Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Am J Pathol ; 193(8): 1116-1128, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37169340

RESUMO

Epithelial-mesenchymal transition is a hallmark of uterine carcinosarcoma (UCS). Here, shotgun proteomics analysis used to identify biomarkers associated with blebbistatin-mediated epithelial-mesenchymal transition in UCS indicated up-regulation of nucleobindin-2 (NUCB2) in endometrial carcinoma (Em Ca) cells. Expression of N-cadherin, Snail, Slug, and ZEB1 was reduced in NUCB2 knockout Em Ca cells, whereas ZEB1, Twist1, and vimentin were up-regulated in NUCB2-overexpressing Em Ca cells. NUCB2 knockout reduced cell proliferation and migration, whereas NUCB2 overexpression had the opposite effect. Treatment of Em Ca cells with transforming growth factor (TGF)-ß1 dramatically altered morphology toward a fibroblastic appearance; concomitantly, expression of NUCB2 and ZEB1 increased. The NUCB2 promoter was also activated by transfection of Smad2. In UCS tissues, NUCB2 expression was significantly higher in sarcomatous compared with carcinomatous components, which was consistent with increased TGF-ß1 mRNA expression in stromal and sarcomatous components compared with carcinomatous components. In addition, NUCB2 score correlated positively with ZEB1 and vimentin scores, whereas ZEB1 score correlated positively with Slug and vimentin scores and inversely with the E-cadherin score. Collectively, these data indicate that TGF-ß-dependent up-regulation of NUCB2 and ZEB1 contributes to the phenotypic characteristics of sarcomatous components in UCS.


Assuntos
Carcinossarcoma , Neoplasias Uterinas , Humanos , Feminino , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo , Nucleobindinas/genética , Nucleobindinas/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Vimentina/metabolismo , Genes Homeobox , Caderinas/genética , Caderinas/metabolismo , Neoplasias Uterinas/genética , Neoplasias Uterinas/patologia , Fenótipo , Carcinossarcoma/genética , Carcinossarcoma/patologia , Dedos de Zinco , Transição Epitelial-Mesenquimal/genética , Linhagem Celular Tumoral
2.
Am J Pathol ; 191(10): 1837-1850, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34214505

RESUMO

Deregulated full-length anaplastic lymphoma kinase (ALK) overexpression has been found in some primary solid tumors, but little is known about its role in ovarian high-grade serous carcinoma (HGSC). The current study focused on the functional roles of ALK in HGSC. Cytoplasmic ALK immunoreactivity without chromosomal rearrangement and gene mutations was significantly higher in HGSC compared with non-HGSC-type ovarian carcinomas, and was significantly associated with several unfavorable clinicopathologic factors and poor prognosis. HGSC cell lines stably overexpressing ALK exhibited increased cell proliferation, enhanced cancer stem cell features, and accelerated cell mobility, whereas these phenotypes were abrogated in ALK-knockdown cells. Expression of the nervous system-associated gene, ELAVL3, and the corresponding protein (commonly known as HuC) was significantly increased in cells overexpressing ALK. Expression of SRY-box transcription factor (Sox)2 and Sox3 (genes associated with the neural progenitor population) increased in ALK-overexpressing but not ALK-knockdown cells. Furthermore, overexpression of Sox2 or Sox3 enhanced both ALK and ELAVL3 promoter activities, suggesting the existence of ALK/Sox/HuC signaling loops. Finally, ALK overexpression was attributed to increased expression of neuroendocrine markers, including synaptophysin, CD56, and B-cell lymphoma 2, in HGSC tissues. These findings suggest that overexpression of full-length ALK may influence the biological behavior of HGSC through cooperation with ELAVL3 and Sox factors, leading to the establishment and maintenance of the aggressive phenotypic characteristics of HGSC.


Assuntos
Quinase do Linfoma Anaplásico/metabolismo , Cistadenocarcinoma Seroso/enzimologia , Cistadenocarcinoma Seroso/patologia , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/patologia , Adulto , Idoso , Diferenciação Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Citoplasma/enzimologia , Proteína Semelhante a ELAV 3/metabolismo , Feminino , Humanos , Pessoa de Meia-Idade , Modelos Biológicos , Análise Multivariada , Gradação de Tumores , Células-Tronco Neoplásicas/patologia , Células Neuroendócrinas/metabolismo , Células Neuroendócrinas/patologia , Fenótipo , Prognóstico , Intervalo Livre de Progressão , Fatores de Transcrição SOX/metabolismo
3.
Carcinogenesis ; 42(9): 1162-1170, 2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34323956

RESUMO

Patients with ovarian clear cell carcinoma (OCCC) experience frequent recurrence, which is most likely due to chemoresistance. We used shotgun proteomics analysis and identified upregulation of ezrin-binding phosphoprotein 50 (EBP50) in recurrent OCCC samples. Cytoplasmic and/or nuclear (Cyt/N), but not membranous, EBP50 immunoreactivity was significantly higher in recurrent OCCC as compared with that of primary tumors. OCCC cells expressing cytoplasmic EBP50 were significantly less susceptible to cisplatin (CDDP)-induced apoptosis compared with cells expressing membranous EBP50. Abrogation of resistance following knockdown of cytoplasmic EBP50 was accompanied by decreased XIAP and BCL2, increased BAX and increased caspase-3 cleavage. We found that poly (ADP-ribose) polymerase1 (PARP1), which is involved in DNA damage detection and repair, binds to EBP50 through its PDZ1 domain. CDDP treatment of cells expressing cytoplasmic (but not membranous) EBP50 increased nuclear PARP1 expression, whereas knockdown of EBP50 cells decreased PARP1 expression and activity following CDDP treatment. Finally, OCCC patients with a combination of Cyt/N EBP50 and high PARP1 score had worst the prognosis for overall and progression-free survival. Together, our data suggest that cytoplasmic EBP50 inhibits apoptosis and promotes OCCC survival through stabilization of PARP1 activity and modulation of the XIAP/BCL2/BAX axis. This may increase the likelihood of tumor recurrence, and we therefore suggest a combined analysis for EBP50 and PARP1 may have great utility in OCCC prediction and prognosis.


Assuntos
Biomarcadores Tumorais/metabolismo , Citoplasma/metabolismo , Neoplasias Ovarianas/metabolismo , Fosfoproteínas/metabolismo , Poli(ADP-Ribose) Polimerase-1/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose , Caspase 3/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Feminino , Humanos , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/metabolismo , Neoplasias Ovarianas/patologia , Prognóstico , Ligação Proteica , Proteômica/métodos , Análise de Sobrevida
4.
Am J Pathol ; 190(11): 2304-2316, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32805233

RESUMO

S100A4 is a small calcium-binding protein that exerts its biological functions by interacting with nonmuscle myosin IIA (NMIIA) and p53. Although S100A4 promotes metastasis in several tumors, little is known about its involvement in the progression of ovarian high-grade serous carcinomas (HGSCs). Herein, we focused on functional roles of the S100A4/NMIIA/p53 axis in these tumors. In HGSC cell lines harboring mutant p53, knockdown (KD) of S100A4 reduced the expression of several epithelial-mesenchymal transition/cancer stem cell markers and the ALDH1high population, consistent with an inhibition of stemness features. S100A4-KD also increased apoptosis, decreased cell proliferation, and accelerated cell mobility. This was accompanied by increased Snail expression, which, in turn, was likely due to loss of p53 function. In contrast, specific inhibition of NMIIA by blebbistatin induced phenotypes that-with the exception of cell proliferation and mobility-were opposite to those observed in S100A4-KD cells. In clinical samples, cytoplasmic and/or nuclear interactions between S100A4, NMIIA, and mutant p53 were observed. In addition, high expression of S100A4, but not NMIIA or p53, was a significant and independent unfavorable prognostic factor in HGSC patients. These findings suggest that, via its interaction with NMIIA and p53, overexpressed S100A4 may induce epithelial-mesenchymal transition/cancer stem cell properties in HGSC and elicit several other tumor-associated phenotypes.


Assuntos
Cistadenocarcinoma Seroso/metabolismo , Células-Tronco Neoplásicas/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Neoplasias Ovarianas/metabolismo , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Cistadenocarcinoma Seroso/patologia , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células-Tronco Neoplásicas/patologia , Neoplasias Ovarianas/patologia , Fatores de Transcrição da Família Snail/biossíntese
5.
Lab Invest ; 100(5): 682-695, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31857700

RESUMO

Uterine carcinosarcoma (UCS) represents a true example of cancer associated with epithelial-mesenchymal transition (EMT), which exhibits cancer stem cell (CSC)-like traits. Although S100A4 is an inducer of EMT, little is known about its involvement in UCS tumorigenesis. Herein, we focused on the functional role of S100A4 during development of UCS. Expression of S100A4 and molecules associated with its function were also examined in 35 UCS cases. In endometrial carcinoma cell lines, S100A4 promoter activity and mRNA levels were significantly increased by the transfection of NF-κB/p65, independent of a putative κB-binding site in the promoter. Cells stably overexpressing S100A4 showed enhancement of CSC properties, along with decreased cell proliferation and acceleration of cell migration. These phenotypes were abrogated in S100A4-knockdown cells. A combination of S100A4 antibody-mediated co-immunoprecipitation and shotgun proteomics analysis revealed that S100A4 strongly interacted with non-muscle myosin II (NMII) heavy chains, including myosin 9 and myosin 14. Specific inhibition of NMII by blebbistatin phenocopied S100A4 overexpression and induced a fibroblast-like morphology. In clinical samples, S100A4 score was significantly higher in sarcomatous as compared with carcinomatous components of UCS, and was positively correlated with ALDH1, Slug, and vimentin scores, and inversely with Ki-67 labeling indices. These findings suggest that an S100A4/NMII-related signaling cascade may contribute to the establishment and maintenance of EMT/CSC properties, along with changes in cell proliferation and migration capability. These events may be initiated in carcinomatous components in UCS and lead to divergent sarcomatous differentiation.


Assuntos
Carcinossarcoma/patologia , Transição Epitelial-Mesenquimal/fisiologia , Proteína A4 de Ligação a Cálcio da Família S100 , Transdução de Sinais/fisiologia , Neoplasias Uterinas/patologia , Carcinossarcoma/química , Linhagem Celular Tumoral , Feminino , Técnicas de Silenciamento de Genes , Humanos , Proteína A4 de Ligação a Cálcio da Família S100/genética , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo , Neoplasias Uterinas/química , Útero/química , Útero/patologia
6.
Mol Carcinog ; 59(12): 1409-1419, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33111989

RESUMO

Glioblastomas (GBM) contain numerous hypoxic foci associated with a rare fraction of glioma stem cells (GSCs). Left-right determination factor (LEFTY) and Nodal, members of the transforming growth factor ß (TGF-ß) superfamily, have glycogen synthase kinase 3ß (GSK-3ß) phosphorylation motifs and are linked with stemness in human malignancies. Herein, we investigated the roles of LEFTY and Nodal in GBM hypoxic foci. In clinical samples, significantly higher expression of LEFTY, Nodal, phospho (p) GSK-3ß, pSmad2, and Nestin, as well as higher apoptotic and lower proliferation rates, were observed in nonpseudopalisading (non-Ps) perinecrotic lesions as compared to Ps and non-necrotic tumor lesions, with a positive correlation between LEFTY, Nodal, pGSK-3ß, or pSmad2 scores. In KS-1, a GBM cell line that lacks endogenous Nodal expression, treatment with the hypoxic mimetic CoCl2 increased LEFTY, pGSK-3ß, and pSmad2 levels, but decreased pAkt levels. Moreover, the promoter for LEFTY, but not Nodal, was activated by Smad2 or TGF-ß1, suggesting that overexpression of LEFTY and Nodal may be due to Akt-independent GSK-3ß inactivation, with or without cooperation of the TGF-ß1/Smad2 axis. LEFTY and Nodal overexpression increased proliferation rates and reduced susceptibility to CoCl2 -induced apoptosis, and increased the expression of epithelial-mesenchymal transition (EMT)/GSC-related markers. An increased ALDH1high population and more efficient spheroid formation was also observed in LEFTY-overexpressing cells. These findings suggest that LEFTY and Nodal may contribute to cell survival in non-Ps GBM perinecrotic lesions, leading to alterations in apoptosis, proliferation, or EMT/GCS features.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Fatores de Determinação Direita-Esquerda/metabolismo , Proteína Nodal/metabolismo , Regulação para Cima , Adolescente , Adulto , Idoso , Neoplasias Encefálicas/genética , Hipóxia Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Cobalto/efeitos adversos , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Humanos , Fatores de Determinação Direita-Esquerda/genética , Masculino , Pessoa de Meia-Idade , Proteína Nodal/genética , Fosforilação , Transdução de Sinais , Adulto Jovem
7.
Cell Commun Signal ; 18(1): 103, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32635925

RESUMO

BACKGROUND: We previously demonstrated that ovarian high grade serous carcinomas (OHGSeCa) and ovarian clear cell carcinomas (OCCCa) with an HNF-1ß+/p53+/ARID1A+ immunophenotype were associated with the worst unfavorable prognosis. To clarify the molecular mechanisms underlying this finding, we focused on alterations in the p53 signaling pathway in these tumors. METHODS: Changes in cell phenotype and function following knockdown of wild-type p53 (p53-KD) were assessed using OCCCa cells expressing endogenous HNF-1ß and ARID1A. The prognostic significance of molecules that were deregulated following p53-KD was also examined using 129 OCCCa/OHGSeCa cases. RESULTS: p53-KD cells had increased expression of Snail, phospho-Akt (pAkt), and pGSK3ß, and decreased E-cadherin expression, leading to epithelial-mesenchymal transition (EMT)/cancer stem cell (CSC) features. The cells also exhibited acceleration of cell motility and inhibition of cell proliferation and apoptosis. Next generation sequencing revealed that fibronectin (FN) expression was significantly increased in the p53 KD-cells, in line with our observation that wild-type p53 (but not mutant p53) repressed FN1 promoter activity. In addition, treatment of OCCCa cells with FN significantly increased cell migration capacity and decreased cell proliferation rate, independent of induction of EMT features. In clinical samples, FN/p53 scores were significantly higher in OCCCa/OHGSeCa with the HNF-1ß+/p53+/ARID1A+ immunophenotype when compared to others. Moreover, high FN/high p53 expression was associated with the worst overall survival and progression-free survival in OCCCa/OHGSeCa patients. CONCLUSION: These findings suggest that upregulation of FN following loss of p53 function may impact the biological behavior of OCCCa/OHGSeCa, particularly in tumors with an HNF-1ß+/p53+/ARID1A+ immunophenotype, through alterations in cell mobility and cell proliferation. The accompanying induction of EMT/CSC properties and inhibition of apoptosis due to p53 abnormalities also contribute to the establishment and maintenance of tumor phenotypic characteristics. Video Abstract.


Assuntos
Fibronectinas/genética , Imunofenotipagem , Neoplasias Ovarianas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima/genética , Linhagem Celular Tumoral , Movimento Celular , Proteínas de Ligação a DNA/metabolismo , Transição Epitelial-Mesenquimal/genética , Feminino , Fibronectinas/metabolismo , Regulação Neoplásica da Expressão Gênica , Fator 1-beta Nuclear de Hepatócito/metabolismo , Humanos , Cinética , Pessoa de Meia-Idade , Modelos Biológicos , Análise Multivariada , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neoplasias Ovarianas/patologia , Fenótipo , Prognóstico , Intervalo Livre de Progressão , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo
8.
BMC Cancer ; 19(1): 308, 2019 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-30943930

RESUMO

BACKGROUND: Expression of Nodal, a member of the TGF-ß superfamily, is commonly absent in differentiated tissues, while its re-expression occurs in a variety of human malignancy. However, little is known about its involvement in ovarian tumorigenesis. Herein, we focused on the functional roles of Nodal in ovarian endometriosis-carcinoma lesions. METHODS: Regulation and function of Nodal and its associated molecules, including Smad2, GSK-3ß, and several cell kinetics-related molecules, were assessed using clinical samples consisting of 108 ovarian carcinomas and 33 endometriotic lesions, as well as ES-2 (ovarian clear cell carcinoma; OCCCa) and Ishikawa (endometrial carcinoma) cell lines. RESULTS: Nodal expression was significantly higher in endometriosis and OCCCa lesions as compared to that of non-OCCCas, with positive correlations to phosphorylated forms of both Smad2 (pSmad2) and GSK-3ß. When compared to endometriotic lesions, the expression of Nodal and pSmad2 was significantly decreased in OCCCa. Treatment of Ishikawa cells with TGF-ß1 resulted in transcriptional upregulation of Nodal, along with increased pSmad2 expression, while inhibition of GSK-3ß also induced an increase in Nodal expression at the posttranslational level. Both ES-2 and Ishikawa cells stably overexpressing Nodal had increased susceptibility to apoptosis in response to treatment with cisplatin and doxorubicin, respectively, together with higher cleaved caspase-3 expression and decreased Bcl2/Bax ratio. Moreover, the stable Nodal-overexpressing cells showed reduced cell proliferation, along with increased expression of p27kip1 and p21waf1. In clinical samples, a significantly higher number of apoptotic cells and lower Ki-67 labeling indices were observed in Nodal-positive as compared to Nodal-negative OCCCa. CONCLUSIONS: These findings suggest that Nodal is a multifunctional cytokine involved in the modulation of cell kinetics in ovarian endometriosis-OCCCa lesions.


Assuntos
Adenocarcinoma de Células Claras/metabolismo , Endometriose/metabolismo , Proteína Nodal/metabolismo , Neoplasias Ovarianas/metabolismo , Regulação para Cima , Adenocarcinoma de Células Claras/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose , Proliferação de Células , Endometriose/genética , Feminino , Humanos , Pessoa de Meia-Idade , Proteína Nodal/genética , Neoplasias Ovarianas/genética , Fosforilação , Transdução de Sinais , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/metabolismo
9.
Gan To Kagaku Ryoho ; 45(9): 1353-1355, 2018 Sep.
Artigo em Japonês | MEDLINE | ID: mdl-30237380

RESUMO

The patient was a 42-year-old man who presented with dysphagia.Upper gastrointestinal endoscopy revealed a protruding lesion in the lower thoracic esophagus.Pathological analysis of the lesion showed squamous cell carcinoma.Laboratory data showed leukocytosis(21,200/mL)despite no evidence of infection, and the serum levels of granulocyte colony-stimu- lating factor(G-CSF)were elevated to 283 pg/mL.We diagnosed him with esophageal squamous cell carcinoma(Lt, type 1, cT4N4M0, cStage IV a).After administering 2 courses of docetaxel plus cisplatin plus S-1(DCS)as neoadjuvant chemotherapy, the patient underwent surgery.The pathological diagnosis was pType 2, T2, N4, M0, pStage IV a. G-CSF immunostaining was positive in tumor cells.After the surgery, the number of leukocytes and serum G-CSF levels decreased to within normal limits.Adjuvant chemotherapy was administered.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Esofágicas/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/biossíntese , Adulto , Terapia Combinada , Transtornos de Deglutição/etiologia , Neoplasias Esofágicas/complicações , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/cirurgia , Carcinoma de Células Escamosas do Esôfago/complicações , Carcinoma de Células Escamosas do Esôfago/cirurgia , Humanos , Masculino
10.
Gan To Kagaku Ryoho ; 43(8): 999-1002, 2016 Aug.
Artigo em Japonês | MEDLINE | ID: mdl-27539044

RESUMO

The first patient was a man in his eighties who visited our department because of anemia. Gastrofiberscopy revealed a bleeding submucosal tumor, approximately 50mm in diameter, in the cardia ofthe stomach. Considering that he underwent coronary-artery bypass surgery and received 3 oral antithrombotic medicines, his bleeding tendency was so high that we decided to choose partial gastrectomy. A postoperative histopathological examination revealed that the tumor was a small cell endocrine carcinoma. The second patient was a woman in her seventies. She had consulted her personal physician because of gastric ulcers; periodic gastrofiberscopy revealed a type 3 gastric cancer, approximately 40mm in diameter, on the posterior wall ofthe middle section ofher stomach. It was histologically diagnosed as a poorly differentiated neuroendocrine carcinoma. On a preoperative blood examination, the levels ofhormones such as glucagon, serotonin, and gastrin were within their respective normal limits. Total gastrectomy was performed, and she received oral S-1 for adjuvant chemotherapy since her discharge from the hospital.


Assuntos
Carcinoma Neuroendócrino/tratamento farmacológico , Neoplasias Gástricas/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Carcinoma Neuroendócrino/cirurgia , Quimioterapia Adjuvante , Evolução Fatal , Feminino , Humanos , Masculino , Estadiamento de Neoplasias , Neoplasias Gástricas/patologia , Neoplasias Gástricas/cirurgia
11.
Hum Pathol ; 85: 210-220, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30448218

RESUMO

Although proteomic profiles for ovarian epithelial carcinoma (OECa) have been widely investigated, no single marker or set of predictors has been clinically implemented mainly because their reliability and validity have not yet been well established. To establish immunohistochemical (IHC) panels for prognosis prediction of OECa for use in daily pathology practice, the expression patterns of 12 IHC markers, p53, HNF-1ß, ARID1A, estrogen receptor-α, progesterone receptor, vimentin, PTEN, PIK3CA, WT1, left-right determination factor, ß-catenin, and Ki-67 were investigated using 282 OECas. Hierarchical clustering analysis revealed 7 major immunoprofile groups (IPGs I-VII) that could be used to categorize OECa tumors independent of histotypes. Based on the results of the cluster analysis and protein expression statuses, we further demonstrated the effective classification of OECa tumors into simplified immunoprofile panels using only 4 IHC markers including HNF-1ß, p53, ARID1A, and WT1. The tumors in IPG VII with HNF1ß+/p53+/ARID1A+ immunophenotype demonstrated a significantly worse overall survival and progression-free survival as compared with the other IPGs. Multivariate Cox regression analysis also revealed that the immunophenotype (HNF1ß+/p53+/ARID1A+) and clinical stage were significant and independent prognostic factors for overall survival and progression-free survival in advanced OECa. In conclusion, we identified immunoprofiles in OECa using a panel of 4 IHC markers, which could identify tumors by the immunophenotype that is associated with the most unfavorable prognosis and thus facilitate prognosis prediction of advanced OECa.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Epitelial do Ovário/metabolismo , Neoplasias Ovarianas/metabolismo , Carcinoma Epitelial do Ovário/mortalidade , Carcinoma Epitelial do Ovário/patologia , Feminino , Humanos , Imunofenotipagem , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/patologia , Prognóstico , Intervalo Livre de Progressão , Proteômica , Taxa de Sobrevida
12.
Hum Pathol ; 89: 10-23, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31022415

RESUMO

We focused on specific molecular events during the development of endometriosis-associated ovarian endometrioid carcinoma (OEmCa) and ovarian clear cell carcinoma (OCCCa). Alterations in ß-catenin (encoded by CTNNB1) and microsatellite instability (MSI), as well as changes in the expression levels of HNF-1ß and DNA mismatch repair (MMR) proteins were investigated in 50 OEmCas and 21 OCCCas with endometriotic lesions. Mutations of CTNNB1 were identified in 28 (56%) of the 50 OEmCa cases and 26 (41.9%) of the 62 coexisting endometriosis lesions. MSI-high (H) was observed in 7 (14.6%) of the 48 OEmCa and 14 (23.3%) of the 60 coexisting endometriosis, and was significantly associated with loss of MMR protein expression, but not CTNNB1 mutations. Nonidentical CTNNB1 status between 2 different epithelial lesions within endometriosis was observed in 8 of 10 informative endometriosis cases that had adjacent OEmCa. Similar findings for MSI features were also found in 2 of 3 informative cases, suggesting that endometriotic lesions may predominantly consist of polyclonal cells. In contrast, high HNF-1ß expression was significantly associated with SLC3A1 expression, which plays a major role in HNF-1ß-triggered induction of reactive oxygen species in OCCCas, independent of abnormalities in both ß-catenin and MSI/MMR status. Finally, 4 inflammatory parameters associated with repeated hemorrhaging in endometriosis were significantly higher in endometriosis with MSI-high when compared with that with MSS, independent of both ß-catenin and HNF-1ß status. In conclusion, different molecular pathways including alterations in ß-catenin, MSI, and HNF-1ß levels may contribute to tumorigenesis in endometriosis-associated carcinoma.


Assuntos
Carcinogênese/metabolismo , Endometriose/complicações , Neoplasias Ovarianas/etiologia , Neoplasias Ovarianas/metabolismo , Adenocarcinoma de Células Claras/etiologia , Adenocarcinoma de Células Claras/genética , Adenocarcinoma de Células Claras/metabolismo , Adulto , Idoso , Carcinogênese/genética , Carcinoma Endometrioide/etiologia , Carcinoma Endometrioide/genética , Carcinoma Endometrioide/metabolismo , Endometriose/metabolismo , Feminino , Fator 1-beta Nuclear de Hepatócito/genética , Fator 1-beta Nuclear de Hepatócito/metabolismo , Humanos , Instabilidade de Microssatélites , Pessoa de Meia-Idade , Neoplasias Ovarianas/genética , Adulto Jovem , beta Catenina/genética , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA