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










Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2255: 197-212, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34033105

RESUMEN

The rapid, efficient detection of cell death is critical for characterizing the underlying biology of in vitro disease models and, in particular, immunotherapy products used for preclinical therapeutic research. Traditional endpoint assays are laborious to perform for mass screening of therapeutic candidates and may fail to fully capture the kinetics of events surrounding the initiation, duration, and mechanisms of cell death-important events that may affect translational relevance and impact therapeutic decision-making during development. Here, we describe simple, efficient methods to measure apoptosis and immune cell killing in both adherent and nonadherent cell populations using the Incucyte® Live-Cell Analysis system and associated nonperturbing reagents, cells, and protocols. Assays are performed in the user's own incubator with minimal disturbance and may be readily incorporated into existing workflows. Users may multiplex to maximize data collection from each sample. The integrated, user-friendly software does not require advanced technical training, enabling rapid analysis. Taken together, this method provides essential kinetic insight for greater understanding of cell death and the dynamic interactions between immune cells and their targets.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Adhesión Celular , Procesamiento de Imagen Asistido por Computador/métodos , Imagen Molecular/métodos , Neoplasias/patología , Linfocitos T/patología , Humanos , Cinética , Neoplasias/metabolismo , Linfocitos T/inmunología , Células Tumorales Cultivadas
2.
Cancer Immunol Immunother ; 64(4): 479-91, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25655760

RESUMEN

The need for novel, effective adjuvants that are capable of eliciting stronger cellular and humoral adaptive immune responses to antigenic targets is well understood in the vaccine development field. Unfortunately, many adjuvants investigated thus far are either too toxic for human application or too weak to induce a substantial response against difficult antigens, such as tumor-associated antigens (TAAs). In spite of this trend, clinical investigations of recombinant Eimeria antigen (rEA) have revealed this protein to be a non-toxic immunogenic agent with the ability to trigger a Th1-predominant response in both murine and human subjects. Our past studies have shown that the injection of a rEA-encoding adenovirus (rAd5-rEA) alongside an HIV antigen-encoding adenovirus greatly improves the adaptive immune response against this pathogen-derived transgene. In this report, we investigated whether rAd5-rEA could promote and/or alter cytotoxic memory responses toward carcinoembryonic antigen (CEA), a colorectal cancer-related TAA. We found that the addition of rAd5-rEA to an Ad-based CEA vaccine induced a dose-dependent increase in several anti-CEA T and B cell responses. Moreover, inclusion of rAd5-rEA increased the number of CEA-derived antigenic epitopes that elicited significant cell-mediated and IgG-mediated recognition. These enhanced anti-CEA immune responses also translated into superior CEA-targeted cell killing, as evaluated by an in vivo cytotoxic T lymphocyte assay. Overall, these results suggest that co-administration of rAd5-rEA with a tumor antigen vaccine can substantially boost and broaden the TAA-specific adaptive memory response, thereby validating the potential of rAd5-rEA to be a beneficial adjuvant during therapeutic cancer vaccination.


Asunto(s)
Adenoviridae/genética , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/farmacología , Antígeno Carcinoembrionario/inmunología , Neoplasias del Colon/inmunología , Eimeria/inmunología , Linfocitos T Citotóxicos/inmunología , Adenoviridae/inmunología , Adyuvantes Inmunológicos , Animales , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Antígeno Carcinoembrionario/genética , Neoplasias del Colon/prevención & control , Eimeria/genética , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Vectores Genéticos/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Prohibitinas , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Vacunación
3.
Methods Mol Biol ; 1219: 35-42, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25308260

RESUMEN

In vitro cell proliferation and apoptosis assays are widely used to study cancer cell biology. Commonly used methodologies are however performed at a single, user-defined endpoint. We describe a kinetic multiplex assay incorporating the CellPlayer(TM) NucLight Red reagent to measure proliferation and the CellPlayer(TM) Caspase-3/7 reagent to measure apoptosis using the two-color, live-content imaging platform, IncuCyte(TM) ZOOM. High-definition phase-contrast images provide an additional qualitative validation of cell death based on morphological characteristics. The kinetic data generated using this strategy can be used to derive informed pharmacology measurements to screen potential cancer therapeutics.


Asunto(s)
Apoptosis , Proliferación Celular , Imagen Molecular/métodos , Animales , Caspasa 3 , Caspasa 7 , Técnicas de Cultivo de Célula , Línea Celular , Células HeLa , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Indicadores y Reactivos , Cinética , Imagen Molecular/instrumentación
4.
PLoS One ; 6(8): e24147, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21912619

RESUMEN

BACKGROUND: Malaria greatly impacts the health and wellbeing of over half of the world's population. Promising malaria vaccine candidates have attempted to induce adaptive immune responses to Circumsporozoite (CS) protein. Despite the inclusion of potent adjuvants, these vaccines have limited protective efficacy. Conventional recombinant adenovirus (rAd) based vaccines expressing CS protein can induce CS protein specific immune responses, but these are essentially equivalent to those generated after use of the CS protein subunit based vaccines. In this study we combined the use of rAds expressing CS protein along with rAds expressing novel innate immune response modulating proteins in an attempt to significantly improve the induction of CS protein specific cell mediated immune (CMI) responses. METHODS AND FINDINGS: BALB/cJ mice were co-vaccinated with a rAd vectors expressing CS protein simultaneous with a rAd expressing either TLR agonist (rEA) or SLAM receptors adaptor protein (EAT-2). Paradoxically, expression of the TLR agonist uncovered a potent immunosuppressive activity inherent to the combined expression of the CS protein and rEA. Fortunately, use of the rAd vaccine expressing EAT-2 circumvented CS protein's suppressive activity, and generated a fivefold increase in the number of CS protein responsive, IFNγ secreting splenocytes, as well as increased the breadth of T cells responsive to peptides present in the CS protein. These improvements were positively correlated with the induction of a fourfold improvement in CS protein specific CTL functional activity in vivo. CONCLUSION: Our results emphasize the need for caution when incorporating CS protein into malaria vaccine platforms expressing or containing other immunostimulatory compounds, as the immunological outcomes may be unanticipated and/or counter-productive. However, expressing the SLAM receptors derived signaling adaptor EAT-2 at the same time of vaccination with CS protein can overcome these concerns, as well as significantly improve the induction of malaria antigen specific adaptive immune responses in vivo.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Inmunidad Celular/inmunología , Inmunidad Innata/inmunología , Factores Inmunológicos/inmunología , Proteínas Protozoarias/inmunología , Vacunas/genética , Vacunas/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Adenoviridae/genética , Animales , Especificidad de Anticuerpos , Presentación de Antígeno/inmunología , Linfocitos B/inmunología , Tolerancia Inmunológica/inmunología , Factores Inmunológicos/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas Protozoarias/genética , Linfocitos T/inmunología , Receptores Toll-Like/agonistas
5.
PLoS One ; 6(7): e22064, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21760953

RESUMEN

Despite recent advances in developing and licensing adjuvants, there is a great need for more potent formulations to enhance immunogenicity of vaccines. An Eimeria tenella derived antigen (rEA) augments immune responses against several pathogens in animal models and recently was confirmed to be safe for human use. In this study, we have analyzed the molecular mechanisms underlying rEA activity in mice, and confirmed that rEA activates multiple immune cell types, including DCs, macrophages, NK, B, and T cells. The rEA adjuvant also elicits the induction of pleiotropic pro-inflammatory cytokines, responses that completely depend upon the presence of the TLR adaptor protein MyD88. Surprisingly, we also found that the TRIF adaptor protein acts as a potent negative regulator of TLR agonist-triggered immune responses. For example, IL12 production and the induction of co-stimulatory molecule expression by DCs and IFNγ production by NK cells in vivo were significantly increased in rEA-treated TRIF-KO mice. Importantly, however, TRIF suppressive effects were not restricted to rEA-mediated responses, but were apparent in LPS- or ODN2006-activated DCs as well. Taken together, our findings confirm that rEA is a potent adjuvant, triggering robust activation of the innate immune system, in a manner that is augmented by MyD88 and inhibited by TRIF; thereby unveiling the potential complexities of modulating TLR activity to augment vaccine efficacy.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Células Dendríticas/inmunología , Receptores Toll-Like/agonistas , Animales , Antígenos de Protozoos/inmunología , Linfocitos B/inmunología , Quimiocinas/metabolismo , Células Dendríticas/citología , Células Dendríticas/parasitología , Eimeria tenella/inmunología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Mediadores de Inflamación , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Macrófagos/citología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Modelos Inmunológicos , Factor 88 de Diferenciación Mieloide/metabolismo , Células T Asesinas Naturales/inmunología , Fosforilación , Prohibitinas , Receptores Toll-Like/inmunología
6.
Hum Gene Ther ; 22(9): 1083-94, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21388344

RESUMEN

Adenovirus (Ad)-based vectors are attractive candidates for a variety of gene-transfer applications. In this study, we found that decay-accelerating factor (DAF)-displaying Ads induce significantly decreased cellular immune responses to transgenes expressed from the vectors in both Ad5-naive and Ad5-immune mice. Specifically, we found a diminished ability of splenocytes to secrete interferon-γ after recall exposure to multiple peptides derived from antigens expressed by DAF-displaying Ads. We also confirmed that DAF-displaying Ads induce decreased numbers of antigen-specific, CD8(+) effector memory and central memory CD8(+) T cells, thereby uncovering a unique role of complement in modulating the induction of robust memory T-cell responses. We also confirmed that DAF-displaying Ads generate significantly reduced titers of Ad capsid-specific neutralizing antibodies after gene transfer in vivo. In conclusion, DAF-displaying Ad5-based vectors exhibit decreased induction of complement-dependent, innate immune responses, resulting in both an improved safety profile and a decreased propensity to induce humoral and cellular adaptive immune responses to Ad capsid proteins and Ad vector-expressed transgene products. This attractive combination of features will be beneficial in a variety of clinically relevant gene-transfer applications.


Asunto(s)
Inmunidad Adaptativa , Adenoviridae/genética , Adenoviridae/inmunología , Antígenos CD55/genética , Vectores Genéticos/inmunología , Transgenes , Animales , Epítopos/inmunología , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Células HEK293 , Humanos , Inmunidad Celular , Inmunidad Humoral , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Transducción Genética
7.
J Cell Physiol ; 226(5): 1323-33, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20945396

RESUMEN

G-protein coupled receptor kinase-5 (GRK5) is a serine/threonine kinase discovered for its role in the regulation of G-protein coupled receptor signaling. Recent studies have shown that GRK5 is also an important regulator of signaling pathways stimulated by non-GPCRs. This study was undertaken to determine the physiological role of GRK5 in Toll-like receptor-4-induced inflammatory signaling pathways in vivo and in vitro. Using mice genetically deficient in GRK5 (GRK5(-/-) ) we demonstrate here that GRK5 is an important positive regulator of lipopolysaccharide (LPS, a TLR4 agonist)-induced inflammatory cytokine and chemokine production in vivo. Consistent with this role, LPS-induced neutrophil infiltration in the lungs (assessed by myeloperoxidase activity) was markedly attenuated in the GRK5(-/-) mice compared to the GRK5(+/+) mice. Similar to the in vivo studies, primary macrophages from GRK5(-/-) mice showed attenuated cytokine production in response to LPS. Our results also identify TLR4-induced NFκB pathway in macrophages to be selectively regulated by GRK5. LPS-induced IκBα phosphorylation, NFκB p65 nuclear translocation, and NFκB binding were markedly attenuated in GRK5(-/-) macrophages. Together, our findings demonstrate that GRK5 is a positive regulator of TLR4-induced IκBα-NFκB pathway as well as a key modulator of LPS-induced inflammatory response.


Asunto(s)
Quinasa 5 del Receptor Acoplado a Proteína-G/efectos de los fármacos , Inflamación/enzimología , Lipopolisacáridos/farmacología , Pulmón/efectos de los fármacos , Macrófagos Peritoneales/efectos de los fármacos , FN-kappa B/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Femenino , Quinasa 5 del Receptor Acoplado a Proteína-G/deficiencia , Quinasa 5 del Receptor Acoplado a Proteína-G/genética , Quinasa 5 del Receptor Acoplado a Proteína-G/metabolismo , Proteínas I-kappa B/metabolismo , Inflamación/inducido químicamente , Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Pulmón/enzimología , Pulmón/inmunología , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidor NF-kappaB alfa , Infiltración Neutrófila/efectos de los fármacos , Fosforilación , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/metabolismo
8.
J Cell Physiol ; 226(3): 627-37, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20717897

RESUMEN

G-protein-coupled receptor kinase 2 (GRK2) is a member of a kinase family originally discovered for its role in the phosphorylation and desensitization of G-protein-coupled receptors. It is expressed in high levels in myeloid cells and its levels are altered in many inflammatory disorders including sepsis. To address the physiological role of myeloid cell-specific GRK2 in inflammation, we generated mice bearing GRK2 deletion in myeloid cells (GRK2▵mye). GRK2▵mye mice exhibited exaggerated inflammatory cytokine/chemokine production, and organ injury in response to lipopolysaccharide (LPS, a TLR4 ligand) when compared to wild-type littermates (GRK2fl/fl). Consistent with this, peritoneal macrophages from GRK2▵mye mice showed enhanced inflammatory cytokine levels when stimulated with LPS. Our results further identify TLR4-induced NF-κB1p105-ERK pathway to be selectively regulated by GRK2. LPS-induced activation of NF-κB1p105-MEK-ERK pathway is significantly enhanced in the GRK2▵mye macrophages compared to GRK2fl/fl cells and importantly, inhibition of the p105 and ERK pathways in the GRK2▵mye macrophages, limits the enhanced production of LPS-induced cytokines/chemokines. Taken together, our studies reveal previously undescribed negative regulatory role for GRK2 in TLR4-induced p105-ERK pathway as well as in the consequent inflammatory cytokine/chemokine production and endotoxemia in mice.


Asunto(s)
Endotoxemia/enzimología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Sistema de Señalización de MAP Quinasas , Células Mieloides/enzimología , Subunidad p50 de NF-kappa B/metabolismo , Choque Séptico/enzimología , Animales , Células Cultivadas , Quimiocina CCL3/metabolismo , Endotoxemia/complicaciones , Endotoxemia/patología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Inflamación/complicaciones , Inflamación/enzimología , Inflamación/patología , Interleucina-10/metabolismo , Subunidad p40 de la Interleucina-12/metabolismo , Lipopolisacáridos/farmacología , Quinasas Quinasa Quinasa PAM/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Modelos Biológicos , Células Mieloides/efectos de los fármacos , Células Mieloides/patología , Subunidad p50 de NF-kappa B/antagonistas & inhibidores , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Especificidad de Órganos/efectos de los fármacos , Choque Séptico/complicaciones , Choque Séptico/patología , Análisis de Supervivencia
9.
J Immunol ; 186(2): 722-32, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21149608

RESUMEN

Recent studies have shown that activation of the signaling lymphocytic activation molecule (SLAM) family of receptors plays an important role in several aspects of immune regulation. However, translation of this knowledge into a useful clinical application has not been undertaken. One important area where SLAM-mediated immune regulation may have keen importance is in the field of vaccinology. Because SLAM signaling plays such a critical role in the innate and adaptive immunity, we endeavored to develop a strategy to improve the efficacy of vaccines by incorporation of proteins known to be important in SLAM-mediated signaling. In this study, we hypothesized that coexpression of the SLAM adapter EWS-FLI1-activated transcript 2 (EAT-2) along with a pathogen-derived Ag would facilitate induction of beneficial innate immune responses, resulting in improved induction of Ag-specific adaptive immune responses. To test this hypothesis, we used rAd5 vector-based vaccines expressing murine EAT-2, or the HIV-1-derived Ag Gag. Compared with appropriate controls, rAd5 vectors expressing EAT-2 facilitated bystander activation of NK, NKT, B, and T cells early after their administration into animals. EAT-2 overexpression also augments the expression of APC (macrophages and dendritic cells) surface markers. Indeed, this multitiered activation of the innate immune system by vaccine-mediated EAT-2 expression enhanced the induction of Ag-specific cellular immune responses. Because both mice and humans express highly conserved EAT-2 adapters, our results suggest that human vaccination strategies that specifically facilitate SLAM signaling may improve vaccine potency when targeting HIV Ags specifically, as well as numerous other vaccine targets in general.


Asunto(s)
Adenovirus Humanos/inmunología , Péptidos y Proteínas de Señalización Intracelular/administración & dosificación , Péptidos y Proteínas de Señalización Intracelular/genética , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/genética , Vacunas contra el SIDA/inmunología , Proteínas Adaptadoras Transductoras de Señales , Infecciones por Adenoviridae/genética , Infecciones por Adenoviridae/inmunología , Infecciones por Adenoviridae/terapia , Adenovirus Humanos/genética , Animales , Línea Celular , Células Cultivadas , Ingeniería Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Humanos , Inmunidad Celular/genética , Inmunidad Innata/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Familia de Multigenes/inmunología , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria , Factores de Transcripción/administración & dosificación , Factores de Transcripción/fisiología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología
10.
Clin Vaccine Immunol ; 18(1): 150-60, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21084461

RESUMEN

HIV/AIDS continue to devastate populations worldwide. Recent studies suggest that vaccines that induce beneficial immune responses in the mucosal compartment may improve the efficacy of HIV vaccines. Adenovirus serotype 5 (Ad5)-based vectors remain a promising platform for the development of effective vaccines. In an effort to improve the efficacy of Ad5-based vaccines, even in the presence of preexisting Ad5 immunity, we evaluated the potential for an Ad5-based HIV vaccine to induce antigen-specific immune responses following sublingual (s.l.) administration, a route not previously tested in regard to Ad-based vaccines. s.l. vaccination with an Ad5-based HIV-Gag vaccine resulted in a significant induction of Gag-specific cytotoxic T-lymphocyte (CTL) responses in both the systemic and the mucosal compartment. We also show that s.l. immunization not only avoided preexisting Ad5 immunity but also elicited a broad repertoire of antigen-specific CTL clones. Additionally, we confirm for the first time that oral delivery of a vaccine expressing a potent Toll-like receptor (TLR) agonist can stimulate innate immune responses through induction of cytokines and chemokines and activation of NK cells, NKT cells, and macrophages in vivo. These results positively correlated with improved antigen-specific CTL responses. These results could be achieved both in Ad5-naïve mice and in mice with preexisting immunity to Ad5. The simplicity of the s.l. vaccination regimen coupled with augmentation of TLR-dependent pathways active in the oral cavity makes s.l. delivery a promising method for HIV vaccine development specifically, as well as for many other vaccine applications in general.


Asunto(s)
Vacunas contra el SIDA/administración & dosificación , Adenovirus Humanos/genética , Vectores Genéticos/genética , Antígenos VIH/inmunología , Inmunidad Mucosa , Linfocitos T Citotóxicos/inmunología , Receptores Toll-Like/agonistas , Vacunas contra el SIDA/inmunología , Adenovirus Humanos/clasificación , Administración Sublingual , Animales , Productos del Gen gag/genética , Productos del Gen gag/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Boca/inmunología , Receptores Toll-Like/metabolismo , Vacunación
11.
J Cell Physiol ; 225(2): 406-16, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20589830

RESUMEN

Beta-arrestins are scaffolding proteins implicated as negative regulators of TLR4 signaling in macrophages and fibroblasts. Unexpectedly, we found that beta-arrestin-1 (beta-arr-1) and -2 knockout (KO) mice are protected from TLR4-mediated endotoxic shock and lethality. To identify the potential mechanisms involved, we examined the plasma levels of inflammatory cytokines/chemokines in the wild-type (WT) and beta-arr-1 and -2 KO mice after lipopolysaccharide (LPS, a TLR4 ligand) injection. Consistent with lethality, LPS-induced inflammatory cytokine levels in the plasma were markedly decreased in both beta-arr-1 and -2 KO, compared to WT mice. To further explore the cellular mechanisms, we obtained splenocytes (separated into CD11(b+) and CD11(b-) populations) from WT, beta-arr-1, and -2 KO mice and examined the effect of LPS on cytokine production. Similar to the in vivo observations, LPS-induced inflammatory cytokines were significantly blocked in both splenocyte populations from the beta-arr-2 KO compared to the WT mice. This effect in the beta-arr-1 KO mice, however, was restricted to the CD11(b-) splenocytes. Our studies further indicate that regulation of cytokine production by beta-arrestins is likely independent of MAPK and IkappaBalpha-NFkappaB pathways. Our results, however, suggest that LPS-induced chromatin modification is dependent on beta-arrestin levels and may be the underlying mechanistic basis for regulation of cytokine levels by beta-arrestins in vivo. Taken together, these results indicate that beta-arr-1 and -2 mediate LPS-induced cytokine secretion in a cell-type specific manner and that both beta-arrestins have overlapping but non-redundant roles in regulating inflammatory cytokine production and endotoxic shock in mice.


Asunto(s)
Arrestinas/metabolismo , Endotoxemia/metabolismo , Inflamación/inducido químicamente , Lipopolisacáridos/toxicidad , Animales , Arrestinas/genética , Antígenos CD11/metabolismo , Técnicas de Cultivo de Célula , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Noqueados , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Transducción de Señal , Bazo/citología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , beta-Arrestina 1 , beta-Arrestinas
12.
Blood ; 116(10): 1669-77, 2010 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-20511542

RESUMEN

Adenovirus (Ad) vectors are widely used in human clinical trials. However, at higher dosages, Ad vector-triggered innate toxicities remain a major obstacle to many applications. Ad interactions with the complement system significantly contribute to innate immune responses in several models of Ad-mediated gene transfer. We constructed a novel class of Ad vectors, genetically engineered to "capsid-display" native and retro-oriented versions of the human complement inhibitor decay-accelerating factor (DAF), as a fusion protein from the C-terminus of the Ad capsid protein IX. In contrast to conventional Ad vectors, DAF-displaying Ads dramatically minimized complement activation in vitro and complement-dependent immune responses in vivo. DAF-displaying Ads did not trigger thrombocytopenia, minimized endothelial cell activation, and had diminished inductions of proinflammatory cytokine and chemokine responses. The retro-oriented display of DAF facilitated the greatest improvements in vivo, with diminished activation of innate immune cells, such as dendritic and natural killer cells. In conclusion, Ad vectors can capsid-display proteins in a manner that not only retains the functionality of the displayed proteins but also potentially can be harnessed to improve the efficacy of this important gene transfer platform for numerous gene transfer applications.


Asunto(s)
Adenoviridae/genética , Antígenos CD55/genética , Proteínas de la Cápside/genética , Proteínas Recombinantes de Fusión/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Activación de Complemento/inmunología , Complemento C3/deficiencia , Complemento C3/genética , Complemento C3/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunidad Innata/inmunología , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Hígado/inmunología , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
13.
J Innate Immun ; 2(4): 353-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20375551

RESUMEN

Adenovirus (Ad) vectors are currently the most commonly utilized gene transfer vectors in humans worldwide. Unfortunately, upon contact with the circulatory system, Ads induce several, innate, complement-dependent toxicities that limit the full potential for Ad-based gene transfer applications. Therefore, we have constructed several novel Ad5-based vectors, 'capsid-displaying' as fiber or pIX fusion proteins, a complement-regulatory peptide (COMPinh). These novel Ads dramatically minimize Ad-dependent activation of the human and non-human primate complement systems, as determined by several assays. In summary, our work has shown that a novel COMPinh-displaying Ad5 has the potential for broadening the safe use of Ad vectors in future human applications.


Asunto(s)
Adenovirus Humanos/genética , Cápside/metabolismo , Inactivadores del Complemento/metabolismo , Vectores Genéticos , Péptidos/metabolismo , Proteínas Recombinantes de Fusión , Adenovirus Humanos/metabolismo , Animales , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Activación de Complemento/efectos de los fármacos , Vía Alternativa del Complemento , Proteínas del Sistema Complemento/efectos de los fármacos , Humanos , Inmunidad Innata , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción Genética
14.
PLoS One ; 5(3): e9579, 2010 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-20221448

RESUMEN

BACKGROUND: Adenoviral based vectors remain promising vaccine platforms for use against numerous pathogens, including HIV. Recent vaccine trials utilizing Adenovirus based vaccines expressing HIV antigens confirmed induction of cellular immune responses, but these responses failed to prevent HIV infections in vaccinees. This illustrates the need to develop vaccine formulations capable of generating more potent T-cell responses to HIV antigens, such as HIV-Gag, since robust immune responses to this antigen correlate with improved outcomes in long-term non-progressor HIV infected individuals. METHODOLOGY/PRINCIPAL FINDINGS: In this study we designed a novel vaccine strategy utilizing an Ad-based vector expressing a potent TLR agonist derived from Eimeria tenella as an adjuvant to improve immune responses from a [E1-]Ad-based HIV-Gag vaccine. Our results confirm that expression of rEA elicits significantly increased TLR mediated innate immune responses as measured by the influx of plasma cytokines and chemokines, and activation of innate immune responding cells. Furthermore, our data show that the quantity and quality of HIV-Gag specific CD8(+) and CD8(-) T-cell responses were significantly improved when coupled with rEA expression. These responses also correlated with a significantly increased number of HIV-Gag derived epitopes being recognized by host T cells. Finally, functional assays confirmed that rEA expression significantly improved antigen specific CTL responses, in vivo. Moreover, we show that these improved responses were dependent upon improved TLR pathway interactions. CONCLUSION/SIGNIFICANCE: The data presented in this study illustrate the potential utility of Ad-based vectors expressing TLR agonists to improve clinical outcomes dependent upon induction of robust, antigen specific immune responses.


Asunto(s)
Adenoviridae/genética , Eimeria tenella/genética , Antígenos VIH/química , Receptores Toll-Like/metabolismo , Vacunas contra el SIDA/química , Animales , Linfocitos T CD8-positivos/metabolismo , Eimeria tenella/metabolismo , Citometría de Flujo/métodos , Vectores Genéticos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Sistemas de Lectura Abierta , Prohibitinas , Linfocitos T Citotóxicos/metabolismo , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/química
15.
BMC Cancer ; 10: 13, 2010 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-20067638

RESUMEN

BACKGROUND: The activities of Rac1 and Cdc42 are essential for HRas-induced transformation of rodent fibroblasts. What is more, expression of constitutively activated mutants of Rac1 and/or Cdc42 is sufficient for their malignant transformation. The role for these two Rho GTPases in HRas-mediated transformation of human fibroblasts has not been studied. Here we evaluated the contribution of Rac1 and Cdc42 to maintaining HRas-induced transformation of human fibroblasts, and determined the ability of constitutively activated mutants of Rac1 or Cdc42 to induce malignant transformation of a human fibroblast cell strain. METHODS: Under the control of a tetracycline regulatable promoter, dominant negative mutants of Rac1 and Cdc42 were expressed in a human HRas-transformed, tumor derived fibroblast cell line. These cells were used to determine the roles of Rac1 and/or Cdc42 proteins in maintaining HRas-induced transformed phenotypes. Similarly, constitutively active mutants were expressed in a non-transformed human fibroblast cell strain to evaluate their potential to induce malignant transformation. Affymetrix GeneChip arrays were used for transcriptome analyses, and observed expression differences were subsequently validated using protein assays. RESULTS: Expression of dominant negative Rac1 and/or Cdc42 significantly altered transformed phenotypes of HRas malignantly transformed human fibroblasts. In contrast, expression of constitutively active mutants of Rac1 or Cdc42 was not sufficient to induce malignant transformation. Microarray analysis revealed that the expression of 29 genes was dependent on Rac1 and Cdc42, many of which are known to play a role in cancer. The dependence of two such genes, uPA and VEGF was further validated in both normoxic and hypoxic conditions. CONCLUSION(S): The results presented here indicate that expression of both Rac1 and Cdc42 is necessary for maintaining several transformed phenotypes in oncogenic HRas transformed human cells, including their ability to form tumors in athymic mice. Our data also indicate that expression of either activated Rac1 or Cdc42 alone is not sufficient for malignant transformation of human fibroblasts, although each is required for specific transformed phenotypes. Furthermore, our study elucidates that the expression of several highly significant cancer related genes require the activities of Rac1 and/or Cdc42 which may also play a critical role in cellular transformation.


Asunto(s)
Transformación Celular Neoplásica , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Neovascularización Patológica , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas ras/metabolismo , Inductores de la Angiogénesis/metabolismo , Humanos , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Tetraciclina/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Unión al GTP rho/metabolismo
16.
Virus Res ; 147(1): 123-34, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19896992

RESUMEN

Adenovirus (Ad)-based vectors have been utilized in human gene transfer clinical trials since 1993. Unfortunately, innate immune responses directed against the Ad capsid and/or its genetic cargo can significantly limit the usage of Ad vectors. Previous studies have demonstrated that several signaling pathways are triggered by Ads, inclusive of TLR-dependent pathways. The G-protein-coupled receptor adaptors beta-arrestin-1 (beta-Arr1) and beta-arrestin-2 (beta-Arr2) are known to have pivotal roles in regulating TLR4 triggered signaling and inflammatory responses. In this study, we examined the role of beta-arrestins in Ad5-vector-induced inflammatory responses. Our studies reveal that both beta-arrestins are capable of modulating Ad5-vector-induced inflammatory responses in vivo and in vitro. Importantly, our studies divulge another level of complexity to these responses, as our results demonstrate beta-Arr1 to be a positive regulator, and beta-Arr2 a negative regulator of Ad5 induced innate immune responses. These data may allow gene therapy biologists to more accurately study the mechanisms underlying Ad5-vector-induced immune responses, and may also direct future efforts to modulate these mechanisms to improve the safety and/or efficacy of this important gene transfer vector.


Asunto(s)
Adenoviridae/inmunología , Adenovirus Humanos/inmunología , Arrestinas/inmunología , Regulación de la Expresión Génica , Vectores Genéticos , Inmunomodulación , Animales , Citocinas/biosíntesis , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/virología , Ratones , Ratones Endogámicos C57BL , beta-Arrestina 1 , Arrestina beta 2 , beta-Arrestinas
17.
Infect Immun ; 77(6): 2499-507, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19332531

RESUMEN

Campylobacter jejuni is an important cause of human enteritis and has been linked to the development of autoimmune diseases. Recently we showed that infection of murine dendritic cells (DCs) with C. jejuni resulted in DC activation and induction of Campylobacter-specific Th1-effector responses. Toll-like receptor (TLR) signaling through myeloid differentiation factor 88 (MyD88) and/or Toll-interleukin 1 (IL-1) receptor domain-containing adaptor-inducing beta interferon (IFN-beta) (TRIF) is critical in inducing immunity against pathogens. In this study, we investigated the role of TLR2, TLR4, MyD88, and TRIF signaling in C. jejuni-induced inflammatory activation of DCs. DC upregulation of major histocompatibility complex class II and costimulatory molecules after C. jejuni challenge was profoundly impaired by TLR2, TLR4, MyD88, and TRIF deficiencies. Similarly, C. jejuni-induced secretion of IL-12, IL-6, and tumor necrosis factor alpha was significantly inhibited in TLR2(-/-), TLR4(-/-), MyD88(-/-), and TRIF(-/-) DCs compared to that in wild-type DCs; however, the magnitude of inhibition was greater in MyD88(-/-), TRIF(-/-), and TLR4(-/-) DCs than in TLR2(-/-) DCs. Furthermore, C. jejuni induced interferon regulatory factor 3 phosphorylation and IFN-beta secretion by DCs in a TLR4-TRIF-dependent fashion, further demonstrating activation of this pathway by C. jejuni. Importantly, TLR2, TLR4, MyD88, and TRIF deficiencies all markedly impaired the Th1-priming ability of C. jejuni-infected DCs. Thus, our results show that cooperative signaling through the TLR4-MyD88 and TLR4-TRIF axes represents a novel mechanism mediating C. jejuni-induced inflammatory responses of DCs. To our knowledge, such a mechanism has not been demonstrated previously for an intact bacterium.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/inmunología , Campylobacter jejuni/inmunología , Células Dendríticas/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Receptor Toll-Like 4/inmunología , Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Animales , Citocinas/metabolismo , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Fosforilación , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/deficiencia
18.
Mol Ther ; 17(4): 685-96, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19174760

RESUMEN

More than 300 human clinical trials utilize recombinant adenoviruses (rAds) as a gene transfer vector, confirming that rAds continue to be of high clinical interest. A primary weakness of rAds is their known propensity to trigger an innate, proinflammatory immune response rapidly after high-dose, systemic administration. In this study, we investigated what affects that pre-emptive treatment with anti-inflammatory glucocorticoids might have upon Ad vector-triggered inflammatory immune responses. We found that a simple pretreatment regimen with Dexamethasone (DEX) can significantly reduce most Ad-induced innate immune responses. DEX prevented rAd induction of systemic cytokine/chemokine releases in a dose-dependent fashion, with higher dosages preventing rAd induction of acute thrombocytopenia, endothelial cell activation, proinflammatory gene induction, and leukocyte infiltration into transduced organs. Transient glucocorticoid pretreatment also significantly reduced rAd-induced adaptive immune responses, including a decreased induction of Ad-neutralizing antibodies (NAbs). Importantly, use of DEX did not reduce the efficacy of rAd-mediated gene transduction nor rAd-derived transgene expression. Our results demonstrate that a simple, pre-emptive and transient glucocorticoid pretreatment is a viable approach to reduce rAd-associated acute toxicities that currently limit the use of Ad vectors in systemic clinical applications.


Asunto(s)
Adenoviridae/genética , Dexametasona/farmacología , Vectores Genéticos/efectos adversos , Inmunidad Innata/efectos de los fármacos , Animales , Expresión Génica/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Macrófagos del Hígado/citología , Hígado/citología , Hígado/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Transducción Genética
19.
J Cell Physiol ; 218(3): 575-83, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19006181

RESUMEN

Type I diabetes is associated with bone loss and marrow adiposity. To identify early events involved in the etiology of diabetic bone loss, diabetes was induced in mice by multiple low dose streptozotocin injections. Serum markers of bone metabolism and inflammation as well as tibial gene expression were examined between 1 and 17 days post-injection (dpi). At 3 dpi, when blood glucose levels were significantly elevated, body, fat pad and muscle mass were decreased. Serum markers of bone resorption and formation significantly decreased at 5 dpi in diabetic mice and remained suppressed throughout the time course. An osteoclast gene, TRAP5 mRNA, was suppressed at early and late time points. Suppression of osteogenic genes (runx2 and osteocalcin) and induction of adipogenic genes (PPARgamma2 and aP2) were evident as early as 5 dpi. These changes were associated with an elevation of serum cytokines, but more importantly we observed an increase in the expression of cytokines in bone, supporting the idea that bone, itself, exhibits an inflammatory response during diabetes induction. This inflammation could in turn contribute to diabetic bone pathology. IFN-gamma (one of the key cytokines elevated in bone and known to be involved in bone regulation) deficiency did not prevent diabetic bone pathology. Taken together, our findings indicate that bone becomes inflamed with the onset of T1-diabetes and during this time bone phenotype markers become altered. However, inhibition of one cytokine, IFN-gamma was not sufficient to prevent the rapid bone phenotype changes.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/genética , Regulación de la Expresión Génica , Osteítis/complicaciones , Osteítis/genética , Animales , Biomarcadores/metabolismo , Resorción Ósea/complicaciones , Resorción Ósea/prevención & control , Citocinas/sangre , Citocinas/genética , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Tipo 1/sangre , Femenino , Ratones , Ratones Endogámicos BALB C , Osteítis/sangre , Osteoclastos/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo
20.
J Biol Chem ; 284(2): 848-54, 2009 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-19008219

RESUMEN

We have reported that expression of Sprouty 2 (Spry2) is necessary for tumor formation by HRas(V12)-transformed fibroblasts. We now report on the role of Spry2 in the inhibition of UV(254 nm) radiation-induced apoptosis in HRas(V12)-transformed human fibroblasts. Silencing Spry2 in this context resulted in increased apoptosis, associated with decreased Akt activation and decreased phosphorylation of HDM2 at Ser-166, which has been shown to stabilize HDM2. As a consequence, when cells with silenced Spry2 were UV-irradiated, they exhibited diminished levels of HDM2 and elevated levels of p53. In agreement with these findings, overexpression of Spry2 in the parental non-transformed fibroblasts led to increased Akt activation and to the stabilization of HDM2. It also led to diminished expression of p53 and decreased apoptosis following UV irradiation. Silencing Spry2 in HRas-transformed cells decreased Rac1 activation, but independent expression of Spry2 in the non-transformed parental cells had no effect on Rac1, suggesting a specific involvement in the activation of Rac1 by Ras. Silencing Spry2 in HRas(V12)-transformed cells resulted in diminished interaction between HRas and Tiam1, a Rac1-specific nucleotide exchange factor. Expression of constitutively active Rac1 in cells with silenced Spry2 partly reversed the effect of Spry2 down-regulation. Furthermore, loss of Spry2 expression in HRas(V12)-transformed cells augmented the cytotoxicity of the DNA-damaging, chemotherapeutic agent cisplatin, a process that was also reversed by active Rac1. Together, these data show that Spry2 inhibits apoptosis in response to DNA damage by regulating Akt, HDM2, and p53, by a process mediated partly by Rac1.


Asunto(s)
Apoptosis , Daño del ADN/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína Oncogénica p21(ras)/metabolismo , Transgenes/genética , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Línea Celular , Cisplatino/toxicidad , Regulación hacia Abajo , Activación Enzimática , Fibroblastos , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana , Proteína Oncogénica p21(ras)/genética , Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...