Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 9(1): 15555, 2019 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-31645573

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
Sci Rep ; 9(1): 4299, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30862797

RESUMEN

Creation of a hemodialysis arteriovenous fistula (AVF) causes aberrant vascular mechanics at and near the AVF anastomosis. When inadequately regulated, these aberrant mechanical factors may impede AVF lumen expansion to cause AVF maturation failure, a significant clinical problem with no effective treatments. The endothelial nitric oxide synthase (NOS3) system is crucial for vascular health and function, but its effect on AVF maturation has not been fully characterized. We hypothesize that NOS3 promotes AVF maturation by regulating local vascular mechanics following AVF creation. Here we report the first MRI-based fluid-structure interaction (FSI) study in a murine AVF model using three mouse strains: NOS3 overexpression (NOS3 OE) and knockout (NOS3-/-) on C57BL/6 background, with C57BL/6 as the wild-type control (NOS3+/+). When compared to NOS3+/+ and NOS3-/-, AVFs in the OE mice had larger lumen area. AVFs in the OE mice also had smoother blood flow streamlines, as well as lower blood shear stress at the wall, blood vorticity, inner wall circumferential stretch, and radial wall thinning at the anastomosis. Our results demonstrate that overexpression of NOS3 resulted in distinct hemodynamic and wall mechanical profiles associated with favorable AVF remodeling. Enhancing NOS3 expression may be a potential therapeutic approach for promoting AVF maturation.


Asunto(s)
Fístula Arteriovenosa/metabolismo , Fístula Arteriovenosa/fisiopatología , Hemodinámica/fisiología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Velocidad del Flujo Sanguíneo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Mecánico
4.
Sci Rep ; 7(1): 8678, 2017 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-28819116

RESUMEN

Transforming growth factor (TGF)-ß1 contributes to autocrine and paracrine functions in the tumor microenvironment (TME). The present study examined the effects of TGF-ß1 crosstalk in TME and its role in mediating tumor formation and progression by targeted abrogation of TGF-ß1 expression in metastatic cells in situ. Using species-specific primers, we found a significant increase in MMP-9 gene expression in the tumor-reactive stroma during late-stage metastasis in the lung. This effect was also confirmed in cancer-associated fibroblasts (CAFs) when co-cultured with the tumor cells. Knockdown of TGF-ß1 expression in the tumor cells negatively affected matrix metalloproteinase (MMP)-9 gene expression. Fibroblasts, cultured in the presence of tumor cells with intact TGF-ß1, showed a significant increase in proliferation rate, as well as expression of VEGF, bFGF, and SDF-1, which was not seen when TGF-ß1 expression was abrogated in tumor cells. Absence of TGF-ß1 in tumor cells also failed to result in myofibroblast differentiation. Co-implantation of CAFs and tumor cells with either intact TGF-ß1 expression or devoid of TGF-ß1 in vivo showed a significant increase in tumor growth kinetics in both cell types, suggesting a possible activation TGF-ß receptor signaling in tumor cells in response to TGF-ß from the TME.


Asunto(s)
Silenciador del Gen , Metaloproteinasa 9 de la Matriz/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Factor de Crecimiento Transformador beta1/genética , Microambiente Tumoral/genética , Biomarcadores , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Línea Celular Tumoral , Proliferación Celular , Quimiocinas/genética , Quimiocinas/metabolismo , Técnicas de Cocultivo , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias/patología , Comunicación Paracrina , Transducción de Señal/efectos de los fármacos , Células del Estroma/metabolismo
5.
Curr Pharm Des ; 23(32): 4729-4744, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28571552

RESUMEN

Head and neck cancer is one of the leading malignancies worldwide. Due to the lack of symptoms in the early stage of the disease, about two thirds of patients present with locally advanced disease at the time of diagnosis. Even with significantly improved survival rates over the past two decades due to advanced imaging and treatment modalities, locoregional recurrence rates in patients with advanced disease ranges from 16% to 35%. Alternative therapeutic targets are being developed to improve survival outcomes. MicroRNAs (miRNA or miRs) are a family of small non-coding RNA species that have been demonstrated to regulate all cellular, physiological and developmental processes. Recently, there has been an exponential increase in the number of studies suggesting that miRNA is involved in regulating tumor metastasis, chemoresistance, radioresistance and survival outcomes. MiRNA candidates have been identified as potential prognostic biomarkers to diagnose cancer stages and progression, as well as to monitor follow-up treatment. In this review, we will discuss the miRNA profile in each stage of head and neck patients' therapy, with an emphasis on its application to clinical outcome prognosis.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de Cabeza y Cuello/genética , MicroARNs/genética , Animales , Progresión de la Enfermedad , Neoplasias de Cabeza y Cuello/patología , Neoplasias de Cabeza y Cuello/terapia , Humanos , Recurrencia Local de Neoplasia , Estadificación de Neoplasias , Evaluación de Resultado en la Atención de Salud , Pronóstico
6.
Theor Biol Med Model ; 14(1): 5, 2017 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-28320412

RESUMEN

BACKGROUND: Arteriovenous fistula (AVF) maturation failure remains a major cause of morbidity and mortality in hemodialysis patients. The two major etiologies of AVF maturation failure are early neointimal hyperplasia development and persistent inadequate outward remodeling. Although hemodynamic changes following AVF creation may impact AVF remodeling and contribute to neointimal hyperplasia development and impaired outward remodeling, detailed AVF hemodynamics are not yet fully known. Since murine AVF models are valuable tools for investigating the pathophysiology of AVF maturation failure, there is a need for a new approach that allows the hemodynamic characterization of murine AVF at high resolutions. METHODS: This methods paper presents a magnetic resonance imaging (MRI)-based computational fluid dynamic (CFD) method that we developed to rigorously quantify the evolving hemodynamic environment in murine AVF. The lumen geometry of the entire murine AVF was reconstructed from high resolution, non-contrast 2D T2-weighted fast spin echo MRI sequence, and the flow rates of the AVF inflow and outflow were extracted from a gradient echo velocity mapping sequence. Using these MRI-obtained lumen geometry and inflow information, CFD modeling was performed and used to calculate blood flow velocity and hemodynamic factors at high resolutions (on the order of 0.5 µm spatially and 0.1 ms temporally) throughout the entire AVF lumen. We investigated both the wall properties (including wall shear stress (WSS), wall shear stress spatial gradient, and oscillatory shear index (OSI)) and the volumetric properties (including vorticity, helicity, and Q-criterion). RESULTS: Our results demonstrate increases in AVF flow velocity, WSS, spatial WSS gradient, and OSI within 3 weeks post-AVF creation when compared to pre-surgery. We also observed post-operative increases in flow disturbances and vortices, as indicated by increased vorticity, helicity, and Q-criterion. CONCLUSIONS: This novel protocol will enable us to undertake future mechanistic studies to delineate the relationship between hemodynamics and AVF development and characterize biological mechanisms that regulate local hemodynamic factors in transgenic murine AVF models.


Asunto(s)
Fístula Arteriovenosa/diagnóstico por imagen , Velocidad del Flujo Sanguíneo/fisiología , Hemodinámica/fisiología , Hidrodinámica , Imagen por Resonancia Magnética/métodos , Animales , Fístula Arteriovenosa/fisiopatología , Biología Computacional/métodos , Masculino , Ratones , Ratones Endogámicos C57BL
7.
Cancer Control ; 24(1): 102-110, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28178722

RESUMEN

BACKGROUND: The molecular signature of ductal carcinoma in situ (DCIS) in the breast is not well understood. Erb-b2 receptor tyrosine kinase 2 (ERBB2 [formerly known as HER2/neu]) positivity in DCIS is predictive of coexistent early invasive breast carcinoma. The aim of this study is to identify the gene-expression signature profiles of estrogen receptor (ER)/progesterone receptor (PR)-positive, ERBB2, and triple-negative subtypes of DCIS. METHODS: Based on ER, PR, and ERBB2 status, a total of 18 high nuclear grade DCIS cases with no evidence of invasive breast carcinoma were selected along with 6 non-neoplastic controls. The 3 study groups were defined as ER/PR-positive, ERBB2, and triple-negative subtypes. RESULTS: A total of 49 genes were differentially expressed in the ERBB2 subtype compared with the ER/PR-positive and triple-negative groups. PROM1 was overexpressed in the ERBB2 subtype compared with ER/PR-positive and triple-negative subtypes. Other genes differentially expressed included TAOK1, AREG, AGR3, PEG10, and MMP9. CONCLUSIONS: Our study identified unique gene signatures in ERBB2-positive DCIS, which may be associated with the development of invasive breast carcinoma. The results may enhance our understanding of the progression of breast cancer and become the basis for developing new predictive biomarkers and therapeutic targets for DCIS.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/genética , Carcinoma Ductal de Mama/genética , Carcinoma Intraductal no Infiltrante/genética , Perfilación de la Expresión Génica , Receptor ErbB-2/metabolismo , Adulto , Anciano , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patología , Carcinoma Intraductal no Infiltrante/metabolismo , Carcinoma Intraductal no Infiltrante/patología , Femenino , Humanos , Persona de Mediana Edad , Invasividad Neoplásica , Estadificación de Neoplasias , Pronóstico , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Tasa de Supervivencia
8.
Proc Natl Acad Sci U S A ; 112(5): 1392-7, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25605930

RESUMEN

Acquired resistance to androgen receptor (AR)-targeted therapies compels the development of novel treatment strategies for castration-resistant prostate cancer (CRPC). Here, we report a profound effect of endostatin on prostate cancer cells by efficient intracellular trafficking, direct interaction with AR, reduction of nuclear AR level, and down-regulation of AR-target gene transcription. Structural modeling followed by functional analyses further revealed that phenylalanine-rich α1-helix in endostatin-which shares structural similarity with noncanonical nuclear receptor box in AR-antagonizes AR transcriptional activity by occupying the activation function (AF)-2 binding interface for coactivators and N-terminal AR AF-1. Together, our data suggest that endostatin can be recognized as an endogenous AR inhibitor that impairs receptor function through protein-protein interaction. These findings provide new insights into endostatin whose antitumor effect is not limited to inhibiting angiogenesis, but can be translated to suppressing AR-mediated disease progression in CRPC.


Asunto(s)
Antagonistas de Andrógenos/metabolismo , Endostatinas/farmacología , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Sitios de Unión , Núcleo Celular/metabolismo , Humanos , Masculino
9.
Mod Pathol ; 28(5): 631-53, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25523612

RESUMEN

A large body of evidence shows that p16(INK4a) overexpression predicts improved survival and increased radiosensitivity in HPV-mediated oropharyngeal squamous cell carcinomas.(OPSCC). Here we demonstrate that the presence of transcriptionally active HPV16 in oral cavity squamous cell carcinomas does not correlate with p16(INK4a) overexpression, enhanced local tumor immunity, or improved outcome. It is interesting that HPV-mediated oropharyngeal squamous cell carcinomas can be categorized as having a 'nonaggressive' invasion phenotype, whereas aggressive invasion phenotypes are more common in HPV-negative squamous cell carcinomas. We have developed primary cancer cell lines from resections with known pattern of invasion as determined by our validated risk model. Given that cell lines derived from HPV-mediated oropharyngeal squamous cell carcinomas are less invasive than their HPV-negative counterparts, we tested the hypothesis that viral oncoproteins E6, E7, and p16(INK4a) can affect tumor invasion. Here we demonstrate that p16(INK4a) overexpression in two cancer cell lines (UAB-3 and UAB-4), derived from oral cavity squamous cell carcinomas with the most aggressive invasive phenotype (worst pattern of invasion type 5 (WPOI-5)), dramatically decreases tumor invasiveness by altering expression of extracellular matrix remodeling genes. Pathway analysis integrating changes in RNA expression and kinase activities reveals different potential p16(INK4a)-sensitive pathways. Overexpressing p16(INK4a) in UAB-3 increases EGFR activity and increases MMP1 and MMP3 expression, possibly through STAT3 activation. Overexpressing p16(INK4a) in UAB-4 decreases PDGFR gene expression and reduces MMP1 and MMP3, possibly through STAT3 inactivation. Alternatively, ZAP70/Syk might increase MUC1 phosphorylation, leading to the observed decreased MMP1 expression.


Asunto(s)
Carcinoma de Células Escamosas/patología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Neoplasias de la Boca/patología , Western Blotting , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/virología , Línea Celular Tumoral , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/virología , Invasividad Neoplásica , Infecciones por Papillomavirus , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
10.
PLoS One ; 9(2): e89940, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24587138

RESUMEN

Anterior Gradient Protein (AGR-2) is reported to be over-expressed in many epithelial cancers and promotes metastasis. A clear-cut mechanism for its observed function(s) has not been previously identified. We found significant upregulation of AGR-2 expression in a bone metastatic prostate cancer cell line, PC3, following culturing in bone marrow-conditioned medium. Substantial AGR-2 expression was also confirmed in prostate cancer tissue specimens in patients with bone lesions. By developing stable clones of PC3 cells with varying levels of AGR-2 expression, we identified that abrogation of AGR-2 significantly reduced cellular attachment to fibronectin, collagen I, collagen IV, laminin I and fibrinogen. Loss of cellular adhesion was associated with sharp decrease in the expression of α4, α5, αV, ß3 and ß4 integrins. Failure to undergo apoptosis following detachment is a hallmark of epithelial cancer metastasis. The AGR-2-silenced PC3 cells showed higher resistance to Tumor necrosis factor-related apoptosis- inducing ligand (TRAIL) induced apoptosis in vitro. This observation was also supported by significantly reduced Caspase-3 expression in AGR-2-silenced PC3 cells, which is a key effector of both extrinsic and intrinsic death signaling pathways. These data suggest that AGR-2 influence prostate cancer metastasis by regulation of cellular adhesion and apoptosis.


Asunto(s)
Neoplasias de la Próstata/metabolismo , Proteínas/metabolismo , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Análisis por Conglomerados , Expresión Génica , Perfilación de la Expresión Génica , Silenciador del Gen , Humanos , Integrinas/genética , Integrinas/metabolismo , Masculino , Ratones , Mucoproteínas , Metástasis de la Neoplasia , Proteínas Oncogénicas , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología
11.
Hum Pathol ; 45(2): 310-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24355195

RESUMEN

We examined racial disparities among 102 oropharyngeal carcinoma (OPC) patients (30 African Americans and 72 whites) comparing rates of transcriptionally active human papillomavirus (HPV)16/18 and p16(INK4a) overexpression, with times to disease progression and disease-specific survival (DSS). Expression of HPV16/18 transcripts was assessed by reverse transcription and polymerase chain reaction using type-specific E6/E7 primers; p16(INK4a) was evaluated by immunohistochemistry. African Americans were significantly more likely to present with high T stage disease and receive nonsurgical treatment. HPV16/18 was present in 63% of patients; no racial differences were observed. Silenced p16(INK4a) in OPC was significantly more common in African Americans (15/24) than in whites (20/69) (P = .004) and in HPV16+ African Americans (6/24) than in HPV+ whites (2/42) (P = .023). Kaplan-Meier analysis for DSS revealed a protective effect for p16(INK4a) overexpression (P = .0028; hazard ratio [HR], 0.23), HPV16+ (P = .036; HR, 0.38), and whites (P = .0039; HR, 0.27). Shorter DSS was associated with primary definitive chemoradiation (P = .019; HR, 3.49) and T3/T4 disease (P = .0001; HR, 7.75). A protective effect with respect to disease progression was observed for HPV16+ (P = .007; HR, 0.27), whites (P = .0006; HR, 0.197), and p16(INK4a) overexpression (P = .0001; HR, 0.116). African Americans with OPC experience poorer outcomes likely due to p16(INK4a) silencing, higher T stage, and nonsurgical treatment but not lower rates of transcriptionally active HPV16/18.


Asunto(s)
Negro o Afroamericano , Neoplasias Orofaríngeas/mortalidad , Adulto , Anciano , Carcinogénesis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Femenino , Silenciador del Gen , Papillomavirus Humano 16/genética , Papillomavirus Humano 18/genética , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Neoplasias Orofaríngeas/etnología , Neoplasias Orofaríngeas/genética , Neoplasias Orofaríngeas/terapia , Papillomaviridae/metabolismo , Infecciones por Papillomavirus/complicaciones
12.
Rom J Morphol Embryol ; 55(3 Suppl): 1057-62, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25607385

RESUMEN

Merkel cell carcinoma (MCC) is a rare, but highly aggressive primary cutaneous malignancy, showing neuroendocrine differentiation. In 2008, a novel member of the polyomavirus family, named Merkel cell polyomavirus (MCPyV) was identified in the genome of MCC tumors raising the possibility of an involvement in its pathogenesis. Due to the rarity of this tumor and current pathology practices, the most readily available tissue is archival formalin-fixed, paraffin-embedded (FFPE) material. In this study, we evaluated the presence of MCPyV in FFPE tissue and correlated its presence with tumor progression. Representative FFPE specimens from 18 tumors belonging to 14 patients with a diagnosis of MCC spanning the period from 2003 to 2008 were retrieved. Following DNA extraction, we performed PCR amplification and sequencing with four different MCPyV-specific primer pairs mapping within the T antigen and VP1 region. Overall, we detected MCPyV amplicons in 8/18 (44.4%) analyzed tumors from 7/14 (50%) cases. Two-year survival rate and median survival for the MCPyV-positive MCCs were 48% and 22.5 months, respectively and for the negative ones 69% and 51.3 months, respectively; however, the difference did not reach statistical significance (p=0.8). There was no significant correlation between the presence of the virus and the stage at presentation; however, tumors in the head and neck area had a lower frequency of viral positivity compared to those arising in the extremities suggesting a MCPyV-independent oncogenetic pathway perhaps, dependent on UV exposure, in a subset of these cases.


Asunto(s)
Carcinoma de Células de Merkel/patología , Carcinoma de Células de Merkel/virología , Formaldehído/química , Poliomavirus de Células de Merkel/aislamiento & purificación , Adhesión en Parafina , Fijación del Tejido , Anciano , Anciano de 80 o más Años , Electroforesis en Gel de Agar , Femenino , Vectores Genéticos/metabolismo , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Pronóstico
13.
Head Neck Pathol ; 7(2): 135-48, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23233027

RESUMEN

Herein we test the following hypotheses: (1) High-risk Human Papillomavirus (HR-HPV) may be involved in the etiology of mucoepidermoid carcinoma (MEC), and (2) The detection rate of HR-HPV in MEC has been increasing over time. Ninety-eight archival MEC specimens from three institutions spanning three decades were studied for HPV16/18 E6/E7 transcripts. RNA was extracted from formalin-fixed paraffin embedded specimens and HPV16/18 E6/E7 expression assessed by nested reverse transcription polymerase chain reaction (RT-PCR). A subset of MEC were also studied for MECT1-MAML2 fusion transcripts by nested RT-PCR and amplicon sequencing. The HPV expression data was validated by immunofluorescence (IF) with monoclonal HPV16/18 E6 antibody, PCR with the GP5+/6+ consensus primers, and sequencing of RT-PCR amplicons. HPV genome was localized by in-situ hybridization with the Ventana Inform HPVIII Family 16 probe. P16(INK4a) overexpression and aberrant p53 expression were assessed by immunohistochemistry. HPV16 E6/E7 transcripts were demonstrated in (29/98) 30% of MEC by RT-PCR. HPV18 E6/E7 transcripts were demonstrated in 13/98 (13%) of MEC by RT-PCR. Seven of 98 tumors (7%) demonstrated both HPV16/18. No significant association was found between HPV status and gender, age, and tumor site. All 13 HPV18+ MEC were diagnosed between 2001 and 2010, whereas 45 MEC diagnosed from 1977 to 2000 were negative for HPV18 (p = 0.002). By contrast, there was no significant difference with respect to HPV16 detection and date of diagnosis. All MEC that were positive for E6 protein were also HPV16/18 positive by RT-PCR. Sequencing a subset of RT-PCR amplicons confirmed HPV type- and region-specific sequences. PCR using GP5+/6+ consensus primers demonstrated HPV status concordance in 9 of 10 cases. DNA degradation was present in the last case; the RT-PCR amplicons were sequenced from this case which confirmed the presence of HPV type- and region-specific sequences. Strong (+4/+4) and diffuse (>50%) nuclear and cytoplasmic p16 expression was seen in 64% of MEC in the glandular regions, and 18% of MEC in the solid, squamoid regions. No correlation was seen between p16 expression and HPV status. Twenty-nine MEC (22 HPV+ and 7 HPV-negative) were selected for further evaluation for p53 expression. Strong aberrant nuclear p53 expression was present in only 2/22 HPV + MEC (9%, both Grade 3); no HPV-negative MEC demonstrated aberrant p53 expression. MECT1-MAML2 fusion transcripts were demonstrated in 23/37 (62%) MEC. No significant association was found between the presence of the MECT1-MAML2 fusion transcripts and tumor grade, HPV status, gender, era of diagnosis (2000 and earlier vs. 2001-2010) or tumor site. We demonstrate for the first time that transcriptionally active HPV16/18 is common to MEC. These findings were validated by demonstrating concordant results by separate PCR with consensus primers, and/or confirming the presence of HPV type- and region-specific sequences in the RT-PCR amplicons. We also visualized E6 viral oncoprotein and HPV genome within tumor cells. HR-HPV is thus potentially implicated in the pathogenesis of MEC. The frequency of HPV18 detection is significantly increased in MEC diagnosed after 2001, whereas we found no differences in the HPV16 detection rates per era of diagnosis.


Asunto(s)
Carcinoma Mucoepidermoide/patología , Transformación Celular Viral , Papillomavirus Humano 16/aislamiento & purificación , Papillomavirus Humano 18/aislamiento & purificación , Neoplasias de las Glándulas Salivales/patología , Carcinoma Mucoepidermoide/virología , ADN Viral/genética , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/genética , Femenino , Papillomavirus Humano 16/genética , Papillomavirus Humano 18/genética , Humanos , Hibridación in Situ , Masculino , Persona de Mediana Edad , Proteínas Oncogénicas Virales/análisis , Proteínas Oncogénicas Virales/genética , Neoplasias de las Glándulas Salivales/virología
14.
J Cutan Pathol ; 39(11): 1004-9, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22882314

RESUMEN

Trichodysplasia spinulosa (TS) is a rare and only recently characterized cutaneous disease occurring in immunocompromised patients. The disease is characterized by spiny follicular papules on clinical examination and by the presence of viral inclusions at ultrastructural examination. In the last year, this virus has been identified as a new member of the polyomavirus family and designated as TS-associated polyomavirus (TSPyV). We report two organ transplant patients with this disease in which we were able to identify the TSPyV at ultrastructural and molecular level from formalin-fixed paraffin-embedded biopsies of lesional skin. Similar to prior described cases, the patients presented with follicular papules which were concentrated on the central face and associated with alopecia. Histopathology of both cases showed dilated follicular infundibula plugged with cornified eosinophilic cells containing large trichohyaline granules. Transmission electron microscopy on paraffin-embedded tissue in case 1 showed 28-nm intracellular viral particles morphologically consistent with polyoma virus. For both cases the presence of TSPyV was confirmed by polymerase chain reaction with virus-specific primers followed by identification by direct sequencing. These two cases show the presence of the newly described TSPyV in TS further establishing its association with this distinctive disease.


Asunto(s)
Ictiosis , Infecciones por Polyomavirus , Poliomavirus , Piel , Adulto , Femenino , Humanos , Ictiosis/genética , Ictiosis/patología , Ictiosis/virología , Masculino , Persona de Mediana Edad , Trasplante de Órganos , Reacción en Cadena de la Polimerasa , Poliomavirus/genética , Poliomavirus/ultraestructura , Infecciones por Polyomavirus/genética , Infecciones por Polyomavirus/patología , Infecciones por Polyomavirus/virología , Piel/ultraestructura , Piel/virología
15.
Head Neck Pathol ; 6 Suppl 1: S104-20, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22782230

RESUMEN

Perhaps one of the most important developments in head and neck oncology of the past decade is the demonstration that patients with human papillomavirus (HPV)-mediated oropharyngeal cancers have significantly improved outcomes, compared to HPV-negative counterpart patients. This has become the basis for clinical trials investigating the impact on "treatment deintensification" for patients with HPV-mediated oropharyngeal cancers. Unfortunately, the significance of HPV in non-oropharyngeal head and neck cancers is much less certain. Our goal is to systematically review the published data regarding the role HPV in carcinomas of the oral cavity, larynx, sinonasal tract and nasopharynx with respect to HPV detection frequency, viral activity, and association with outcome. We also present preliminary data on HPV16/18 transcriptional status in oral cavity carcinomas, as well as salivary gland neoplasia, as determined by nested reverse transcription PCR for HPV E6/E7 RNA. The weighted prevalence (WP) of HPV DNA detection in 4,195 oral cavity cancer patients is 20.2 %, (95 % CI 16.0 %, 25.2 %). HPV16 is the most common type detected. Importantly, no data currently demonstrates a significant association between the presence of HPV DNA and improved outcome. The WP of HPV DNA in 1,712 laryngeal cancer patients is 23.6 %, (95 % CI 18.7 %, 29.3 %). Similarly, no association has yet been demonstrated between HPV DNA status and outcome. The WP of HPV DNA detection in 120 sinonasal cancer patients is 29.6 % (95 % CI 17.8 %, 44.9 %), and in 154 nasopharyngeal carcinoma patients is 31.1 %, (95 % CI 20.3 %, 44.5 %). Recent preliminary data also suggests an association between HPV and certain salivary gland neoplasms. The clinical significance of these findings is unclear. The published data strongly support the need for studies on patients with oral and laryngeal carcinomas that will be powered to find any differences in clinical outcome with respect to HR-HPV and p16 overexpression.


Asunto(s)
Neoplasias de Cabeza y Cuello/epidemiología , Neoplasias de Cabeza y Cuello/virología , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/epidemiología , Humanos , Pronóstico
16.
J Biol Chem ; 287(15): 12241-9, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22351751

RESUMEN

Noggin is a glycosylated-secreted protein known so far for its inhibitory effects on bone morphogenetic protein (BMP) signaling by sequestering the BMP ligand. We report here for the first time a novel mechanism by which noggin directly induces adipogenesis of mesenchymal stem cells independently of major human adipogenic signals through C/EBPδ, C/EBPα and peroxisome proliferator-activated receptor-γ. Evaluation of a possible mechanism for noggin-induced adipogenesis of mesenchymal stem cells identified the role of Pax-1 in mediating such differentiation. The relevance of elevated noggin levels in obesity was confirmed in a preclinical, immunocompetent mouse model of spontaneous obesity and in human patients with higher body mass index. These data clearly provide a novel role for noggin in inducing adipogenesis and possibly obesity and further indicates the potential of noggin as a therapeutic target to control obesity.


Asunto(s)
Adipogénesis , Proteínas Portadoras/fisiología , Células Madre Mesenquimatosas/fisiología , Obesidad/patología , Adiposidad , Adulto , Animales , Densidad Ósea , Proteína delta de Unión al Potenciador CCAAT/genética , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas Portadoras/sangre , Femenino , Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Obesidad/sangre , Obesidad/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Factores de Transcripción Paired Box/metabolismo , Adulto Joven
17.
Hum Pathol ; 42(12): 1911-22, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21683979

RESUMEN

High-throughput gene expression profiling from formalin-fixed, paraffin-embedded tissues has become a reality, and several methods are now commercially available. The Illumina whole-genome complementary DNA-mediated annealing, selection, extension and ligation assay (Illumina, Inc) is a full-transcriptome version of the original 512-gene complementary DNA-mediated annealing, selection, extension and ligation assay, allowing high-throughput profiling of 24,526 annotated genes from degraded and formalin-fixed, paraffin-embedded RNA. This assay has the potential to allow identification of novel gene signatures associated with clinical outcome using banked archival pathology specimen resources. We tested the reproducibility of the whole-genome complementary DNA-mediated annealing, selection, extension and ligation assay and its sensitivity for detecting differentially expressed genes in RNA extracted from matched fresh and formalin-fixed, paraffin-embedded cells, after 1 and 13 months of storage, using the human breast cell lines MCF7 and MCF10A. Then, using tumor worst pattern of invasion as a classifier, 1 component of the "risk model," we selected 12 formalin-fixed, paraffin-embedded oral squamous cell carcinomas for whole-genome complementary DNA-mediated annealing, selection, extension and ligation assay analysis. We profiled 5 tumors with nonaggressive, nondispersed pattern of invasion, and 7 tumors with aggressive dispersed pattern of invasion and satellites scattered at least 1 mm apart. To minimize variability, the formalin-fixed, paraffin-embedded specimens were prepared from snap-frozen tissues, and RNA was obtained within 24 hours of fixation. One hundred four down-regulated genes and 72 up-regulated genes in tumors with aggressive dispersed pattern of invasion were identified. We performed quantitative reverse transcriptase polymerase chain reaction validation of 4 genes using Taqman assays and in situ protein detection of 1 gene by immunohistochemistry. Functional cluster analysis of genes up-regulated in tumors with aggressive pattern of invasion suggests presence of genes involved in cellular cytoarchitecture, some of which already associated with tumor invasion. Identification of these genes provides biologic rationale for our histologic classification, with regard to tumor invasion, and demonstrates that the whole-genome complementary DNA-mediated annealing, selection, extension and ligation assay is a powerful assay for profiling degraded RNA from archived specimens when combined with quantitative reverse transcriptase polymerase chain reaction validation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Carcinoma de Células Escamosas/genética , Perfilación de la Expresión Génica/métodos , Genoma Humano/genética , Neoplasias de la Boca/genética , Fosfoproteínas/genética , Fijación del Tejido/métodos , Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Análisis por Conglomerados , Regulación hacia Abajo/genética , Estudios de Factibilidad , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias de la Boca/patología , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Estabilidad del ARN/genética , ARN Neoplásico/genética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Transcripción Genética/genética , Regulación hacia Arriba/genética
18.
Clin Cancer Res ; 15(23): 7175-85, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19920103

RESUMEN

PURPOSE: Current evidence indicates that an osteoblast lesion in prostate cancer is preceded by osteolysis. Thus, prevention of osteolysis would reduce complications of bone metastasis. Bone marrow-derived mesenchymal stem cells have the ability to differentiate into osteoblast and produce osteoprotegerin, a decoy receptor for the receptor activator for nuclear factor kappaB ligand, naturally. The present study examined the potential of unmodified mesenchymal stem cells to prevent osteolytic bone lesions in a preclinical mouse model of prostate cancer. EXPERIMENTAL DESIGN: The human prostate cancer cell line PC3 was implanted in tibiae of severe combined immunodeficient mice. After establishment of the tumor, either unmodified or genetically engineered mesenchymal stem cells overexpressing osteoprotegerin was injected at the site of tumor growth. The effects of therapy were monitored by bioluminescence imaging, micro-computed tomography, immunohistochemistry, and histomorphometry. RESULTS: Data indicated significant (P < 0.001) inhibition of tumor growth and restoration of bone in mice treated with unmodified and modified mesenchymal stem cells. Detailed analysis suggested that the donor mesenchymal stem cell inhibited tumor progression by producing woven bone around the growing tumor cells in the tibiae and by preventing osteoclastogenesis. CONCLUSIONS: Overcoming the limitation of the number of mesenchymal stem cells available in the bone can provide significant amelioration for osteolytic damage without further modification.


Asunto(s)
Células de la Médula Ósea/citología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Madre Mesenquimatosas/citología , Neoplasias de la Próstata/patología , Animales , Línea Celular Tumoral , Humanos , Inmunohistoquímica/métodos , Masculino , Ratones , Ratones SCID , Metástasis de la Neoplasia , Osteoclastos/citología , Osteoprotegerina/metabolismo , Neoplasias de la Próstata/terapia , Tibia/patología , Microtomografía por Rayos X/métodos
19.
Int J Oncol ; 34(5): 1425-31, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19360356

RESUMEN

Cks1 plays an essential role in SCFSkp2-mediated ubiquitination, and consequently turnover, of the cdk2 inhibitor and tumor supressor p27Kip1. High Cks1 expression is associated with aggressive breast tumors and correlates with low p27Kip1 levels in some cases, although it is also an independent prognostic marker for survival, and provides predictive information in addition to that provided by p27Kip1 alone. In this report we demonstrate that Cks1 protein and mRNA are elevated to very high levels in mammary tumors initiated by erbB2, c-myc and polyoma middle-T (PyMT) in transgenic mice, whereas Cks1 protein is hardly detectable in the normal mammary epithelium. Cks1 is also highly upregulated in rat mammary tumors initiated by methylnitrosourea (MNU). Despite high levels of Cks1 expression, p27Kip1 levels were not reduced, and were in fact slightly higher in mammary tumors initiated by erbB2, PyMT and MNU. In contrast mammary tumors from MMTV-c-myc mice did exhibit low p27Kip1 and higher levels of Skp2. Together, these data suggest that deregulated Cks1 expression might play roles in oncogene and carcinogen-initiated mammary tumorigenesis independent of p27Kip1 turnover in certain tumors. Stable overexpression of Cks1 in human breast carcinoma MCF-7 cells did not significantly reduce p27Kip1 expression, although it conferred resistance to Faslodex (ICI 182780)-mediated inhibition of colony outgrowth in these cells. In contrast, Cks1-depleted MCF-7 cells formed fewer colonies in estrogen-containing medium. Therefore, our studies also suggest that Cks1 levels regulate the responsiveness of ER+ breast cancers to estrogens and anti-estrogens.


Asunto(s)
Quinasas CDC2-CDC28/genética , Carcinoma/inducido químicamente , Carcinoma/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Neoplasias Mamarias Animales/inducido químicamente , Neoplasias Mamarias Animales/genética , Animales , Antineoplásicos Hormonales/farmacología , Quinasas CDC2-CDC28/metabolismo , Carcinógenos , Carcinoma/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Resistencia a Antineoplásicos/genética , Estradiol/análogos & derivados , Estradiol/farmacología , Estrógenos/farmacología , Femenino , Fulvestrant , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Mamarias Animales/metabolismo , Metilnitrosourea , Ratones , Ratones Transgénicos , Ratas , Ratas Sprague-Dawley , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Células Tumorales Cultivadas
20.
Prostate ; 69(10): 1055-66, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19301304

RESUMEN

BACKGROUND: Although anti-angiogenic therapy is a promising new line of therapy for prostate cancer, we recently reported that stable expression of endostatin arrested the progression of prostate cancer to poorly differentiated state and distant metastasis in TRAMP mice. However, the same therapy failed to provide any benefit when given either during or after the onset of metastatic switch. The present study determined the possible mechanisms behind the selective advantage of endostatin therapy in early-stage disease. METHODS: Angiogenesis-related gene expression analysis was performed to identify target genes and molecular pathways involved in the therapy effects. Based on the results from in vivo studies, and recapitulation of the in vivo data in vitro using tumorigenic and non-tumorigenic human prostate cancer cells that are either androgen-sensitive or androgen-independent, analyses of possible mechanisms of the selective advantage of early treatment were performed using assays for cell proliferation, apoptosis, migration, and cell signaling. The identified mechanisms were further confirmed in vivo. RESULTS: Results indicated that cells with high androgen receptor (AR) expression were more sensitive to endostatin treatment than androgen-independent cells with low or no AR expression. Endostatin was found to significantly downregulate the expression of growth factors, receptor tyrosine kinases, proteases, and AR both in vitro and in vivo only when the cells express high-levels of AR. Cell proliferation was not influenced by endostatin treatment but migration was significantly affected only in androgen-sensitive cells. Targeted downregulation of AR prior to endostatin treatment in androgen-sensitive cells and overexpression of AR in androgen-independent cells indicated that the effect of endostatin via AR downregulation is mediated by a non-genotropic mechanism on Ras and RhoA pathways, and independently of AR on MAPK/ERK pathway. CONCLUSIONS: These data indicate that systemically stable endostatin expression delays the onset of metastatic switch by acting on multiple pathways involving AR.


Asunto(s)
Endostatinas/fisiología , Neovascularización Patológica/genética , Neovascularización Patológica/terapia , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/terapia , Receptores Androgénicos/biosíntesis , Receptores Androgénicos/genética , Angiostatinas/uso terapéutico , Animales , Apoptosis/genética , Línea Celular Tumoral , Regulación hacia Abajo/genética , Endostatinas/administración & dosificación , Endostatinas/biosíntesis , Endostatinas/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Patológica/patología , Neoplasias de la Próstata/patología , Receptores Androgénicos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA