Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Transl Med ; 13: 119, 2015 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-25890290

RESUMEN

OBJECTIVES: In Qataris, a population characterized by a small size and a high rate of consanguinity, between two-thirds to three-quarters of adults are overweight or obese. We investigated the relevance of 23 obesity-related loci in the Qatari population. METHODS: Eight-hundred-four individuals assessed to be third generation Qataris were included in the study and assigned to 3 groups according to their body mass index (BMI): 190 lean (BMI < 25 kg/m(2)); 131 overweight (25 kg/m(2) ≤ BMI < 30 kg/m(2)) and 483 obese (BMI ≥ 30 kg/m(2)). Genomic DNA was isolated from peripheral blood and genotyped by TaqMan. RESULTS: Two loci significantly associated with obesity in Qataris: the TFAP2B variation (rs987237) (A allele versus G allele: chi-square = 10.3; P = 0.0013) and GNPDA2 variation (rs10938397) (A allele versus G allele: chi-square = 6.15; P = 0.013). The TFAP2B GG genotype negatively associated with obesity (OR = 0.21; P = 0.0031). Conversely, the GNDPA2 GG homozygous genotype associated with higher risk of obesity in subjects of age < 32 years (P = 0.0358). CONCLUSION: We showed a different genetic profile associated with obesity in the Qatari population compared to Western populations. Studying the genetic background of Qataris is of primary importance as the etiology of a given disease might be population-specific.


Asunto(s)
Árabes/genética , Consanguinidad , Sitios Genéticos , Predisposición Genética a la Enfermedad , Obesidad/genética , Adulto , Índice de Masa Corporal , Femenino , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Fenotipo , Polimorfismo de Nucleótido Simple/genética , Análisis de Componente Principal , Qatar , Grupos Raciales/genética , Delgadez/genética
2.
Blood ; 125(2): 261-72, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25395422

RESUMEN

Donor-derived immunity against tumor-associated antigens (TAAs) may exert selective antileukemic activity reprieving the allogeneic recipient from graft-versus-host disease. As TAAs are highly expressed in placental tissues we hypothesized that pregnancy could drive respective immunity in healthy individuals. Thus, we investigated the frequency and level of immune responses against clinically relevant TAAs in 114 blood donors and 44 women during their first pregnancy. Quantitative reverse-transcription polymerase chain reaction was employed to detect low levels of interferon-γ after primary peptide stimulation of CD8(+) T lymphocytes. In blood donors, primary immune responses of low and/or high avidity were found against WT1 (15%), MUC1 (14%), PRAME (7%), and HER2/neu (5%) and exerted killing functions against leukemic cells. Men had higher responses than women, likely due to gonadal cancer-testis-antigen expression. Interestingly, a history of prior delivery was not associated with increased responses, whereas the strongest responses during pregnancy were found in early trimesters to disappear after delivery. This boost and loss of TAA-specific immunity suggests that virtually every donor harbors the potential to mount antileukemic immune responses in a recipient. However, in the absence of the driving target and a permissive environment, they are short-lived and thus require supplemental strategies such as vaccination or immunomodulation to facilitate their persistence.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inmunoterapia , Embarazo/inmunología , Adulto , Linfocitos T CD8-positivos/inmunología , Estudios Transversales , Femenino , Enfermedad Injerto contra Huésped/inmunología , Antígeno HLA-A2/inmunología , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Adulto Joven
3.
Mol Oncol ; 9(1): 93-104, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25174651

RESUMEN

BACKGROUND: The existence of a dichotomy between immunologically active and quiescent tumor phenotypes has been recently recognized in several types of cancer. The activation of a Th1 type of immune signature has been shown to confer better prognosis and likelihood to respond to immunotherapy. However, whether such dichotomy depends on the genetic make-up of individual cancers is not known yet. BRAF and NRAS mutations are commonly acquired during melanoma progression. Here we explored the role of BRAF and NRAS mutations in influencing the immune phenotype based on a classification previously identified by our group. METHODS: One-hundred-thirteen melanoma metastases underwent microarray analysis and BRAF and NRAS genotyping. Allele-specific PCR was also performed in order to exclude low-frequency mutations. RESULTS: Comparison between BRAF and NRAS mutant versus wild type samples identified mostly constituents or regulators of MAPK and related pathways. When testing gene lists discriminative of BRAF, NRAS and MAPK alterations, we found that 112 BRAF-specific transcripts were able to distinguish the two immune-related phenotypes already described in melanoma, with the poor phenotype associated mostly with BRAF mutation. Noteworthy, such association was stronger in samples displaying low BRAF mRNA expression. However, when testing NRAS mutations, we were not able to find the same association. CONCLUSION: This study suggests that BRAF mutation-related specific transcripts associate with a poor phenotype in melanoma and provide a nest for further investigation.


Asunto(s)
Melanoma/genética , Melanoma/inmunología , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/inmunología , Línea Celular Tumoral , Femenino , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Humanos , Masculino , Melanoma/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Células TH1/inmunología , Células TH1/patología
4.
PLoS One ; 8(2): e56153, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23441164

RESUMEN

UNLABELLED: Hepatocellular carcinomas (HCCs) are a heterogeneous group of tumors that differ in risk factors and genetic alterations. In Italy, particularly Southern Italy, chronic hepatitis C virus (HCV) infection represents the main cause of HCC. Using high-density oligoarrays, we identified consistent differences in gene-expression between HCC and normal liver tissue. Expression patterns in HCC were also readily distinguishable from those associated with liver metastases. To characterize molecular events relevant to hepatocarcinogenesis and identify biomarkers for early HCC detection, gene expression profiling of 71 liver biopsies from HCV-related primary HCC and corresponding HCV-positive non-HCC hepatic tissue, as well as gastrointestinal liver metastases paired with the apparently normal peri-tumoral liver tissue, were compared to 6 liver biopsies from healthy individuals. Characteristic gene signatures were identified when normal tissue was compared with HCV-related primary HCC, corresponding HCV-positive non-HCC as well as gastrointestinal liver metastases. Pathway analysis classified the cellular and biological functions of the genes differentially expressed as related to regulation of gene expression and post-translational modification in HCV-related primary HCC; cellular Growth and Proliferation, and Cell-To-Cell Signaling and Interaction in HCV-related non HCC samples; Cellular Growth and Proliferation and Cell Cycle in metastasis. Also characteristic gene signatures were identified of HCV-HCC progression for early HCC diagnosis. CONCLUSIONS: A diagnostic molecular signature complementing conventional pathologic assessment was identified.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Perfilación de la Expresión Génica , Hepacivirus/patogenicidad , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Carcinoma Hepatocelular/virología , Análisis por Conglomerados , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Hígado/metabolismo , Hígado/virología , Neoplasias Hepáticas/virología , Metástasis de la Neoplasia , Transducción de Señal
5.
Hum Gene Ther Methods ; 23(5): 285-96, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23131031

RESUMEN

Vaccinia virus (VACV) has emerged as an attractive tool in oncolytic virotherapy. VACV replication efficiency plays a crucial role in the therapeutic outcome. However, little is known about the influence of host factors on viral replication efficiency and permissiveness of a host cell line to infection and oncolysis. In this study, replication of the attenuated VACV GLV-1h68 strain and three wild-type VACV isolates was determined in two autologous human melanoma cell lines (888-MEL and 1936-MEL). Host gene expression and viral gene expression in infected cells were evaluated via respective expression array platforms. Microarray analyses followed by sequential statistical approaches characterized human genes that change specifically due to virus infection. Viral gene transcription correlated with viral replication in a time-dependent manner. A set of human genes revealed strong correlations with the respective viral gene expression. Finally we identified a set of human genes with possible predictive value for viral replication in an independent dataset. The results demonstrate a probable correlation between viral replication, early gene expression, and the respective host response, and thus a possible involvement of human host factors in viral early replication. The characterization of human target genes that influence viral replication could help answer the question of host cell permissiveness to oncolytic virotherapy and provide important information for the development of novel recombinant vaccinia viruses with improved features to enhance replication rate and hence trigger therapeutic outcome.


Asunto(s)
Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Virus Oncolíticos/fisiología , Transcripción Genética , Virus Vaccinia/fisiología , Replicación Viral , Línea Celular , Análisis por Conglomerados , Perfilación de la Expresión Génica , Regulación Viral de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Transducción de Señal
6.
Proc Natl Acad Sci U S A ; 109(2): 570-5, 2012 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-22203985

RESUMEN

Because oncogene MET and EGF receptor (EGFR) inhibitors are in clinical development against several types of cancer, including glioblastoma, it is important to identify predictive markers that indicate patient subgroups suitable for such therapies. We investigated in vivo glioblastoma models characterized by hepatocyte growth factor (HGF) autocrine or paracrine activation, or by MET or EGFR amplification, for their susceptibility to MET inhibitors. HGF autocrine expression correlated with high phospho-MET levels in HGF autocrine cell lines, and these lines showed high sensitivity to MET inhibition in vivo. An HGF paracrine environment may enhance glioblastoma growth in vivo but did not indicate sensitivity to MET inhibition. EGFRvIII amplification predicted sensitivity to EGFR inhibition, but in the same tumor, increased copies of MET from gains of chromosome 7 did not result in increased MET activity and did not predict sensitivity to MET inhibitors. Thus, HGF autocrine glioblastoma bears an activated MET signaling pathway that may predict sensitivity to MET inhibitors. Moreover, serum HGF levels may serve as a biomarker for the presence of autocrine tumors and their responsiveness to MET therapeutics.


Asunto(s)
Comunicación Autocrina/fisiología , Biomarcadores/metabolismo , Receptores ErbB/metabolismo , Glioblastoma/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Biomarcadores/sangre , Western Blotting , Línea Celular Tumoral , Análisis por Conglomerados , Hibridación Genómica Comparativa , Cartilla de ADN/genética , Ensayo de Inmunoadsorción Enzimática , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/sangre , Factor de Crecimiento de Hepatocito/sangre , Humanos , Hibridación Fluorescente in Situ , Análisis por Micromatrices , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Piridazinas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Triazoles/farmacología
7.
BMC Cancer ; 11: 451, 2011 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-22011439

RESUMEN

BACKGROUND: Oncolytic viral therapy represents an alternative therapeutic strategy for the treatment of cancer. We previously described GLV-1h68, a modified Vaccinia Virus with exclusive tropism for tumor cells, and we observed a cell line-specific relationship between the ability of GLV-1h68 to replicate in vitro and its ability to colonize and eliminate tumor in vivo. METHODS: In the current study we surveyed the in vitro permissivity to GLV-1h68 replication of the NCI-60 panel of cell lines. Selected cell lines were also tested for permissivity to another Vaccinia Virus and a vesicular stomatitis virus (VSV) strain. In order to identify correlates of permissity to viral infection, we measured transcriptional profiles of the cell lines prior infection. RESULTS: We observed highly heterogeneous permissivity to VACV infection amongst the cell lines. The heterogeneity of permissivity was independent of tissue with the exception of B cell derivation. Cell lines were also tested for permissivity to another Vaccinia Virus and a vesicular stomatitis virus (VSV) strain and a significant correlation was found suggesting a common permissive phenotype. While no clear transcriptional pattern could be identified as predictor of permissivity to infection, some associations were observed suggesting multifactorial basis permissivity to viral infection. CONCLUSIONS: Our findings have implications for the design of oncolytic therapies for cancer and offer insights into the nature of permissivity of tumor cells to viral infection.


Asunto(s)
Vectores Genéticos/fisiología , Virus Oncolíticos/fisiología , Virus Vaccinia/fisiología , Tropismo Viral , Línea Celular Tumoral , Análisis por Conglomerados , Medios de Cultivo , Perfilación de la Expresión Génica , Regulación Viral de la Expresión Génica , Genes Reporteros , Células HT29 , Humanos , Transcripción Genética , Vesiculovirus/fisiología , Replicación Viral
8.
J Transl Med ; 9: 170, 2011 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-21978632

RESUMEN

BACKGROUND: Over 90% of low risk (LR) neuroblastoma patients survive whereas less than 30% of high risk (HR) patients are long term survivors. Age (children younger than 18 months old) is associated with LR disease. Considering that adaptive immune system is well developed in older children, and that T cells were shown to be involved in tumor escape and progression of cancers, we sought to determine whether HR patients may tend to show a signature of adaptive immune responses compared to LR patients who tend to have diminished T-cell responses but an intact innate immune response. METHODS: We performed microarray analysis of RNA extracted from the tumor specimens of HR and LR patients. Flow cytometry was performed to determine the cellular constituents in the blood while multiplex cytokine array was used to detect the cytokine profile in patients' sera. A HR tumor cell line, SK-N-SH, was also used for detecting the response to IL-1ß, a cytokines which is involved in the innate immune responses. RESULTS: Distinct patterns of gene expression were detected in HR and LR patients indicating an active T-cell response and a diminished adaptive immune response, respectively. A diminished adaptive immune response in LR patients was evident by higher levels of IL-10 in the sera. In addition, HR patients had lower levels of circulating myeloid derived suppressor cells (MDSC) compared with a control LR patient. LR patients showed slightly higher levels of cytokines of the innate immune responses. Treatment of the HR tumor line with IL-1ß induced expression of cytokines of the innate immune responses. CONCLUSIONS: This data suggests that adaptive immune responses may play an important role in the progression of HR disease whereas innate immune responses may be active in LR patients.


Asunto(s)
Perfilación de la Expresión Génica , Inmunidad/genética , Neuroblastoma/genética , Neuroblastoma/inmunología , Inmunidad Adaptativa/genética , Línea Celular Tumoral , Niño , Preescolar , Citocinas/sangre , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Lactante , Recién Nacido , Mediadores de Inflamación/metabolismo , Masculino , Células Mieloides/inmunología , Neuroblastoma/sangre , Factores de Riesgo
9.
BMC Cancer ; 11: 68, 2011 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-21320309

RESUMEN

BACKGROUND: In principle, the elimination of malignancies by oncolytic virotherapy could proceed by different mechanisms--e.g. tumor cell specific oncolysis, destruction of the tumor vasculature or an anti-tumoral immunological response. In this study, we analyzed the contribution of these factors to elucidate the responsible mechanism for regression of human breast tumor xenografts upon colonization with an attenuated vaccinia virus (VACV). METHODS: Breast tumor xenografts were analyzed 6 weeks post VACV infection (p.i.; regression phase) by immunohistochemistry and mouse-specific expression arrays. Viral-mediated oncolysis was determined by tumor growth analysis combined with microscopic studies of intratumoral virus distribution. The tumor vasculature was morphologically characterized by diameter and density measurements and vessel functionality was analyzed by lectin perfusion and extravasation studies. Immunological aspects of viral-mediated tumor regression were studied in either immune-deficient mouse strains (T-, B-, NK-cell-deficient) or upon cyclophosphamide-induced immunosuppression (MHCII+-cell depletion) in nude mice. RESULTS: Late stage VACV-infected breast tumors showed extensive necrosis, which was highly specific to cancer cells. The tumor vasculature in infected tumor areas remained functional and the endothelial cells were not infected. However, viral colonization triggers hyperpermeability and dilatation of the tumor vessels, which resembled the activated endothelium in wounded tissue. Moreover, we demonstrated an increased expression of genes involved in leukocyte-endothelial cell interaction in VACV-infected tumors, which orchestrate perivascular inflammatory cell infiltration. The immunohistochemical analysis of infected tumors displayed intense infiltration of MHCII-positive cells and colocalization of tumor vessels with MHCII+/CD31+ vascular leukocytes. However, GI-101A tumor growth analysis upon VACV-infection in either immunosuppressed nude mice (MHCII+-cell depleted) or in immune-deficient mouse strains (T-, B-, NK-cell-deficient) revealed that neither MHCII-positive immune cells nor T-, B-, or NK cells contributed significantly to VACV-mediated tumor regression. In contrast, tumors of immunosuppressed mice showed enhanced viral spreading and tumor necrosis. CONCLUSIONS: Taken together, these results indicate that VACV-mediated oncolysis is the primary mechanism of tumor shrinkage in the late regression phase. Neither the destruction of the tumor vasculature nor the massive VACV-mediated intratumoral inflammation was a prerequisite for tumor regression. We propose that approaches to enhance viral replication and spread within the tumor microenvironment should improve therapeutical outcome.


Asunto(s)
Neoplasias Mamarias Experimentales/terapia , Neoplasias Mamarias Experimentales/virología , Viroterapia Oncolítica/métodos , Virus Vaccinia/fisiología , Animales , Vasos Sanguíneos/patología , Vasos Sanguíneos/fisiopatología , Permeabilidad Capilar/fisiología , Línea Celular , Línea Celular Tumoral , Análisis por Conglomerados , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Leucocitos/metabolismo , Leucocitos/patología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Desnudos , Virus Oncolíticos/fisiología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Trasplante Heterólogo , Vasodilatación
10.
Breast Cancer Res Treat ; 126(2): 385-94, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20480224

RESUMEN

Studies conducted in animal model of infectious diseases or H-Y antigen model suggest a crucial role for CD4+ T cells in providing help for CD8+ T-cell memory responses. This concept suggests that inclusion of T helper epitopes in vaccine formulation will result in improved CD8+ T-cell responses. Although this concept has been applied to cancer vaccine design, the role of CD4+ T cells in the memory differentiation of CD8+ T cells and retention of their anti-tumor function have never been tested in breast cancer model. Using the FVB mouse model of neu-positive breast carcinoma we report for the first time that helpless T cells showed cytostatic or tumor inhibitory effects during primary tumor challenge whereas, helped T cells showed cytotoxic effects and resulted in complete tumor rejection. Such differential effects, in vivo, were associated with higher frequency of CD8+PD-L1+ and CD8+PD-1+ T cells in animals harboring helpless T cells as well as higher titer of IL-2 in the sera of animals harboring helped T cells. However, depletion of CD4+ T cells did not alter the ability of neu-specific CD8+ T cells to differentiate into memory cells and to retain their effector function against the tumor during recall challenge. These results suggest the inhibitory role of CD4+ T cells on CD8+ T-cell exhaustion without substantial effects on the differentiation of memory T cells during priming phase of the immune responses against breast cancer.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Reactividad Cruzada , Receptor ErbB-2/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/fisiología , Supervivencia Celular/inmunología , Técnicas de Cocultivo , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Receptores de Hialuranos/metabolismo , Interferón gamma/metabolismo , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Fenotipo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Telómero/metabolismo , Carga Tumoral/inmunología
11.
Adv Exp Med Biol ; 684: 178-88, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20795549

RESUMEN

Over the past several years, progress in the field of tumor immunology has lead to advances in active immunotherapy and vaccination as a means ofeliciting tumor-specific immune responses to mediate tumor regression and clearance. Developing vaccines targeted against cancer became an important focus as a therapy following the success of viral vaccines in preventing infection and disease. In humans with cancer, similar to viral infections, the host immune system is capable of recognizing antigens expressed on tumor cells. This similarity allows the immunological framework of the viral vaccine to be adapted to the cancer setting in hopes of enhancing human T-cell reactivity against tumor. It is generally believed that a requirement for tumor destruction to occur is the induction of sufficient levels of immune cells with high avidity for recognition of tumor antigens. Moreover, the cells must be targeted to the tumor site and be capable of infiltrating tumor stroma.2 Several tumor-associated antigens (TAA) have been identified in the melanoma model which has allowed for immunization trials to evaluate therapeutic potential of tumor-specific T-cell induction. Some clinical trials reported limited success ofT-cell mediated tumor rejection, reportingpartial or complete regression in 10 to 30% of patients. Although tumor regression was not observed following active immunization in vivo, ex vivo assays evaluating TAA-specific T cells demonstrated tumor recognition and subsequent T-cell activation suggesting that tumor-specific T-cell induction indeed occurs but alone is not adequate to induce tumor regression. Recently, the usefulness and success of active-specific immunization (ASI) against TAAs as a means ofeliciting a tumor-specific immune response leading to tumor regression and clearance has been a topic of debate and discussion.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Activación de Linfocitos/inmunología , Neoplasias/inmunología , Linfocitos T/inmunología , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/uso terapéutico , Ensayos Clínicos como Asunto , Humanos , Inmunoterapia Activa , Neoplasias/patología , Neoplasias/prevención & control
12.
J Exp Med ; 207(8): 1625-36, 2010 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-20624890

RESUMEN

Signaling through the adaptor protein myeloid differentiation factor 88 (MyD88) promotes carcinogenesis in several cancer models. In contrast, MyD88 signaling has a protective role in the development of azoxymethane (AOM)/dextran sodium sulfate (DSS) colitis-associated cancer (CAC). The inability of Myd88(-/-) mice to heal ulcers generated upon injury creates an altered inflammatory environment that induces early alterations in expression of genes encoding proinflammatory factors, as well as pathways regulating cell proliferation, apoptosis, and DNA repair, resulting in a dramatic increase in adenoma formation and progression to infiltrating adenocarcinomas with frequent clonal mutations in the beta-catenin gene. Others have reported that toll-like receptor (Tlr) 4-deficient mice have a similar susceptibility to colitis to Myd88-deficient mice but, unlike the latter, are resistant to CAC. We have observed that mice deficient for Tlr2 or Il1r do not show a differential susceptibility to colitis or CAC. However, upon AOM/DSS treatment Il18(-/-) and Il18r1(-/-) mice were more susceptible to colitis and polyp formation than wild-type mice, suggesting that the phenotype of Myd88(-/-) mice is, in part, a result of their inability to signal through the IL-18 receptor. This study revealed a previously unknown level of complexity surrounding MyD88 activities downstream of different receptors that impact tissue homeostasis and carcinogenesis.


Asunto(s)
Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Interleucina-18/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal/fisiología , Adenocarcinoma/inducido químicamente , Adenocarcinoma/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Azoximetano/farmacología , Proliferación Celular/efectos de los fármacos , Colon/efectos de los fármacos , Colon/metabolismo , Colon/patología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/genética , Pólipos del Colon/patología , Ciclooxigenasa 2/genética , Enzimas Reparadoras del ADN/genética , Sulfato de Dextran/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad/genética , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Interleucina-18/genética , Subunidad alfa del Receptor de Interleucina-18/genética , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Fosforilación/efectos de los fármacos , Receptores Tipo I de Interleucina-1/genética , Factor de Transcripción STAT3/genética , Organismos Libres de Patógenos Específicos , beta Catenina/genética
13.
J Transl Med ; 8: 10, 2010 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-20113473

RESUMEN

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) remains a leading cause of cancer mortality for which novel gene therapy approaches relying on tumor-tropic adenoviruses are being tested. METHODS: We obtained the global transcriptional profiling of primary PDAC using RNA from eight xenografted primary PDAC, three primary PDAC bulk tissues, three chronic pancreatitis and three normal pancreatic tissues. The Affymetrix GeneChip HG-U133A was used. The results of the expression profiles were validated applying immunohistochemical and western blot analysis on a set of 34 primary PDAC and 10 established PDAC cell lines. Permissivity to viral vectors used for gene therapy, Adenovirus 5 and Adeno-Associated Viruses 5 and 6, was assessed on PDAC cell lines. RESULTS: The analysis of the expression profiles allowed the identification of two clearly distinguishable phenotypes according to the expression of interferon-stimulated genes. The two phenotypes could be readily recognized by immunohistochemical detection of the Myxovirus-resistance A protein, whose expression reflects the activation of interferon dependent pathways. The two molecular phenotypes discovered in primary carcinomas were also observed among established pancreatic adenocarcinoma cell lines, suggesting that these phenotypes are an intrinsic characteristic of cancer cells independent of their interaction with the host's microenvironment. The two pancreatic cancer phenotypes are characterized by different permissivity to viral vectors used for gene therapy, as cell lines expressing interferon stimulated genes resisted to Adenovirus 5 mediated lysis in vitro. Similar results were observed when cells were transduced with Adeno-Associated Viruses 5 and 6. CONCLUSION: Our study identified two molecular phenotypes of pancreatic cancer, characterized by a differential expression of interferon-stimulated genes and easily recognized by the expression of the Myxovirus-resistance A protein. We suggest that the detection of these two phenotypes might help the selection of patients enrolled in virally-mediated gene therapy trials.


Asunto(s)
Adenocarcinoma , Adenoviridae , Carcinoma Ductal Pancreático , Regulación Neoplásica de la Expresión Génica , Terapia Genética/métodos , Inmunidad/fisiología , Neoplasias Pancreáticas , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma/terapia , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/terapia , Línea Celular Tumoral , Dependovirus/genética , Dependovirus/metabolismo , Perfilación de la Expresión Génica , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Interferones/inmunología , Ratones , Ratones Desnudos , Análisis por Micromatrices/métodos , Trasplante de Neoplasias , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia , Selección de Paciente , Fenotipo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción Genética , Trasplante Heterólogo
14.
Proc Natl Acad Sci U S A ; 107(2): 803-8, 2010 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-20080756

RESUMEN

Limited responsiveness to IFN-alpha in hepatitis C virus (HCV)-infected African-Americans compared to European Americans (AAs vs. EAs) hinders the management of HCV. Here, we studied healthy non-HCV-infected AA and EA subjects to test whether immune cell response to IFN-alpha is determined directly by race. We compared baseline and IFN-alpha-induced signal transducer and activator of transcription (STAT)-1, STAT-2, STAT-3, STAT-4, and STAT-5 protein and phosphorylation levels in purified T cells, global transcription, and a genomewide single-nucleotide polymorphism (SNP) profile of healthy AA and EA blood donors. In contrast to HCV-infected individuals, healthy AAs displayed no evidence of reduced STAT activation or IFN-alpha-stimulated gene expression compared to EAs. Although >200 genes reacted to IFN-alpha treatment, race had no impact on any of them. The only gene differentially expressed by the two races (NUDT3, P < 10(-7)) was not affected by IFN-alpha and bears no known relationship to IFN-alpha signaling or HCV pathogenesis. Genomewide analysis confirmed the self-proclaimed racial attribution of most donors, and numerous race-associated SNPs were identified within loci involved in IFN-alpha signaling, although they clearly did not affect responsiveness in the absence of HCV. We conclude that racial differences observed in HCV-infected patients in the responsiveness to IFN-alpha are unrelated to inherent racial differences in IFN-alpha signaling and more likely due to polymorphisms affecting the hosts' response to HCV, which in turn may lead to a distinct disease pathophysiology responsible for altered IFN signaling and treatment response.


Asunto(s)
Hepatitis C/genética , Interferón-alfa/genética , Grupos Raciales/genética , Adulto , Población Negra/genética , Secuencia Conservada , Femenino , Citometría de Flujo , Amplificación de Genes , Perfilación de la Expresión Génica , Genoma Humano , Hepacivirus/genética , Hepacivirus/inmunología , Humanos , Interferón-alfa/fisiología , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , ARN Viral/genética , Valores de Referencia , Transducción de Señal/genética , Población Blanca/genética
15.
BMC Genomics ; 10: 301, 2009 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-19583830

RESUMEN

BACKGROUND: GLV-1h68 is an attenuated recombinant vaccinia virus (VACV) that selectively colonizes established human xenografts inducing their complete regression. RESULTS: Here, we explored xenograft/VACV/host interactions in vivo adopting organism-specific expression arrays and tumor cell/VACV in vitro comparing VACV replication patterns. There were no clear-cut differences in vitro among responding and non-responding tumors, however, tumor rejection was associated in vivo with activation of interferon-stimulated genes (ISGs) and innate immune host's effector functions (IEFs) correlating with VACV colonization of the xenografts. These signatures precisely reproduce those observed in humans during immune-mediated tissue-specific destruction (TSD) that causes tumor or allograft rejection, autoimmunity or clearance of pathogens. We recently defined these common pathways in the "immunologic constant of rejection" hypothesis (ICR). CONCLUSION: This study provides the first prospective validation of a universal mechanism associated with TSD. Thus, xenograft infection by oncolytic VACV, beyond offering a promising therapy of established cancers, may represent a reliable pre-clinical model to test therapeutic strategies aimed at modulating the central pathways leading to TSD; this information may lead to the identification of principles that could refine the treatment of cancer and chronic infection by immune stimulation or autoimmunity and allograft rejection through immune tolerance.


Asunto(s)
Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Virus Vaccinia/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Línea Celular Tumoral , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Desnudos , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de Secuencia de ADN
16.
IDrugs ; 12(5): 297-301, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19431094

RESUMEN

Anticancer vaccines have not matched the clinical expectations projected from their ability to induce consistently systemic anticancer T-cell responses. Thus, a dichotomy is observed between the immunological and clinical endpoints of anticancer immunization. Anticancer vaccines have clearly demonstrated that highly specific T-cell responses can be induced that can recognize autologous cancer antigens in patients with cancer. This ability is an outstanding achievement of modern biotechnology, yielding one of the most specific types of potential anticancer reagents. However, systemic, vaccine-induced anticancer responses exemplify a broader immunological paradox: cytotoxic T-cells can coexist within the same organism with their target cells not only in the context of cancer, but also in the context of chronic infections, well-controlled allo-transplant reactions and autoimmunity. According to this view, anticancer immune responses are a facet of a tissue-specific autoimmune phenomenon specific for cancer tissue that may or may not result in the successful immune-destruction of target cells, depending on an assortment of genetic factors related to the background of the host or evolving phenotypes of a heterogeneous cancer environment. This feature article summarizes the current understanding of the mechanisms leading to tumor rejection in humans as well as in experimental models, in the context of the broader immunological phenomenon leading to tissue-specific destruction. Anticancer vaccines that may not induce clinically significant anticancer responses independently could function as a unique tool to enhance the specificity of the response of the host against cancer, provided that strategies are implemented to amplify the immune reaction initiated by vaccine-induced antibodies and/or T-cells.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Inmunoterapia , Neoplasias , Animales , Formación de Anticuerpos/efectos de los fármacos , Vacunas contra el Cáncer/farmacología , Humanos , Melanoma/inmunología , Melanoma/prevención & control , Melanoma/terapia , Modelos Inmunológicos , Neoplasias/inmunología , Neoplasias/prevención & control , Neoplasias/terapia , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología
17.
Cancer Immunol Immunother ; 58(9): 1355-62, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19266198

RESUMEN

The concept of using replicating oncolytic viruses in cancer therapy dates to the beginning of the twentieth century. However, in the last few years, an increasing number of pre-clinical and clinical trials have been carried out with promising preliminarily results. Novel, indeed, is the suggestion that viral oncolytic therapy might not operate exclusively through an oncolysis-mediated process but additionally requires the "assistance" of the host's immune system. Originally, the host's immune response was believed to play a predominant obstructive role against viral replication, hence limiting the anti-tumor efficacy of viral vectors. Recent data, however, suggest that the immune response may also play a key role in promoting tumor destruction in association with the oncolytic process. In fact, immune effector pathways activated during oncolytic virus-induced tumor rejection seem to follow a similar pattern to those observed when the broader phenomenon of immune-mediated tissue-specific rejection occurs in other immune-related pathologies. We recently formulated the "Immunologic Constant of Rejection" hypothesis, emphasizing commonalties in transcriptional patterns observed when tissue-destruction occurs: whether with a favorable outcome, such as in tumor rejection and pathogen clearance; or a destructive one, such as in allograft rejection or autoimmunity. Here, we propose that a similar mechanism induces clearance of virally infected tumors and that such a mechanism is primarily dependent on innate immune functions.


Asunto(s)
Efecto Citopatogénico Viral/inmunología , Terapia Genética , Neoplasias/inmunología , Neoplasias/terapia , Viroterapia Oncolítica , Poxviridae/genética , Efecto Citopatogénico Viral/genética , Humanos , Neoplasias/genética
18.
J Transl Med ; 7: 22, 2009 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-19321006

RESUMEN

BACKGROUND: Resveratrol (RES), an estrogen analog, is considered as a potential cancer chemo-preventive agent. However, it remains unclear how RES is transported into cells. In this study, we observed that Caveolin-1(CAV1) expression can increase the cytotoxic and pro-apoptotic activity of RES in a dose- and time-dependent manner both in vitro and in vivo in a Hepatocellular Carcinoma animal model. METHODS: High performance liquid chromatography (HPLC) demonstrated that RES intra-cellular concentration is increased about 2-fold in cells stably expressing CAV1 or CAVM1 (a scaffolding domain (81-101AA)-defective CAV1 mutant) compared to the untransduced human Hepatoblastoma cell line (HepG2) or after transduction with the green fluorescent protein (GFP) control vector. The increased intra-cellular transport of RES was abolished in cells stably expressing CAVM2 (a cholesterol shuttle domain (143-156AA)-defective CAV1 mutant) or CAVRNAi. In order to further characterize CAV1-dependent RES transport, we synthesized RES-dansyl chloride derivatives as fluorescent probes to visualize the transport process, which demonstrated a distribution consistent with that of CAV1 in HepG2 cells. RESULTS: In addition, RES endocytosis was not mediated by estrogen receptor (ER) alpha and beta, as suggested by lack of competitive inhibition by estrogen or Tamoxifen. Pathway analysis showed that RES can up-regulate the expression of endogenous CAV1; this activates further the MAPK pathway and caspase-3 expression. DISCUSSION: This study provides novel insights about the role played by CAV1 in modulating cellular sensitivity to RES through enhancement of its internalization and trafficking.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Caveolina 1/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Estilbenos/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Sinergismo Farmacológico , Endocitosis/efectos de los fármacos , Genes Reporteros , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Ratones Desnudos , Resveratrol , Eliminación de Secuencia , Transfección
19.
Trends Immunol ; 29(6): 256-62, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18457994

RESUMEN

The complexity underlying a pathologic process does not necessarily require a complex explanation. The biology determining allograft or cancer rejection, autoimmunity or tissue damage during pathogen infections is complex; however, common patterns are emerging that lead to a common final outcome. For instance, tissue destruction occurs with resolution of the pathogenic process (cancer, infection) or tissue damage and organ failure (autoimmunity, allograft rejection). Observations in humans based on transcriptional profiling converge into what we call an 'immunologic constant of rejection' that characterizes such occurrences. This constant includes the coordinate activation of interferon-stimulated genes (ISGs) and immune effector functions (IEFs). Understanding this final effector pathway may suggest novel strategies for the induction or inhibition of tissue-specific destruction with therapeutic intent in cancer and other immune pathologies.


Asunto(s)
Quimiocinas/inmunología , Citotoxicidad Inmunológica/inmunología , Rechazo de Injerto/inmunología , Hipersensibilidad Tardía , Infecciones/inmunología , Factores Reguladores del Interferón/inmunología , Animales , Quimiocinas/genética , Quimiocinas/metabolismo , Rechazo de Injerto/prevención & control , Humanos , Inmunidad/fisiología , Inmunoterapia/tendencias , Infecciones/microbiología , Infecciones/virología , Inflamación , Factores Reguladores del Interferón/biosíntesis , Factores Reguladores del Interferón/genética , Neoplasias/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T/genética , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo , Activación Transcripcional/inmunología
20.
Cancer Res ; 68(7): 2436-46, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18381452

RESUMEN

We have previously shown T-cell-mediated rejection of the neu-overexpressing mammary carcinoma cells (MMC) in wild-type FVB mice. However, following rejection of primary tumors, a fraction of animals experienced a recurrence of a neu antigen-negative variant (ANV) of MMC (tumor evasion model) after a long latency period. In the present study, we determined that T cells derived from wild-type FVB mice can specifically recognize MMC by secreting IFN-gamma and can induce apoptosis of MMC in vitro. Neu transgenic (FVBN202) mice develop spontaneous tumors and cannot reject it (tumor tolerance model). To dissect the mechanisms associated with rejection or tolerance of MMC tumors, we compared transcriptional patterns within the tumor microenvironment of MMC undergoing rejection with those that resisted it either because of tumor evasion/antigen loss recurrence (ANV tumors) or because of intrinsic tolerance mechanisms displayed by the transgenic mice. Gene profiling confirmed that immune rejection is primarily mediated through activation of IFN-stimulated genes and T-cell effector mechanisms. The tumor evasion model showed combined activation of Th1 and Th2 with a deviation toward Th2 and humoral immune responses that failed to achieve rejection likely because of lack of target antigen. Interestingly, the tumor tolerance model instead displayed immune suppression pathways through activation of regulatory mechanisms that included in particular the overexpression of interleukin-10 (IL-10), IL-10 receptor, and suppressor of cytokine signaling (SOCS)-1 and SOCS-3. These data provide a road map for the identification of novel biomarkers of immune responsiveness in clinical trials.


Asunto(s)
Neoplasias Mamarias Experimentales/inmunología , Recurrencia Local de Neoplasia/inmunología , Receptor ErbB-2/inmunología , Animales , Apoptosis/inmunología , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/inmunología , Línea Celular Tumoral , Femenino , Tolerancia Inmunológica , Inmunoterapia Adoptiva , Interleucina-10/biosíntesis , Interleucina-10/inmunología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/terapia , Ratones , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Ratas , Receptor ErbB-2/genética , Receptores de Interleucina-10/biosíntesis , Receptores de Interleucina-10/inmunología , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/biosíntesis , Proteínas Supresoras de la Señalización de Citocinas/inmunología , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA