Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Oncoimmunology ; 1(5): 659-669, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22934258

RESUMEN

Adenosine deaminase (ADA) is responsible for the deamination of immunosuppressive adenosine to inosine. In human T lymphocytes, ADA is associated with dipeptidyl peptidase IV (CD26). ADA expression and activity were evaluated in regulatory T cells (Treg) and CD4(+) T effector cells (Teff) of patients with head and neck squamous cell cancer (HNSCC). CD4(+)CD39(+) and CD4(+)CD39(neg) T cells were isolated by single-cell sorting from the peripheral blood of 15 HNSCC patients and 15 healthy donors (NC). CD26/ADA expression in these cells was studied by multicolor flow cytometry, confocal microscopy, RT-PCR and immunohistochemistry in tumor tissues. ADA activity was evaluated by mass spectrometry, suppression of Teff proliferation in CFSE assays and cytokine production by Luminex. CD4(+)CD39(+) Treg had low and CD4(+)CD39(neg) Teff high CD26/ADA expression and ADA activity in NC or HNSCC. The frequency and suppressor activity of CD39(+)CD26(neg) Treg were elevated in patients relative to NC (p < 0.01). However, ADA activity in patients' CD4(+)CD39(neg) Teff was decreased (p < 0.05), resulting in extracellular adenosine accumulation. Also, patients' Teff were more sensitive to inhibitory signals delivered via adenosine receptors. IL-2, IL12 and INFγ upregulated ADA expression and activity in CD4(+)CD39(neg) Teff, whereas IL-10, PGE(2) and CADO downregulated it. The differentially expressed CD26/ADA can serve as surface markers for functionally-active CD39(+)CD26(neg) Treg.

2.
Clin Dev Immunol ; 2012: 173029, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22654951

RESUMEN

The role of the tryptophan-catabolizing enzyme, indoleamine 2,3-dioxygenase (IDO1), in tumor escape and metastasis formation was analyzed using two pairs of Ido1+ and Ido1- murine breast cancer cell lines. Ido1 expression in 4T1 cells was knocked down by shRNA, and Ido1 expression in NT-5 cells was upregulated by stable transfection. Growth of Ido1- tumors and spontaneous metastasis formation were inhibited in immunocompetent mice. A higher level of cytotoxic T lymphocytes was generated by spleen cells from mice bearing Ido1- tumors than Ido1+ tumors. Tumor and metastatic growth was enhanced in immunodeficient mice, confirming an intensified immune response in the absence of Ido1 expression. However, Ido1+ tumors grow faster than Ido1- tumors in immunodeficient SCID/beige mice (lacking T, B, and NK cells) suggesting that some Ido1-controlled nonimmunological mechanisms may be involved in tumor cell growth regulation. In vitro experiments demonstrated that downregulation of Ido1 in tumor cells was associated with decreased cell proliferation, increased apoptosis, and changed expression of cell cycle regulatory genes, whereas upregulation of Ido1 in the cells had the opposite effects. Taken together, our findings indicate that Ido1 expression could exert immunological and nonimmunological effects in murine breast tumor cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/patología , Escape del Tumor , Animales , Procesos de Crecimiento Celular , Línea Celular Tumoral , Células Cultivadas , Citotoxicidad Inmunológica , Femenino , Huésped Inmunocomprometido , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Neoplasias Mamarias Experimentales/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones SCID , ARN Interferente Pequeño/genética , Transgenes/genética
3.
Cancer Immunol Immunother ; 60(1): 111-22, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20960188

RESUMEN

Previous studies document that PGE(2) and adenosine suppress production of inflammatory cytokines. The present study demonstrates for the first time that (1) PGE(2) and 2-chloroadenosine (CADO; a stable analog of adenosine) directly inhibit the cytolytic function of human tumor-infiltrating lymphocytes (TILs); (2) the combination PGE(2) and CADO have additive suppressive effects; and (3) the cooperative immunosuppressive actions of PGE(2) and CADO are mediated via EP2 receptors (EP2Rs) and A(2A) receptors (A(2A)Rs) and are due to amplification of cAMP production, activation of protein kinase A (PKA) and T cell receptor (TCR) inhibitor Csk leading to inhibition of Lck, ZAP-70 and Akt phosphorylation. (4) During ex vivo expansion, TILs undergo three stages of differentiation converting from TILs with high cytotoxic activity and relative resistance to combined EP2R/A(2A)R suppression (stage I) to TILs retaining high cytotoxicity and gaining sensitivity to combined suppression (stage II) and then to TILS that are less cytotoxic and very sensitive to combined suppression (stage III). (5) Finally, we find that pretreatment of TILs with non-inhibitory concentrations of EP2R agonists (such as PGE(2) or butaprost) or A(2A)R agonists (such as CADO or CGS21680) increases their cytotoxic activity and induces resistance to EP2R and A(2A)R inhibitory signaling (cross-resistance) due to homologous and heterologous desensitization and internalization of EP2Rs and A(2A)Rs, thus preventing their inhibitory signaling. We conclude that inducing resistance of TILs to the suppressive effects of PGE(2) and adenosine in the tumor microenvironment could represent a novel strategy for improving the efficacy of adoptive immunotherapy.


Asunto(s)
Inmunoterapia Adoptiva , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Melanoma/terapia , Neoplasias Cutáneas/terapia , Subgrupos de Linfocitos T/efectos de los fármacos , Adenosina/análogos & derivados , Adenosina/farmacología , Alprostadil/análogos & derivados , Alprostadil/farmacología , Células Cultivadas , Citotoxicidad Inmunológica/efectos de los fármacos , Dinoprostona/farmacología , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Melanoma/inmunología , Melanoma/patología , Fenetilaminas/farmacología , Receptor Cross-Talk , Receptor de Adenosina A2A/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/agonistas , Transducción de Señal , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología
4.
J Biol Chem ; 285(36): 27571-80, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20558731

RESUMEN

Adaptive regulatory T cells (Tr1) are induced in the periphery upon encountering cognate antigens. In cancer, their frequency is increased; however, Tr1-mediated suppression mechanisms are not yet defined. Here, we evaluate the simultaneous involvement of ectonucleotidases (CD39/CD73) and cyclooxygenase 2 (COX-2) in Tr1-mediated suppression. Human Tr1 cells were generated from peripheral blood mononuclear cell-derived, sorted CD4(+)CD25(-) T cells and incubated with autologous immature dendritic cells, irradiated COX-2(+) or COX-2(-) tumor cells, and IL-2, IL-10, and IL-15 (each at 10-15 IU/ml) for 10 days as described (Bergmann, C., Strauss, L., Zeidler, R., Lang, S., and Whiteside, T. L. (2007) Cancer Immunol. Immunother. 56, 1429-1442). Tr1 were phenotyped by multicolor flow cytometry, and suppression of proliferating responder cells was assessed in carboxyfluorescein diacetate succinimidyl ester-based assays. ATP hydrolysis was measured using a luciferase detection assay, and levels of adenosine or prostaglandin E(2) (PGE(2)) in cell supernatants were analyzed by mass spectrometry or ELISA, respectively. Intracellular cAMP levels were measured by enzyme immunoassay. The COX-2(+) tumor induced a greater number of Tr1 than COX-2(-) tumor (p < 0.05). Tr1 induced by COX-2(+) tumor were more suppressive, hydrolyzed more exogenous ATP (p < 0.05), and produced higher levels of adenosine and PGE(2) (p < 0.05) than Tr1 induced by COX-2(-) tumor. Inhibitors of ectonucleotidase activity, A(2A) and EP(2) receptor antagonists, or an inhibitor of the PKA type I decreased Tr1-mediated suppression (p < 0.05), whereas rolipram, a PDE(4) inhibitor, increased the intracellular cAMP level in responder cells and their susceptibility to Tr1-mediated suppression. Tr1 present in tumors or the peripheral blood of head and neck squamous cell carcinoma patients co-expressed COX-2, CD39, and CD73. A concomitant inhibition of PGE(2) and adenosine via the common intracellular cAMP pathway might be a novel approach for improving results of immune therapies for cancer.


Asunto(s)
Inmunidad Adaptativa/inmunología , Adenosina/inmunología , Dinoprostona/inmunología , Tolerancia Inmunológica/inmunología , Linfocitos T Reguladores/inmunología , 5'-Nucleotidasa/metabolismo , Adenosina/biosíntesis , Aminoácidos Cíclicos/metabolismo , Antígenos CD/metabolismo , Apirasa/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/inmunología , Línea Celular Tumoral , Ciclooxigenasa 2/deficiencia , Ciclooxigenasa 2/metabolismo , Dinoprostona/biosíntesis , Regulación hacia Abajo/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/inmunología , Humanos , Espacio Intracelular/metabolismo , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo
5.
J Clin Oncol ; 28(13): 2159-66, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20368574

RESUMEN

PURPOSE: Early detection of ovarian cancer has great promise to improve clinical outcome. PATIENTS AND METHODS: Ninety-six serum biomarkers were analyzed in sera from healthy women and from patients with ovarian cancer, benign pelvic tumors, and breast, colorectal, and lung cancers, using multiplex xMAP bead-based immunoassays. A Metropolis algorithm with Monte Carlo simulation (MMC) was used for analysis of the data. RESULTS: A training set, including sera from 139 patients with early-stage ovarian cancer, 149 patients with late-stage ovarian cancer, and 1,102 healthy women, was analyzed with MMC algorithm and cross validation to identify an optimal biomarker panel discriminating early-stage cancer from healthy controls. The four-biomarker panel providing the highest diagnostic power of 86% sensitivity (SN) for early-stage and 93% SN for late-stage ovarian cancer at 98% specificity (SP) was comprised of CA-125, HE4, CEA, and VCAM-1. This model was applied to an independent blinded validation set consisting of sera from 44 patients with early-stage ovarian cancer, 124 patients with late-stage ovarian cancer, and 929 healthy women, providing unbiased estimates of 86% SN for stage I and II and 95% SN for stage III and IV disease at 98% SP. This panel was selective for ovarian cancer showing SN of 33% for benign pelvic disease, SN of 6% for breast cancer, SN of 0% for colorectal cancer, and SN of 36% for lung cancer. CONCLUSION: A panel of CA-125, HE4, CEA, and VCAM-1, after additional validation, could serve as an initial stage in a screening strategy for epithelial ovarian cancer.


Asunto(s)
Biomarcadores de Tumor/sangre , Inmunoensayo , Tamizaje Masivo/métodos , Neoplasias Ováricas/diagnóstico , Anciano , Anciano de 80 o más Años , Algoritmos , Antígeno Ca-125/sangre , Antígeno Carcinoembrionario/sangre , Interpretación Estadística de Datos , Detección Precoz del Cáncer , Proteínas Secretorias del Epidídimo/análisis , Femenino , Humanos , Inmunoensayo/estadística & datos numéricos , Tamizaje Masivo/estadística & datos numéricos , Persona de Mediana Edad , Método de Montecarlo , Estadificación de Neoplasias , Neoplasias Ováricas/sangre , Neoplasias Ováricas/patología , Valor Predictivo de las Pruebas , Pronóstico , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Molécula 1 de Adhesión Celular Vascular/sangre , beta-Defensinas
6.
Leuk Res ; 34(7): 917-24, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20116850

RESUMEN

We used a novel high-throughput drug screening assay, based on Luminex technology, to identify anti-myeloma agents capable of inhibiting cytokines and growth factors essential for multiple myeloma (MM) from a chemical library of 1120 compounds provided by MMRF. Tetracycline derivatives inhibited MM cell proliferation and osteoclast activating factors without obvious effect on cell viability. Steroid compounds specifically decreased angiogenesis-related factors, but stimulated osteoclast activating factors. Antihelmintic drugs potently inhibited cytokines and were cytotoxic. The screen identified potential candidates with potent anti-MM properties that need further investigation.


Asunto(s)
Antineoplásicos/farmacología , Citocinas/antagonistas & inhibidores , Ensayos de Selección de Medicamentos Antitumorales/métodos , Inhibidores de Crecimiento/farmacología , Ensayos Analíticos de Alto Rendimiento/métodos , Inmunoensayo/métodos , Mieloma Múltiple/tratamiento farmacológico , Corticoesteroides/farmacología , Antihelmínticos/farmacología , Antiinfecciosos/farmacología , Antineoplásicos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Glicósidos Cardíacos/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Citocinas/metabolismo , Inhibidores de Crecimiento/aislamiento & purificación , Humanos , Microesferas , Osteoclastos/citología
7.
Anticancer Agents Med Chem ; 10(2): 164-71, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20184538

RESUMEN

Despite significant efforts in diagnosing and treating lung cancer, therapeutic resistance remains a major unresolved clinical and scientific problem. Cancer stem cells (CSCs) are thought to be responsible for the failure of current chemotherapy of lung cancer. The concept of CSCs has radically changed the view of cancer therapy. Today a majority of current treatment modalities target the differentiated cancer cells and avoid the drug resistant cancer-initiating stem cells. This review summarizes our understanding of lung CSCs and their role in metastasis formation and growth of non- small-cells lung cancer (NSCLC). High tumorigenic and metastatic properties of lung CSCs are associated with the efficient cytokine network production and with the specific signaling pathways. This review underlines the experimental evidence indicating that the stem cell factor (SCF) and its receptor c-kit (CD117) play an important role in survival and proliferation of lung CSCs. Thus, molecularly targeting key cytokine network axes of such highly tumorigenic and metastatic CSCs must be considered for improving the current anti-cancer strategy efficacy. Standard chemotherapy in combination with specific axis of cytokine network targeting, such as SCF-c-kit, could eliminate both bulk tumor cells and CSCs, and therefore to be truly curative therapies. This review provides a summary of some of the developments in the field of lung CSCs targeting and highlights aspects which could help in the drug discovery process.


Asunto(s)
Antineoplásicos/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Neoplasias Pulmonares/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Citocinas/genética , Citocinas/metabolismo , Citocinas/fisiología , Sistemas de Liberación de Medicamentos/tendencias , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Neoplasias Pulmonares/patología , Células Madre Neoplásicas/patología , Transducción de Señal/genética
8.
J Biol Chem ; 285(10): 7176-86, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-19858205

RESUMEN

Naturally occurring regulatory T cells (nTreg) are crucial for maintaining tolerance to self and thus preventing autoimmune diseases and allograft rejections. In cancer, Treg down-regulate antitumor responses by several distinct mechanisms. This study analyzes the role the adenosinergic pathway plays in suppressive activities of human nTreg. Human CD4(+)CD25(high)FOXP3(+) Treg overexpress CD39 and CD73, ectonucleotidases sequentially converting ATP into AMP and adenosine, which then binds to A(2a) receptors on effector T cells, suppressing their functions. CD4(+)CD39(+) and CD4(+)CD25(high) T cells express low levels of adenosine deaminase (ADA), the enzyme responsible for adenosine breakdown, and of CD26, a surface-bound glycoprotein associated with ADA. In contrast, T effector cells are enriched in CD26/ADA but express low levels of CD39 and CD73. Inhibitors of ectonucleotidase activity (e.g. ARL67156) and antagonists of the A(2a) receptor (e.g. ZM241385) blocked Treg-mediated immunosuppression. The inhibition of ADA activity on effector T cells enhanced Treg-mediated immunosuppression. Thus, human nTreg characterized by the presence of CD39 and the low expression of CD26/ADA are responsible for the generation of adenosine, which plays a major role in Treg-mediated immunosuppression. The data suggest that the adenosinergic pathway represents a potential therapeutic target for regulation of immunosuppression in a broad variety of human diseases.


Asunto(s)
Adenosina/inmunología , Linfocitos T CD4-Positivos/inmunología , Factores de Transcripción Forkhead/inmunología , Inmunosupresores/inmunología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , 5'-Nucleotidasa/inmunología , Adenosina/química , Adenosina/farmacología , Adenosina Desaminasa/metabolismo , Adulto , Animales , Antígenos CD/inmunología , Apirasa/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de los fármacos , Dipeptidil Peptidasa 4/inmunología , Femenino , Citometría de Flujo , Proteínas Ligadas a GPI , Humanos , Masculino , Persona de Mediana Edad , Receptor de Adenosina A2A/metabolismo , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/efectos de los fármacos , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/efectos de los fármacos , Adulto Joven
9.
Cancer Res ; 70(1): 338-46, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20028869

RESUMEN

Cancer stem cells (CSC) are thought to be responsible for tumor initiation and tumor regeneration after chemotherapy. Previously, we showed that chemotherapy of non-small cell lung cancer (NSCLC) cells lines can select for outgrowth of highly tumorigenic and metastatic CSCs. The high malignancy of lung CSCs was associated with an efficient cytokine network. In this study, we provide evidence that blocking stem cell factor (SCF)-c-kit signaling is sufficient to inhibit CSC proliferation and survival promoted by chemotherapy. CSCs were isolated from NSCLC cell lines as tumor spheres under CSC-selective conditions and their stem properties were confirmed. In contrast to other tumor cells, CSCs expressed c-kit receptors and produced SCF. Proliferation of CSCs was inhibited by SCF-neutralizing antibodies or by imatinib (Gleevec), an inhibitor of c-kit. Although cisplatin treatment eliminated the majority of tumor cells, it did not eliminate CSCs, whereas imatinib or anti-SCF antibody destroyed CSCs. Significantly, combining cisplatin with imatinib or anti-SCF antibody prevented the growth of both tumor cell subpopulations. Our findings reveal an important role for the SCF-c-kit signaling axis in self-renewal and proliferation of lung CSCs, and they suggest that SCF-c-kit signaling blockade could improve the antitumor efficacy of chemotherapy of human NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal/fisiología , Factor de Células Madre/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Comunicación Autocrina/efectos de los fármacos , Comunicación Autocrina/fisiología , Benzamidas , Células Cultivadas , Cisplatino/administración & dosificación , Citometría de Flujo , Humanos , Procesamiento de Imagen Asistido por Computador , Mesilato de Imatinib , Células Madre Neoplásicas/efectos de los fármacos , Piperazinas/administración & dosificación , Proteínas Proto-Oncogénicas c-kit/efectos de los fármacos , Pirimidinas/administración & dosificación , Transducción de Señal/efectos de los fármacos , Factor de Células Madre/efectos de los fármacos
10.
Clin Cancer Res ; 15(20): 6348-57, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19825957

RESUMEN

PURPOSE: Regulatory T cell (Treg) frequency and activity are increased in cancer patients and play a major role in tumor escape. Although disease progression is favored by the presence of Treg, mechanisms used by Treg to suppress antitumor immunity are unknown. The ectonucleotidases CD39 and CD73 are expressed in Treg and convert ATP into immunosuppressive adenosine. In this study, the involvement of the adenosinergic pathway in Treg-mediated suppression in head and neck squamous cell carcinoma (HNSCC) patients was evaluated. EXPERIMENTAL DESIGN: HNSCC patients with an active disease (n = 19) and patients with no evident disease after therapy (n = 14) were studied. Ectonucleotidase expression on CD4(+) T cells and CD4(+)CD25(high) Treg was evaluated by flow cytometry and compared with normal controls. Ectonucleotidase activity was also compared within these three groups. The data were analyzed for associations of ectonucleotidase expression/function with disease stage. RESULTS: The percentages and expression levels of CD39 and CD73 in CD4(+) T cells and Treg were greater in HNSCC than in normal controls and highest in patients with no evident disease. Patients' Treg hydrolyzed ATP at higher rates and produced higher levels of adenosine than normal controls' Treg. The increased frequency and enzymatic activity of CD4(+)CD39(+) cells corresponded to increased adenosine-mediated suppression of effector T cells, which was partly inhibited by ARL67156, an ectonucleotidase inhibitor, and by ZM241385, a selective A(2a)/A(2b) receptor antagonist. CONCLUSIONS: CD39(+) Treg frequency and adenosine-mediated suppression are significantly increased in HNSCC patients. The adenosinergic pathway is involved in Treg-mediated immunosuppression in cancer and its attenuation could be a promising immunotherapeutic strategy for patients with HNSCC.


Asunto(s)
5'-Nucleotidasa/metabolismo , Antígenos CD/metabolismo , Apirasa/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Linfocitos T Reguladores/metabolismo , Adenosina Trifosfato/metabolismo , Adulto , Anciano , Femenino , Humanos , Terapia de Inmunosupresión , Masculino , Persona de Mediana Edad , N-Glicosil Hidrolasas/metabolismo
11.
Cancer Res ; 69(12): 5226-33, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19491263

RESUMEN

There is increasing evidence that prolactin (PRL), a hormone/cytokine, plays a role in breast, prostate, and colorectal cancers via local production or accumulation. Elevated levels of serum PRL in ovarian and endometrial cancers have been reported, indicating a potential role for PRL in endometrial and ovarian carcinogenesis. In this study, we show that serum PRL levels are significantly elevated in women with a strong family history of ovarian cancer. We show dramatically increased expression of PRL receptor in ovarian and endometrial tumors as well as in endometrial hyperplasia, signifying the importance of PRL signaling in malignant and premalignant conditions. PRL mRNA was expressed in ovarian and endometrial tumors, indicating the presence of an autocrine loop. PRL potently induced proliferation in several ovarian and endometrial cancer cell lines. Binding of PRL to its receptor was followed by rapid phosphorylation of extracellular signal-regulated kinase (ERK) 1/2, mitogen-activated protein kinase/ERK kinase 1, signal transducer and activator of transcription 3, CREB, ATF-2, and p53 and activation of 37 transcription factors in ovarian and endometrial carcinoma cells. PRL also activated Ras oncogene in these cells. When human immortalized normal ovarian epithelial cells were chronically exposed to PRL, a malignant transformation occurred manifested by the acquired ability of transformed cells to form clones, grow in soft agar, and form tumors in severe combined immunodeficient-beige mice. Transformation efficiency was diminished by a Ras inhibitor, providing proof that PRL-induced transformation uses the Ras pathway. In summary, we present findings that indicate an important role for PRL in ovarian and endometrial tumorigenesis. PRL may represent a risk factor for ovarian and endometrial cancers.


Asunto(s)
Neoplasias de los Genitales Femeninos/fisiopatología , Prolactina/fisiología , Animales , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular , Cartilla de ADN , Femenino , Citometría de Flujo , Neoplasias de los Genitales Femeninos/metabolismo , Neoplasias de los Genitales Femeninos/patología , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Prolactina/sangre , Prolactina/genética , ARN Mensajero/genética , Transducción de Señal
12.
J Immunol Methods ; 346(1-2): 55-63, 2009 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-19450601

RESUMEN

Human regulatory T cells (Treg) have been variously defined as CD4(+)CD25(+), CD4(+)CD25(high) or CD4(+)CD25(high)FOXP3(+) cells which are responsible for maintaining peripheral tolerance. Their isolation from human peripheral blood or tissues depends on the expression level of CD25(IL-2Ralpha) - a surface marker which is also expressed on activated effector helper T cells. CD39, a cell surface associated ectonucleotidase, can be used to purify Treg with strong suppressor functions. The CD4(+)CD39(+) T cells catalyze cleavage of adenosine triphosphate (ATP) to adenosine monophosphate (AMP), which is then further cleaved to adenosine. CD4(+)CD39(+) T cells largely overlap with CD4(+)CD25(high)FOXP3(+) but not CD4(+)CD25(+) T cell subset, and mediate equally potent immune suppression. Thus, CD39 surface marker can be successfully used for routine isolation of functionally-active human Treg from the peripheral blood of healthy donors or patients with cancer for studies of their role in health and disease.


Asunto(s)
Antígenos CD/análisis , Apirasa/análisis , Carcinoma de Células Escamosas/inmunología , Citometría de Flujo , Neoplasias de Cabeza y Cuello/inmunología , Separación Inmunomagnética , Linfocitos T Reguladores/inmunología , Adulto , Carcinoma de Células Escamosas/sangre , Estudios de Casos y Controles , Células Cultivadas , Femenino , Factores de Transcripción Forkhead/análisis , Neoplasias de Cabeza y Cuello/sangre , Humanos , Inmunofenotipificación , Subunidad alfa del Receptor de Interleucina-2/análisis , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados , Adulto Joven
13.
Clin Cancer Res ; 15(8): 2647-56, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19351767

RESUMEN

PURPOSE: Tumor cell growth and migration can be directly regulated by chemokines. In the present study, the association of CCL11 with ovarian cancer has been investigated. EXPERIMENTAL DESIGN AND RESULTS: Circulating levels of CCL11 in sera of patients with ovarian cancer were significantly lower than those in healthy women or women with breast, lung, liver, pancreatic, or colon cancer. Cultured ovarian carcinoma cells absorbed soluble CCL11, indicating that absorption by tumor cells could be responsible for the observed reduction of serum level of CCL11 in ovarian cancer. Postoperative CCL11 levels in women with ovarian cancer negatively correlated with relapse-free survival. Ovarian tumors overexpressed three known cognate receptors of CCL11, CC chemokine receptors (CCR) 2, 3, and 5. Strong positive correlation was observed between expression of individual receptors and tumor grade. CCL11 potently stimulated proliferation and migration/invasion of ovarian carcinoma cell lines, and these effects were inhibited by neutralizing antibodies against CCR2, CCR3, and CCR5. The growth-stimulatory effects of CCL11 were likely associated with activation of extracellular signal-regulated kinase 1/2, MEK1, and STAT3 phosphoproteins and with increased production of multiple cytokines, growth factors, and angiogenic factors. Inhibition of CCL11 signaling by the combination of neutralizing antibodies against the ligand and its receptors significantly increased sensitivity to cisplatin in ovarian carcinoma cells. CONCLUSION: We conclude that CCL11 signaling plays an important role in proliferation and invasion of ovarian carcinoma cells and CCL11 pathway could be targeted for therapy in ovarian cancer. Furthermore, CCL11 could be used as a biomarker and a prognostic factor of relapse-free survival in ovarian cancer.


Asunto(s)
Quimiocina CCL11/fisiología , Neoplasias Ováricas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimiocina CCL11/sangre , Quimiocina CCL11/farmacología , Citocinas/análisis , Femenino , Humanos , Neoplasias Ováricas/sangre , Neoplasias Ováricas/patología , Fosfoproteínas/análisis , Receptores CCR2/metabolismo , Receptores CCR3/metabolismo , Receptores CCR5/metabolismo , Transducción de Señal , Factores de Transcripción/análisis
14.
Science ; 323(5912): 393-6, 2009 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-19150848

RESUMEN

Retroviruses have the potential to acquire host cell-derived genetic material during reverse transcription and can integrate into the genomes of larger, more complex DNA viruses. In contrast, RNA viruses were believed not to integrate into the host's genome under any circumstances. We found that illegitimate recombination between an exogenous nonretroviral RNA virus, lymphocytic choriomeningitis virus, and the endogenous intracisternal A-type particle (IAP) retrotransposon occurred and led to reverse transcription of exogenous viral RNA. The resulting complementary DNA was integrated into the host's genome with an IAP element. Thus, RNA viruses should be closely scrutinized for any capacity to interact with endogenous retroviral elements before their approval for therapeutic use in humans.


Asunto(s)
ADN Complementario/genética , Genes de Partícula A Intracisternal/genética , Virus de la Coriomeningitis Linfocítica/genética , ARN Viral/genética , Recombinación Genética , Transcripción Reversa , Integración Viral , Animales , Infecciones por Arenaviridae/virología , Secuencia de Bases , Línea Celular , Glicoproteínas/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Transfección , Proteínas Virales/genética
15.
Int J Cancer ; 123(9): 2031-40, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18697197

RESUMEN

The ability of human tumor cell lines to produce various cytokines, chemokines, angiogenic and growth factors was investigated using Luminex multiplex technology. Media conditioned by tumor cells protected tumor cells from drug-induced apoptosis and stimulated tumor cell proliferation. Antibodies neutralizing IL-6, CXCL8, CCL2 and CCL5 blocked this stimulation. Treatment of tumor cells with doxorubicin and cisplatin resulted in a substantial increase in the production of IL-6, CXCL8, CCL2, CCL5, BFGF, G-CSF and VEGF. This stimulation was associated with drug-induced activation of NF-kappaB, AP-1, AP-2, CREB, HIF-1, STAT-1, STAT-3, STAT-5 and ATF-2 transcription factors and upregulation of IL-6, CXCL8, FGF-2, CSF-3 and CCL5 gene expression. Treatment of tumor cells with doxorubicin and antibodies neutralizing G-CSF, CCL2 or CCL5 had higher inhibitory effects than each modality used alone. These results indicate that chemokines and growth factors produced by tumor by binding to the cognate receptors on tumor and stroma cells could provide proliferative and antiapoptotic signals helping tumor to escape drug-mediated destruction. Clinical studies showed that antibodies neutralizing VEGF (Avastin/Bevacizumab) or blocking HER2/neu signaling (Herceptin/Trastuzumab) could increase the efficacy of chemotherapy, although these beneficial effects have been limited. It is possible that drug-stimulated production of growth and proangiogenic factors could counterbalance the effects of antibody therapy. In addition, numerous growth factors and chemokines share angiogenic and growth-stimulating properties, and thus reduction of a single factor is insufficient to completely block tumor growth. Thus, a broad disruption of tumor cytokine network is needed to further increase the efficacy of cancer therapy.


Asunto(s)
Citocinas/biosíntesis , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Inductores de la Angiogénesis/metabolismo , Línea Celular Tumoral , Proliferación Celular , Quimiocinas/biosíntesis , Cisplatino/farmacología , Medios de Cultivo Condicionados , Doxorrubicina/farmacología , Sustancias de Crecimiento/biosíntesis , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Factores de Transcripción/metabolismo , Factor A de Crecimiento Endotelial Vascular/fisiología
16.
PLoS One ; 3(8): e3077, 2008 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-18728788

RESUMEN

BACKGROUND: Cancer stem cells (CSCs) are thought to be responsible for tumor regeneration after chemotherapy, although direct confirmation of this remains forthcoming. We therefore investigated whether drug treatment could enrich and maintain CSCs and whether the high tumorogenic and metastatic abilities of CSCs were based on their marked ability to produce growth and angiogenic factors and express their cognate receptors to stimulate tumor cell proliferation and stroma formation. METHODOLOGY/FINDINGS: Treatment of lung tumor cells with doxorubicin, cisplatin, or etoposide resulted in the selection of drug surviving cells (DSCs). These cells expressed CD133, CD117, SSEA-3, TRA1-81, Oct-4, and nuclear beta-catenin and lost expression of the differentiation markers cytokeratins 8/18 (CK 8/18). DSCs were able to grow as tumor spheres, maintain self-renewal capacity, and differentiate. Differentiated progenitors lost expression of CD133, gained CK 8/18 and acquired drug sensitivity. In the presence of drugs, differentiation of DSCs was abrogated allowing propagation of cells with CSC-like characteristics. Lung DSCs demonstrated high tumorogenic and metastatic potential following inoculation into SCID mice, which supported their classification as CSCs. Luminex analysis of human and murine cytokines in sonicated lysates of parental- and CSC-derived tumors revealed that CSC-derived tumors contained two- to three-fold higher levels of human angiogenic and growth factors (VEGF, bFGF, IL-6, IL-8, HGF, PDGF-BB, G-CSF, and SCGF-beta). CSCs also showed elevated levels of expression of human VEGFR2, FGFR2, CXCR1, 2 and 4 receptors. Moreover, human CSCs growing in SCID mice stimulated murine stroma to produce elevated levels of angiogenic and growth factors. CONCLUSIONS/SIGNIFICANCE: These findings suggest that chemotherapy can lead to propagation of CSCs and prevention of their differentiation. The high tumorigenic and metastatic potentials of CSCs are associated with efficient cytokine network production that may represent a target for increased efficacy of cancer therapy.


Asunto(s)
Citocinas/fisiología , Neoplasias Pulmonares/patología , Células Madre/patología , Células Tumorales Cultivadas , Antígenos CD/análisis , Antineoplásicos/farmacología , Automatización , Supervivencia Celular/efectos de los fármacos , Quimiocinas/biosíntesis , Quimiocinas/fisiología , Citocinas/biosíntesis , Citometría de Flujo , Sustancias de Crecimiento/biosíntesis , Sustancias de Crecimiento/fisiología , Humanos , Metástasis de la Neoplasia , Células Madre/efectos de los fármacos
17.
Exp Cell Res ; 314(7): 1605-16, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18313665

RESUMEN

TRAIL is a death ligand that induces apoptosis in malignant but not normal cells. Recently the ability of TRAIL to induce proliferation in apoptosis-resistant normal and malignant cells was reported. In this study, we analyzed TRAIL effects in apoptosis sensitive MCF7, OVCAR3 and H460 human tumor cell lines. TRAIL at low concentrations preferentially induced cell proliferation. At 100 ng/ml, apoptotic death was readily observed, however surviving cells acquired higher proliferative capacity. TRAIL-stimulated production of several cytokines, IL-8, RANTES, MCP-1 and bFGF, and activation of caspases 1 and 8 was essential for this effect. Antibodies to IL-8, RANTES, and bFGF blocked TRAIL-induced cell proliferation and further stimulated apoptosis. For the first time, we report that high TRAIL concentrations induced cell senescence as determined by the altered morphology and expression of several senescence markers: SA-beta-gal, p21Waf1/Cip1, p16INK4a, and HMGA. Caspase 9 inhibition protected TRAIL-treated cells from senescence, whereas inhibition of caspases 1 and 8 increased the yield of SLP cells. In conclusion, in cultured human carcinoma cells, TRAIL therapy results in three functional outcomes, apoptosis, proliferation and senescence. TRAIL-induced proapoptotic and prosurvival responses correlate with the strength of signaling. TRAIL-induced cytokine production is responsible for its proliferative and prosurvival effects.


Asunto(s)
Apoptosis/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Citocinas/biosíntesis , Neoplasias/patología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Inhibidores de Caspasas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Medios de Cultivo Condicionados , Inhibidores Enzimáticos/farmacología , Humanos , Modelos Biológicos , Neoplasias/enzimología , Pruebas de Neutralización , Fenotipo , Transducción de Señal/efectos de los fármacos
18.
Cancer Immunol Immunother ; 57(11): 1611-23, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18327580

RESUMEN

INTRODUCTION: We hypothesize that adenosine and PGE2 could have a complementary immunosuppressive effect that is mediated via common cAMP-PKA signaling. MATERIALS AND METHODS: To test this hypothesis, the effect of adenosine and PGE2 on the cytotoxic activity and cytokine production of lymphokine activated killer (LAK) cells was investigated. RESULTS: PGE2 and adenosine inhibited LAK cells cytotoxic activity and production of INF-gamma, GM-CSF and TNF-alpha. In combination they showed substantially higher inhibition than each modality used alone. Using agonists and antagonists specific for PGE2 and adenosine receptors we found that cooperation of PGE2 and adenosine in their inhibitory effects are mediated via EP2 and A2A receptors, respectively. LAK cells have 35-fold higher expression of EP2 than A2A. Combined PGE2 and adenosine treatment resulted in augmentation of cAMP production, PKA activity, CREB phosphorylation and inhibition of Akt phosphorylation. Wortmannin and LY294002 enhanced the suppressive effects of adenosine and PGE2. In contrast, Rp-8-Br-cAMPS, an inhibitor of PKA type I blocked their immunosuppressive effects, suggesting that the inhibitory effects of PGE2 and adenosine are mediated via common pathway with activation of cAMP-PKA and inhibition of Akt. CONCLUSION: In comparison to other immunosuppressive molecules (TGF-beta and IL-10), adenosine and PGE2 are unique in their ability to inhibit the executive function of highly cytotoxic cells. High intratumor levels of adenosine and PGE2 could protect tumor from immune-mediated destruction by inactivation of the tumor infiltrating functionally active immune cells.


Asunto(s)
Adenosina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dinoprostona/farmacología , Células Asesinas Activadas por Linfocinas/efectos de los fármacos , Animales , Western Blotting , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Proteínas Quinasas Dependientes de AMP Cíclico/inmunología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Tolerancia Inmunológica , Terapia de Inmunosupresión , Interferón gamma/metabolismo , Interleucina-10/farmacología , Células Asesinas Activadas por Linfocinas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Oxitócicos/farmacología , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/farmacología , Factor de Necrosis Tumoral alfa/farmacología
19.
Gynecol Oncol ; 107(1): 58-65, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17659325

RESUMEN

OBJECTIVE: Endometrial carcinoma is the most common gynecologic cancer. Although the prognosis for endometrial cancer is generally good, cancers identified at late stages are associated with high levels of morbidity and mortality. Therefore, prevention and early detection may further reduce the burden of this challenging disease. METHODS: A panel of 64 serum biomarkers was analyzed in sera of patients with stages I-III endometrial cancer and age-matched healthy women, utilizing a multiplex xMAP bead-based immunoassay. For multivariate analysis, four different statistical classification methods were used: logistic regression (LR), separating hyperplane (SHP), k nearest neighbors (KNN), and classification tree (CART). For each of these classifiers, a diagnostic model was created based on the cross-validation set consisting of sera from 115 patients with endometrial cancer and 135 healthy women. RESULTS: Our data have demonstrated that patients with endometrial cancer have significantly different expression patterns of several serum biomarkers as compared to healthy controls. Prolactin was the strongest discriminative biomarker for endometrial cancer providing 98.3% sensitivity and 98.0% specificity alone. Our results have revealed that serum concentration of cancer antigens, including CA 125, CA 15-3, and CEA are higher in patients with Stage III endometrial cancer as compared to those with Stage I. In addition, we have shown that the expression of CA 125, AFP, and ACTH is elevated in women with tumor grade 3 vs. grade 1. Furthermore, five-biomarker panel (prolactin, GH, Eotaxin, E-selectin, and TSH) identified in this study was able to discriminate endometrial cancer from ovarian and breast cancers with high sensitivity and specificity. CONCLUSIONS: The ability of prolactin to accurately discriminate between cancer and control groups indicates that this biomarker could potentially be used for development of blood-based test for the early detection of endometrial cancer in high-risk populations. Combining the information on multiple serum markers using flexible statistical methods allows for achieving high cancer selectivity.


Asunto(s)
Biomarcadores de Tumor/sangre , Proteínas Sanguíneas/análisis , Neoplasias Endometriales/diagnóstico , Prolactina/sangre , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Inmunoensayo/métodos , Persona de Mediana Edad , Sensibilidad y Especificidad
20.
Cancer Res ; 67(12): 5949-56, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17575165

RESUMEN

The goal of this study was to investigate the effects of adenosine and its stable analogue 2-chloroadenosine (CADO) on the cytotoxic activity and cytokine production by human antimelanoma specific CD8+ and CD4+ T-helper type 1 (Th1) clones. The cytotoxic activity of CD8+ T cells was inhibited by adenosine and CADO. Using Lab MAP multiplex technology, we found that adenosine inhibits production of various cytokines and chemokines by CD8+ and CD4+ T cells. Studies with CGS21680, a specific agonist of adenosine A2A receptor (AdoRA2A), and ZM241385, an AdoRA2-selective antagonist, indicate that the inhibitory effects of adenosine are mediated via cyclic AMP (cAMP)-elevating AdoRA2A, leading to protein kinase A (PKA) activation. Using cAMP analogues with different affinities for the A and B sites of the regulatory subunits of PKAI and PKAII, we found that activation of PKAI, but not of PKAII, mimicked the inhibitory effects of adenosine on T-cell cytotoxic activity and cytokine production. Inhibitors of the PKA catalytic subunits (H89 and PKA inhibitor peptide 14-22) failed to abrogate the inhibitory effects of CADO. In contrast, Rp-8-Br-cAMPS that antagonizes binding of cAMP to the regulatory I subunit and PKA activation was efficient in blocking the inhibitory effect of adenosine on the functional activity of T cells. Our findings on the ability of adenosine to inhibit the effector function of antimelanoma specific T cells suggest that intratumor-produced adenosine could impair the function of tumor-infiltrating T lymphocytes. Thus, blocking the inhibitory activity of tumor-produced adenosine might represent a new strategy for improvement of cancer immunotherapy.


Asunto(s)
Adenosina/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citocinas/biosíntesis , Melanoma/inmunología , 2-Cloroadenosina/metabolismo , Adenosina/análogos & derivados , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Citotoxicidad Inmunológica , Humanos , Receptor de Adenosina A2A/metabolismo , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...