Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncol Rep ; 34(2): 833-42, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26081937

RESUMO

We investigated the clinical significance of podoplanin expression in relation to clinicopathological variables in head and neck squamous cell carcinoma (HNSCC), to determine its effectiveness as a marker for high-risk HNSCC patients. Upregulation of podoplanin in HNSCC tissues was examined using immunohistochemistry and clinicopathological analyses. Wound healing and invasion assays were performed using HNSCC cells that were transfected with podoplanin siRNA, podoplanin vector and cotransfection with both the podoplanin vector and VEGF-C siRNA. High podoplanin expression was significantly associated with ~3- and 5-fold increases in the presence of positive lymph node metastasis and poor histological grade, respectively (P<0.05). High levels of podoplanin expression were significantly associated with decreased overall and disease-specific survival rates (P<0.05). Furthermore, upregulation of podoplanin induced cell wound repair activity and invasiveness in the FaDu and SNU-1041 cells, respectively, while downregulation of podoplanin expression through VEGF-C silencing using co-transfection of both the podoplanin vector and VEGF-C siRNA suppressed cell wound healing and invasion abilities in the HNSCC cells. Our findings suggest that high podoplanin expression is associated with aggressive tumor behavior, poor prognosis and metastatic regulation through interaction with VEGF-C, suggesting that podoplanin may be used as a potential prognostic biomarker for HNSCC.


Assuntos
Biomarcadores Tumorais/biossíntese , Carcinoma de Células Escamosas/genética , Neoplasias de Cabeça e Pescoço/genética , Glicoproteínas de Membrana/biossíntese , Fator C de Crescimento do Endotélio Vascular/genética , Adulto , Idoso , Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/patologia , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Metástase Linfática , Masculino , Glicoproteínas de Membrana/genética , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Prognóstico , Fator C de Crescimento do Endotélio Vascular/biossíntese , Cicatrização
2.
Med Oncol ; 31(11): 236, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25267569

RESUMO

Smad3 functions as an integrator of diverse signaling, including transforming growth factor ß signaling and the function of Smad3 is complexly regulated by differential phosphorylation at various sites of Smad3. Despite the importance of Smad3 and its various phosphoisoforms, their prognostic significance has rarely been studied. In this study, we demonstrated the prognostic significance of Smad3, its phosphoisoforms, and Smad4 expression by immunohistochemistry in 126 esophageal squamous cell carcinomas. The phosphoisoforms of Smad3 studied in this article included phosphorylation at C-terminal (pSmad3C)(Ser(423/425)) and phosphorylation at the linker region (pSmad3L)(Ser(213)). High expression of Smad3 was associated with shorter overall survival. Co-existence of high expression of pSmad3L(S213) and low expression of pSmad3C(S423/425) were associated with advanced N stage and an independent prognostic factor for overall [hazard ratio (HR) 2.03, 95 % confidence interval (CI) (1.10-3.75), p = 0.023] and disease-free survival [HR 2.41, 95 % CI (1.32-4.39), p = 0.004]. In conclusion, co-existence of high pSmad3L(Ser(213)) expression and low pSmad3C(Ser(423/425)) expression can be considered as immunohistochemical biomarkers for predicting prognosis as well as future therapeutic targets. In addition, our results of combinatory effect of differential phosphorylation of Smad3 on prognosis suggest the mode of action of Smad3 might be logically determined by its phosphorylation pattern.


Assuntos
Biomarcadores Tumorais/biossíntese , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteína Smad3/biossíntese , Proteína Smad4/biossíntese , Idoso , Carcinoma de Células Escamosas/mortalidade , Neoplasias Esofágicas/mortalidade , Carcinoma de Células Escamosas do Esôfago , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Isoformas de Proteínas/biossíntese , Taxa de Sobrevida/tendências
3.
Stem Cells ; 32(4): 998-1011, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24715692

RESUMO

Redox balance has been suggested as an important determinant of "stemness" in embryonic stem cells (ESCs). In this study, we demonstrate that peroxiredoxin (Prx) plays a pivotal role in maintenance of ESC stemness during neurogenesis through suppression of reactive oxygen species (ROS)-sensitive signaling. During neurogenesis, Prx I and Oct4 are expressed in a mutually dependent manner and their expression is abruptly downregulated by an excess of ROS. Thus, in Prx I(-/-) or Prx II(-/-) ESCs, rapid loss of stemness can occur due to spontaneous ROS overload, leading to their active commitment into neurons; however, stemness is restored by the addition of an antioxidant or an inhibitor of c-Jun N-terminal kinase (JNK). In addition, Prx I and Prx II appear to have a tight association with the mechanism underlying the protection of ESC stemness in developing teratomas. These results suggest that Prx functions as a protector of ESC stemness by opposing ROS/JNK cascades during neurogenesis. Therefore, our findings have important implications for understanding of maintenance of ESC stemness through involvement of antioxidant enzymes and may lead to development of an alternative stem cell-based therapeutic strategy for production of high-quality neurons in large quantity.


Assuntos
Células-Tronco Embrionárias/enzimologia , MAP Quinase Quinase 4/metabolismo , Neurogênese/fisiologia , Peroxirredoxinas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Células-Tronco Embrionárias/citologia , MAP Quinase Quinase 4/genética , Camundongos , Camundongos Knockout , Peroxirredoxinas/genética
4.
Nat Commun ; 4: 2062, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23797602

RESUMO

Constitutive NF-κB activation in cancer cells is caused by defects in the signalling network responsible for terminating the NF-κB response. Here we report that plant homeodomain finger protein 20 (PHF20) maintains NF-κB in an active state in the nucleus by inhibiting the interaction between PP2A and p65. We show that PHF20 induces canonical NF-κB signalling by increasing the DNA-binding activity of NF-κB subunit p65. In PHF20 overexpressing cells, the termination of tumour necrosis factor-induced p65 phosphorylation is impaired whereas upstream signalling events triggered by tumour necrosis factor are unaffected. This effect strictly depends on the interaction between PHF20 and methylated lysine residues of p65, which hinders recruitment of PP2A to p65, thereby maintaining p65 in a phosphorylated state. We further show that PHF20 levels correlate with p65 phosphorylation levels in human glioma specimens. Our work identifies PHF20 as a novel regulator of NF-κB activation and suggests that elevated expression of PHF20 may drive constitutive NF-κB activation in some cancers.


Assuntos
Antígenos de Neoplasias/metabolismo , Biomarcadores Tumorais/metabolismo , Proteína Fosfatase 2/metabolismo , Transdução de Sinais , Fator de Transcrição RelA/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Proteínas de Ligação a DNA , Glioma/metabolismo , Glioma/patologia , Células HEK293 , Células HeLa , Humanos , Imuno-Histoquímica , Lisina/metabolismo , Metilação/efeitos dos fármacos , Modelos Biológicos , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Fatores de Transcrição , Fator de Necrose Tumoral alfa/farmacologia
5.
Histol Histopathol ; 28(11): 1483-90, 2013 11.
Artigo em Inglês | MEDLINE | ID: mdl-23673988

RESUMO

Yes-associated protein (YAP) is a transcriptional co-activator and functions as a nuclear downstream effector of the Hippo pathway. Differential expression of YAP and phosphorylated Yes-associated protein (pYAP), which are involved in the expression of Ki-67 and phosphorylated extracellular signal-regulated kinase (pERK) in colorectal adenocarcinoma (CRAC), is not clear. Herein, we hypothesized that nuclear expression of YAP could predict cell proliferation and poor prognosis, while cytoplasmic expression of pYAP would show a reverse correlation with cell proliferation. Paraffin-embedded samples from 144 CRAC patients were studied using immunohistochemistry for YAP, pYAP, Ki-67 and pERK. Frozen samples from 20 CRAC patients were examined for YAP mRNA in tumor and non-tumor tissues, using quantitative real-time PCR. High nuclear YAP expression coincided with high Ki-67 expression (P=0.002). The high nuclear YAP expression group tended to display a poor overall and disease-free survival (P=0.089 and P=0.089, respectively), but YAP mRNA levels in the 20 CRAC tissues were not significantly different in comparison with the 20 non-tumor tissues (P=0.929). We observed an inverse correlation between high cytoplasmic pYAP expression and high Ki-67 expression (P=0.001). Nuclear pERK expression was positively correlated with nuclear YAP expression, but negatively correlated with cytoplasmic pYAP expression (P=0.017 and P=0.020, respectively). Activated nuclear YAP and inactivated cytoplasmic pYAP in CRAC showed a positive correlation with Ki-67 and nuclear pERK expression, suggesting that the expression of YAP and pYAP is a possible predictor of tumor cell proliferation and prognosis in CRAC.


Assuntos
Adenocarcinoma/metabolismo , Neoplasias Colorretais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Antígeno Ki-67/biossíntese , Proteínas Nucleares/biossíntese , Fatores de Transcrição/biossíntese , Adenocarcinoma/mortalidade , Adenocarcinoma/patologia , Biomarcadores Tumorais/análise , Proteínas de Ciclo Celular , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Fosforilação , Prognóstico , Modelos de Riscos Proporcionais , Reação em Cadeia da Polimerase em Tempo Real
6.
Biochem Biophys Res Commun ; 434(4): 861-6, 2013 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-23611781

RESUMO

The testes of most mammals are sensitive to temperature. To survive and adapt under conditions that promote endoplasmic reticulum (ER) stress such as heat shock, cells have a self-protective mechanism against ER stress that has been termed the "Unfolded Protein Response" (UPR). However, the cellular and molecular events underlying spermatogenesis with testicular hyperthermia involved in the UPR signaling pathway under ER stress remain poorly understood. In the present study, we verified that UPR signaling via phospho-eIF2α/ATF4/GADD34, p90ATF6, and phospho-IRE1α/XBP-1 is activated with testicular hyperthermia (43 °C, 15 min/day) and induced ER stress-mediated apoptosis associated with CHOP, phospho-JNK, and caspase-3 after repetitive periods of hyperthermia. Levels of phospho-eIF2α protein of mouse spermatocytes in the testis were rapidly increased by one cycle of testicular hyperthermia. ATF4/GADD34 and p90ATF6 expression gradually increased and decreased, respectively, with repetitive cycles of hyperthermia. Spliced XBP1 mRNA as a marker of IRE1 activity was increased after one, three cycles of hyperthermia and decreased by five cycles of hyperthermia. Although the levels of anti-apoptotic phospho-JNK (p54) were gradually decreased after three cycles of hyperthermia, CHOP expression was rapidly increased. After five cycles of testicular hyperthermia, the levels of cleaved caspase-3 and TUNEL-positive apoptotic spermatocytes cells were significantly increased. Our data demonstrated that testicular hyperthermia induces UPR signaling and repetitive cycles of hyperthermia lead to apoptosis of spermatocytes in mouse testis. These results suggest a link between the UPR signaling pathway and testicular hyperthermia.


Assuntos
Temperatura Alta , Transdução de Sinais/fisiologia , Espermatócitos/metabolismo , Testículo/metabolismo , Resposta a Proteínas não Dobradas/fisiologia , Fator 4 Ativador da Transcrição/metabolismo , Fator 6 Ativador da Transcrição/metabolismo , Processamento Alternativo , Animais , Apoptose/genética , Apoptose/fisiologia , Western Blotting , Caspase 3/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Estresse do Retículo Endoplasmático/genética , Estresse do Retículo Endoplasmático/fisiologia , Endorribonucleases/metabolismo , Fator de Iniciação 2 em Eucariotos/metabolismo , Imuno-Histoquímica , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Fosfatase 1/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição de Fator Regulador X , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Fator de Transcrição CHOP/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Resposta a Proteínas não Dobradas/genética , Proteína 1 de Ligação a X-Box
7.
Oncol Lett ; 5(3): 813-818, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23426273

RESUMO

Gallbladder carcinoma (GBC) is a lethal neoplasm, and new prognostic markers are required. Deregulation of E3 ligases contributes to cancer development and is associated with poor prognosis. Carboxyl terminus of heat shock protein 70-interacting protein (CHIP) is a U-box-type E3 ubiquitin ligase, the role of which has not been evaluated in GBC. Therefore, the present study investigated CHIP expression in GBC and its prognostic significance. In the present study, CHIP expression was measured in 78 tumor specimens of GBC by immunohistochemistry and the correlation between CHIP expression and clinicopathological factors was analyzed. Of the tumor specimens, 26.9% showed high staining intensity [the CHIP high expression group (HEG)]. The CHIP-HEG was not associated with other common clinicopathological parameters, including T stage, and lymph node and distant metastases. CHIP-HEG patients had a significantly worse prognosis than patients with low CHIP expression with median cancer-specific survival times of 8.0 months (range, 1-34 months) and 13.0 months (range, 1-110 months), respectively (P=0.023). Multivariate analyses showed that CHIP expression was close to being an independent risk factor for predicting patient survival. CHIP expression may be associated with a poor prognosis in GBC. Since CHIP is not associated with other clinicopathological prognostic factors, it may serve as an ideal molecular marker for predicting patient outcomes.

8.
Anticancer Res ; 32(9): 3835-40, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22993326

RESUMO

Yes-associated protein (YAP) is a nuclear downstream effector of the Hippo pathway, a regulator of cell growth. The phosphorylated form of YAP (pYAP), located in the cytoplasm, prevents cellular proliferation by spatially segregating YAP from the nucleus. This study aimed at investigating the relationships of pYAP and YAP with clinicopathological variables in esophageal squamous cell carcinoma (ESCC). Samples of ESCC from 142 patients were studied using immunohistochemistry for YAP, pYAP and Ki-67. In all cases of ESCC, higher nuclear expression of YAP was correlated with Ki-67 expression, tumor diameter, histological grade (1-2 versus 3), and pathological TNM stage (I versus II-IV) in univariate analyses (p=0.036, p=0.025, p=0.021, and p=0.033, respectively). Higher nuclear expression of YAP was associated with worse overall and disease-free survival (p=0.006 and p=0.008, respectively). Multivariate analysis showed higher nuclear expression of YAP to be an independent prognostic marker for overall survival (p=0.034). We observed a trend towards inverse correlation of cytoplasmic pYAP expression and histological grade (1-2 versus 3) (p=0.087). Our results suggest that YAP shifts from the nucleus to the cytoplasm as a consequence of phosphorylation, which occurs in the presence of high tumor cell density in the case of ESCC, and may be a potential indication of histological differentiation. Nuclear expression of YAP is correlated with tumor cell proliferation and is an independent predictor of worse prognosis of ESCC.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Fatores de Transcrição/metabolismo , Idoso , Carcinoma de Células Escamosas/patologia , Neoplasias Esofágicas/patologia , Feminino , Humanos , Imuno-Histoquímica , Antígeno Ki-67/biossíntese , Masculino , Estadiamento de Neoplasias , Proteínas Nucleares/biossíntese , Fosfoproteínas/biossíntese , Fosforilação , Fatores de Transcrição/biossíntese , Proteínas de Sinalização YAP
9.
Exp Ther Med ; 3(5): 811-817, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22969974

RESUMO

CD44 has been implicated in tumor development and progression in several types of cancer. CD44 expression is altered in renal cell carcinoma (RCC) and has been suggested as a useful prognostic marker, but its prognostic role in RCC remains controversial. We investigated the expression of CD44 in a large homogeneous set of localized clear cell RCC to determine its potential prognostic value. We examined 110 patients with localized clear cell RCC who underwent nephrectomy. The clinicopathological data were obtained retrospectively and the expression level of CD44 was studied by immunohistochemistry. Correlations between CD44 expression and clinical parameters as well as survival were determined. The CD44-high expression group (HEG) was significantly associated with a higher nuclear grade (P=0.014) and tumor recurrence (P<0.001) when compared with the CD44-low expression group (LEG). Concerning survival, the 5-year recurrence-free survival (RFS) rates for the CD44-HEG and CD44-LEG groups were 38.9 and 91.3%, respectively (P<0.001), and the 5-year disease-specific survival (DSS) rates for the CD44-HEG and CD44-LEG groups were 55.6 and 94.6%, respectively (P<0.001). Multivariate analyses showed that CD44 expression [hazard ratio (HR), 9.204; P<0.001] was an independent risk factor predicting RFS in patients with clear cell RCC. CD44 expression remained an independent prognostic factor for DSS (P=0.002). In conclusion, these data indicate that CD44 expression is associated with the progression of clear cell RCC and is an independent poor prognostic factor for tumor recurrence and survival, suggesting that CD44 may serve as a useful molecular marker.

10.
Oncol Lett ; 3(2): 490-496, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22740938

RESUMO

Interleukin-32 (IL-32) is a proinflammatory cytokine that acts as a significant pathogenetic factor in various diseases and malignancies. However, the clinical effect of IL-32 expression in renal cell carcinoma (RCC) has not previously been investigated. The aim of the present study was to examine the significance of IL-32 overexpression in localized clear cell RCC (CCRCC). We examined 112 patients with localized CCRCC who underwent nephrectomy. The clinicopathological data were obtained by retrospective review and the expression levels of IL-32 were studied by immunohistochemistry. Tumors were classified according to staining intensity (0, no staining intensity; 1, weak; 2, intermediate; 3, strong). The cases with staining intensities from 0 to 2 comprised the IL-32 low-expression group (LEG), whereas those with a staining intensity of 3 comprised the IL-32 high-expression group (HEG). Correlations between IL-32 expression and clinicopathological parameters were determined. Staining intensities were determined for all cases as follows: 26 cases (23.2%) (score 0), 43 cases (38.4%) (score 1), 31 cases (27.7%) (score 2) and 12 cases (10.7%) (score 3). IL-32 HEG exhibited a higher recurrence rate compared to the IL-32 LEG (50 vs. 13%, P=0.001). For survival rates, the 5-year recurrence-free survival (RFS), disease-specific survival (DSS) and overall survival (OS) rates were lower in the IL-32 HEG group compared with the IL-32 LEG group (RFS, P=0.001; DSS, P<0.001; OS, P=0.026, respectively). Univariate analyses revealed that Fuhrman nuclear grade and a high IL-32 expression were significant prognostic factors for predicting RFS, DSS and OS in CCRCC, whereas multivariate analyses indicated that Fuhrman nuclear grade and high IL-32 expression were still independent risk factors. In conclusion, IL-32 overexpression was associated with high recurrence rates and low RFS, DSS and OS, indicating that it may be a novel prognostic factor for predicting outcomes in patients with CCRCC.

11.
Clin Cancer Res ; 18(12): 3270-80, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22553346

RESUMO

PURPOSE: Although recent studies have suggested that chemokine (C-X-C motif) ligand 12 (CXCL12) is important in the progression of various malignancies, its role in gallbladder carcinoma (GBC) remains unknown. We investigated CXCL12 expression in GBC and its biologic and prognostic role in GBC tumorigenesis. EXPERIMENTAL DESIGN: We examined CXCL12 expression in tumor specimens from 72 patients with GBC by immunohistochemistry and analyzed the correlation between CXCL12 expression and clinicopathologic factors or survival. The functional significance of CXCL12 expression was investigated by CXCL12 treatment and suppression of CXCR4, a major receptor of CXCL12, as well as by CXCL12 overexpression in in vitro and in vivo studies. RESULTS: CXCL12 was differentially expressed in GBC tissues. CXCL12 expression was significantly associated with a high histologic grade (P = 0.042) and nodal metastasis (P = 0.015). Multivariate analyses showed that CXCL12 expression (HR, 8.675; P = 0.014) was an independent risk factor for patient survival. CXCL12 significantly increased anchorage-dependent and -independent growth, migration, invasion, adhesiveness, and survival of GBC cells in vitro, and these effects were dependent on CXCR4. Consistent with these results, overexpression of CXCL12 significantly promoted GBC tumorigenicity in a xenograft model. CONCLUSIONS: Our results indicate that GBC cells express both CXCL12 and its receptor CXCR4, and CXCL12 may have a role in GBC progression through an autocrine mechanism. In addition, CXCL12 is a novel independent poor prognostic factor in patients with GBCs. Thus, targeting CXCL12 and CXCR4 may provide a novel therapeutic strategy for GBC treatment.


Assuntos
Quimiocina CXCL12/metabolismo , Progressão da Doença , Neoplasias da Vesícula Biliar/metabolismo , Neoplasias da Vesícula Biliar/fisiopatologia , Receptores CXCR4/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Transformação Celular Neoplásica , Quimiocina CXCL12/genética , Feminino , Vesícula Biliar/metabolismo , Neoplasias da Vesícula Biliar/patologia , Humanos , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Invasividade Neoplásica , Metástase Neoplásica , Prognóstico , Interferência de RNA , RNA Interferente Pequeno , Receptores CXCR4/genética , Transplante Heterólogo
12.
Histol Histopathol ; 27(5): 581-91, 2012 05.
Artigo em Inglês | MEDLINE | ID: mdl-22419022

RESUMO

Epithelial-mesenchymal transition (EMT) is characterized by the loss of epithelial cell junction proteins and the gain of mesenchymal markers. The aim of this study was to analyze the associations between the EMT-related markers vimentin, E-cadherin, ß-catenin, slug, snail, and twist1 and clinicopathologic parameters as well as epidermal growth factor receptor (EGFR) gene copy number and protein expression in non-small cell lung carcinoma (NSCLC). Fifty-nine squamous cell carcinomas (SCC) and 43 adenocarcinomas (AD) were immunohistochemically examined for respective EMT markers and for EGFR, using the EGFR PharmDx kit (Dako) for protein expression and automated silver enhanced in situ hybridization (SISH) for copy number. Vimentin expression in tumor epithelia was significantly higher in AD samples than in SCC samples (P=0.015). Among AD samples, vimentin expression was positively correlated with histologic grade (2 vs. 3; P=0.021) and exhibited a tendency toward a positive correlation with pTNM stage (I vs. II-IV; P=0.052). EGFR gene copy number was positively correlated with EGFR protein expression among both AD samples (P=0.008) and SCC samples (P=0.042), with EGFR protein expression being significantly higher in SCC samples compared with AD (P=0.038). Among AD samples, EGFR protein expression was associated with higher cytoplasmic expression of ß-catenin (P=0.031). Among SCC samples, EGFR protein expression was negatively correlated with nuclear expression of ß-catenin (P=0.033) but positively with nuclear slug (P=0.021). The expression pattern of EMT markers in AD suggests that vimentin is a possible immunohistochemical predictor of tumor progression.


Assuntos
Adenocarcinoma/patologia , Carcinoma de Células Escamosas/patologia , Transição Epitelial-Mesenquimal , Neoplasias Pulmonares/patologia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Idoso , Caderinas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Transição Epitelial-Mesenquimal/genética , Transição Epitelial-Mesenquimal/fisiologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Dosagem de Genes , Humanos , Imuno-Histoquímica , Hibridização In Situ , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Pessoa de Meia-Idade , Mutação , Proteínas Nucleares/metabolismo , Fatores de Transcrição da Família Snail , Fatores de Transcrição/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Vimentina/metabolismo , beta Catenina/metabolismo
13.
Ann Surg Oncol ; 19(8): 2763-72, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22246425

RESUMO

BACKGROUND: Although NDRG2 is a candidate tumor suppressor, its exact role in renal cell carcinoma (RCC) is not fully understood. We investigated the functional role of NDRG2 and its clinical relevance in RCC tumorigenesis. METHODS: NDRG2 expression and its clinical implications in clear cell RCC were evaluated. Biological function was assessed by a proliferation assay, anchorage-independent growth assay, and wound healing and transwell migration assays in RCC cell lines overexpressing NDRG2 coupled with an investigation of the effects of NDRG2 expression on the epithelial-mesenchymal transition (EMT). RESULTS: NDRG2 was differentially expressed in patients with RCC. A loss of NDRG2 was significantly associated with a higher proportion of tumors >10 cm and a high nuclear grade. Furthermore, multivariate analyses indicated that a loss of NDRG2 was an independent poor prognostic factor for patient survival (recurrence-free survival, hazard ratio 7.901; disease-specific survival, hazard ratio 15.395; overall survival, hazard ratio 11.339; P < 0.001 for all parameters). NDRG2 expression inhibited the anchorage-independent growth and migration of RCC cells. NDRG2 expression also modulated the expression of EMT-related genes such as Snail, Slug, and SIP1, and it decreased EMT signaling in RCC cells. Finally, NDRG2 recovered E-cadherin expression in E-cadherin-negative RCC cells. CONCLUSIONS: These results indicate that a lack of NDRG2 is associated with oncogenic properties through the loss of its role as a tumor suppressor, and that NDRG2 is an independent poor prognostic factor predicting survival in clear cell RCC, suggesting that it can serve as a novel prognostic biomarker.


Assuntos
Carcinoma de Células Renais/patologia , Transformação Celular Neoplásica/patologia , Neoplasias Renais/patologia , Recidiva Local de Neoplasia/patologia , Nefrectomia/mortalidade , Complicações Pós-Operatórias , Proteínas Supressoras de Tumor/metabolismo , Idoso , Western Blotting , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/mortalidade , Carcinoma de Células Renais/cirurgia , Movimento Celular , Proliferação de Células , Transição Epitelial-Mesenquimal , Feminino , Seguimentos , Humanos , Técnicas Imunoenzimáticas , Neoplasias Renais/metabolismo , Neoplasias Renais/mortalidade , Neoplasias Renais/cirurgia , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/mortalidade , Recidiva Local de Neoplasia/cirurgia , Estadiamento de Neoplasias , Prognóstico , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Taxa de Sobrevida , Análise Serial de Tecidos , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/genética , Cicatrização
14.
Nat Immunol ; 12(8): 742-51, 2011 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-21725320

RESUMO

The orphan nuclear receptor SHP (small heterodimer partner) is a transcriptional corepressor that regulates hepatic metabolic pathways. Here we identified a role for SHP as an intrinsic negative regulator of Toll-like receptor (TLR)-triggered inflammatory responses. SHP-deficient mice were more susceptible to endotoxin-induced sepsis. SHP had dual regulatory functions in a canonical transcription factor NF-κB signaling pathway, acting as both a repressor of transactivation of the NF-κB subunit p65 and an inhibitor of polyubiquitination of the adaptor TRAF6. SHP-mediated inhibition of signaling via the TLR was mimicked by macrophage-stimulating protein (MSP), a strong inducer of SHP expression, via an AMP-activated protein kinase-dependent signaling pathway. Our data identify a previously unrecognized role for SHP in the regulation of TLR signaling.


Assuntos
NF-kappa B/imunologia , Receptores Citoplasmáticos e Nucleares/imunologia , Sepse/imunologia , Receptores Toll-Like/imunologia , Proteínas Quinases Ativadas por AMP/imunologia , Animais , Imunoprecipitação da Cromatina , Feminino , Immunoblotting , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Fator 6 Associado a Receptor de TNF/imunologia , Ubiquitinação/imunologia
15.
Pathol Res Pract ; 207(7): 428-32, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21680106

RESUMO

Dual specificity phosphatase 6 (DUSP6) is a member of the MAP kinase phophatase family. DUSP6 inactivates extracellular signal-regulated kinase (ERK), belonging to the MAP kinase family, and can act in tumor suppressive pathways. The aim of this study was to investigate associations of DUSP6 expression with expression of ERK and Ki-67 and with clinicopathological parameters in lung adenocarcinoma and squamous cell carcinoma. A total of 102 squamous cell carcinomas and 66 adenocarcinomas were studied using immunohistochemistry for DUSP6, ERK1/2, and Ki-67. In 66 adenocarcinomas, high DUSP6 expression was positively correlated with ERK1/2 expression. High DUSP6 expression was correlated with lower histological grade and lower Ki-67 index in the adenocarcinomas. In 102 squamous cell carcinomas, high DUSP6 expression was correlated with lower ERK expression, with greater smoking pack-years, but not with the Ki-67 index. These results indicate that DUSP6 acts as a negative feedback regulator of ERK in adenocarcinoma progression, but that DUSP6 does not play a role in the downregulation of ERK in squamous cell carcinoma. The differential expression of DUSP6 correlated with Ki-67 index, suggesting that DUSP6 plays an important role in cancer resistance in different subtypes of non-small cell lung carcinoma.


Assuntos
Adenocarcinoma/patologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Escamosas/patologia , Fosfatase 6 de Especificidade Dupla/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Antígeno Ki-67/metabolismo , Neoplasias Pulmonares/patologia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , DNA de Neoplasias/análise , Fosfatase 6 de Especificidade Dupla/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , Humanos , Antígeno Ki-67/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Pneumonectomia
16.
Hum Pathol ; 42(10): 1476-83, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21496863

RESUMO

Gallbladder carcinoma is a lethal malignancy and is hard to cure by current treatment. Thus, identification of molecular prognostic markers to predict gallbladder carcinoma as therapeutic targets is urgently needed. Recent studies have demonstrated that L1 cell adhesion molecule is associated with the prognosis of variable malignancy. Here, we investigated L1 cell adhesion molecule expression in gallbladder carcinoma and its prognostic significance. In this study, we examined L1 cell adhesion molecule expression in tumor specimens from 69 patients with gallbladder carcinoma by immunohistochemistry and analyzed the correlation between L1 cell adhesion molecule expression and clinicopathologic factors or survival. L1 cell adhesion molecule was not expressed in the normal epithelium of the gallbladder but in 63.8% of gallbladder carcinomas, remarkably at the invasive front of the tumors. In addition, L1 cell adhesion molecule expression was significantly associated with high histologic grade, advanced pathologic T stage and clinical stage, and positive venous/lymphatic invasion. Multivariate analyses showed that L1 cell adhesion molecule expression (hazard ratio, 3.503; P = .028) and clinical stage (hazard ratio, 3.091; P = .042) were independent risk factor for disease-free survival. L1 cell adhesion molecule expression in gallbladder carcinoma was significantly correlated with tumor progression and unfavorable clinicopathologic features. L1 cell adhesion molecule expression was an independent poor prognostic factor for disease-free survival in patients with gallbladder carcinoma. Taken together, our findings suggest that L1 cell adhesion molecule expression could be used as a novel prognostic factor for patient survival and might be a potential therapeutic target in gallbladder carcinomas.


Assuntos
Adenocarcinoma/metabolismo , Neoplasias da Vesícula Biliar/metabolismo , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Adenocarcinoma/diagnóstico , Adenocarcinoma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Biomarcadores Tumorais/metabolismo , Feminino , Neoplasias da Vesícula Biliar/diagnóstico , Neoplasias da Vesícula Biliar/patologia , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Metástase Neoplásica , Estadiamento de Neoplasias , Prognóstico , Estudos Retrospectivos , beta Catenina/metabolismo
17.
J Immunol ; 186(2): 1248-58, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21160040

RESUMO

The scaffolding adaptor protein p62/SQSTM1 (p62) has been shown to be an autophagy receptor that acts as a link between the ubiquitination and autophagy machineries. However, the roles of autophagy and p62 in human keratinocytes are not well understood. In this study, we show that keratinocyte autophagy negatively regulates p62 expression, which is essential for the prevention of excessive inflammation and the induction of cathelicidin in human keratinocytes. Stimulation of TLR2/6 or TLR4 in primary human keratinocytes robustly activated autophagy pathways and up-regulated p62 expression through induction of NADPH oxidases 2 and 4 and the generation of reactive oxygen species. MyD88 and TNFR-associated factor 6, key signaling molecules that mediate TLR activation, played an essential role in the induction of autophagy and p62 expression. Additionally, blockade of autophagy significantly increased the generation of inflammatory cytokines and expression of p62 in primary human keratinocytes. Notably, silencing hp62 through RNA interference resulted in a significant decrease in NF-κB activation, inflammatory cytokine production, cathelicidin expression, and cell proliferation (as well as cyclin D1 expression) in keratinocytes. Epidermal expression of p62 was further found to be significantly higher in psoriatic skin than in skin affected by atopic dermatitis or from healthy controls. Collectively, our data provide new insights into the roles of autophagy and p62 in controlling cutaneous inflammation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Autofagia/imunologia , Regulação para Baixo/imunologia , Mediadores da Inflamação/fisiologia , Queratinócitos/imunologia , Queratinócitos/patologia , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Peptídeos Catiônicos Antimicrobianos/biossíntese , Linhagem Celular , Citocinas/biossíntese , Dermatite Atópica/imunologia , Dermatite Atópica/metabolismo , Dermatite Atópica/patologia , Células HEK293 , Humanos , Queratinócitos/metabolismo , NF-kappa B/fisiologia , Psoríase/imunologia , Psoríase/metabolismo , Psoríase/patologia , Espécies Reativas de Oxigênio/metabolismo , Proteína Sequestossoma-1 , Receptor 2 Toll-Like/metabolismo , Receptor 2 Toll-Like/fisiologia , Receptor 6 Toll-Like/metabolismo , Receptor 6 Toll-Like/fisiologia , Catelicidinas
18.
Hum Pathol ; 42(3): 315-23, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21190720

RESUMO

Yes-associated protein, a downstream effector of the Hippo signaling pathway, has been linked to progression of non-small-cell lung carcinoma. The aim of this study was to investigate expression of Yes-associated protein in lung adenocarcinoma and squamous cell carcinoma. Associations of Yes-associated protein expression with clinicopathologic parameters, expression of cell cycle-specific markers, and epidermal growth factor receptor gene amplification were also analyzed. In a univariate analysis of the 66 adenocarcinomas, high nuclear expression of Yes-associated protein was significantly correlated with expression of cyclin A and mitogen-activated protein kinase. Multivariate analysis, including age and sex, showed that cyclin A expression was independently correlated with nuclear expression of Yes-associated protein in adenocarcinomas. Furthermore, high nuclear expression of Yes-associated protein was also a significant predictor of epidermal growth factor receptor gene amplification for adenocarcinoma. For the 102 squamous cell carcinomas, univariate analysis revealed that high cytoplasmic expression of Yes-associated protein was correlated with the low pathologic TNM staging (stage I) and histologic grading. Multivariate analysis, including age and sex, showed that cytoplasmic expression of Yes-associated protein was an independent predictor of low pathologic TNM staging. These results indicate that nuclear overexpression of Yes-associated protein contributes to pulmonary adenocarcinoma growth and that high cytoplasmic expression of Yes-associated protein is an independent predictor of low pathologic TNM staging and histologic grading. The differential effects of Yes-associated protein expression patterns in adenocarcinomas and squamous cell carcinomas suggest that Yes-associated protein may play important roles in different pathways in distinct tumor subtypes. These observations may, therefore, lead to new perspectives on therapeutic targeting of these tumor types.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Ciclinas/metabolismo , Receptores ErbB/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogênicas c-yes/metabolismo , Carcinoma Pulmonar de Células não Pequenas/diagnóstico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/genética , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Diagnóstico por Imagem , Receptores ErbB/genética , Feminino , Amplificação de Genes , Dosagem de Genes , Humanos , Hibridização In Situ , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Masculino , Pessoa de Meia-Idade , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Estadiamento de Neoplasias , Prognóstico , Proteínas Proto-Oncogênicas c-yes/genética , Análise Serial de Tecidos
19.
Exp Ther Med ; 2(3): 499-504, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-22977531

RESUMO

Chemokine stromal cell-derived factor (SDF)-1α and its receptor CXC chemokine receptor 4 (CXCR4) have been shown to impact cancer progression. Accumulating evidence suggests that CXCR4 and SDF-1α expression is useful for evaluating the risk of gastric cancer progression. Thus, combined analysis of SDF-1α and CXCR4 should have high prognostic potential as a molecular marker for gastric cancer. We investigated the expression of SDF-1α and CXCR4 using immunohistochemistry in relation to prognosis, clinicopathological features and clinical outcomes in 221 cases of primary gastric cancer. Patients were categorized into three groups according to CXCR4 and SDF-1α expression: high CXCR4/high SDF-1α, low CXCR4/low SDF-1α, and high CXCR4/low SDF-1α - low CXCR4/high SDF-1α. No significant differences were noted in age, gender, histology, tumor location, lymphovascular invasion or proportion of tumor size >5 cm among the three groups. However, high CXCR4/high SDF-1α expression in tumor cells was significantly associated with depth of invasion of the tumor, lymph node involvement, and higher tumor stage compared to tumors with low CXCR4/low SDF-1α expression or high CXCR4/low SDF-1α - low CXCR4/high SDF-1α expression. Furthermore, patients with high CXCR4/high SDF-1α expression had the worst patient prognosis, whereas patients who had low CXCR4/low SDF-1α expression showed the most favorable prognosis. In conclusion, CXCR4 and SDF-1α are useful prognostic factors in gastric cancer, and the combination of high CXCR4 protein expression with high SDF-1α expression suggests a dismal prognosis.

20.
Oncol Lett ; 2(6): 1197-1202, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22848288

RESUMO

Lymph node metastasis is considered to be a significant prognostic factor for early gastric cancer (EGC). However, no real consensus exists on which patient and/or tumor characteristics are associated with lymph node metastasis. We investigated whether stromal cell-derived factor (SDF)-1α expression correlates with lymph node metastasis in patients with EGC by immunohistochemically examining the expression of SDF-1α in 138 archival tissue specimens of EGC. Of these specimens, 59 (42.8%) and 79 (57.2%) were grouped into SDF-1α-positive and SDF-1α-negative groups, respectively. No significant differences existed with respect to age, gender, tumor location, proportion of tumors >20 mm in size, macroscopic type, depth of invasion or histology between the SDF-1α-positive and -negative groups. However, the SDF-1α-positive group was significantly correlated with lymphovascular invasion and lymph node metastasis. Results of the univariate analyses indicated that lymphovascular invasion, undifferentiated histology and SDF-1α positivity were statistically significant risk factors affecting lymph node metastasis in patients with EGC. Multivariate analyses showed that lymphovascular invasion [hazard ratio (HR), 8.595; 95% confidence interval (CI), 1.694-43.595; P=0.009], undifferentiated histology (HR, 2.965; 95% CI, 1.037-8.471; P=0.043) and SDF-1α positivity (HR, 2.108; 95% CI, 1.316-10.135; P=0.013) were independent risk factors predicting lymph node metastasis in EGC. In conclusion, these results suggest that SDF-1α expression in tumor cells is a predictive marker of lymph node metastasis in EGC.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...