Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Immunol ; 207(12): 2952-2965, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34810226

RESUMEN

Hundreds of genes have been linked to multiple sclerosis (MS); yet, the underlying mechanisms behind these associations have only been investigated in a fraction of cases. Endoplasmic reticulum aminopeptidase 1 (ERAP1) is an endoplasmic reticulum-localized aminopeptidase with important roles in trimming peptides destined for MHC class I and regulation of innate immune responses. As such, genetic polymorphisms in ERAP1 have been linked to multiple autoimmune diseases. In this study, we present, to our knowledge, the first mechanistic studies performed to uncover why polymorphisms in ERAP1 are associated with increased susceptibility to MS. Combining multiple mouse models of CNS autoimmunity with high-dimensional single-cell spectral cytometry, adoptive transfer studies, and integrative analysis of human single-cell RNA-sequencing datasets, we identify an intrinsic defect in B cells as being primarily responsible. Not only are mice lacking ERAP1 more susceptible to CNS autoimmunity, but adoptive transfer of B cells lacking ERAP1 into B cell-deficient mice recapitulates this susceptibility. We found B cells lacking ERAP1 display decreased proliferation in vivo and express higher levels of activation/costimulatory markers. Integrative analysis of single-cell RNA sequencing of B cells from 36 individuals revealed subset-conserved differences in gene expression and pathway activation in individuals harboring the MS-linked K528R ERAP1 single-nucleotide polymorphism. Finally, our studies also led us to create, to our knowledge, the first murine protein-level map of the CNS IL-10+ immune compartment at steady state and during neuroinflammation. These studies identify a role for ERAP1 in the modulation of B cells and highlight this as one reason why polymorphisms in this gene are linked to MS.


Asunto(s)
Enfermedades Autoinmunes , Linfocitos B , Esclerosis Múltiple , Aminopeptidasas/genética , Aminopeptidasas/metabolismo , Animales , Autoinmunidad/genética , Sistema Nervioso Central , Ratones , Antígenos de Histocompatibilidad Menor/genética , Esclerosis Múltiple/genética , Polimorfismo de Nucleótido Simple
2.
J Immunol ; 206(1): 193-205, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33288545

RESUMEN

T cell exhaustion represents one of the most pervasive strategies tumors employ to circumvent the immune system. Although repetitive, cognate TCR signaling is recognized as the primary driving force behind this phenomenon, and it remains unknown what other forces drive T cell exhaustion in the tumor microenvironment (TME). In this study, we show that activation of the self-ligand SLAMF7 immune receptor on T cells induced STAT1 and STAT3 phosphorylation, expression of multiple inhibitory receptors, and transcription factors associated with T cell exhaustion. Analysis of The Cancer Genome Atlas revealed that SLAMF7 transcript levels were strongly correlated with various inhibitory receptors and that high SLAMF7 expression was indicative of poor survival in clear cell renal cell carcinoma (ccRCC). Targeted reanalysis of a CyTOF dataset, which profiled the TME in 73 ccRCC patients, revealed cell-type-specific SLAMF7 expression patterns, strong correlations between exhausted T cells and SLAMF7+ tumor-associated macrophages (TAMs), and a unique subset of SLAMF7highCD38high TAMs. These SLAMF7highCD38high TAMs showed the strongest correlations with exhausted T cells and were an independent prognostic factor in ccRCC. Confirmatory ex vivo coculture studies validated that SLAMF7-SLAMF7 interactions between murine TAMs and CD8+ T cells induce expression of multiple inhibitory receptors. Finally, mice lacking SLAMF7 show restricted growth of B16-F10 tumors, and CD8+ T cells from these mice express less PD-1 and TOX and exhibited an impaired ability to progress through the exhaustion developmental trajectory to terminal exhaustion. These findings suggest that SLAMF7 might play an important role in modulating T cell function in the TME.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Neoplasias Renales/metabolismo , Melanoma/metabolismo , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Neoplasias Cutáneas/metabolismo , Linfocitos T/inmunología , Animales , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/mortalidad , Células Cultivadas , Reprogramación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Tolerancia Inmunológica , Neoplasias Renales/inmunología , Neoplasias Renales/mortalidad , Masculino , Melanoma/inmunología , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Experimentales , Transducción de Señal , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Neoplasias Cutáneas/inmunología , Análisis de Supervivencia , Microambiente Tumoral
3.
BMC Immunol ; 23(1): 9, 2022 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-35246034

RESUMEN

BACKGROUND: In addition to its role in antigen presentation, recent reports establish a new role for endoplasmic reticulum aminopeptidase 1 (ERAP1) in innate immunity; however, the mechanisms underlying these functions are not fully defined. We previously confirmed that loss of ERAP1 functions resulted in exaggerated innate immune responses in a murine in vivo model. Here, we investigated the role of ERAP1 in suppressing inflammasome pathways and their dependence on ER stress responses. RESULTS: Using bone marrow-derived macrophages (BMDMs), we found that loss of ERAP1 in macrophages resulted in exaggerated production of IL-1ß and IL-18 and augmented caspase-1 activity, relative to wild type macrophages. Moreover, an in vivo colitis model utilizing dextran sodium sulfate (DSS) confirmed increased levels of proinflammatory cytokines and chemokines in the colon of DSS treated ERAP1-/- mice as compared to identically stimulated WT mice. Interestingly, stimulated ERAP1-/- BMDMs and CD4+ T cells simultaneously demonstrated exaggerated ER stress, assessed by increased expression of ER stress-associated genes, a state that could be reverted to WT levels with use of the ER stress inhibitor Tauroursodeoxycholic acid (TUDCA). CONCLUSIONS: Together, these results not only suggest that ERAP1 is important for regulating inflammasome dependent innate immune response pathways in vivo, but also propose a mechanism that underlies these changes, that may be associated with increased ER stress due to lack of normal ERAP1 functions.


Asunto(s)
Aminopeptidasas , Estrés del Retículo Endoplásmico , Inflamasomas , Aminopeptidasas/genética , Aminopeptidasas/metabolismo , Animales , Inmunidad Innata/genética , Ratones , Ratones Noqueados , Antígenos de Histocompatibilidad Menor/genética
4.
J Neuroinflammation ; 19(1): 241, 2022 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-36199066

RESUMEN

BACKGROUND: Multiple sclerosis (MS) is a chronic, debilitating condition characterized by CNS autoimmunity stemming from a complex etiology involving both environmental and genetic factors. Our current understanding of MS points to dysregulation of the immune system as the pathogenic culprit, however, it remains unknown as to how the many genes associated with increased susceptibility to MS are involved. One such gene linked to MS susceptibility and known to regulate immune function is the self-ligand immune cell receptor SLAMF7. METHODS: We subjected WT and SLAMF7-/- mice to multiple EAE models, compared disease severity, and comprehensively profiled the CNS immune landscape of these mice. We identified all SLAMF7-expressing CNS immune cells and compared the entire CNS immune niche between genotypes. We performed deep phenotyping and in vitro functional studies of B and T cells via spectral cytometry and BioPlex assays. Adoptive transfer studies involving the transfer of WT and SLAMF7-/- B cells into B cell-deficient mice (µMT) were also performed. Finally, B-T cell co-culture studies were performed, and a comparative cell-cell interaction network derived from scRNA-seq data of SLAMF7+ vs. SLAMF7- human CSF immune cells was constructed. RESULTS: We found SLAMF7-/- mice to be more susceptible to EAE compared to WT mice and found SLAMF7 to be expressed on numerous CNS immune cell subsets. Absence of SLAMF7 did not grossly alter the CNS immune landscape, but allowed for altered immune cell subset infiltration during EAE in a model-dependent manner. Global lack of SLAMF7 expression increased myeloid cell activation states along with augmented T cell anti-MOG immunity. B cell profiling studies revealed increased activation states of specific plasma and B cell subsets in SLAMF7-/- mice during EAE, and functional co-culture studies determined that SLAMF7-/- B cells induce exaggerated T cell activation. Adoptive transfer studies revealed that the increased susceptibility of SLAMF7-/- mice to EAE is partly B cell dependent and reconstruction of the human CSF SLAMF7-interactome found B cells to be critical to cell-cell communication between SLAMF7-expressing cells. CONCLUSIONS: Our studies have identified novel roles for SLAMF7 in CNS immune regulation and B cell function, and illuminate underpinnings of the genetic association between SLAMF7 and MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Inmunidad Adaptativa , Animales , Autoinmunidad , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/metabolismo , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/patología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo
5.
J Immunol ; 202(1): 228-238, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30530590

RESUMEN

Current advances in combined antiretroviral therapy have rendered HIV infection a chronic, manageable disease; however, the problem of persistent immune activation still remains despite treatment. The immune cell receptor SLAMF7 has been shown to be upregulated in diseases characterized by chronic immune activation. In this study, we studied the function of the SLAMF7 receptor in immune cells of HIV patients and the impacts of SLAMF7 signaling on peripheral immune activation. We observed increased frequencies of SLAMF7+ PBMCs in HIV+ individuals in a clinical phenotype-dependent manner, with discordant and long-term nonprogressor patients showing elevated SLAMF7 levels, and elite controllers showing levels comparable to healthy controls. We also noted that SLAMF7 was sensitive to IFN-⍺ stimulation, a factor elevated during HIV infection. Further studies revealed SLAMF7 to be a potent inhibitor of the monocyte-derived proinflammatory chemokine CXCL10 (IP-10) and other CXCR3 ligands, except in a subset of HIV+ patients termed SLAMF7 silent (SF7S). Studies utilizing small molecule inhibitors revealed that the mechanism of CXCL10 inhibition is independent of known SLAMF7 binding partners. Furthermore, we determined that SLAMF7 activation on monocytes is able to decrease their susceptibility to HIV-1 infection in vitro via downregulation of CCR5 and upregulation of the CCL3L1 chemokine. Finally, we discovered that neutrophils do not express SLAMF7, are CXCL10+ at baseline, are able to secrete CXCL10 in response to IFN-⍺ and LPS, and are nonresponsive to SLAMF7 signaling. These findings implicate the SLAMF7 receptor as an important regulator of IFN-⍺-driven innate immune responses during HIV infection.


Asunto(s)
Infecciones por VIH/inmunología , VIH-1/fisiología , Interferón-alfa/metabolismo , Neutrófilos/inmunología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Células Cultivadas , Quimiocina CCL3/metabolismo , Quimiocina CXCL10/metabolismo , Progresión de la Enfermedad , Susceptibilidad a Enfermedades , Humanos , Fenotipo , Receptores CCR5/metabolismo , Transducción de Señal , Regulación hacia Arriba
6.
J Immunol ; 196(4): 1741-52, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26792800

RESUMEN

There is a compelling need for more effective vaccine adjuvants to augment induction of Ag-specific adaptive immune responses. Recent reports suggested the bacterial second messenger bis-(3'-5')-cyclic-dimeric-guanosine monophosphate (c-di-GMP) acts as an innate immune system modulator. We recently incorporated a Vibrio cholerae diguanylate cyclase into an adenovirus vaccine, fostering production of c-di-GMP as well as proinflammatory responses in mice. In this study, we recombined a more potent diguanylate cyclase gene, VCA0848, into a nonreplicating adenovirus serotype 5 (AdVCA0848) that produces elevated amounts of c-di-GMP when expressed in mammalian cells in vivo. This novel platform further improved induction of type I IFN-ß and activation of innate and adaptive immune cells early after administration into mice as compared with control vectors. Coadministration of the extracellular protein OVA and the AdVCA0848 adjuvant significantly improved OVA-specific T cell responses as detected by IFN-γ and IL-2 ELISPOT, while also improving OVA-specific humoral B cell adaptive responses. In addition, we found that coadministration of AdVCA0848 with another adenovirus serotype 5 vector expressing the HIV-1-derived Gag Ag or the Clostridium difficile-derived toxin B resulted in significant inhibitory effects on the induction of Gag and toxin B-specific adaptive immune responses. As a proof of principle, these data confirm that in vivo synthesis of c-di-GMP stimulates strong innate immune responses that correlate with enhanced adaptive immune responses to concomitantly administered extracellular Ag, which can be used as an adjuvant to heighten effective immune responses for protein-based vaccine platforms against microbial infections and cancers.


Asunto(s)
Inmunidad Adaptativa/inmunología , Adyuvantes Inmunológicos/farmacología , Antígenos/inmunología , GMP Cíclico/análogos & derivados , Inmunoterapia/métodos , Adenoviridae/inmunología , Animales , Western Blotting , GMP Cíclico/biosíntesis , GMP Cíclico/inmunología , Ensayo de Inmunoadsorción Enzimática , Ensayo de Immunospot Ligado a Enzimas , Citometría de Flujo , Vectores Genéticos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Transducción Genética
7.
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
8.
Int Immunol ; 26(5): 291-303, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24374770

RESUMEN

The signaling lymphocytic activation molecule (SLAM) receptor-associated adaptor Ewing's sarcoma-associated transcript-2 (EAT-2) is primarily expressed in innate immune cells including dendritic cells (DCs), macrophages and NK cells. A recent human HIV vaccine study confirmed that EAT-2 expression was associated with the enhanced immunogenicity induced by the MRKAd5/HIV vaccine. We previously harnessed the capability of EAT-2 to modulate signaling mediated by SLAM receptors and demonstrated that by incorporating EAT-2 expression into vaccines, one could enhance innate and adaptive immune responses in mice, even in the face of pre-existing immunity to the vaccine vectors. Herein, we investigated the innate immune responses of human cells exposed to EAT-2-over-expressing vaccines. Our results demonstrate that EAT-2 over-expression can significantly alter the kinetics of critical pro-inflammatory cytokine and chemokine responses elaborated by human PBMCs. In addition, enhanced DC maturation and increased monocyte phagocytosis were observed in EAT-2-transduced human cells. We also found that EAT-2 over-expression improved antigen presentation by human cells. Moreover, EAT-2 over-expression increased the anti-tumor activity of human NK cells against K562 tumor cell targets. Many of these responses were extinguished with use of an EAT-2 variant carrying a mutant SH2 domain (R31Q), suggesting a critical role for the interaction between EAT-2 and SLAM receptors in mediating these responses. In conclusion, these results provide evidence that EAT-2 interacts with key components of multiple arms of the human innate immune system, and that this role highlights the potential for targeting EAT-2 functions so as to improve a number of human immunotherapeutic approaches, including vaccine development.


Asunto(s)
Sistema Inmunológico/inmunología , Inmunidad Innata/inmunología , Inmunomodulación/inmunología , Factores de Transcripción/inmunología , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Células Cultivadas , Técnicas de Cocultivo , Citocinas/inmunología , Citocinas/metabolismo , Citotoxicidad Inmunológica/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/metabolismo , Inmunidad Innata/genética , Inmunomodulación/genética , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Células K562 , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Microscopía Fluorescente , Monocitos/inmunología , Monocitos/metabolismo , Mutación , Receptor 2 Gatillante de la Citotoxidad Natural/inmunología , Receptor 2 Gatillante de la Citotoxidad Natural/metabolismo , Fagocitosis/inmunología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Dominios Homologos src/genética , Dominios Homologos src/inmunología
9.
Int Immunol ; 26(12): 685-95, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25087231

RESUMEN

Endoplasmic reticulum aminopeptidase 1 (ERAP1) is a critical component of the adaptive immune system that has been shown to increase or decrease the presentation of specific peptides on MHC class I molecules. Here, we have demonstrated that ERAP1 functions are not only important during the presentation of antigen-derived peptides, but these functions can also completely change which antigen-derived peptides ultimately become selected as immunodominant T-cell epitopes. Our results suggest that ERAP1 may do this by destroying epitopes that would otherwise become immunodominant in the absence of adequate ERAP1 functionality. We further establish that ERAP1-mediated influences on T-cell functions are both qualitative and quantitative, by demonstrating that loss of ERAP1 function redirects CTL killing toward a different set of antigen-derived epitopes and increases the percent of antigen-specific memory T cells elicited by antigen exposure. As a result, our studies suggest that normal ERAP1 activity can act to suppress the numbers of T effector memory cells that respond to a given antigen. This unique finding may shed light on why certain ERAP1 single nucleotide polymorphisms are associated with several autoimmune diseases, for example, by significantly altering the robustness and quality of CD8+ T-cell memory responses to antigen-derived peptides.


Asunto(s)
Aminopeptidasas/metabolismo , Antígenos/inmunología , Citotoxicidad Inmunológica , Epítopos Inmunodominantes/inmunología , Memoria Inmunológica , Péptidos/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Inmunidad Adaptativa , Aminopeptidasas/genética , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Selección Clonal Mediada por Antígenos , Citocinas/biosíntesis , Citotoxicidad Inmunológica/genética , Epítopos Inmunodominantes/química , Epítopos Inmunodominantes/genética , Memoria Inmunológica/genética , Ratones , Ratones Noqueados , Antígenos de Histocompatibilidad Menor
10.
J Immunol ; 189(3): 1349-59, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22745373

RESUMEN

The mixed results from recent vaccine clinical trials targeting HIV-1 justify the need to enhance the potency of HIV-1 vaccine platforms in general. Use of first-generation recombinant adenovirus serotype 5 (rAd5) platforms failed to protect vaccinees from HIV-1 infection. One hypothesis is that the rAd5-based vaccine failed due to the presence of pre-existing Ad5 immunity in many vaccines. We recently confirmed that EAT-2-expressing rAd5 vectors uniquely activate the innate immune system and improve cellular immune responses against rAd5-expressed Ags, inclusive of HIV/Gag. In this study, we report that use of the rAd5-EAT-2 vaccine can also induce potent cellular immune responses to HIV-1 Ags despite the presence of Ad5-specific immunity. Compared to controls expressing a mutant SH2 domain form of EAT-2, Ad5 immune mice vaccinated with an rAd5-wild-type EAT-2 HIV/Gag-specific vaccine formulation significantly facilitated the induction of several arms of the innate immune system. These responses positively correlated with an improved ability of the vaccine to induce stronger effector memory T cell-biased, cellular immune responses to a coexpressed Ag despite pre-existing anti-Ad5 immunity. Moreover, inclusion of EAT-2 in the vaccine mixture improves the generation of polyfunctional cytolytic CD8(+) T cell responses as characterized by enhanced production of IFN-γ, TNF-α, cytotoxic degranulation, and increased in vivo cytolytic activity. These data suggest a new approach whereby inclusion of EAT-2 expression in stringent human vaccination applications can provide a more effective vaccine against HIV-1 specifically in Ad5 immune subjects.


Asunto(s)
Vacunas contra el SIDA/farmacología , Vacunas contra el Cáncer/farmacología , Inmunidad Innata , Memoria Inmunológica , Subgrupos de Linfocitos T/inmunología , Factores de Transcripción/fisiología , Vacunas contra el SIDA/genética , Vacunas contra el SIDA/inmunología , Inmunidad Adaptativa/genética , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Línea Celular , Células Cultivadas , Vectores Genéticos , Inmunidad Innata/genética , Memoria Inmunológica/genética , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/farmacología
11.
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
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.
Mol Ther Oncolytics ; 24: 180-193, 2022 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-35036523

RESUMEN

Targeted modulation of the immune system against tumors can achieve responses in otherwise refractory cancers, which has spurred efforts aimed at optimizing such strategies. To this end, we have previously investigated cancer immunotherapy approaches using recombinant adenovirus vectors, as well as via modulation of the self-ligand receptor SLAMF7. Here, we present a gene transfer-based immunotherapy approach using targeted expression of a SLAMF7-Fc fusion construct directly into tumors at high concentrations via a recombinant adenoviral vector (Ad-SF7-Fc). Using multiple murine cancer models, we show that Ad-SF7-Fc can induce tumor control via augmentation of innate immunity; specifically, induction of type I interferons and activation of dendritic cells (DCs) and macrophages. Analogously, we find that modulating SLAMF7 signaling via an adenoviral vector expressing its intracellular adaptor, EAT-2, is also capable of inducing tumor control. Finally, we employ a novel in vivo prediction approach and dataset integration with machine learning to dissect how Ad-SF7-Fc modulates cell-type-specific responses in the tumor microenvironment to achieve tumor control. Thus, our novel combinatorial cancer immunotherapy highlights the benefit of multimodal immune modulation and lays a framework for combination with complementary approaches capable of inducing adaptive immune responses.

14.
J Immunol ; 181(3): 2134-44, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18641352

RESUMEN

Adenovirus (Ad) vectors are promising candidates for both gene transfer and vaccine applications. In this study, we investigated the role of TLR2 in innate and adaptive immune responses to Ad and/or the transgene it expresses following systemic injection. We found that Ad directly activates ERK1/2 in vivo, but that initiation of ERK1/2 activation is primarily a MyD88/TLR2-independent, but Kupffer cell-dependent, event. The complexity of Ad-induced innate immune responses was confirmed when we also found that both TLR2 and MyD88 functions are required for the sustained activation of ERK1/2. Although we found that the initial activation of NF-kappaB by Ads is dependent upon MyD88, but independent of TLR2 in (non-Kupffer cells) the liver, TLR2 significantly influenced the Ad-induced late phase NF-kappaB activation. These very rapid responses were positively correlated with subsequent innate immune responses to the Ad vector, as our results confirmed that the induction of several cytokines and chemokines, and the expression of innate immune response genes following Ad injection were TLR2 dependent in vivo. The requirement of TLR2 in Ad-induced innate responses also correlated with significantly altered adaptive immune responses. For example, our results demonstrate that the generation of Ad-neutralizing Abs, and anti-transgene-specific Abs elicited subsequent to Ad vector treatments, are both dependent upon TLR2 functionality. Finally, we found that several Ad-induced innate immune responses are dependent on both TLR2 and TLR9. Therefore, this study confirms that several (but not all) Ad-induced innate and adaptive immune responses are TLR dependent.


Asunto(s)
Adaptación Fisiológica/inmunología , Adenoviridae/inmunología , Citocinas/inmunología , Inmunidad Innata/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 9/metabolismo , Adenoviridae/genética , Animales , Anticuerpos/inmunología , Células Cultivadas , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Macrófagos del Hígado/inmunología , Macrófagos del Hígado/metabolismo , Hígado/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética
15.
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
16.
Clin Vaccine Immunol ; 24(6)2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28381403

RESUMEN

Recombinant adenovirus serotype 5 (Ad5) vectors are promising vaccine candidates due to their intrinsic immunogenicity and potent transgene expression; however, widespread preexisting Ad5 immunity has been considered a developmental impediment to the use of traditional, or conventional, E1 and E3 gene-deleted Ad5 (Ad5[E1-]) vaccines. Even in the presence of anti-Ad5 immunity, recent murine and human studies have confirmed E2b gene-deleted Ad5 (Ad5[E1-,E2b-]) vaccines to be highly efficacious inducers of transgene-specific memory responses and significantly less toxic options than Ad5[E1-] vaccines. While these findings have been substantially confirmed, the molecular mechanisms underlying the different reactions to these vaccine platforms are unknown. Using cultures of human peripheral blood mononuclear cells (hPBMCs) derived from multiple human donors, we found that Ad5[E1-,E2b-] vaccines trigger higher levels of hPBMC proinflammatory cytokine secretion than Ad5[E1-] vaccines. Interestingly, these responses were generated regardless of the donors' preexisting anti-Ad5 humoral and cell-mediated immune response status. In vitro hPBMC infection with the Ad5[E1-,E2b-] vaccine also provoked greater Th1-dominant gene responses yet smaller amounts of Ad-derived gene expression than Ad5[E1-] vaccines. These results suggest that Ad5[E1-,E2b-] vaccines, in contrast to Ad5[E1-] vaccines, do not promote activities that suppress innate immune signaling, thereby allowing for improved vaccine efficacy and a superior safety profile independently of previous Ad5 immunity.


Asunto(s)
Vacunas contra el Adenovirus/inmunología , Adenovirus Humanos/inmunología , Portadores de Fármacos , Expresión Génica , Vectores Genéticos , Vacunas contra el Adenovirus/genética , Adenovirus Humanos/genética , Células Cultivadas , Citocinas/metabolismo , Humanos , Leucocitos Mononucleares/inmunología , Serogrupo , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
17.
Vaccine ; 34(27): 3109-3118, 2016 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-27151882

RESUMEN

The CD2-like receptor activating cytotoxic cell (CRACC) receptor is a member of the SLAM family of receptors that are found on several types of immune cells. We previously demonstrated that increasing the abundance of the adaptor protein EAT-2 during vaccination enhanced innate and adaptive immune responses to vaccine antigens. Engagement of the CRACC receptor in the presence of the EAT-2 adaptor generally results in immune cell activation, while activating CRACC signaling in cells that lack EAT-2 adaptor inhibits their effector and regulatory functions. As EAT-2 is the only SAP adaptor that interacts with the CRACC receptor, we hypothesized that technologies that specifically modulate CRACC signaling during vaccination may also improve antigen specific adaptive immune responses. To test this hypothesis, we constructed a CRACC-targeting Fc fusion protein and included it in vaccination attempts. Indeed, mice co-vaccinated with the CRACC-Fc fusion protein and an adenovirus vaccine expressing the HIV-Gag protein had improved Gag-specific T cell responses, as compared to control mice. These responses are characterized by increased numbers of Gag-specific tetramer+ CD8+ T cells and increases in production of IFNγ, TNFα, and IL2, by Gag-specific CD8+ T cells. Moreover, our results revealed that use of the CRACC-Fc fusion protein enhances vaccine-elicited innate immune responses, as characterized by increased dendritic cells (DCs) maturation and IFNγ production from NK cells. This study highlights the importance of CRACC signaling during the induction of an immune response generally, and during vaccinations specifically, and also lends insight into the mechanisms underlying our prior results noting EAT-2-dependent improvements in vaccine efficacy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Células Asesinas Naturales/inmunología , Transporte de Proteínas , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/inmunología , Vacunas contra el SIDA/inmunología , Animales , Citocinas/inmunología , Inmunidad Innata , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células RAW 264.7 , Proteínas Recombinantes de Fusión/inmunología , Factores de Transcripción/inmunología
18.
J Innate Immun ; 7(3): 275-89, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25591727

RESUMEN

Endoplasmic reticulum aminopeptidase 1 (ERAP1) gene polymorphisms have been linked to several autoimmune diseases; however, the molecular mechanisms underlying these associations are not well understood. Recently, we demonstrated that ERAP1 regulates key aspects of the innate immune response. Previous studies show ERAP1 to be endoplasmic reticulum-localized and secreted during inflammation. Herein, we investigate the possible roles that ERAP1 polymorphic variants may have in modulating the innate immune responses of human peripheral blood mononuclear cells (hPBMCs) using two experimental methods: extracellular exposure of hPBMCs to ERAP1 variants and adenovirus (Ad)-based ERAP1 expression. We found that exposure of hPBMCs to ERAP1 variant proteins as well as ERAP1 overexpression by Ad5 vectors increased inflammatory cytokine and chemokine production, and enhanced immune cell activation. Investigating the molecular mechanisms behind these responses revealed that ERAP1 is able to activate innate immunity via multiple pathways, including the NLRP3 (NOD-like receptor, pyrin domain-containing 3) inflammasome. Importantly, these responses varied if autoimmune disease-associated variants of ERAP1 were examined in the assay systems. Unexpectedly, blocking ERAP1 cellular internalization augmented IL-1ß production. To our knowledge, this is the first report identifying ERAP1 as being involved in modulating innate responses of human immune cells, a finding that may explain why ERAP1 has been genetically associated with several autoimmune diseases.


Asunto(s)
Aminopeptidasas/inmunología , Enfermedades Autoinmunes/inmunología , Inmunidad Innata , Leucocitos Mononucleares/inmunología , Adenoviridae , Aminopeptidasas/genética , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Línea Celular , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Leucocitos Mononucleares/patología , Ratones , Antígenos de Histocompatibilidad Menor , Proteína con Dominio Pirina 3 de la Familia NLR , Transducción Genética
19.
Clin Vaccine Immunol ; 21(11): 1550-9, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25230938

RESUMEN

The bacterial second messenger cyclic di-GMP (c-di-GMP) stimulates inflammation by initiating innate immune cell recruitment and triggering the release of proinflammatory cytokines and chemokines. These properties make c-di-GMP a promising candidate for use as a vaccine adjuvant, and numerous studies have demonstrated that administration of purified c-di-GMP with different antigens increases protection against infection in animal models. Here, we have developed a novel approach to produce c-di-GMP inside host cells as an adjuvant to exploit a host-pathogen interaction and initiate an innate immune response. We have demonstrated that c-di-GMP can be synthesized in vivo by transducing a diguanylate cyclase (DGC) gene into mammalian cells using an adenovirus serotype 5 (Ad5) vector. Expression of DGC led to the production of c-di-GMP in vitro and in vivo, and this was able to alter proinflammatory gene expression in murine tissues and increase the secretion of numerous cytokines and chemokines when administered to animals. Furthermore, coexpression of DGC modestly increased T-cell responses to a Clostridium difficile antigen expressed from an adenovirus vaccine, although no significant differences in antibody titers were observed. This adenovirus c-di-GMP delivery system offers a novel method to administer c-di-GMP as an adjuvant to stimulate innate immunity during vaccination.


Asunto(s)
Adenoviridae/enzimología , Adyuvantes Inmunológicos/metabolismo , GMP Cíclico/análogos & derivados , Proteínas de Escherichia coli/metabolismo , Inmunidad Innata/efectos de los fármacos , Liasas de Fósforo-Oxígeno/metabolismo , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/genética , Vacunas Bacterianas/inmunología , Clostridioides difficile/genética , Clostridioides difficile/inmunología , GMP Cíclico/metabolismo , Proteínas de Escherichia coli/genética , Masculino , Ratones Endogámicos BALB C , Liasas de Fósforo-Oxígeno/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción Genética
20.
Hum Immunol ; 74(8): 916-26, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23639554

RESUMEN

The safe and effective activation of the innate and adaptive immune systems are crucial in the implementation of immunotherapeutic modalities for the prevention and treatment of human diseases. Eimeria antigen (EA) and its recombinantly expressed analog (rEA) are extremely effective activators of innate immunity in mice. The effects of rEA in the mouse are primarily mediated through the TLR11/12 MyD88 signaling system. Human cells lack functional TLR11 and TLR12, suggesting that rEA would not be effective in providing beneficial immune activation in humans. In the current report we provide definitive evidence that the treatment of human peripheral blood mononuclear cell (PBMC) cultures with rEA significantly up regulates CD69, CD107, NKG2D levels on NK cells. Furthermore, rEA stimulates human NK cell effector functions including increasing intracellular levels of IFNγ and Granzyme B. These responses are positively correlated with an improved capacity of rEA stimulated human PBMCs to kill NK cell-sensitive human K562 tumor cells. Importantly, rEA-triggered innate immune responses was not associated with increased pro-inflammatory cytokines and chemokines production. These data confirm a previously unidentified role for rEA in human immune cell activation, and suggests the utilization of rEA in immunotherapies against a variety of infectious diseases and cancers.


Asunto(s)
Antígenos de Protozoos/inmunología , Eimeria/inmunología , Inmunidad Innata/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Proteínas Recombinantes/inmunología , Antígenos CD/metabolismo , Antígenos de Protozoos/farmacología , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Regulación de la Expresión Génica , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunofenotipificación , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Activación de Linfocitos/efectos de los fármacos , Subfamília C de Receptores Similares a Lectina de Células NK/metabolismo , Fenotipo , Prohibitinas , Proteínas Recombinantes/farmacología , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA