Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 9(11): 1225-35, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18820683

RESUMEN

The involvement of complement-activation products in promoting tumor growth has not yet been recognized. Here we show that the generation of complement C5a in a tumor microenvironment enhanced tumor growth by suppressing the antitumor CD8(+) T cell-mediated response. This suppression was associated with the recruitment of myeloid-derived suppressor cells into tumors and augmentation of their T cell-directed suppressive abilities. Amplification of the suppressive capacity of myeloid-derived suppressor cells by C5a occurred through regulation of the production of reactive oxygen and nitrogen species. Pharmacological blockade of the C5a receptor considerably impaired tumor growth to a degree similar to the effect produced by the anticancer drug paclitaxel. Thus, our study demonstrates a therapeutic function for complement inhibition in the treatment of cancer.


Asunto(s)
Complemento C5a/inmunología , Terapia de Inmunosupresión , Neoplasias/inmunología , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Linfocitos T Citotóxicos/inmunología , Animales , Activación de Complemento , Convertasas de Complemento C3-C5/genética , Complemento C5a/antagonistas & inhibidores , Complemento C5a/farmacología , Regulación hacia Abajo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Células Mieloides/inmunología , Células Mieloides/metabolismo , Neoplasias/terapia , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor de Anafilatoxina C5a/inmunología , Receptor de Anafilatoxina C5a/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Adv Exp Med Biol ; 1223: 81-97, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32030686

RESUMEN

The involvement of inflammation in cancer progression is well-established. The immune system can play both tumor-promoting and -suppressive roles, and efforts to harness the immune system to help fight tumor growth are at the forefront of research. Of particular importance is the inflammatory profile at the site of the tumor, with respect to both the leukocyte population numbers, the phenotype of these cells, as well as the contribution of the tumor cells themselves. In this regard, the pro-inflammatory effects of pattern recognition receptor expression and activation in the tumor microenvironment have emerged as a relevant issue both for therapy and to understand tumor development.Pattern recognition receptors (PRRs) were originally recognized as components of immune cells, particularly innate immune cells, as detectors of pathogens. PRR signaling in immune cells activates them, inducing robust antimicrobial responses. In particular, toll-like receptors (TLRs) constitute a family of membrane-bound PRRs which can recognize pathogen-associated molecular patterns (PAMPs) carried by bacteria, virus, and fungi. In addition, PRRs can recognize products generated by stressed cells or damaged tissues, namely damage-associated molecular patterns or DAMPS. Taking into account the role of the immune system in fighting tumors together with the presence of immune cells in the microenvironment of different types of tumors, strategies to activate immune cells via PRR ligands have been envisioned as an anticancer therapeutic approach.In the last decades, it has been determined that PRRs are present and functional on nonimmune cells and that their activation in these cells contributes to the inflammation in the tumor microenvironment. Both tumor-promoting and antitumor effects have been observed when tumor cell PRRs are activated. This argues against nonspecific activation of PRR ligands in the tumor microenvironment as a therapeutic approach. Therefore, the use of PRR ligands for anticancer therapy might benefit from strategies that specifically deliver these ligands to immune cells, thus avoiding tumor cells in some settings. This review focuses on these aspects of TLR signaling in the tumor microenvironment.


Asunto(s)
Neoplasias/inmunología , Neoplasias/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo , Microambiente Tumoral , Humanos , Receptores de Reconocimiento de Patrones/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal/inmunología , Receptores Toll-Like/inmunología , Microambiente Tumoral/inmunología
3.
Psychosom Med ; 79(3): 306-310, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28002383

RESUMEN

OBJECTIVE: It has been proposed that the inflammatory cytokine system is regulated through the vagus nerve, where vagal activation inhibits release of inflammatory cytokines and, therefore, inflammation. Thus, loss of vagal activation (i.e., reduced high-frequency heart rate variability [HF-HRV]) should result in greater inflammation. Evidence to date for this relationship has relied on animal models and resting states in humans. The present study used a psychosocial stressor to test whether stress-induced decreases in HF-HRV predict increases in circulating inflammatory markers. METHODS: Thirty healthy young women completed a speech stressor. HF-HRV was assessed before and during the stressor while circulating plasma interleukin 6, tumor necrosis factor α, and C-reactive protein were assessed before and 1 hour after the stressor. RESULTS: Consistent with the neural reflex for immunity, greater reductions in HF-HRV during the stressor were associated with greater increases in tumor necrosis factor α (ß = -0.29 to -0.47) and interleukin 6 (ß = -0.40 to -0.68) but not C-reactive protein (ß = 0.10 to 0.29) 1 hour after the stressor. CONCLUSIONS: These findings expand on the current literature by showing that changes in HF-HRV predict and precede changes in circulating inflammatory cytokines in humans and may have implications for treatment of inflammatory diseases.


Asunto(s)
Frecuencia Cardíaca/fisiología , Inflamación/sangre , Sistema Nervioso Parasimpático/fisiopatología , Estrés Psicológico/sangre , Estrés Psicológico/fisiopatología , Adulto , Proteína C-Reactiva/metabolismo , Femenino , Humanos , Interleucina-6/sangre , Factor de Necrosis Tumoral alfa/sangre , Adulto Joven
4.
Semin Cancer Biol ; 35 Suppl: S199-S223, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25865775

RESUMEN

Cancer arises in the context of an in vivo tumor microenvironment. This microenvironment is both a cause and consequence of tumorigenesis. Tumor and host cells co-evolve dynamically through indirect and direct cellular interactions, eliciting multiscale effects on many biological programs, including cellular proliferation, growth, and metabolism, as well as angiogenesis and hypoxia and innate and adaptive immunity. Here we highlight specific biological processes that could be exploited as targets for the prevention and therapy of cancer. Specifically, we describe how inhibition of targets such as cholesterol synthesis and metabolites, reactive oxygen species and hypoxia, macrophage activation and conversion, indoleamine 2,3-dioxygenase regulation of dendritic cells, vascular endothelial growth factor regulation of angiogenesis, fibrosis inhibition, endoglin, and Janus kinase signaling emerge as examples of important potential nexuses in the regulation of tumorigenesis and the tumor microenvironment that can be targeted. We have also identified therapeutic agents as approaches, in particular natural products such as berberine, resveratrol, onionin A, epigallocatechin gallate, genistein, curcumin, naringenin, desoxyrhapontigenin, piperine, and zerumbone, that may warrant further investigation to target the tumor microenvironment for the treatment and/or prevention of cancer.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Microambiente Tumoral/genética , Antineoplásicos/uso terapéutico , Carcinogénesis/genética , Proliferación Celular/efectos de los fármacos , Humanos , Terapia Molecular Dirigida , Neoplasias/genética , Neoplasias/prevención & control , Neovascularización Patológica/genética , Neovascularización Patológica/prevención & control , Transducción de Señal , Microambiente Tumoral/efectos de los fármacos
5.
Semin Cancer Biol ; 35 Suppl: S276-S304, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26590477

RESUMEN

Targeted therapies and the consequent adoption of "personalized" oncology have achieved notable successes in some cancers; however, significant problems remain with this approach. Many targeted therapies are highly toxic, costs are extremely high, and most patients experience relapse after a few disease-free months. Relapses arise from genetic heterogeneity in tumors, which harbor therapy-resistant immortalized cells that have adopted alternate and compensatory pathways (i.e., pathways that are not reliant upon the same mechanisms as those which have been targeted). To address these limitations, an international task force of 180 scientists was assembled to explore the concept of a low-toxicity "broad-spectrum" therapeutic approach that could simultaneously target many key pathways and mechanisms. Using cancer hallmark phenotypes and the tumor microenvironment to account for the various aspects of relevant cancer biology, interdisciplinary teams reviewed each hallmark area and nominated a wide range of high-priority targets (74 in total) that could be modified to improve patient outcomes. For these targets, corresponding low-toxicity therapeutic approaches were then suggested, many of which were phytochemicals. Proposed actions on each target and all of the approaches were further reviewed for known effects on other hallmark areas and the tumor microenvironment. Potential contrary or procarcinogenic effects were found for 3.9% of the relationships between targets and hallmarks, and mixed evidence of complementary and contrary relationships was found for 7.1%. Approximately 67% of the relationships revealed potentially complementary effects, and the remainder had no known relationship. Among the approaches, 1.1% had contrary, 2.8% had mixed and 62.1% had complementary relationships. These results suggest that a broad-spectrum approach should be feasible from a safety standpoint. This novel approach has potential to be relatively inexpensive, it should help us address stages and types of cancer that lack conventional treatment, and it may reduce relapse risks. A proposed agenda for future research is offered.


Asunto(s)
Heterogeneidad Genética , Terapia Molecular Dirigida , Neoplasias/terapia , Medicina de Precisión , Antineoplásicos Fitogénicos/uso terapéutico , Resistencia a Antineoplásicos/genética , Humanos , Neoplasias/genética , Neoplasias/patología , Neoplasias/prevención & control , Transducción de Señal , Microambiente Tumoral/genética
6.
Brain Behav Immun ; 52: 27-31, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26526647

RESUMEN

Past work has linked negative repetitive thought (worry, rumination) about stressors to sustained stress responses. Less is known about the effects of neutral types of repetitive thought (e.g., reflection) on physiological stress responses. The present study examined whether greater trait reflection was associated with a lower inflammatory response to an acute psychosocial stressor. Thirty-four healthy undergraduate women completed a speech stressor, and plasma interleukin-6 (IL-6), C-reactive protein, and tumor necrosis factor-α levels were assessed before and after the stressor. Higher levels of reflection predicted lower IL-6 responses 1h after the stressor. Stressor appraisal was not a significant mediator. These preliminary findings stand in contrast to existing evidence that other forms of repetitive thought like worry and rumination may exacerbate or prolong the inflammatory stress response and indicate that reflection is a notable factor worth considering when examining the relationship between stress, inflammation, and health.


Asunto(s)
Interleucina-6/sangre , Estrés Psicológico/sangre , Estrés Psicológico/psicología , Adolescente , Adulto , Ansiedad/sangre , Ansiedad/psicología , Proteína C-Reactiva/metabolismo , Cognición/fisiología , Citocinas/sangre , Femenino , Humanos , Hidrocortisona/sangre , Inflamación/sangre , Factor de Necrosis Tumoral alfa/sangre , Adulto Joven
7.
Immunol Invest ; 45(8): 712-720, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27420920

RESUMEN

In the last 15 years, it has become apparent that ovarian cancer is recognized by the immune system, taking into account that T cell infiltration can be associated with increased overall survival. Several studies indicate that a correct combination of cluster of differentiation 8 and cluster of differentiation 4 T cells is key to fight tumor progression and that the presence of regulatory T cells (Tregs) infiltrating ovarian solid tumors (or present in ascites) is deleterious. Several markers that characterize Tregs include glucocorticoid-induced tumor necrosis factor receptor, cytotoxic T lymphocyte antigen-4, and forkhead box protein 3 (Foxp3). Research has shown that Tregs can infiltrate cancerous tissue and contribute to tumor growth by secreting immunosuppressive cytokines such as transforming growth factor beta and interleukin (IL)-10. Importantly, these cells might hamper the efficacy of immunotherapeutic approaches, thus strategies involving depletion or regulation of this population have been proposed and tested in experimental models. In this Minireview, we will discuss the relevance of Tregs in ovarian cancer and the experimental approaches destined to impair their immunosuppressive effects.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Inmunoterapia/métodos , Neoplasias Ováricas/terapia , Linfocitos T Citotóxicos/patología , Linfocitos T Reguladores/patología , Antígeno CTLA-4/genética , Antígeno CTLA-4/inmunología , Movimiento Celular , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Humanos , Tolerancia Inmunológica , Interleucina-10/genética , Interleucina-10/inmunología , Activación de Linfocitos , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Transducción de Señal , Análisis de Supervivencia , Linfocitos T Citotóxicos/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología
8.
Am J Physiol Cell Physiol ; 308(1): C68-78, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25339657

RESUMEN

Adhesion of circulating tumor cells to vascular endothelium is mediated by specialized molecules that are functional under shear forces exerted by hematogenous flow. Endothelial E-selectin binding to glycoforms of CD44 mediates shear-resistant cell adhesion in numerous physiological and pathological conditions. However, this pathway is poorly understood in breast cancer and is the focus of the present investigation. All breast cancer cell lines used in this study strongly expressed CD44. In particular, BT-20 cells expressed CD44s and multiple CD44v isoforms, whereas MDA-MB-231 cells predominantly expressed CD44s but weakly expressed CD44v isoforms. CD44 expressed by BT-20, but not MDA-MB-231, cells possessed E-selectin ligand activity as detected by Western blotting and antigen capture assays. Importantly, CD44 expressed by intact BT-20 cells were functional E-selectin ligands, regulating cell rolling and adhesion under physiological flow conditions, as found by shRNA-targeted silencing of CD44. Antigen capture assays strongly suggest greater shear-resistant E-selectin ligand activity of BT-20 cell CD44v isoforms than CD44s. Surprisingly, CD44 was not recognized by the HECA-452 MAb, which detects sialofucosylated epitopes traditionally expressed by selectin ligands, suggesting that BT-20 cells express a novel glycoform of CD44v as an E-selectin ligand. The activity of this glycoform was predominantly attributed to N-linked glycans. Furthermore, expression of CD44v as an E-selectin ligand correlated with high levels of fucosyltransferase-3 and -6 and epithelial, rather than mesenchymal, cell phenotype. Together, these data demonstrate that expression of CD44 as a functional E-selectin ligand may be important in breast cancer metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Adhesión Celular , Selectina E/metabolismo , Receptores de Hialuranos/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Células CHO , Línea Celular Tumoral , Movimiento Celular , Cricetulus , Selectina E/genética , Células Endoteliales/metabolismo , Transición Epitelial-Mesenquimal , Femenino , Fucosiltransferasas/metabolismo , Glicosilación , Humanos , Receptores de Hialuranos/genética , Ligandos , Metástasis de la Neoplasia , Fenotipo , Isoformas de Proteínas , Interferencia de ARN , Flujo Sanguíneo Regional , Transfección
9.
Exp Cell Res ; 323(1): 7-27, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24569142

RESUMEN

Dendritic cells (DCs) are immune cells found in the peripheral tissues where they sample the organism for infections or malignancies. There they take up antigens and migrate towards immunological organs to contact and activate T lymphocytes that specifically recognize the antigen presented by these antigen presenting cells. In the steady state there are several types of resident DCs present in various different organs. For example, in the mouse, splenic DC populations characterized by the co-expression of CD11c and CD8 surface markers are specialized in cross-presentation to CD8 T cells, while CD11c/SIRP-1α DCs seem to be dedicated to activating CD4 T cells. On the other hand, DCs have also been associated with the development of various diseases such as cancer, atherosclerosis, or inflammatory conditions. In such disease, DCs can participate by inducing angiogenesis or immunosuppression (tumors), promoting autoimmune responses, or exacerbating inflammation (atherosclerosis). This change in DC biology can be prompted by signals in the microenvironment. We have previously shown that the interaction of DCs with various extracellular matrix components modifies the immune properties and angiogenic potential of these cells. Building on those studies, herewith we analyzed the angiogenic profile of murine myeloid DCs upon interaction with 2D and 3D type-I collagen environments. As determined by PCR array technology and quantitative PCR analysis we observed that interaction with these collagen environments induced the expression of particular angiogenic molecules. In addition, DCs cultured on collagen environments specifically upregulated the expression of CXCL-1 and -2 chemokines. We were also able to establish DC cultures on type-IV collagen environments, a collagen type expressed in pathological conditions such as atherosclerosis. When we examined DC populations in atherosclerotic veins of Apolipoprotein E deficient mice we observed that they expressed adhesion molecules capable of interacting with collagen. Finally, to further investigate the interaction of DCs with collagen in other pathological conditions, we determined that both murine ovarian and breast cancer cells express several collagen molecules that can contribute to shape their particular tumor microenvironment. Consistently, tumor-associated DCs were shown to express adhesion molecules capable of interacting with collagen molecules as determined by flow cytometry analysis. Of particular relevance, tumor-associated DCs expressed high levels of CD305/LAIR-1, an immunosuppressive receptor. This suggests that signaling through this molecule upon interaction with collagen produced by tumor cells might help define the poorly immunogenic status of these cells in the tumor microenvironment. Overall, these studies demonstrate that through interaction with collagen proteins, DCs can be capable of modifying the microenvironments of inflammatory disease such as cancer or atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Neoplasias de la Mama/metabolismo , Células Dendríticas/metabolismo , Neoplasias Ováricas/metabolismo , Receptores de Colágeno/metabolismo , Animales , Apolipoproteínas E/genética , Aterosclerosis/inmunología , Neoplasias de la Mama/inmunología , Antígeno CD11c/metabolismo , Moléculas de Adhesión Celular/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Proliferación Celular , Quimiocina CXCL1/biosíntesis , Quimiocina CXCL2/biosíntesis , Quimiotaxis , Colágeno/metabolismo , Femenino , Integrina alfa1beta1/biosíntesis , Integrina alfa1beta1/metabolismo , Integrina alfa2beta1/biosíntesis , Integrina alfa2beta1/metabolismo , Integrina alfa3beta1/biosíntesis , Integrina alfa3beta1/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Neovascularización Fisiológica , Neoplasias Ováricas/inmunología , Receptores de Colágeno/biosíntesis , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/metabolismo , Receptores Depuradores de Clase A/biosíntesis , Receptores Depuradores de Clase A/metabolismo , Microambiente Tumoral , Regulación hacia Arriba
10.
J Cell Physiol ; 229(1): 68-78, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23765988

RESUMEN

Although recent evidence indicates that several chemokines and defensins, well-known as inflammatory mediators, are expressed in the male and female reproductive tracts, the location and functional significance of chemokine networks in sperm physiology and sperm reproductive tract interactions are poorly understood. To address this deficiency in our knowledge, we examined the expression and function in sperm of CCR6, a receptor common to several chemoattractant peptides, and screened several reproductive tract fluids for the presence of specific ligands. CCR6 protein is present in mouse and human sperm and mainly localized in the sperm tail with other minor patterns in sperm from mice (neck and acrosomal region) and men (neck and midpiece regions). As expected from the protein immunoblotting and immunofluorescence results, mouse Ccr6 mRNA is expressed in the testis. Furthermore, the Defb29 mRNA encoding the CCR6 ligand, ß-defensin DEFB29, is expressed at high levels in the epididymis. As determined by protein chip analysis, several chemokines (including some that act through CCR6, such as CCL20/MIP-3α (formerly macrophage inflammatory protein 3α) and protein hormones were present in human follicular fluid, endometrial secretions, and seminal plasma. In functional chemotaxis assays, capacitated human sperm exhibited a directional movement towards CCL20, and displayed modifications in motility parameters. Our data indicate that chemokine ligand/receptor interactions in the male and female genital tracts promote sperm motility and chemotaxis under non-inflammatory conditions. Therefore, some of the physiological reactions mediated by CCR6 ligands in male reproduction extend beyond a pro-inflammatory response and might find application in clinical reproduction and/or contraception.


Asunto(s)
Quimiotaxis/genética , Receptores CCR6/biosíntesis , Motilidad Espermática/genética , Espermatozoides/metabolismo , Animales , Quimiocina CCL20/biosíntesis , Quimiocina CCL20/genética , Epidídimo/citología , Epidídimo/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ratones , Receptores CCR6/genética , Espermatozoides/crecimiento & desarrollo , Espermatozoides/ultraestructura , beta-Defensinas/biosíntesis , beta-Defensinas/genética
11.
Semin Immunol ; 22(3): 132-43, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20356763

RESUMEN

Although cancer vaccines with defined antigens are commonly used, the use of whole tumor cell preparations in tumor immunotherapy is a very promising approach and can obviate some important limitations in vaccine development. Whole tumor cells are a good source of TAAs and can induce simultaneous CTLs and CD4(+) T helper cell activation. We review current approaches to prepare whole tumor cell vaccines, including traditional methods of freeze-thaw lysates, tumor cells treated with ultraviolet irradiation, and RNA electroporation, along with more recent methods to increase tumor cell immunogenicity with HOCl oxidation or infection with replication-incompetent herpes simplex virus.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/citología , Neoplasias Ováricas/inmunología , Animales , Apoptosis , Femenino , Humanos , Inmunoterapia/métodos , Ratones , Necrosis , Neoplasias Ováricas/terapia , Simplexvirus/inmunología
12.
Molecules ; 18(4): 3841-58, 2013 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-23535518

RESUMEN

Accumulating evidence supports a role for viruses in the pathogenesis of type 1 diabetes mellitus (T1DM). Activation of dsRNA-sensing pathways by viral dsRNA induces the production of inflammatory cytokines and chemokines that trigger beta cell apoptosis, insulitis, and autoimmune-mediated beta cell destruction. This study was designed to evaluate and describe potential protective effects of phenylmethimazole (C10), a small molecule which blocks dsRNA-mediated signaling, on preventing dsRNA activation of beta cell apoptosis and the inflammatory pathways important in the pathogenesis of T1DM. We first investigated the biological effects of C10, on dsRNA-treated pancreatic beta cells in culture. Cell viability assays, quantitative real-time PCR, and ELISAs were utilized to evaluate the effects of C10 on dsRNA-induced beta cell cytotoxicity and cytokine/chemokine production in murine pancreatic beta cells in culture. We found that C10 significantly impairs dsRNA-induced beta cell cytotoxicity and up-regulation of cytokines and chemokines involved in the pathogenesis of T1DM, which prompted us to evaluate C10 effects on viral acceleration of T1DM in NOD mice. C10 significantly inhibited viral acceleration of T1DM in NOD mice. These findings demonstrate that C10 (1) possesses novel beta cell protective activity which may have potential clinical relevance in T1DM and (2) may be a useful tool in achieving a better understanding of the role that dsRNA-mediated responses play in the pathogenesis of T1DM.


Asunto(s)
Diabetes Mellitus Tipo 1/prevención & control , Enterovirus/patogenicidad , Células Secretoras de Insulina/efectos de los fármacos , Metimazol/análogos & derivados , ARN Bicatenario/efectos adversos , Tionas/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Citocinas/sangre , Diabetes Mellitus Tipo 1/virología , Enterovirus/metabolismo , Femenino , Inflamación/tratamiento farmacológico , Inflamación/patología , Metimazol/farmacología , Ratones , Ratones Endogámicos NOD , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo , Regulación hacia Arriba
13.
J Biomed Biotechnol ; 2012: 425476, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22505809

RESUMEN

Many clinical trials have been carried out or are in progress to assess the therapeutic potential of dendritic-cell- (DC-) based vaccines on cancer patients, and recently the first DC-based vaccine for human cancer was approved by the FDA. Herewith, we describe the general characteristics of DCs and different strategies to generate effective antitumor DC vaccines. In recent years, the relevance of the tumor microenvironment in the progression of cancer has been highlighted. It has been shown that the tumor microenvironment is capable of inactivating various components of the immune system responsible for tumor clearance. In particular, the effect of the tumor microenvironment on antigen-presenting cells, such as DCs, does not only render these immune cells unable to induce specific immune responses, but also turns them into promoters of tumor growth. We also describe strategies likely to increase the efficacy of DC vaccines by reprogramming the immunosuppressive nature of the tumor microenvironment.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacología , Células Dendríticas/inmunología , Microambiente Tumoral/inmunología , Animales , Humanos
14.
BMC Immunol ; 12: 35, 2011 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-21645356

RESUMEN

BACKGROUND: Dendritic cells (DCs) are antigen presenting cells capable of inducing specific immune responses against microbial infections, transplant antigens, or tumors. Interestingly, microenvironment conditions such as those present in tumor settings might induce a DC phenotype that is poorly immunogenic and with the capability of promoting angiogenesis. We hypothesize that this plasticity may be caused not only by the action of specific cytokines or growth factors but also by the properties of the surfaces with which they interact, such as extracellular matrix (ECM) components. RESULTS: Herewith we studied the effect of different surfaces and soluble factors on the biology of DCs. To accomplish this, we cultured murine myeloid(m) DCs on surfaces coated with fibronectin, collagen I, gelatin, and Matrigel using poly-D-lysine and polystyrene as non-biological surfaces. Further, we cultured these cells in the presence of regular DC medium (RPMI 10% FBS) or commercially available endothelial medium (EGM-2). We determined that mDCs could be kept in culture up to 3 weeks in these conditions, but only in the presence of GM-CSF. We were able to determine that long-term DC cultures produce an array of angiogenic factors, and that some of these cultures still retain the capability to induce T cell responses. CONCLUSIONS: Altogether these data indicate that in order to design DC-based vaccines or treatments focused on changing the phenotype of DCs associated with diseases such as cancer or atherosclerosis, it becomes necessary to fully investigate the microenvironment in which these cells are present or will be delivered.


Asunto(s)
Células Dendríticas/fisiología , Células Mieloides/fisiología , Proteínas Angiogénicas/metabolismo , Animales , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Colágeno , Colágeno Tipo I/farmacología , Células Dendríticas/efectos de los fármacos , Combinación de Medicamentos , Femenino , Fibronectinas/farmacología , Gelatina/farmacología , Laminina , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Células Mieloides/efectos de los fármacos , Polilisina , Poliestirenos , Proteoglicanos , Linfocitos T/inmunología
15.
J Transl Med ; 9: 77, 2011 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-21609494

RESUMEN

BACKGROUND: Time-dependent chemotherapeutic agents can selectively target tumor cells in susceptible phases of the cell cycle however a fraction of tumor cells in non-vulnerable cell cycle phases remain drug-resistant. Immunotherapy represents a promising approach to overcome the limitation of phase-specific drugs and improve their clinical efficacy. Here, we investigated the potential use of anticancer chemotherapeutic drugs in combination with IL-18, a cytokine with strong immunostimulatory properties. METHODS: Four chemotherapeutic drugs commonly used in ovarian cancer were first tested for the ability to increase the immunogenicity and killing of the murine ovarian cancer cell line ID8 in vitro. Chemotherapeutric agents with measured time-dependent immune-enhancing effects were then tested for antitumor effectiveness in vivo in combination with IL-18 immunotherapy using the ID8-Vegf ovarian cancer model. RESULTS: Paclitaxel or topotecan exposure alone mediated incomplete, time-dependent killing against the murine ovarian cancer cell line ID8 in vitro, whereas carboplatin or gemcitabine mediated comprehensive, dose-dependent killing. In the plateau phase of the time-dependent killing by topotecan or paclitaxel, drug-resistant ID8 cells were more immunogenic with elevated expression of MHC-I and Fas, and increased sensitivity to CTL and Fas agonistic antibody in vitro. Moreover, the antitumor effectiveness of time-dependent agents in vivo was significantly improved with the addition of IL-18 through a T cell-dependent mechanism, while the effectiveness of drugs without significant phase specificity were not. CONCLUSIONS: Tumor immunotherapy with IL-18 can significantly augment the killing fraction of phase-specific chemotherapeutic drugs and provide survival benefit. The safety profile of IL-18 and its positive interactions with select anticancer chemotherapeutic agents strongly supports the clinical investigation of this combinatorial approach.


Asunto(s)
Antineoplásicos/uso terapéutico , Inmunoterapia , Interleucina-18/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Animales , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Interleucina-18/farmacología , Ratones , Ratones Endogámicos C57BL , Neoplasias/inmunología , Paclitaxel/farmacología , Linfocitos T/efectos de los fármacos , Factores de Tiempo , Topotecan/farmacología , Regulación hacia Arriba/efectos de los fármacos , Receptor fas/metabolismo
16.
Nat Med ; 10(9): 950-8, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15334073

RESUMEN

The involvement of immune mechanisms in tumor angiogenesis is unclear. Here we describe a new mechanism of tumor vasculogenesis mediated by dendritic cell (DC) precursors through the cooperation of beta-defensins and vascular endothelial growth factor-A (Vegf-A). Expression of mouse beta-defensin-29 recruited DC precursors to tumors and enhanced tumor vascularization and growth in the presence of increased Vegf-A expression. A new leukocyte population expressing DC and endothelial markers was uncovered in mouse and human ovarian carcinomas coexpressing Vegf-A and beta-defensins. Tumor-infiltrating DCs migrated to tumor vessels and independently assembled neovasculature in vivo. Bone marrow-derived DCs underwent endothelial-like differentiation ex vivo, migrated to blood vessels and promoted the growth of tumors expressing high levels of Vegf-A. We show that beta-defensins and Vegf-A cooperate to promote tumor vasculogenesis by carrying out distinct tasks: beta-defensins chemoattract DC precursors through CCR6, whereas Vegf-A primarily induces their endothelial-like specialization and migration to vessels, which is mediated by Vegf receptor-2.


Asunto(s)
Inductores de la Angiogénesis/inmunología , Células Dendríticas/inmunología , Neovascularización Patológica/inmunología , Factor A de Crecimiento Endotelial Vascular/metabolismo , beta-Defensinas/inmunología , Animales , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Colágeno , Células Dendríticas/metabolismo , Combinación de Medicamentos , Citometría de Flujo , Técnicas Histológicas , Immunoblotting , Laminina , Ratones , Proteoglicanos , Receptores de Quimiocina/metabolismo , beta-Defensinas/metabolismo
17.
Cell Immunol ; 261(1): 42-50, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19931043

RESUMEN

Herewith we investigated the role of nitric oxide synthase (NOS)-II in the establishment of oral tolerance induced by low antigen dose. To accomplish this, we used a rat model of oral tolerance induced by intragastric administration of low doses of ovalbumin (OVA). NOS-II was inhibited in vivo during the onset of tolerance by intraperitoneal (i.p.) treatment with aminoguanidine (AMG), a selective NOS-II inhibitor. Four experimental groups were generated: (TOL), tolerised rats, receiving OVA but no AMG; (TAG), rats tolerised with OVA and simultaneously receiving AMG i.p.; (CAG), controls treated with AMG but no oral antigen; and (CONT), controls receiving neither OVA nor AMG treatment. The state of oral tolerance was evaluated in all groups by analysing several immune parameters upon subcutaneous administration of OVA in Freund's complete adjuvant. First, we were able to determine that NOS-II inhibition altered the TH1/TH2 balance in tolerised rats, driving the TH2 anti-OVA response in TOL rats towards TH1 in TAG animals, which showed enhanced delayed hypersensitivity responses. Second, splenocyte cultures from TAG rats showed lower levels of IL-10 production compared to TOL samples as determined by ELISA analysis. Last, we detected the presence of a functional distinct Tr1 regulatory T cell population in spleen samples recovered from TAG animals. Contrary to what happened with TOL Tr1 cells, the levels of Tr1 cells in TAG samples were modified by in vitro stimulation with OVA. All together, these data indicate a preponderant role for NOS-II in the process of oral tolerance induced by low antigen dose.


Asunto(s)
Guanidinas/administración & dosificación , Tolerancia Inmunológica/efectos de los fármacos , Administración Oral , Animales , Células Cultivadas , Pollos , Inhibidores Enzimáticos/farmacología , Femenino , Inyecciones Intraperitoneales , Modelos Animales , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Ovalbúmina/inmunología , Ratas , Ratas Wistar , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células Th2/efectos de los fármacos , Células Th2/inmunología
19.
J Neuroendocrinol ; 32(11): e12854, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32350959

RESUMEN

Growth hormone (GH) excess in bovine (b)GH transgenic mice has been shown to alter white adipose tissue (WAT) immune cell populations. The present study aimed to evaluate the effects of GH resistance on WAT immune cell populations using GH receptor knockout (GHR-/- ) mice. Eight- and 24-month-old, male GHR-/- and wild-type mice were used. Body composition and tissue weights were determined, and systemic inflammation was assessed by measuring serum cytokine levels. The stromal vascular fraction (SVF) was isolated from three distinct WAT depots, and immune cell populations were quantified using flow cytometry. GHR-/- mice at both ages had decreased body weight but were obese. Although no significant changes were observed in serum levels of the measured cytokines, SVF cell alterations were seen and differed from depot to depot. Total SVF cells were decreased in epidydimal (Epi) depots, whereas SVF cells per gram adipose tissue weight were increased in mesenteric (Mes) depots of GHR-/- mice relative to controls. T cells and T helper cells were increased in Mes at 8 months old, whereas cytotoxic T cells were decreased in subcutaneous (SubQ) at 24 months old. Other cells were unchanged at both ages measured. The present study demonstrates that removal of GH action results in modest and depot-specific changes to several immune cell populations in WAT of intra-abdominal depots (Epi and Mes), which are somewhat surprising results because the SubQ has the largest change in size, whereas the Mes has no size change. Taken together with previous results from bovine GH transgenic mice, these data suggest that GH induces changes in the immune cell population of WAT in a depot-specific manner. Notably, GHR-/- mice appear to be protected from age-related WAT inflammation and immune cell infiltration despite obesity.


Asunto(s)
Tejido Adiposo Blanco/patología , Proteínas Portadoras/genética , Inflamación/genética , Inflamación/patología , Obesidad/genética , Obesidad/patología , Grasa Abdominal/inmunología , Grasa Abdominal/patología , Tejido Adiposo Blanco/inmunología , Envejecimiento , Animales , Composición Corporal , Citocinas/sangre , Epidídimo/patología , Hormona del Crecimiento/genética , Hormona del Crecimiento/metabolismo , Inmunidad Celular/genética , Inmunidad Celular/inmunología , Inflamación/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Obesidad/inmunología , Tamaño de los Órganos , Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores/inmunología
20.
Immunobiology ; 225(1): 151854, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31753553

RESUMEN

Dendritic cells (DCs) are immune cells that surveil the organism for infections or malignancies and activate specific T lymphocytes initiating specific immune responses. Contrariwise, DCs have been show to participate in the development of diseases, among them some types of cancer by inducing angiogenesis or immunosuppression. The ultimate fate of DC functions regarding their role in disease or health is prompted by signals from the microenvironment. We have previously shown that the interaction of DCs with various extracellular matrix components modifies the immune properties and angiogenic potential of these cells. The objective of the current studies was to investigate the angiogenic and immune profile of murine myeloid DCs upon interaction with laminin environments, with a particular emphasis on ovarian cancer. Our results show that murine ovarian tumors produce several types of laminins, as determined by PCR analysis, and also that tumor-associated DCs, both from ascites or solid tumors express adhesion molecules capable of interacting with these molecules as determined by flow cytometry and PCR analysis. Further, we established that DCs cultured on laminin upregulate both AKT and MEK signaling pathways, and that long-term culture on laminin surfaces decreases the immunological capacities of these cells when compared to the same cells cultured on synthetic substrates. In addition, we observed that tumor conditioned media was able to modify the metabolic status of these cells, and also reprogram the development of DCs from bone marrow precursors towards the generation of myeloid-derived suppressor cells. Overall, these studies demonstrate that the interaction between soluble factors and extracellular matrix components of the ovarian cancer microenvironment shape the biology of DCs and thus help them become co-conspirators of tumor growth.


Asunto(s)
Células Dendríticas/fisiología , Matriz Extracelular/metabolismo , Laminina/metabolismo , Células Mieloides/fisiología , Neoplasias Ováricas/inmunología , Animales , Antígenos de Neoplasias/inmunología , Carcinogénesis , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Laminina/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neovascularización Patológica , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA