exert a significant influence in the pathogenesis of PCOS; however, the relationship between miRNA, PCOS, and GC apoptosis is not entirely understood. METHODS: To clarify the effect of miR-194 in PCOS, CCK-8, Ki67 staining, AO/EB, and flow cytometry assays were used to assess cell growth, proliferation, and apoptosis in KGN cells, which were artificially stimulated to overexpress miR-194. Luciferase reporter assays and rescue experiments were used to elucidate the mechanism underlying miR-194 in PCOS. RESULTS: miR-194 expression was significantly up-regulated in rat models of PCOS and the ovarian GCs of PCOS patients. miR-194 suppression promoted KGN cell growth and proliferation. miR-194 overexpression also induced cell apoptosis, while miR-194 downregulation had an opposite effect. Furthermore, up-regulating heparin-binding EGF-like growth factor (HB-EGF) expression rescued the pro-apoptotic effects of miR-194 upregulation on KGN cells. CONCLUSIONS: miR-194 is increased in PCOS granulosa cell and may function as a novel biomarker and therapeutic target for KGN cells via HB-EGF regulation.


Assuntos
Apoptose/fisiologia , Tumor de Células da Granulosa/metabolismo , Células da Granulosa/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/biossíntese , MicroRNAs/biossíntese , Síndrome do Ovário Policístico/metabolismo , Adulto , Animais , Linhagem Celular , Proliferação de Células/fisiologia , Feminino , Tumor de Células da Granulosa/patologia , Células da Granulosa/patologia , Humanos , Síndrome do Ovário Policístico/patologia , Ratos , Ratos Sprague-Dawley , Adulto Jovem
10.
Reproduction ; 161(5): 581-591, 2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33764899

RESUMO

Humanin (HN) is a short peptide involved in many biological processes such as apoptosis, cell survival, inflammatory response, and reaction to stressors like oxidative stress, between others. In the ovary, a correct balance between pro- and anti-apoptotic factors is crucial for folliculogenesis. In the follicular atresia, survival or death of granulosa cells is a critical process. The goal of this study was to evaluate the action of HN on granulosa cell fate. To explore endogenous HN function in the ovary, we used a recombinant baculovirus (BV) encoding a short-hairpin RNA targeted to silence HN (shHN). HN downregulation modified ovarian histoarchitecture and increased apoptosis of granulosa cells. HN was also detected in a granulosa tumor cell line (KGN). Transduction of KGN cells with BV-shHN resulted in HN downregulation and increased apoptosis. On the other hand, treatment of KGN cells with exogenous HN increased cell viability and decreased apoptosis. In summary, these findings indicate that HN is a cytoprotective factor in granulosa cells of antral follicles, suggesting that this peptide would be involved in the regulation of folliculogenesis. Also, this peptide is a cytoprotective factor in KGN cells, and therefore, it could be involved in granulosa tumor cell behavior.


Assuntos
Citoproteção , Tumor de Células da Granulosa/patologia , Células da Granulosa/citologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Folículo Ovariano/citologia , Ovário/citologia , Fragmentos de Peptídeos/metabolismo , Animais , Feminino , Tumor de Células da Granulosa/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Folículo Ovariano/metabolismo , Ovário/metabolismo , Estresse Oxidativo , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/genética , RNA Interferente Pequeno/genética , Ratos , Ratos Wistar
11.
Toxicology ; 452: 152715, 2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33571556

RESUMO

Granulosa cell tumors (GCT) of the ovary have a good prognosis. Recurrence tends to be late; however, > 66 % of patients with recurrent GCT die from the disease. Most recurrences are abdominopelvic, although distant metastases have been documented. Here, we tested the hypothesis that a mixture of persistent endocrine-disrupting chemicals (EDCs) stimulates the invasion of GCT cells. We selected perfluorooctanoate (PFOA, 2 ng/mL), perfluorooctanesulfonate (PFOS, 8 ng/mL), 2,2-dichlorodiphenyldichloroethylene (p,p'-DDE, 1 ng/mL), polychlorinated biphenyl 153 (PCB153, 100 pg/mL), and hexachlorobenzene (HCB, 50 pg/mL), which have the highest measured concentrations in follicular fluid of women undergoing treatment with assisted reproductive technology. The human GCT cell lines COV434 and KGN have been used as in vitro models of juvenile (JGCT) and adult (AGCT) GCT subtypes, respectively. Cells were treated with a mixture of the test compounds for 15 min prior to analysis of protein phosphorylation; for 4 h prior to analysis in a circular chemorepellent-induced defect assay; for 6 h prior to analysis of matrix metalloproteinase 2 (MMP2) activity; for 24 h prior to analysis of migration, invasion, and gene expression; and for 48 h prior to analysis of protein expression. First, we showed that KGN cells migrated and exhibited invasive behavior. By contrast, COV434 cells lacked migration and invasion potential. Moreover, expression of mesenchymal genes and the gene encoding MMP2 was higher in KGN cells, and that of epithelial genes lower, than that in COV434 cells. Treatment of KGN cells with the EDC mixture stimulated cell migration, invasion, and lymphatic dissemination. The results suggest that the role of the EDC mixture in AGCT invasion is not related to changes in expression of epithelial and mesenchymal genes; rather, it is related to increased expression and activity of MMP2. Additionally, silencing insulin-like growth factor 1 (IGF1R) in AGCT abolished the stimulatory effect of the EDC mixture on KGN spheroid invasion. These results demonstrate that the EDC mixture increased KGN spheroid invasion by stimulating expression and activity of MMP2 via IGF1R.


Assuntos
Regulação Neoplásica da Expressão Gênica , Tumor de Células da Granulosa/metabolismo , Metaloproteinase 2 da Matriz/biossíntese , Poluentes Orgânicos Persistentes/toxicidade , Receptor IGF Tipo 1/biossíntese , Esferoides Celulares/metabolismo , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Relação Dose-Resposta a Droga , Feminino , Tumor de Células da Granulosa/genética , Tumor de Células da Granulosa/patologia , Humanos , Metaloproteinase 2 da Matriz/genética , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Receptor IGF Tipo 1/genética , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/patologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
12.
Cancer Res ; 80(17): 3466-3479, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32641411

RESUMO

The mutant protein FOXL2C134W is expressed in at least 95% of adult-type ovarian granulosa cell tumors (AGCT) and is considered to be a driver of oncogenesis in this disease. However, the molecular mechanism by which FOXL2C134W contributes to tumorigenesis is not known. Here, we show that mutant FOXL2C134W acquires the ability to bind SMAD4, forming a FOXL2C134W/SMAD4/SMAD2/3 complex that binds a novel hybrid DNA motif AGHCAHAA, unique to the FOXL2C134W mutant. This binding induced an enhancer-like chromatin state, leading to transcription of nearby genes, many of which are characteristic of epithelial-to-mesenchymal transition. FOXL2C134W also bound hybrid loci in primary AGCT. Ablation of SMAD4 or SMAD2/3 resulted in strong reduction of FOXL2C134W binding at hybrid sites and decreased expression of associated genes. Accordingly, inhibition of TGFß mitigated the transcriptional effect of FOXL2C134W. Our results provide mechanistic insight into AGCT pathogenesis, identifying FOXL2C134W and its interaction with SMAD4 as potential therapeutic targets to this condition. SIGNIFICANCE: FOXL2C134W hijacks SMAD4 and leads to the expression of genes involved in EMT, stemness, and oncogenesis in AGCT, making FOXL2C134W and the TGFß pathway therapeutic targets in this condition. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/17/3466/F1.large.jpg.


Assuntos
Transição Epitelial-Mesenquimal/genética , Proteína Forkhead Box L2/genética , Regulação Neoplásica da Expressão Gênica/genética , Tumor de Células da Granulosa/patologia , Proteínas Smad/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Proteína Forkhead Box L2/metabolismo , Tumor de Células da Granulosa/genética , Tumor de Células da Granulosa/metabolismo , Humanos , Mutação , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Proteína Smad4/metabolismo
13.
Cancer Res ; 80(17): 3480-3491, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32641414

RESUMO

The somatic missense point mutation c.402C>G (p.C134W) in the FOXL2 transcription factor is pathognomonic for adult-type granulosa cell tumors (AGCT) and a diagnostic marker for this tumor type. However, the molecular consequences of this mutation and its contribution to the mechanisms of AGCT pathogenesis remain unclear. To explore these mechanisms, we engineered V5-FOXL2WT- and V5-FOXL2C134W-inducible isogenic cell lines and performed chromatin immunoprecipitation sequencing and transcriptome profiling. FOXL2C134W associated with the majority of the FOXL2 wild-type DNA elements as well as a large collection of unique elements genome wide. This model enabled confirmation of altered DNA-binding specificity for FOXL2C134W and identification of unique targets of FOXL2C134W including SLC35F2, whose expression increased sensitivity to YM155. Our results suggest FOXL2C134W drives AGCT by altering the binding affinity of FOXL2-containing complexes to engage an oncogenic transcriptional program. SIGNIFICANCE: A mechanistic understanding of FOXL2C134W-induced regulatory state alterations drives discovery of a rationally designed therapeutic strategy.


Assuntos
DNA/metabolismo , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Tumor de Células da Granulosa/genética , Linhagem Celular Tumoral , Feminino , Tumor de Células da Granulosa/metabolismo , Humanos , Mutação de Sentido Incorreto , Mutação Puntual , Ligação Proteica
14.
Mol Biol Rep ; 47(5): 3789-3796, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32277442

RESUMO

We aimed to provide a comparative characterization of DNA damage response elements, survival/apoptosis and cell cycle progression of the malignant granulosa cells exposed to gemcitabine and cisplatin. Malignant granulosa tumor cell lines COV434 and KGN were used for the experiments. Cell viability, proliferation, DNA damage response and apoptosis were investigated. Cell cycle progression was assessed. In vitro estradiol (E2) and AMH productions of the cells were measured. Exposure of asynchronous malignant granulosa cells to gemcitabine caused growth arrest, induced DNA damage and activated cellular stress pathways, cell cycle checkpoint sensors and triggered apoptosis as evidenced by increased expression of phospho-p38, γ-histone H2AX, phospho-Chk-1/phospho-Chk-2, and cleaved forms of PARP and caspase-3 in a dose dependent manner. In vitro E2 and AMH productions of the cells were decreased along with reduction in viable cell mass. Cisplatin treatment produced a similar response but it was associated with JNK activation rather than p38. When the cells were synchronized and treated with gemcitabine at G2/M transition, the degradation of cyclin B1 and dephosphorylation of cdc-2 at Tyr 15 residue did not occur, resulting in cycle arrest. Similar effects on cell cycle progression was also observed in cisplatin. However, it was associated with JNK activation and higher expression of γ-histone H2AX and cleaved forms of caspase-3 and PARP, indicative of more extensive DNA damage and apoptosis in the cells. This descriptive study provides evidence that gemcitabine exerts cytotoxic effects and causes perturbations in cell cycle progression of malignant granulosa cells.


Assuntos
Desoxicitidina/análogos & derivados , Tumor de Células da Granulosa/metabolismo , Células da Granulosa/efeitos dos fármacos , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/metabolismo , Cisplatino/farmacologia , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/fisiologia , Reparo do DNA/fisiologia , Desoxicitidina/metabolismo , Desoxicitidina/farmacologia , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Tumor de Células da Granulosa/tratamento farmacológico , Células da Granulosa/metabolismo , Humanos , Gencitabina
15.
Mol Cell Endocrinol ; 503: 110698, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31891770

RESUMO

Endocrine-disrupting chemicals (EDCs), such as perfluorooctanoate, perfluorooctane sulfonate, 2,2-dichlorodiphenyldichloroethylene, hexachlorobenzene, and polychlorinated biphenyl 153 are persistent pollutants that are found in human follicular fluid (FF). These compounds may affect endocrine function, disrupt steroid secretion by granulosa cells, and play a role in granulosa cell tumor (GCT) development. GCTs demonstrate endocrine activity, expressing aromatase and secreting 17ß-estradiol (E2). We aimed to determine the effects of a mixture of EDCs, similar to that found in human FF, on human granulosa tumor cell lines representing the juvenile (JGCT) and adult (AGCT) forms (COV434 and KGN cells, respectively). We found that all the individual compounds and mixtures tested altered granulosa tumor cell function by disrupting E2 secretion. In KGN cells, which possess significantly higher basal aromatase gene expression, and therefore secrete more E2 than JGCT cells, EDC mixtures activated estrogen receptors (ERs) and G protein-coupled receptor-30 signaling, thereby stimulating E2 secretion, without affecting aromatase expression. By contrast, in COV434 cells, which demonstrate higher CYP1A1 expression, a key mediator of estrogen metabolism, than KGN cells, EDC mixtures reduced E2 secretion in parallel with increases in the 2-hydroxyestrogen 1/E2 ratio and CYP1A1 expression, implying an upregulation of E2 metabolism. These results indicate that the EDC mixture present in FF disrupts E2 secretion in JGCT and AGCT cells according to the estrogen metabolic potential of the cell type, involving both classical and non-classical ER pathways.


Assuntos
Disruptores Endócrinos/farmacologia , Estradiol/metabolismo , Estrogênios/metabolismo , Tumor de Células da Granulosa/metabolismo , Poluentes Orgânicos Persistentes/farmacologia , Linhagem Celular Tumoral , Disruptores Endócrinos/isolamento & purificação , Feminino , Líquido Folicular/química , Tumor de Células da Granulosa/patologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Células da Granulosa/patologia , Humanos , Redes e Vias Metabólicas/efeitos dos fármacos , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Via Secretória/efeitos dos fármacos
16.
Toxicol In Vitro ; 65: 104769, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31923581

RESUMO

Insulin-like growth factor-1 (IGF1) is a hormone involved in cell proliferation. We previously showed that IGF1 directly stimulates cell proliferation in granulosa cell tumors (GCTs). Estrogen regulates IGF1 expression in several reproductive organs including the uterus and ovaries. This study aimed to investigate the effects of a mixture of endocrine-disrupting chemicals (EDCs) on secretion of IGF1 by COV434 and KGN cells, which have been used as in vitro models of juvenile and adult GCTs, respectively. The EDC mixture contained perfluorooctanoate, perfluorooctane sulfonate, 2,2-dichlorodiphenyldichloroethylene, hexachlorobenzene, and polychlorinated biphenyl 153, which are persistent hormonally active environmental toxicants present in ovarian follicular fluid (FF). Expression and secretion of IGF1 were significantly higher in GTCs than in HGrC1 human non-cancer granulosa cells (with the profile HGrC1 < COV434 < KGN). Treatment with the EDC mixture as well as individual test compounds significantly increased IGF1 secretion in KGN cells. Moreover, IGFBP3 gene expression in KGN cells was downregulated after treatment with the EDC mixtures. The estrogen receptor alpha pathway was involved in this effect. Conditioned medium of KGN cells treated with the EDC mixture increased proliferation of HGrC1 human non-cancer granulosa cells. These results indicate that the mixture of EDCs found in FF increases secretion of IGF1 by KGN cells and thus indirectly contributes to progression of adult GCTs, and increases proliferation of non-cancer granulosa cells.


Assuntos
Disruptores Endócrinos/farmacologia , Líquido Folicular/química , Tumor de Células da Granulosa/metabolismo , Células da Granulosa/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/metabolismo , Neoplasias Ovarianas/metabolismo , Esferoides Celulares/efeitos dos fármacos , Adulto , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Feminino , Células da Granulosa/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/genética , PPAR gama/genética , Transdução de Sinais , Esferoides Celulares/metabolismo
17.
Oncogene ; 39(9): 1875-1890, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31745296

RESUMO

Ovarian granulosa cell tumors (GCTs) are indolent tumors of the ovary affecting women at all ages and potentially displaying late recurrence. Even if there is still little information regarding the mechanisms involved in GCT development and progression, FOXL2 would be a major tumor suppressor gene in granulosa cells. We analyzed the mechanisms underlying GCT initiation and progression by using mice with targeted expression of SV40 large T-antigen in granulosa cells (AT mouse), which develop GCTs. Consistent with patients, AT mice with developing GCTs displayed increased levels in circulating anti-Müllerian hormone (AMH), estradiol and androgens, as well as decreased FOXL2 protein abundance. Very few mice developed metastases (1 out of 30). In situ analyses revealed that GCT initiation resulted from both increased granulosa cell survival and proliferation in large antral follicles. Tumorigenesis was associated with the combined inactivation of p53 and Rb pathways, as shown by the impaired expression of respective downstream targets regulating cell apoptosis and proliferation, i.e., Bax, Bak, Gadd45a, Ccna2, Ccne1, E2f1, and Orc1. Importantly, the expression of FOXL2 was still present in newly developed GCTs and its downregulation only started during GCT growth. Collectively, our experiments provide evidence that disrupted p53/Rb signaling can drive tumor initiation and growth. This model challenges the current paradigm that impaired FOXL2 signaling is a major switch of granulosa cell tumorigenesis, albeit possibly contributing to tumor growth.


Assuntos
Carcinogênese/patologia , Proteína Forkhead Box L2/metabolismo , Tumor de Células da Granulosa/patologia , Células da Granulosa/patologia , Proteína do Retinoblastoma/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Carcinogênese/genética , Carcinogênese/metabolismo , Células Cultivadas , Regulação para Baixo , Feminino , Proteína Forkhead Box L2/genética , Tumor de Células da Granulosa/genética , Tumor de Células da Granulosa/metabolismo , Células da Granulosa/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Proteína do Retinoblastoma/genética , Proteína Supressora de Tumor p53/genética
18.
Mol Biol Rep ; 46(6): 6325-6338, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31538301

RESUMO

Apelin and chemerin are adipocytokines that play important roles in many physiological and pathological processes throughout the body. Our previous study demonstrated that these two adipokines are expressed and secreted by epithelial and granulosa cancer cell lines. 17ß-estradiol (E2) and insulin-like growth factor-1 (IGF-1) are important regulators of ovarian functions, and their roles are well known. This study investigated whether apelin and chemerin regulate proliferation and apoptosis of epithelial (OVCAR-3) and granulosa (COV434) ovarian cancer cell lines by interacting with E2 and IGF-1. Apelin and chemerin did not affect caspase-3 activation in either cell line. However, apelin abrogated the stimulatory effects of E2 on proliferation of OVCAR-3 cells and of IGF-1 on proliferation of COV434 cells independently of ERK1/2 and PI3K via crosstalk of apelin receptor with estrogen receptor alpha and IGF-1 receptor, respectively.


Assuntos
Apelina/metabolismo , Carcinoma Epitelial do Ovário/metabolismo , Estradiol/farmacologia , Tumor de Células da Granulosa/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Neoplasias Ovarianas/metabolismo , Apelina/genética , Receptores de Apelina/metabolismo , Carcinoma Epitelial do Ovário/genética , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quimiocinas/genética , Quimiocinas/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Tumor de Células da Granulosa/genética , Humanos , Neoplasias Ovarianas/genética , Receptor IGF Tipo 1/metabolismo
19.
J Steroid Biochem Mol Biol ; 194: 105418, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31376461

RESUMO

Granulosa cell tumors of the ovary (GCTs) are the predominant form of ovarian stromal tumors and can lead to abnormally secreted estrogen hormones. Studies have reported that forkhead transcription factor 2 (FOXL2) inhibits estrogen synthesis and its gene mutation can lead to GCTs. We unexpected found that estrogen also regulates the expression level of FOXL2. High-dose estrogen increased the expression of FOXL2 in ovarian-like granulosa (KGN) cells at both the mRNA and protein levels. However, no research has reported on the molecular regulatory mechanism and function between estrogen and FOXL2 in the development of GCTs. In this research, FOXL2 was highly expressed in KGN cells and ovarian stromal tumor tissues. Deletion of FOXL2 increased the estrogen secretion in KGN cells. In turn, high-dose estrogen increased the FOXL2 expression levels. FOXL2 was phosphorylated by GPR30 (G protein coupled receptor)-Protein kinase C (PKC) signaling pathway upon estrogen stimulation. Estrogen inhibited cell migration and proliferation, while promoting cell apoptosis. Deletion of FOXL2 inhibited the influence of estrogen on cell proliferation, migration, and apoptosis. Results suggest that estrogen via regulating FOXL2 suppresses cell proliferation and induces cell apoptosis.


Assuntos
Estradiol/farmacologia , Estrogênios/farmacologia , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Tumor de Células da Granulosa/metabolismo , Neoplasias Ovarianas/metabolismo , Adulto , Idoso , Apoptose/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , Adulto Jovem
20.
J Ovarian Res ; 12(1): 76, 2019 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-31412918

RESUMO

BACKGROUND: Granulosa cell tumors (GCTs) are derived from proliferating granulosa cells of the ovarian follicle. They are known for their late recurrence and most patients with an aggressive form die from their disease. There are no treatment options for this slowly proliferating tumor besides surgery and chemotherapy. In a number of tumors, analogs of the second mitochondria-derived activator of caspases (SMAC), alone or in combination with other molecules, such as TNFα, are evolving as new treatment options. SMAC mimetics block inhibitor of apoptosis proteins (IAPs), which bind caspases (e.g. XIAP), or activate the pro-survival NF-κB pathway (e.g. cIAP1/2). Expression of IAPs by GCTs is yet not fully elucidated but recently XIAP and its inhibition by SMAC mimetics in a combination therapy was described to induce apoptosis in a GCT cell line, KGN. We evaluated the expression of cIAP1 in GCTs and elucidated the effects of the SMAC mimetic BV-6 using KGN as a model. RESULTS: Employing immunohistochemistry, we observed cIAP1 expression in a tissue microarray (TMA) of 42 GCT samples. RT-PCR confirmed expression of cIAP1/2, as well as XIAP, in primary, patient-derived GCTs and in KGN. We therefore tested the ability of the bivalent SMAC mimetic BV-6, which is known to inhibit cIAP1/2 and XIAP, to induce cell death in KGN. A dose response study indicated an EC50 ≈ 8 µM for both, early (< 8) and advanced (> 80) passages, which differ in growth rate and presumably aggressiveness. Quantitative RT-PCR showed upregulation of NF-κB regulated genes in BV-6 stimulated cells. Blocking experiments with the pan-caspase inhibitor Z-VAD-FMK indicated caspase-dependence. A concentration of 20 µM Z-VAD-FMK was sufficient to significantly reduce apoptosis. This cell death was further substantiated by results of Western Blot studies. Cleaved caspase 3 and cleaved PARP became evident in the BV-6 treated group. CONCLUSIONS: Taken together, the results show that BV-6 is able to induce apoptosis in KGN cells. This approach may therefore offer a promising therapeutic avenue to treat GCTs.


Assuntos
Tumor de Células da Granulosa/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Neoplasias Ovarianas/metabolismo , Adulto , Idoso , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Tumor de Células da Granulosa/genética , Humanos , Proteínas Inibidoras de Apoptose/genética , Pessoa de Meia-Idade , Oligopeptídeos/farmacologia , Neoplasias Ovarianas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...