Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Gynecol Oncol ; 150(3): 426-431, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30126589

RESUMO

OBJECTIVES: Inactivating somatic mutations of ARID1A, a chromatin remodeling gene, are common in endometrioid endometrial carcinoma (EEC) but rare in complex atypical hyperplasia (CAH). Our objectives were to determine the clinical significance of ARID1A loss during tumor progression from CAH to EEC and to assess its role as a predictive cancer biomarker. METHODS: In cohort A, ARID1A immunoreactivity was evaluated in endometrial sampling (biopsy/curettage) specimens showing CAH to determine whether ARID1A expression correlates with the presence of EEC at subsequent hysterectomy. In cohort B, ARID1A immunoreactivity was evaluated in the hysterectomy specimens with concurrent CAH and EEC to assess for the concordance of ARID1A expression in both components. RESULTS: In cohort A, loss of ARID1A immunoreactivity was identified in the endometrial sampling specimen of 31% of patients undergoing hysterectomy for a preoperative diagnosis of CAH. EEC was identified in the hysterectomy specimen of 94% of patients with loss of ARID1A in the endometrial sampling specimen while only 15% of patients with retained ARID1A expression (P < 0.0001). No association was observed between ARID1A expression and demographic characteristics. In cohort B, 14 (31%) of 45 patients with concurrent CAH/EEC in their hysterectomy specimens had complete loss of ARID1A expression in the EEC components. Among these 14 patients, 50% also had loss of ARID1A immunoreactivity in the CAH component. CONCLUSIONS: ARID1A immunostaining may correlate with malignant transformation and the presence of concurrent EEC in patients with CAH identified at pre-hysterectomy endometrial sampling. Further investigation to determine the potential utility of ARID1A expression as a tissue biomarker is warranted.


Assuntos
Carcinoma/metabolismo , Hiperplasia Endometrial/metabolismo , Neoplasias do Endométrio/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Idoso , Biomarcadores Tumorais/metabolismo , Carcinoma/genética , Carcinoma/patologia , Carcinoma/cirurgia , Proteínas de Ligação a DNA , Progressão da Doença , Hiperplasia Endometrial/genética , Hiperplasia Endometrial/patologia , Hiperplasia Endometrial/cirurgia , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Neoplasias do Endométrio/cirurgia , Feminino , Humanos , Histerectomia , Pessoa de Meia-Idade , Mutação , Gradação de Tumores , Estadiamento de Neoplasias , Proteínas Nucleares/genética , Valor Preditivo dos Testes , Estudos Retrospectivos , Fatores de Risco , Fatores de Transcrição/genética
2.
J Obstet Gynaecol Res ; 42(11): 1588-1598, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27718288

RESUMO

AIM: The proper preoperative diagnosis and management of cervical proliferative disorders presenting with multiple cysts, including minimal deviation adenocarcinoma (MDA), lobular endocervical glandular hyperplasia (LEGH), and nabothian cyst (NC), have not been fully established. We previously proposed a management protocol comprising a diagnostic approach using cytology, magnetic resonance imaging, and gastric-type mucin and subsequent treatment. We herein evaluate the usefulness of this protocol and implications of GNAS mutations in LEGH. METHODS: The clinical courses of 94 patients with cervical multicystic lesions who visited our hospital between June 1995 and September 2014 were retrospectively analyzed. GNAS mutations were investigated in 10 LEGH, five LEGH with atypia, and two MDA cases. RESULTS: Of the 94 patients, the conditions of 10, 59, and 25 were clinically diagnosed as suspicious of MDA or carcinoma (S/O MDA-Ca), suspicious of LEGH (S/O LEGH), and NC, respectively. Ten patients each with S/O MDA-Ca and S/O LEGH underwent hysterectomy, and the correct ratio for diagnosis was 90% (18/20). Of the 42 S/O LEGH cases followed-up for more than 12 months, three showed an increase in tumor size. After hysterectomy, two were LEGH with atypia while one was NC. The GNAS mutation was detected in two cases of LEGH with atypia, one of which showed an increase in tumor size during follow-up. CONCLUSION: The management protocol we propose herein will be useful. An increase in tumor size is important to detect potentially malignant LEGH. GNAS mutations may be involved in the tumorigenesis of potentially malignant LEGH.


Assuntos
Adenocarcinoma/diagnóstico por imagem , Adenocarcinoma/genética , Adenocarcinoma/cirurgia , Colo do Útero/patologia , Cromograninas/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Neoplasias do Colo do Útero/diagnóstico por imagem , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/cirurgia , Adulto , Idoso , Colo do Útero/diagnóstico por imagem , Colo do Útero/cirurgia , Conização , Feminino , Humanos , Hiperplasia , Histerectomia , Pessoa de Meia-Idade , Mutação , Estudos Retrospectivos , Resultado do Tratamento
3.
Gynecol Oncol ; 139(2): 338-44, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26343160

RESUMO

OBJECTIVES: Molecular markers associated with tumor progression in uterine carcinoma are poorly defined. In this study, we determine whether upregulation of LAMC1, a gene encoding extracellular matrix protein, laminin γ1, is associated with various uterine carcinoma subtypes and stages of tumor progression. METHODS: An analysis of the immunostaining patterns of laminin γ1 in normal endometrium, atypical hyperplasia, and a total of 150 uterine carcinomas, including low-grade and high-grade endometrioid carcinomas, uterine serous and clear cell carcinoma, was performed. Clinicopathological correlation was performed to determine biological significance. The Cancer Genome Atlas (TCGA) data set was used to validate our results. RESULTS: As compared to normal proliferative and secretory endometrium, for which laminin γ1 immunoreactivity was almost undetectable, increasing laminin C1 staining intensity was observed in epithelial cells from atypical hyperplasia to low-grade endometrioid to high-grade endometrioid carcinoma, respectively. Laminin γ1 expression was significantly associated with FIGO stage, myometrial invasion, cervical/adnexal involvement, angiolymphatic invasion and lymph node metastasis. Similarly, analysis of the endometrial carcinoma data set from TCGA revealed that LAMC1 transcript levels were higher in high-grade than those in low-grade endometrioid carcinoma. Silencing LAMC1 expression by siRNAs in a high-grade endometrioid carcinoma cell line did not affect its proliferative activity but significantly suppressed cell motility and invasion in vitro. CONCLUSIONS: These data suggest that laminin γ1 may contribute to the development and progression of uterine carcinoma, likely through enhancing tumor cell motility and invasion. Laminin γ1 warrants further investigation regarding its role as a biomarker and therapeutic target in uterine carcinoma.


Assuntos
Adenocarcinoma de Células Claras/genética , Carcinoma Endometrioide/genética , Hiperplasia Endometrial/genética , Neoplasias do Endométrio/genética , Endométrio/metabolismo , Regulação Neoplásica da Expressão Gênica , Laminina/genética , Neoplasias Císticas, Mucinosas e Serosas/genética , RNA Mensageiro/metabolismo , Adenocarcinoma de Células Claras/metabolismo , Adenocarcinoma de Células Claras/patologia , Carcinoma Endometrioide/metabolismo , Carcinoma Endometrioide/patologia , Linhagem Celular Tumoral , Movimento Celular , Bases de Dados Factuais , Progressão da Doença , Hiperplasia Endometrial/metabolismo , Hiperplasia Endometrial/patologia , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Laminina/metabolismo , Gradação de Tumores , Invasividade Neoplásica , Neoplasias Císticas, Mucinosas e Serosas/metabolismo , Neoplasias Císticas, Mucinosas e Serosas/patologia , Regulação para Cima
4.
Histopathology ; 60(5): 826-37, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22348356

RESUMO

AIMS: The aim of this study was to investigate the significance of the expression of Notch-related molecules in endometrial carcinoma. METHODS AND RESULTS: The expression of Notch receptors (Notch1 and 3) and Notch ligands [Jagged (JAG) 1 and Delta-like (DLL) 4] was examined immunohistochemically in 37 normal and 76 malignant endometrial tissue samples. For each section, immunohistochemical staining was scored using a positivity index (PI, full score; 200). The effects of a Notch inhibitor, DAPT, on cell proliferation, invasion and motility were investigated using endometrial carcinoma cell lines. The PIs for Notch1 (mean±SD 90.4±15.3), Notch3 (95.6 ± 20.4), JAG1 (95.5±10.0) and DLL4 (88.2±9.6), were significantly higher in endometrial carcinoma than normal endometrium. The PI for Notch1 was associated significantly with advanced International Federation of Gynecologists & Obstetricians (FIGO) stage. In addition, patients with tumours showing high expression of both Notch1 and JAG1 had a poor prognosis compared with those having double-negative carcinomas (P=0.015). DAPT suppressed invasiveness of cells derived from the endometrial carcinoma cell line KLE. CONCLUSIONS: The Notch1-JAG1 axis may enhance the invasive properties of endometrial carcinomas, which suggests the Notch pathway may be a promising target for the treatment of this malignancy.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Carcinoma Endometrioide/metabolismo , Neoplasias do Endométrio/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Receptor Notch1/metabolismo , Receptores Notch/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Biomarcadores Tumorais/metabolismo , Carcinoma Endometrioide/tratamento farmacológico , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dipeptídeos/farmacologia , Neoplasias do Endométrio/tratamento farmacológico , Feminino , Humanos , Proteína Jagged-1 , Invasividade Neoplásica , Prognóstico , Receptor Notch3 , Proteínas Serrate-Jagged
5.
Exp Mol Pathol ; 91(2): 563-8, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21763306

RESUMO

BACKGROUND: We previously reported the overexpression of lipocalin2 (LCN2), a 25kDa secretory protein involved in iron-transportation, in endometrial carcinoma and its possible contribution to endometrial carcinogenesis. Recently, a specific receptor for LCN2, solute carrier family 22 member 17 (SLC22A17), was identified. The present study was undertaken to investigate the expression of SLC22A17 in endometrial carcinoma. METHODS: The expression of the SLC22A17 and LCN2 proteins was examined immunohistochemically using 69 cases of endometrial carcinoma and adjacent normal endometrial tissues. Immunoreactivity was evaluated according to the percentage of positive cells and described as a positivity index (PI, full score 100). RESULTS: The expression of SLC22A17 was negligible in normal endometria, but positive staining for SLC22A17 (PI≧1) was observed in 35 cases of endometrial carcinoma. The PI for SLC22A17 was significantly higher in cases with histological grade 3 (P<0.0005), advanced FIGO stage (P=0.002), deep myometrial invasion (P=0.029), positive lymph-vascular space invasion (P=0.029), positive intraperitoneal cytology (P=0.020) and adnexal metastasis (P=0.029). The expression of SLC22A17 and LCN2 was positively correlated with a significant difference (P=0.002), and the patients who overexpressed both SLC22A17 and LCN2 showed poorer survival than those without the expression of SLC22A17 or LCN2 (P=0.002). Moreover, the overexpression of both SLC22A17 and LCN2 was indicated to be an independent prognostic factor by multivariable analysis. CONCLUSIONS: These results suggested that SLC22A17, in cooperation with LCN2, to be involved in the acquisition of aggressive behavior among endometrial carcinoma cells.


Assuntos
Proteínas de Fase Aguda/metabolismo , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Lipocalinas/metabolismo , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias do Endométrio/diagnóstico , Endométrio/metabolismo , Endométrio/patologia , Feminino , Humanos , Imuno-Histoquímica , Lipocalina-2 , Pessoa de Meia-Idade , Prognóstico , Análise de Sobrevida
6.
Int J Gynecol Cancer ; 21(7): 1287-96, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21685796

RESUMO

OBJECTIVE: To clarify the preoperative differential diagnosis and management of minimal deviation adenocarcinoma (MDA) and lobular endocervical glandular hyperplasia (LEGH), a multicenter study was performed. METHODS: A total of 112 patients who underwent conization or a hysterectomy for suspected MDA were collected from 24 hospitals. The pathological diagnosis in each case was determined by a central pathological review board. The diagnostic significance of clinicopathologic findings including results of magnetic resonance imaging (MRI), Papanicolaou (Pap) smears, and testing for gastric mucin was analyzed. RESULTS: The central pathological review identified 37 cases of Nabothian cyst or tunnel cluster, 54 cases of LEGH, 6 cases of MDA, 11 cases of adenocarcinoma, and 4 cases of benign disease. Lobular endocervical glandular hyperplasia was often associated with adenocarcinoma in situ, MDA, and mucinous adenocarcinoma. Three MDA patients had a recurrence, whereas none of LEGH patients had a recurrence irrespective of the type of surgery. On MRI, LEGH appeared as a characteristic multicystic lesion with an inner solid component, whereas MDA showed a predominantly solid pattern. A Pap smear or gastric mucin alone had limited diagnostic power. However, a combination of these findings is useful; that is, a cystic structure with inner solid components on MRI associated with mild glandular atypia and gastric mucin strongly suggested LEGH (24/26, 92%). A solid structure with atypical glandular cells was indicative of MDA or adenocarcinoma (5/5, 100%). CONCLUSIONS: The combination of MRI, Pap smears, and gastric mucin will improve the accuracy of the preoperative diagnosis of MDA and LEGH. Patients suspected of having LEGH may need to be treated with less aggressive methods.


Assuntos
Adenocarcinoma/diagnóstico , Colo do Útero/patologia , Neoplasias do Colo do Útero/diagnóstico , Adulto , Idoso , Diagnóstico Diferencial , Feminino , Mucinas Gástricas/análise , Humanos , Hiperplasia , Imageamento por Ressonância Magnética , Pessoa de Meia-Idade , Teste de Papanicolaou , Cuidados Pré-Operatórios , Estudos Retrospectivos , Esfregaço Vaginal
7.
J Cell Mol Med ; 14(9): 2305-17, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19583808

RESUMO

Although overexpression of cyclin A2 is reportedly an indicator of a poor prognosis of various malignancies including endometrial carcinoma, its molecular mechanism remains undetermined. To address this issue, we examined the effect of cyclin A2 on the development of resistance to chemotherapeutic drugs. The expression of cyclin A2 protein was increased in advanced-stage and chemotherapy-refractory stage endometrial carcinomas compared with that in early-stage tumours. The expression levels of cyclin A2 in endometrial carcinoma cell lines correlated positively with the IC(50) for cisplatin. Endometrial carcinoma HHUA cells that overexpressed cyclin A2 showed increased resistance to cisplatin in vitro and in vivo, via the activation of a survival pathway, the inositol-3 phosphate kinase (PI3K) cascade. The use of a cDNA microarray identified an Akt-binding protein, periplakin, as a novel target of cyclin A2. The cyclin A2-induced up-regulation of periplakin was mediated via direct binding of Sp1 to the promoter that was activated by cyclin A2 along with chromatin remodelling involving CBP/p300, and the siRNA-mediated silencing of periplakin suppressed the PI3K pathway. These results indicate cyclin A2 to be involved in the acquisition of aggressive behaviour of tumour cells through the activation of PI3K by cyclin A2-induced periplakin, and to be a promising therapeutic target.


Assuntos
Cisplatino/farmacologia , Ciclina A2/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias do Endométrio/patologia , Plaquinas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Regulação para Cima/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias do Endométrio/enzimologia , Neoplasias do Endométrio/genética , Ativação Enzimática/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sequência com Séries de Oligonucleotídeos , Fosfatidilinositol 3-Quinases/metabolismo , Plaquinas/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Reprodutibilidade dos Testes , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição Sp1/metabolismo
8.
Gynecol Endocrinol ; 26(3): 220-9, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19724954

RESUMO

Cyclin-dependent-kinase (cdk) inhibitor, p27(Kip1) (p27), has been shown to participate in progestin-induced growth suppression of normal endometrial glands. To analyse the molecular mechanisms regulating p27 protein, we examined immunohistochemical expression of the SCF(Skp2) (Skp1-Cullin-F-box protein) complex factors, i.e. Skp1, Cul1 and Skp2, and compared them with that of p27, steroid receptors and Ki-67. In normal endometrial glands, the expression of Skp2 was observed in the proliferative phase, whereas that of p27 was observed in the secretory phase. Cultured normal endometrial glandular cells showed that progesterone induced the down-regulation of Skp2 along with up-regulation of p27. In endometrial carcinomas, the inverse topological correlation between Skp2 and p27 was evident in 39/66 (59%) cases, and the expression of Skp2 showed a strong correlation with Ki-67. These findings suggest that the expression of SCF(Skp2) complex changes during the menstrual cycle in normal endometrium and the SCF(Skp2) ubiquitin-proteasome pathway may also work in endometrial carcinomas.


Assuntos
Carcinoma/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Neoplasias do Endométrio/metabolismo , Endométrio/metabolismo , Proteínas Quinases Associadas a Fase S/metabolismo , Adulto , Idoso , Proteínas Culina/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Pessoa de Meia-Idade , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo
9.
Endocr Relat Cancer ; 16(1): 113-22, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18852162

RESUMO

To examine estrogen-induced growth mechanisms of endometrial carcinoma, we investigated the estrogen-induced activation of the mitogen-activated protein kinase (MAPK) pathway and cell cycle regulators. Estradiol (E(2)) treatment at concentrations of 10(-8) M and 10(-6) M to estrogen receptor (ER)-positive endometrial carcinoma Ishikawa cells for 24 h resulted in increased cell proliferation by 20% and 28% respectively. The E(2)-induced proliferation was associated with the activation of extracellular signal-regulated kinase (MAPK)3/1 and up-regulation of cyclin D1 and E, which were suppressed by the addition of an MAP2K inhibitor (U0126) or an ER antagonist (ICI 182 780). Then, our screening for estrogen-inducible growth factors identified that IGF1 was up-regulated remarkably by E(2). Immunoprecipitation using conditioned medium of Ishikawa cells after E(2) treatment confirmed the E(2)-induced secretion of IGF1 protein. Treatment with recombinant IGF1 stimulated cell proliferation in a dose-dependent fashion, in association with MAPK3/1 phosphorylation and up-regulation of cyclin D1 and E. These IGF1-induced responses were suppressed by treatment with MAP2K inhibitor or anti-IGF1 receptor antibody. Immunohistochemical staining confirmed the expression of activated MAPK3/1 in normal proliferative phase endometria and endometrial carcinomas, indicating the involvement of this pathway in actively proliferating endometrial tissues in vivo. These findings suggest that E(2)-induced proliferation of endometrial carcinoma cells is mediated by the MAPK3/1 pathway via autocrine stimulation of IGF1.


Assuntos
Ciclina D1/metabolismo , Ciclina E/metabolismo , Neoplasias do Endométrio/metabolismo , Estradiol/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Comunicação Autócrina/fisiologia , Divisão Celular/fisiologia , Linhagem Celular Tumoral , Ciclina D1/genética , Ciclina E/genética , Relação Dose-Resposta a Droga , Neoplasias do Endométrio/patologia , Endométrio/metabolismo , Endométrio/patologia , Estradiol/farmacologia , Feminino , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Humanos , Fator de Crescimento Insulin-Like I/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
10.
Anticancer Res ; 29(4): 1023-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19414341

RESUMO

BACKGROUND: Although progestins have been used for the treatment of endometrial neoplasias, the mechanisms of progestin-induced growth suppression remain undetermined. MATERIALS AND METHODS: Immunostaining for steroid receptor coactivators (SRC-1, p300/CBP), corepressors (NCoR, SMRT) and Ki-67 in 15 neoplastic endometria before and after the treatment with medroxyprogesterone acetate (MPA) was performed. The effect of progestin on cell proliferation and cofactors expression were examined using T47D cells. RESULTS: Of the 15 cases, 10 showed good histological responses to MPA (Responder) and 5 poor responses (Non-responder). In Responders, MPA treatment resulted in reduced expression of Ki-67 by 78% (p=0.0076) along with increased NCoR expression by 158 % (p=0.0077). Progestin treatment for T47D cells resulted in up-regulation of NCoR mRNA and protein with the suppression of cell proliferation. Immunoprecipitation revealed that NCoR was bound to estrogen receptor alpha, but not to progesterone receptor in T47D cells. CONCLUSION: The up-regulation of NCoR by progestins is associated with the suppression of estrogen-induced growth of neoplastic cells.


Assuntos
Hiperplasia Endometrial/tratamento farmacológico , Neoplasias do Endométrio/tratamento farmacológico , Proteínas Nucleares/metabolismo , Progestinas/farmacologia , Proteínas Repressoras/metabolismo , Adulto , Western Blotting , Proliferação de Células , Proteínas de Ligação a DNA/metabolismo , Hiperplasia Endometrial/metabolismo , Hiperplasia Endometrial/patologia , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Feminino , Histona Acetiltransferases/metabolismo , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Proteínas Nucleares/genética , Correpressor 1 de Receptor Nuclear , Correpressor 2 de Receptor Nuclear , Coativador 1 de Receptor Nuclear , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas , Regulação para Cima , Adulto Jovem , Fatores de Transcrição de p300-CBP/metabolismo
11.
Placenta ; 85: 56-62, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31327484

RESUMO

OBJECTIVES: Cell senescence is irreversible cell cycle arrest. The human placenta is a unique organ that grows and matures during pregnancy until 40 weeks of gestation. However, the role of senescence in placental villi, particularly in the two types of trophoblast, has not yet been elucidated in detail. Therefore, we herein investigated the expression of cell senescence-related markers in trophoblast. METHODS: Seventy normal placental tissues were used. The expression of senescence-associated beta-galactosidase (SA-ß-gal), cell senescence-related markers (p16, p21, and promyelocytic leukemia; PML), and a growth marker (MCM2) was immunohistochemically examined. The expression of these markers in BeWo cells before and after cell fusion using forskolin was also investigated. RESULTS: The expression of MCM2 is detected in cytotrophoblast (CT). The expression of SA-ß-gal in CT is strong in the first and second trimesters, but weaker in the third trimester. Syncytiotrophoblast (ST) are negative in the first and second trimesters, but become positive in the third trimester. The immunohistochemical expression of p16, p21, and PML is stronger in CT than in ST throughout pregnancy. Furthermore, the expression of these markers in ST significantly increases as pregnancy advances. The expression of SA-ß-gal, PML, and p21 in BeWo cells is stronger after than before cell fusion. CONCLUSIONS: The proliferation and senescence of CT occurred in early to mid-pregnancy in association with syncytial fusion, while senescence was observed in ST in late pregnancy. This coordinated trophoblastic senescence may be essential for maintaining placental function.


Assuntos
Senescência Celular , Trofoblastos/fisiologia , Biomarcadores/metabolismo , Feminino , Humanos , Gravidez
12.
Clin Cancer Res ; 13(5): 1389-98, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17332280

RESUMO

PURPOSE: Research has revealed abnormal activation of the hedgehog pathway in human malignancies. The present study was undertaken to examine the expression and functional involvement of the hedgehog pathway in endometrial tissues. EXPERIMENTAL DESIGN: The expression of sonic hedgehog (Shh), patched (Ptch), Smoothened (Smo), and Gli1 was examined in various endometrial tissues and endometrial carcinoma cell lines. The effect of hedgehog signaling on the proliferation of endometrial carcinoma cell lines was also examined. RESULTS: The expression of Shh, Ptch, Smo, and Gli1 was very weak in normal endometrium, but was increased in endometrial hyperplasia and carcinoma stepwisely with significant differences. There was no marked difference in the expression of these molecules in carcinomas according to stages and histologic grades. Treatment with cyclopamine, a specific inhibitor of the hedgehog pathway, for endometrial carcinoma Ishikawa and HHUA cells suppressed growth by 56% and 67%, respectively, compared with the control. The addition of recombinant Shh peptide to HHUA cells enhanced their proliferation by 41%. The silencing of Gli1 using small interfering RNA (siGli1) resulted in the growth suppression and down-regulation of Ptch expression. In addition, the cyclopamine/siGli1-induced growth suppression was associated with the down-regulation of cyclins D1 and A and N-myc. No somatic mutations for ptch and smo genes were detected in the endometrial carcinoma cases examined. CONCLUSIONS: The abnormal activation of this pathway is involved in the proliferation of endometrial carcinoma cells possibly in an auto-/paracrine fashion, suggesting the possibility of the hedgehog pathway being a novel candidate for molecular targeting.


Assuntos
Neoplasias do Endométrio/metabolismo , Endométrio/metabolismo , Proteínas Hedgehog/metabolismo , Transdução de Sinais/fisiologia , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células , Análise Mutacional de DNA , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Gynecol Oncol Rep ; 22: 89-91, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29159261

RESUMO

•Pure-type ovarian squamous cell carcinoma (POSCC) is extremely rare.•This is the first report of G-CSF-producing POSCC.•This case was successfully treated with primary surgery and standard chemotherapy.•A tumor with uninfected neutrophilia may be a G-CSF-producing tumor.•18F-FDG-PET/CT and MRI may be useful for diagnosing G-CSF-producing tumors.

14.
Horm Cancer ; 8(4): 257-267, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28516379

RESUMO

Although progestin has been used to treat endometrial hyperplasia and endometrial carcinoma (EC), its therapeutic efficacy is limited. In order to improve this, the underlining mechanisms of the effects of progestin need to be elucidated in more detail. In the present study, we examined the involvement of mitogen-inducible gene-6 (MIG6), a negative regulator of the EGF receptor, in the progestin-mediated growth suppression of endometrial epithelia. The immunohistochemical expression of MIG6 was elevated in the early to mid-secretory phases of normal endometrium and also with endometrial hyperplasia after medroxyprogesterone acetate (MPA) therapy. The addition of progesterone (P4) to progesterone receptor (PR)-positive EC cells reduced the viability and induced MIG6 messenger RNA (mRNA) and protein expression. The silencing of MIG6 using siRNA eliminated the P4-mediated reduction of EC cell viability, indicating that MIG6 is an essential downstream component of PR-mediated growth suppression. In order to enhance PR-driven signals, we examined the effects of histone deacetylase (HDAC) inhibitors because histone acetylation has been shown to increase the expression of PR. The addition of three HDAC inhibitors (panobinostat, LBH589; trichostatin A, TSA; suberoylanilide hydroxamic acid, SAHA) decreased the viability of EC cells and up-regulated the expression of PR and MIG6, and these effects were the strongest with LBH589. The addition of LBH589 and MPA synergistically decreased the viability and increased apoptosis in EC cells. These results indicate that LBH589 has potential as an enhancer of progestin therapy via the up-regulation of PR and MIG6.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Neoplasias do Endométrio/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Indóis/farmacologia , Progestinas/farmacologia , Receptores de Progesterona/genética , Proteínas Supressoras de Tumor/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Biomarcadores , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Neoplasias do Endométrio/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Panobinostat , Receptores de Progesterona/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Supressoras de Tumor/metabolismo
15.
Appl Immunohistochem Mol Morphol ; 25(6): 415-421, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-26862948

RESUMO

Sirtuin 1 (SIRT1), originally identified as a longevity gene, regulates DNA repair and metabolism by deacetylating target proteins such as p53. SIRT1 plays a key role in the pathophysiology of metabolic diseases and neurodegenerative disorders, and is considered to protect against age-related diseases including cancer. In contrast, SIRT1 may be oncogenic because its overexpression has been detected in many cancers. The aim of the present study was to clarify the expression and the role of SIRT1 in ovarian carcinoma (OvCa). The expression of SIRT1 was evaluated immunohistochemically in 16 cases of normal ovaries, 35 cases of endometriosis with/without carcinoma, and 68 cases of OvCa (endometrioid, 16; clear cell, 20; mucinous, 16; serous, 16). Staining results were evaluated semiquantitatively by the Immunoreactive Scoring System, and the relationships with clinicopathologic features and outcomes of patients were analyzed. The expression of SIRT1 was higher in endometrioid, mucinous, and clear-cell carcinomas than in the inclusion cysts of normal ovaries, but not in serous carcinoma (P=0.038). The expression of SIRT1 on OvCa did not correlate with age, stage, location of metastasis, or capsular penetration. However, elevated SIRT1 expression was a significant predictor of shorter survival in univariate (P=0.038) and multivariate (P=0.037) survival analyses, regardless of the tumor stage. Results of the present study suggest a positive role for SIRT1 in the development of OvCa and its potential as a novel therapeutic target.


Assuntos
Neoplasias Ovarianas/patologia , Sirtuína 1/metabolismo , Feminino , Humanos , Pessoa de Meia-Idade , Neoplasias Ovarianas/metabolismo , Resultado do Tratamento
16.
Transl Oncol ; 10(4): 621-631, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28667895

RESUMO

BACKGROUND: SIRT1 is a longevity gene that forestalls aging and age-related diseases including cancer, and has recently attracted widespread attention due to its overexpression in some cancers. We previously identified the overexpression of SIRT1 in ovarian carcinoma (OvCa) as a poor prognostic factor. However, mechanistic insights into the function of SIRT1 in OvCa have yet to be elucidated. METHODS: Quantitative real-time reverse PCR (qRT-PCR) and Western blotting were employed to examine the expression of SIRT1 in a panel of human OvCa cell lines. si-RNA or sh-RNA and cDNA technologies were utilized to knockdown or overexpress SIRT1, respectively. The effects of SIRT1 on proliferation and chemoresistance were examined using a WST-1 assay, and the underlying mechanisms were confirmed using an apoptotic assay, and the quantification of glutathione (GSH), and reactive oxygen species (ROS). The aggressiveness of SIRT1 was analyzed using in vitro invasion and migration assays. RESULTS: SIRT1 was more strongly expressed in OvCa cell lines than in the immortalized ovarian epithelium at the gene and protein levels. Stress up-regulated the expression of SIRT1 in dose- and time-dependent manners. SIRT1 significantly enhanced the proliferation (P<.05), chemoresistance (P<.05), and aggressiveness of OvCa cells by up-regulating multiple antioxidant pathways to inhibit oxidative stress. Further study into the overexpression of SIRT1 demonstrated the up-regulation of several stemness-associated genes and enrichment of CD44v9 via an as-yet-unidentified pathway. CONCLUSIONS: Our results suggest that SIRT1 plays a role in the acquisition of aggressiveness and chemoresistance by OvCa, and has potential as a therapeutic target for OvCa.

17.
Oncotarget ; 8(42): 72147-72156, 2017 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-29069775

RESUMO

Drug repositioning is an alternative strategy redirecting existing drugs for new disease. We have previously reported an antitumor effect of statins, antidyslipidemic drugs, on ovarian cancer in vitro and in vivo. In this study, we investigated the antitumor effects of other mevalonate pathway inhibitors and the mechanism of the antitumor effect from a metabolic perspective. The effects of inhibitors of the mevalonate pathway on tumor cell growth were evaluated in vitro. Bisphosphonates that inhibit this pathway are commonly used as antiosteoporotic drugs, and antitumor effects of the bisphosphonate were examined in vitro and in vivo. Metabolites in SKOV3 ovarian cancer cells were analyzed before and after lovastatin treatment, using capillary electrophoresis-mass spectrometry. All mevalonate pathway inhibitors showed concentration-dependent inhibitory effects on tumor cell growth. Particularly marked effects were obtained with inhibitors of farnesyltransferase and geranylgeranyltransferase. The bisphosphonate was also shown to have an antitumor effect in vivo. The expression of autophagy marker LC3A/3B was increased in cells after treatment. In metabolomics analysis, lovastatin treatment increased the metabolites involved in the tricarboxylic acid cycle while reducing the metabolites associated with glycolysis. Also it decreased glutathione and resulted to work with chemotherapeutic agents synergistically. Inhibition at any point in the mevalonate pathway, and especially of farnesyl pyrophosphate and geranylgeranyl pyrophosphate, suppresses growth of ovarian cancer cells. Inhibition of this pathway may induce autophagy, cause a shift to activation of the tricarboxylic acid cycle and enhance susceptibility to chemotherapy. Drug repositioning targeting mevalonate pathway for ovarian cancer deserves consideration for clinical application.

18.
Endocrinology ; 147(10): 4863-70, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16825317

RESUMO

Impaired mismatch repair (MMR) is reportedly crucial in the early stages of endometrial carcinogenesis. Although estrogen exposure is considered an important risk factor for endometrial carcinoma, the relationship between estrogen and MMR activity remains undetermined. The present study was undertaken to elucidate the effect of estrogen on MMR activity in normal and malignant endometrial cells. The expression of MMR proteins, hMLH1 and hMSH2, and its correlation with estrogen was examined using immunohistochemical and immunofluorescent techniques. The effect of estradiol (E2) on the expression of hMLH1/hMSH2 protein/mRNA and in vitro MMR activity using two types of heteroduplex (G/T mismatches, 2-base insertion-deletion loops) was examined in cultured normal endometrial glandular cells and estrogen receptor-positive endometrial carcinoma Ishikawa cells. Immunohistochemical expression of hMLH1 and hMSH2 in normal endometrial glands was positively correlated with the serum E2 levels. The expression of hMLH1/hMSH2 protein and mRNA was increased in normal endometrial glandular and Ishikawa cells by E2 treatment. In vitro MMR activity was up-regulated by E2 in both types of cell and heteroduplex. Immunofluorescent analysis demonstrated that E2 enhanced proliferation and hMLH1/hMSH2 expression in both cells; however, proliferating cells without hMLH1/hMSH2 expressions implying high-risk cells were more frequently observed under low E2 concentrations. Collectively, the E2-induced up-regulation of MMR activity in endometrial cells suggests that high estrogen levels act as an intrinsic defense against endometrial carcinogenesis, whereas the imbalance between cell growth and MMR under low E2 environment as seen at postmenopause is vulnerable to carcinogenesis.


Assuntos
Pareamento Incorreto de Bases/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Neoplasias do Endométrio/metabolismo , Endométrio/metabolismo , Estrogênios/farmacologia , Proteínas Adaptadoras de Transdução de Sinal , Adulto , Western Blotting , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Células Cultivadas , Neoplasias do Endométrio/patologia , Endométrio/citologia , Endométrio/efeitos dos fármacos , Estrogênios/sangue , Feminino , Técnica Direta de Fluorescência para Anticorpo , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Proteína 1 Homóloga a MutL , Proteína 2 Homóloga a MutS/biossíntese , Proteína 2 Homóloga a MutS/genética , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima/efeitos dos fármacos
19.
Clin Cancer Res ; 11(17): 6133-8, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16144912

RESUMO

PURPOSE: Although several gene abnormalities have been reported in endometrial carcinoma, the genetic alterations have not fully been elucidated. Recent studies have revealed frequent activating mutations of the gene for BRAF, an effector of Ras protein in the mitogen-activated protein kinase pathway, in several malignancies. However, the prevalence and significance of BRAF mutations in endometrial carcinoma remain unclear. EXPERIMENTAL DESIGN: We examined BRAF mutations in exons 11 and 15 in 97 cases of endometrial carcinoma (endometrioid type, 78; nonendometrioid type, 19), 9 cases of atypical endometrial hyperplasia, and 20 cases of normal endometrium by direct sequencing. In addition, mutations of KRAS and p53 and the immunohistochemical expression of hMLH1 and hMSH2 were also examined. RESULTS: Of the 97 carcinomas and 9 hyperplasias, 20 (21%) and 1 (11%) had BRAF mutations, most of them at previously unreported sites. Twenty samples of normal endometrium and 21 samples of normal endometrium obtained from sites adjacent to neoplastic lesions had no BRAF mutations. There was no apparent difference in the prevalence of BRAF mutation among stages, histologic subtypes, or grades. Mutations of KRAS and p53 were found in 18 (19%) and 22 (23%) cases, and 65 (67%) and 92 (95%) cases showed positive immunostaining for hMLH1 and hMSH2, respectively. BRAF mutation was more frequently found in hMLH1-negative cases (12 of 32, 41%) than in hMLH1-positive cases (7 of 65, 11%; P = 0.008), suggesting that it is associated with an abnormal mismatch repair function. CONCLUSIONS: These findings suggest that mutations of the BRAF gene are partly involved in the malignant transformation of the endometrium.


Assuntos
Hiperplasia Endometrial/genética , Neoplasias do Endométrio/genética , Mutação/genética , Proteínas Proto-Oncogênicas B-raf/genética , Proteína Supressora de Tumor p53/genética , Proteínas Adaptadoras de Transdução de Sinal , Adulto , Idoso , Proteínas de Transporte , Análise Mutacional de DNA , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Endométrio/fisiologia , Feminino , Genes ras/fisiologia , Humanos , Técnicas Imunoenzimáticas , Masculino , Pessoa de Meia-Idade , Proteína 1 Homóloga a MutL , Proteína 2 Homóloga a MutS , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas/metabolismo
20.
PLoS One ; 11(5): e0155220, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27168162

RESUMO

PURPOSE: Lipocalin 2 (LCN2) is a secretory protein that is involved in various physiological processes including iron transport. We previously identified LCN2 as an up-regulated gene in endometrial carcinoma, and found that the overexpression of LCN2 and its receptor, SLC22A17, was associated with a poor prognosis. However, the functions and mechanism of action of LCN2 currently remain unclear. METHODS: The LCN2-overexpressing endometrial carcinoma cell lines, HHUA and RL95-2, and LCN2-low-expressing one, HEC1B, were used. The effects of LCN2 on cell migration, cell viability, and apoptosis under various stresses, including ultraviolet (UV) irradiation and cisplatin treatment, were examined using the scratch wound healing assay, WST-1 assay, and Apostrand assay, respectively. RESULTS: LCN2-silencing using shRNA method significantly reduced the migration ability of cells (p<0.05). Cytotoxic stresses significantly decreased the viability of LCN2-silenced cells more than that of control cells. In contrast, LCN2 overexpression was significantly increased cisplatin resistance. These effects were canceled by the addition of the iron chelator, deferoxamine. After UV irradiation, the expression of phosphorylated Akt (pAkt) was decreased in LCN2-silenced cells, and the PI3K inhibitor canceled the difference induced in UV sensitivity by LCN2. The cisplatin-induced expression of pAkt was not affected by LCN2; however, the expression of p53 and p21 was increased by LCN2-silencing. CONCLUSIONS: These results indicated that LCN2 was involved in the migration and survival of endometrial carcinoma cells under various stresses in an iron-dependent manner. The survival function of LCN2 may be exerted through the PI3K pathway and suppression of the p53-p21 pathway. These functions of LCN2 may increase the malignant potential of endometrial carcinoma cells.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/genética , Células Epiteliais/metabolismo , Regulação Neoplásica da Expressão Gênica , Lipocalina-2/genética , Fosfatidilinositol 3-Quinases/genética , Proteína Supressora de Tumor p53/genética , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/efeitos da radiação , Cisplatino/farmacologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Endométrio/patologia , Endométrio/efeitos da radiação , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Células Epiteliais/efeitos da radiação , Feminino , Humanos , Lipocalina-2/antagonistas & inibidores , Lipocalina-2/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA