Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Diagn Pathol ; 19(1): 46, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38429827

RESUMO

AIMS: Primary mucoepidermoid carcinomas (MECs) of the sinonasal tract and nasopharynx are rare entities that represent a diagnostic challenge, especially in biopsy samples. Herein, we present a case series of MECs of the sinonasal and skull base and its mimics to evaluate the clinicopathological and molecular characteristics in order to avoid misdiagnosis. METHODS: We reviewed the pathology records of patients diagnosed from 2014 to 2022. Thirty MECs were consecutively diagnosed during that period. RESULTS: Based on morphological and fluorescence in situ hybridization (FISH) analyses, 30 tumors originally diagnosed as MECs were separated into MAML2 fusion-positive (7 cases) and MAML2 fusion-negative groups (23 cases), in which 14 tumors were positive for the EWSR1::ATF1 fusion; these tumors were reclassified to have hyalinizing clear cell carcinoma (HCCC). The remaining nine MAML2 FISH negative cases were reconfirmed as squamous cell carcinoma (SCC, 3 cases) which showed keratinization and high Ki-67 expression; DEK::AFF2 carcinomas (2 cases), in which DEK gene rearrangement was detected by FISH; and MECs as previously described (4 cases) with typical morphological features. Including 7 MAML2 rearrangements tumors, 11 MEC cases had a male-to-female ratio of 4.5:1, and 6 tumors arose from the nasopharyngeal region, while 5 tumors arose from the sinonasal region. The prognosis of this series of salivary gland-type MECs was favorable. CONCLUSIONS: Our study confirmed that HCCC runs the risk of being misdiagnosed as MEC in the sinonasal tract and nasopharynx, particularly with biopsy specimens. Careful histological evaluation with supporting molecular testing can facilitate pathological diagnoses.


Assuntos
Carcinoma Mucoepidermoide , Carcinoma de Células Escamosas , Neoplasias das Glândulas Salivares , Humanos , Masculino , Feminino , Carcinoma Mucoepidermoide/diagnóstico , Carcinoma Mucoepidermoide/genética , Carcinoma Mucoepidermoide/patologia , Hibridização in Situ Fluorescente , Fatores de Transcrição/genética , Neoplasias das Glândulas Salivares/patologia
2.
Am J Surg Pathol ; 48(5): 588-595, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38357912

RESUMO

Nasal chondromesenchymal hamartoma (NCMH) is a rare benign polypoid mesenchymal tumor arising in the nasal cavity and/or paranasal sinuses. Recognizing these sporadic, rare lesions is crucial, as surgical complete removal of the mass is the common treatment approach. This retrospective study analyzed the demographics, symptoms, and imaging data of 9 patients diagnosed with NCMH between January 2017 and June 2023, possibly representing the largest single-center adult case cohort to date. Diagnostic techniques included nasal endoscopy, CT/MRI scan, immunohistological studies, and morphologic comparisons. Pathologic specimens were subjected to Sanger sequencing of exons 24 and 25 of DICER1. The average age of 9 cases was 24.4 years, and the oldest was 55 years. Four of the patients were children, ranging from 1 year old to 11 years old, with an average of 4.5 years. Nasal congestion is the most common registered symptom. Endoscopic findings showed that most patients had smooth pink neoplasms or polypoid masses in the nasal meatus. Radiologic scanning revealed soft-tissue density masses that occupied the nasal cavity. Histologically, the characteristic structure of NCMHs is immature cellular cartilage nodules and mature cartilage nodules distributed in a loose mucoid matrix. Five of the 9 patients had somatic DICER1 missense mutations. Four of the patients with DICER1-mutated NCMH exhibited a p.E1813 missense hotspot mutation. We also report a case of a rare p.P1836H missense mutation. The detected DICER1 somatic mutations provide compelling evidence of an association with the DICER1 tumor family. We emphasize the importance of pathologic consultation and the need for pathologists to accumulate experience in NCMH diagnosis to avoid misdiagnosis.


Assuntos
Hamartoma , Neoplasias de Tecido Conjuntivo e de Tecidos Moles , Doenças Nasais , Criança , Lactente , Adulto , Humanos , Adulto Jovem , Estudos Retrospectivos , Doenças Nasais/genética , Doenças Nasais/diagnóstico , Doenças Nasais/patologia , Cavidade Nasal/patologia , Hamartoma/genética , Hamartoma/patologia , Ribonuclease III/genética , Neoplasias de Tecido Conjuntivo e de Tecidos Moles/patologia , Mutação , RNA Helicases DEAD-box/genética
3.
Am J Surg Pathol ; 47(10): 1168-1175, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37377124

RESUMO

The aim of this study was to evaluate the clinicopathologic features, molecular characteristics, treatment strategy, and prognosis of nasopharyngeal hyalinizing clear cell carcinoma (HCCC). Retrospective observational case series. Institutional pathology records between 2006 and 2022 were searched for all cases of nasopharyngeal HCCC. We included 10 male and 16 female patients aged 30 to 82 years (median: 60.5 y, mean: 54.6 y). The most common symptoms were blood-stained rhinorrhea and nasal obstruction. Tumors most often involved the lateral wall of the nasopharynx, followed by the superior posterior wall. Microscopically, all tumor cells were arranged in sheets, nests, cords, and single cells in a hyaline/myxoid/fibrous stroma. The tumor cells were polygonal, with or without distinct cell borders, and displayed abundant clear-to-eosinophilic cytoplasm. All 26 cases were positive for pancytokeratin, CK7, p40, and p63 but negative for myoepithelial differentiation markers. Ki-67 labeling was low and ranged from 1% to 10%. All 26 cases demonstrated EWSR1 and EWSR1-ATF1 rearrangements, and no case demonstrated MAML2 rearrangement. Complete follow-up data were available for 23 patients: 14 patients underwent endoscopic surgery alone, 5 underwent radiation therapy followed by endoscopic surgery, 3 underwent radiation therapy followed by biopsy, and 1 underwent cisplatin chemotherapy before endoscopic surgery. Clinical follow-up ranged from 6 to 195 months; 13 patients (56.5%) were alive without tumor, 5 patients (21.7%) died of disease, 5 patients (21.7%) survived with tumor. HCCCs of the nasopharynx are rare tumors. The definitive diagnosis depends on histopathology, immunohistochemistry, and molecular studies. The optimal treatment for patients with nasopharyngeal HCCC is wide local excision. Radiation and chemotherapy might be good options for managing locally advanced cases. Nasopharyngeal HCCC is less indolent than previously thought. Tumor stage and the choice of treatment are key factors affecting the prognosis of nasopharyngeal HCCC patients.


Assuntos
Adenocarcinoma de Células Claras , Carcinoma , Neoplasias das Glândulas Salivares , Humanos , Masculino , Feminino , Estudos Retrospectivos , Nasofaringe/química , Nasofaringe/patologia , Neoplasias das Glândulas Salivares/patologia , Fatores de Transcrição , Carcinoma/patologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/análise , Adenocarcinoma de Células Claras/patologia
4.
Laryngoscope ; 131(10): E2662-E2668, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33847383

RESUMO

OBJECTIVE: Chromogranin (CHG), synaptophysin (Syn), and CD56 are generally used in a panel to support diagnoses of laryngeal neuroendocrine carcinomas (NECs). However, the absence of expression of these markers does not completely exclude the diagnosis. INSM1 is a novel marker that is considered sufficiently sensitive and specific for NE differentiation. The aim of this study is not only to detect its sensitivity and specificity, but also to evaluate its application in grading for laryngeal NECs. METHODS: The clinicopathological characteristics of the 25 cases with laryngeal NECs were retrospectively analyzed. The expressions of INSM1, CHG, Syn, and CD56 were detected by immunohistochemistry. RESULTS: Of the 25 laryngeal NECs, INSM1 had higher sensitivity (92%) than Syn (84%), CHG (76%) and CD56 (76%). The average H scores of INSM1, CD56, Syn, and CHG were 160, 37.5, 300, 300 for well-differentiated neuroendocrine carcinoma (WD-NEC); 190, 149, 209, 215 for moderately differentiated neuroendocrine carcinoma (MD-NEC); 251, 208, 104, 25 for poorly differentiated neuroendocrine carcinoma with small cell (SCNEC); 109, 160, 98, 26 for large cell types (LCNEC), respectively. Of these 98 non-neuroendocrine tumors, INSM1 expression was seen in nine (9%) tumors, all were squamous cell carcinoma. And INSM1 staining was generally focal. CONCLUSION: INSM1 has high sensitivity and specificity in diagnosis of laryngeal NECs. For grading laryngeal NECs, Syn and CHG showed significant advantages in the diagnosis of WD-NEC and MD-NEC, whereas INSM1 and CD56 showed greater diagnostic value in the diagnosis of SCNEC and LCNEC, especially in SCNEC. LEVEL OF EVIDENCE: 4 Laryngoscope, 131:E2662-E2668, 2021.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias Laríngeas/patologia , Tumores Neuroendócrinos/patologia , Proteínas Repressoras/análise , Idoso , Antígeno CD56/análise , Cromograninas/análise , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Estudos Retrospectivos , Sensibilidade e Especificidade , Sinaptofisina/análise
5.
Cancer Biol Med ; 17(4): 953-969, 2020 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-33299646

RESUMO

Objective: Pancreatic ductal adenocarcinoma (PDAC) is a disease with high mortality. Many so-called "junk" noncoding RNAs need to be discovered in PDAC. The purpose of this study was therefore to investigate the function and regulatory mechanism of the long noncoding RNA MEG3 in PDAC. Methods: The Gene Expression Omnibus database (GEO database) was used to determine the differential expression of long noncoding RNAs in PDAC, and MEG3 was selected for subsequent verification. Tissue and cell samples were used to verify MEG3 expression, followed by functional detection in vitro and in vivo. Microarrays were used to characterize long noncoding RNA and mRNA expression profiles. Competing endogenous RNA analyses were used to detect differential MEG3 and relational miRNA expression in PDAC. Finally, promoter analyses were conducted to explain the downregulation of MEG3 PDAC. Results: We generated a catalogue of PDAC-associated long noncoding RNAs in the GEO database. The ectopic expression of MEG3 inhibited PDAC growth and metastasis in vitro and in vivo, which was statistically significant (P < 0.05). Microarray analysis showed that multiple microRNAs interacted with MEG3. We also showed that MEG3, as a competing endogenous RNA, directly sponged miR-374a-5p to regulate PTEN expression. The transcription factor, Sp1, recruited EZH2 and HDAC3 to the promoter and transcriptionally repressed MEG3 expression. Finally, clinical data showed that MEG3 and miR-374a-5p expressions were correlated with clinicopathological features. Statistically, Sp1, EZH2, HDAC3, and miR-374a-5p were negatively correlated with MEG3 (P < 0.05). Conclusions: Reduced MEG3 levels played a crucial role in the PDAC malignant phenotype, which provided insight into novel and effective molecular targets of MEG3 for pancreatic cancer treatment.


Assuntos
Carcinoma Ductal Pancreático/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Histona Desacetilases/genética , Neoplasias Pancreáticas/genética , RNA Longo não Codificante/genética , Animais , Carcinoma Ductal Pancreático/patologia , Proliferação de Células , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Neoplasias Pancreáticas/patologia , Prognóstico , Regiões Promotoras Genéticas , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Cancer Cell Int ; 20: 55, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32099528

RESUMO

BACKGROUND: The bromodomain and extra-terminal domain (BET) family of proteins, especially BRD4 play an important role in epigenetic regulation, and are essential for cell survival and also are promising anticancer targets. This study aims to analyze the effect of BRD4 on the cell growth and progression of pancreatic cancer and novel mechanisms involved. METHODS: Expression of BRD4 in pancreatic cancer and paired adjacent noncancerous tissues from 76 patients was analyzed by western blotting, immunohistochemistry, and real time PCR. Its correlation with the clinicopathological characteristics and prognosis of pancreatic cancer patients was analyzed. The effects of BRD4 on the cell proliferation were detected by colony formation assay and sulforhodamine B assay. Migration and invasion were determined by Transwell assays, and the effect of BRD4 on subcutaneous tumor formation was verified in nude mice. Cell cycle analysis was detected by flow cytometry. The potential downstream targets of BRD4 and related molecular mechanisms were clarified by RNA sequencing, chromatin immunoprecipitation and dual luciferase reporter assay. RESULTS: BRD4 was overexpressed in pancreatic cancer. Biological results showed that BRD4 functioned as tumor promoter, facilitated cell proliferation, migration and invasion in vitro and in vivo. Further, caveolin-2 was selected as the downstream gene of BRD4 by RNA sequencing. Caveolin-2 overexpression can partially reverse the decreased cell growth ability caused by BRD4 knockdown, but did not affect cell migration and invasion. Chromatin immunoprecipitation assay and dual luciferase reporter assay revealed BRD4 could bind to the promoter region of caveolin-2 and upregulate caveolin-2 expression. Clinical data further indicated a positive correlation between BRD4 and caveolin-2 expression. BRD4 (high)/caveolin-2 (high) correlated with shorter overall survival of patients with pancreatic cancer. Multivariate analysis revealed that both BRD4 and caveolin-2 were independent factors. CONCLUSIONS: Our findings reveal the oncogenic effects of BRD4 in pancreatic cancer and elucidate a possible mechanism by which BRD4 and caveolin-2 act to enhance cell growth. Targeting the BRD4-caveolin-2 interaction by development of BET inhibitors will be a therapeutic strategy for pancreatic cancer.

7.
Front Oncol ; 9: 588, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31312615

RESUMO

Background: NLRP3 inflammasome is an inflammatory mediator. The expression of NLRP3 inflammasome is associated with the development of various tumors and is closely related to the prognosis of tumors. However, the role of NLRP3 inflammasome in laryngeal squamous cell carcinoma (LSCC) remains unclear. This study aim to investigate the influence of NLPR3 inflammasome expression in LSCC, and especially the NLRP3 inflammasome expression level and the prognosis of LSCC after surgery in a Chinese population. Methods: We used quantitative real-time PCR and immunohistochemical (IHC) staining to calculate the mRNA (20 patients, fresh tissue) and protein expression (104 patients, paraffin tissue microarray) levels of the NLRP3 inflammasome (NLRP3/IL-18/IL-1ß/ASC/caspase-1), respectively. We also analyzed the relationship between NLRP3 inflammasome expression levels and LSCC cancer tissues compared with adjacent normal tissues and the clinical features of LSCC. Kaplan-Meier survival curves of overall survival (OS) and disease-free survival (DFS) in LSCC patients were compared and analyzed under different expression levels of the NLRP3 inflammasome. Results: Our results indicated that the mRNA expression of the NLRP3 inflammasome was higher in LSCC cancer tissues compared with adjacent normal tissues (p < 0.001). The IHC staining score also demonstrated that the expression of the NLRP3 inflammasome was higher than in the adjacent normal tissues (p < 0.001). The NLRP3 inflammasome expression also exhibited a close relationship with the clinicopathological characteristics (especially the stage of LSCC) of LSCC. Univariate Cox regression analysis and multivariate Cox regression analysis revealed that both NLRP3 and IL-1ß had an increased risk of LSCC progression (p < 0.05). The Kaplan-Meier log rank test (OS and DFS) demonstrated that high expression of NLRP3/IL-18/IL-1ß/ASC was statistically different than the low expression group (p < 0.05) of LSCC patients after surgery. Conclusion: The high expression group of the NLRP3 inflammasome (NLRP3/IL-18/IL-1ß/ASC) had a poorer prognosis (OS and DFS) than the low expression group of LSCC patients 5 years after surgery. The NLRP3 inflammasome (NLRP3/IL-18/IL-1ß/ASC) may be used as an auxiliary indicator to predict LSCC patient prognosis after surgery.

8.
Am J Transl Res ; 11(2): 1030-1039, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30899402

RESUMO

Vasculogenic mimicry (VM) is an alternative type of blood and nutrition supply that is associated with more aggressive tumor biology and increased cancer-related mortality. However, the clinical implications of VM remain unclear in patients with pancreatic ductal adenocarcinoma (PDAC). The aim of this study was to investigate the clinical significance of VM in PDAC patients and to seek a novel and more efficient treatment strategy by targeting this unique process. Here, cluster of differentiation 34 (CD34)/periodic acid-Schiff (PAS) double-staining of 76 PDAC clinical specimens revealed that VM expression was related to clinical stage (P=0.049) and lymph node metastasis (P=0.023). Notably, VM expression was correlated with a poor prognosis in patients with PDAC. Additionally, we discovered that there was a positive correlation between the expressions of VM and phosphorylated extracellular signal regulated kinase (p-ERK1/2) in 76 clinical samples (P<0.001). Moreover, our results further indicated that treatment with the ERK1/2 inhibitor SCH772984 effectively blocked VM formation by repressing the production of p-ERK1/2-MMP-2/9, which have been established as classical markers of VM. Further, JQ1, a bromodomain and extraterminal domain (BET) inhibitor, also exerted significant inhibitory efficiency against VM formation by decreasing the activation of ERK1/2-MMP-2/9. In conclusion, our work suggests that VM is a marker of poor prognosis in patients with PDAC and that JQ1 can inhibit VM formation via the ERK1/2-MMP-2/9 signaling pathway.

9.
BMC Cancer ; 18(1): 1032, 2018 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-30352575

RESUMO

BACKGROUND: It was demonstrated that long non-coding RNAs occupied an important position in tumor pathogenesis and progression. We have previously found that the metastasis-associated lung adenocarcinoma transcript 1 (MALAT-1) promotes cell proliferation and metastases in pancreatic ductal adenocarcinoma (PDAC). The present study was aimed to discuss the underlying mechanisms. METHODS: Bioinformatics method was used to identify the miRNA target of MALAT-1. Expressions of relative genes were assessed by quantitative real-time PCR and western blotting, respectively. Sulforhodamine B assay and Transwell assay were employed to detect cell proliferation, migration and invasion, respectively. Moreover, RNA immunoprecipitation was performed to determine whether RNA-induced silencing complex contained MALAT-1 and its potential binding miRNA. Luciferase assays was used to confirm potential binding site. RESULTS: Bioinformatics search predicted that miR-200c-3p was a direct target of MALAT-1. Further, we found a reciprocal suppression between MALAT-1 and miR-200c-3p expression. In terms of mechanisms, high MALAT-1 and low miR-200c-3p may form a novel feedback loop. On the one hand, MALAT-1 functioned as a competing endogenous RNA to suppress miR-200c-3p expression, leading to upregulation of ZEB1 expression. On the other hand, miR-200c-3p inhibited the level of MALAT-1 expression was in a way similar to miRNA-mediated downregulation of target genes. Clinical data further indicated that MALAT-1 and ZEB1 expression was negatively correlated with miR-200c-3p transcript level of PDAC tissues. There was a positive correlation between MALAT-1 and ZEB1 level. MALAT-1 (high)/miR-200c-3p (low) correlated with shorter overall survival of PDAC patients. Multivariate analysis revealed that both MALAT-1 and miR-200c-3p levels were independent prognostic factors. CONCLUSION: Our findings firstly revealed a novel feedback loop between high MALAT-1 and low miR-200c-3p. Targeting the feedback loop between high MALAT-1 and low miR-200c-3p will be a therapeutic strategy for PDAC.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , MicroRNAs/biossíntese , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , RNA Longo não Codificante/biossíntese , Biomarcadores Tumorais , Carcinoma Ductal Pancreático/genética , Linhagem Celular , Movimento Celular/genética , Proliferação de Células/genética , Humanos , MicroRNAs/genética , Neoplasias Pancreáticas/genética , Prognóstico , RNA Longo não Codificante/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/biossíntese
10.
Sci Rep ; 7(1): 470, 2017 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-28352075

RESUMO

Cyclooxygenase-2 (COX-2) was stated to be overexpression in various human malignancies associating with angiogenesis, metastasis and chemoresistence. Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease displaying many of these characteristics. A common abnormality of PDAC is overexpression of specificity protein-1 (Sp1), which was said to correlate with malignant phenotypes of human cancers. Using RNA-seq data from The Cancer Genome Atlas (TCGA), we found that Sp1 expression was positively correlated with that of COX-2 in PDAC, and that the inhibition or overexpression of Sp1 in PDAC cells leads to decreased or elevated COX-2 expression. Luciferase reporter gene and chromatin immunoprecipitation (ChIP) assays revealed that elevated transcription of COX-2 requires Sp1 binding to sequence positions around -245/-240 of COX-2 promoter. Activated epidermal growth factor receptor (EGFR) and downstream p38 mitogen-activated protein kinase (p38-MAPK) were also profoundly altered in PDAC. The inhibition of EGFR/p38-MAPK signaling resulted in reduced Sp1 activation, decreased COX-2 and vascular endothelial growth factor (VEGF) expression. Thus, Sp1 could transcriptionally activate COX-2 expression in a process relies on activated EGFR/p38-MAPK signaling. Finally, we found that the inhibition of COX-2 leads to decreased angiogenesis in a process dependent on VEGF, which link COX-2 to angiogenesis in PDAC.


Assuntos
Ciclo-Oxigenase 2/genética , Regulação Neoplásica da Expressão Gênica , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Transdução de Sinais , Fator de Transcrição Sp1/metabolismo , Biomarcadores , Linhagem Celular Tumoral , Biologia Computacional/métodos , Ciclo-Oxigenase 2/metabolismo , Receptores ErbB/metabolismo , Genes Reporter , Humanos , Modelos Biológicos , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/patologia , Ativação Transcricional , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
Oncotarget ; 7(10): 11194-207, 2016 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-26848980

RESUMO

Enhancer of zeste homolog 2 (EZH2) is an essential component of the polycomb repressive complex 2 (PRC2), which is required for epigenetic silencing of target genes, including those affecting cancer progression. Its role in pancreatic cancer remains to be clarified; therefore, we investigated the effects of aberrantly expressed EZH2 on pancreatic cancer. We found that EZH2 expression is up-regulated in pancreatic cancer tissues and positively correlated with lymph node metastasis and advanced clinical stage in pancreatic cancer patients. EZH2 knockdown in pancreatic cancer cell lines inhibited cell migration and invasion, but did not alter cell proliferation. Silencing of EZH2 also increased E-cadherin expression in vitro, and E-cadherin expression was inversely correlated with EZH2 expression in pancreatic cancer tissue samples. Patients with high EZH2 and low E-cadherin expression had the worst prognosis. RIP and ChIP assays suggest that EZH2 is recruited to the E-cadherin promoter by the long non-coding RNA, MALAT-1 (metastasis associated in lung adenocarcinoma transcript 1), where it represses E-cadherin expression. Our results show that EZH2-based therapies may be an option for the treatment of pancreatic cancer.


Assuntos
Caderinas/metabolismo , Movimento Celular , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias Pancreáticas/patologia , RNA Longo não Codificante/metabolismo , Adulto , Idoso , Antígenos CD , Proliferação de Células , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/patologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/mortalidade
12.
Oncotarget ; 7(13): 16505-16, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26918727

RESUMO

Previous studies showed that aberrant CDH1 or/and HDAC3 localization is essential for the progression of some human cancers. Here, we investigate the prognostic significance of aberrant CDH1 and HDAC3 localization in 84 pancreatic cancer patients. Our results show that increases in both membrane and cytoplasmic CDH1 correlate with lymph node metastasis (P = 0.026 and P < 0.001, respectively) and clinical stage (P = 0.020 and P < 0.001, respectively). Increased nuclear HDAC3 correlates with lymph node metastasis (P < 0.001) and advanced clinical stage (P < 0.001), but increased cytoplasmic HDAC3 does not (P > 0.05). Multivariate analysis showed that nuclear HDAC3 and cytoplasmic CDH1 (P = 0.001 and P = 0.010, respectively), as well as tumor differentiation (P = 0.009) are independent prognostic factors. Most importantly, patients with high co-expression of nuclear HDAC3 and cytoplasmic CDH1 had shorter survival times (P < 0.001), more frequent lymph node metastasis (P < 0.001), and advanced clinical stage (P < 0.001). Our studies provide convincing evidence that nuclear HDAC3 and cytoplasmic CDH1 have independent prognostic value in pancreatic cancer and provide novel targets for prognostic therapeutics.


Assuntos
Biomarcadores Tumorais/metabolismo , Caderinas/metabolismo , Histona Desacetilases/metabolismo , Neoplasias Pancreáticas/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos CD , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Progressão da Doença , Feminino , Humanos , Imuno-Histoquímica , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Estadiamento de Neoplasias , Neoplasias Pancreáticas/patologia , Prognóstico , Análise de Sobrevida
13.
Int J Clin Exp Pathol ; 8(9): 11092-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26617828

RESUMO

Jumonji domain-containing protein 1A (JMJD1A) play a key role in the development and progression of several malignancies. The present study investigated the expression and clinical significance of JMJD1A in gastric cancer. JMJD1A was found to be upregulated in gastric cancer tissues and cell lines. Furthermore, the upregulated expression of JMJD1A was significantly correlated with the results of the invasion depth (P=0.006), lymph node metastatic status (P<0.001), and TNM stage (P<0.001). JMJD1A was also shown to be an independent prognostic predictor of overall survival (HR3.988; 95% CI 1.948-8.167; P<0.001) for patients with gastric cancer. In addition, in vitro experiment revealed that knockdown of JMJD1A expression inhibited the gastric cancer cell proliferation, and further study suggested that JMJD1A knockdown suppressed MAPK pathway via transcriptional downregulation the expression of long noncoding RNA MALAT1. Therefore, we speculated that JMJD1A-MALAT1-MAPK signaling might participate in the JMJD1A-induced cell proliferation of gastric cancer. Collectively, our data demonstrate for the first time that JMJD1A gene has an important regulatory role in gastric carcinogenesis, and could function as a novel prognostic indicator and a potential therapeutic target for gastric cancer.


Assuntos
Biomarcadores Tumorais/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Neoplasias Gástricas/enzimologia , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Gastrectomia , Regulação Neoplásica da Expressão Gênica , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Estimativa de Kaplan-Meier , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Terapia de Alvo Molecular , Invasividade Neoplásica , Estadiamento de Neoplasias , Modelos de Riscos Proporcionais , Interferência de RNA , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Transdução de Sinais , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidade , Neoplasias Gástricas/patologia , Neoplasias Gástricas/terapia , Fatores de Tempo , Transfecção , Regulação para Cima
14.
Int J Mol Sci ; 16(4): 6677-93, 2015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25811929

RESUMO

Cancer stem cells (CSCs) play a vital role in tumor initiation, progression, metastasis, chemoresistance, and recurrence. The mechanisms that maintain the stemness of these cells remain largely unknown. Our previous study indicated that MALAT-1 may serve as an oncogenic long noncoding RNA in pancreatic cancer by promoting epithelial-mesenchymal transition (EMT) and regulating CSCs markers expression. More significantly, there is emerging evidence that the EMT process may give rise to CSCs, or at least cells with stem cell-like properties. Therefore, we hypothesized that MALAT-1 might enhance stem cell-like phenotypes in pancreatic cancer cells. In this study, our data showed that MALAT-1 could increase the proportion of pancreatic CSCs, maintain self-renewing capacity, decrease the chemosensitivity to anticancer drugs, and accelerate tumor angiogenesis in vitro. In addition, subcutaneous nude mouse xenografts revealed that MALAT-1 could promote tumorigenicity of pancreatic cancer cells in vivo. The underlying mechanisms may involve in increased expression of self-renewal related factors Sox2. Collectively, we for the first time found the potential effects of MALAT-1 on the stem cell-like phenotypes in pancreatic cancer cells, suggesting a novel role of MALAT-1 in tumor stemness, which remains to be fully elucidated.


Assuntos
Células-Tronco Neoplásicas/patologia , Neoplasias Pancreáticas/patologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Transplante de Neoplasias , Células-Tronco Neoplásicas/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Regulação para Cima
15.
Oncol Rep ; 32(6): 2485-92, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25269958

RESUMO

Pancreatic cancer is one of the most aggressive solid malignancies with a dismal survival rate. Recent studies have shown that high expression levels of long noncoding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript-1 (MALAT-1) correlate with several solid tumors. However, the underlying molecular mechanisms and its clinical significance in pancreatic cancer remain to be elucidated. In the present study, our results showed that MALAT-1 expression levels were upregulated in pancreatic cancer tissues compared with adjacent noncancerous controls. Consistently, higher expression level of MALAT-1 was found in all seven pancreatic cancer cell lines relative to the human pancreatic ductal epithelial cell. Further function analysis revealed that downregulation of MALAT-1 could inhibit tumor cell proliferation and decrease cell migration and invasion in vitro. The underlying mechanisms are possibly involved in inducing G2/M cell cycle arrest, promoting cell apoptosis, suppressing epithelial-mesenchymal transition and reducing cancer stem-like properties. In conclusion, this study indicated that MALAT-1 may serve as an oncogenic lncRNA that is involved in malignancy phenotypes of pancreatic cancer. Therefore, it may be used as a potential therapeutic target.


Assuntos
Movimento Celular , Proliferação de Células , Neoplasias Pancreáticas/metabolismo , RNA Longo não Codificante/metabolismo , Apoptose , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Pontos de Checagem da Fase G2 do Ciclo Celular , Expressão Gênica , Humanos , Invasividade Neoplásica , Células-Tronco Neoplásicas/metabolismo , Neoplasias Pancreáticas/patologia , RNA Longo não Codificante/genética , Regulação para Cima
16.
Int J Oncol ; 45(4): 1523-30, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25070540

RESUMO

Pancreatic cancer is one of the most aggressive solid malignancies with a dismal survival rate. Recent studies have shown that high expression levels of histone deacetylase 3 (HDAC3) correlate with malignant phenotype. However, the expression patterns and biological role of HDAC3 in pancreatic cancer remain unclear. In this study, our data showed that a higher level of HDAC3 protein expression was found in pancreatic cancer as compared to paired paracancerous tissues. Consistently, higher expression level of HDAC3 was found in all of the eight pancreatic cancer cell lines relative to human pancreatic ductal epithelial cells (HPDE). In addition, further function analysis revealed that HDAC3 can function as oncogenic protein, which could promote pancreatic cancer cell proliferation, migration and invasion, and may increase drug resistance. Moreover, the functional involvement of HDAC3 was partially correlated with post-induction repression of P53, P27 and Bax gene transcription, acting via H3K9 deacetylation. Taken together, our data suggest that HDAC3 participates in the pathogenesis and progression of pancreatic cancer through histone modification, which might be a pivotal epigenetic target against this devastating disease.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Histona Desacetilases/metabolismo , Histonas/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Regulação Neoplásica da Expressão Gênica , Histona Desacetilases/genética , Humanos , Invasividade Neoplásica , Neoplasias Pancreáticas/genética , Regiões Promotoras Genéticas , Complexo de Endopeptidases do Proteassoma/genética , Proteína Supressora de Tumor p53/genética , Proteína X Associada a bcl-2/genética , Gencitabina
17.
Nan Fang Yi Ke Da Xue Xue Bao ; 31(3): 419-22, 2011 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-21421474

RESUMO

OBJECTIVE: To establish a nasopharyngeal carcinoma (NPC) cell line CNE1-pLVTHM/BART7 with stable ebv-miR-BART7 overexpression. METHODS: The recombinant lentivirus pLVTHM/BART7 expression plasmid was packaged into mature lentivirus by 293FT cells and used to infect CNE1 cells. Flow cytometry was employed for sorting the GFP(+) cells. The efficiency of ebv-miR-BART7 overexpression was determined using qRT-PCR. RESULTS: The recombinant lentivirus plasmid pLVTHM/BART7 was successfully constructed and verified by PCR and sequencing. The expression of ebv-miR-BART7 in CNE1 cells infected with the lentivirus pLVTHM/BART7 was significantly increased as compared with the negative control and the blank control cells. CONCLUSION: The recombinant lentivirus vector pLVTHM/BART7 results in high and stable expression of ebv-miR-BART7 in infected CNE1 cells, which provides a useful cell model for further studies of the role of ebv-miR-BART7 in nasopharyngeal carcinoma.


Assuntos
Linhagem Celular Tumoral , Vetores Genéticos , Lentivirus/genética , MicroRNAs , Neoplasias Nasofaríngeas/genética , Carcinoma , Humanos , Carcinoma Nasofaríngeo , Plasmídeos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...