Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 19(1): 241, 2022 Oct 03.
Article in English | MEDLINE | ID: mdl-36199066

ABSTRACT

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.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Adaptive Immunity , Animals , Autoimmunity , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Humans , Ligands , Mice , Mice, Inbred C57BL , Multiple Sclerosis/pathology , Signaling Lymphocytic Activation Molecule Family/genetics , Signaling Lymphocytic Activation Molecule Family/metabolism
2.
BMC Immunol ; 23(1): 9, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35246034

ABSTRACT

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.


Subject(s)
Aminopeptidases , Endoplasmic Reticulum Stress , Inflammasomes , Aminopeptidases/genetics , Aminopeptidases/metabolism , Animals , Immunity, Innate/genetics , Mice , Mice, Knockout , Minor Histocompatibility Antigens/genetics
3.
Mol Ther Oncolytics ; 24: 180-193, 2022 Mar 17.
Article in English | MEDLINE | ID: mdl-35036523

ABSTRACT

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.

4.
J Immunol ; 207(12): 2952-2965, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34810226

ABSTRACT

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.


Subject(s)
Autoimmune Diseases , B-Lymphocytes , Multiple Sclerosis , Aminopeptidases/genetics , Aminopeptidases/metabolism , Animals , Autoimmunity/genetics , Central Nervous System , Mice , Minor Histocompatibility Antigens/genetics , Multiple Sclerosis/genetics , Polymorphism, Single Nucleotide
5.
J Immunol ; 206(1): 193-205, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33288545

ABSTRACT

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.


Subject(s)
Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Melanoma/metabolism , Signaling Lymphocytic Activation Molecule Family/metabolism , Skin Neoplasms/metabolism , T-Lymphocytes/immunology , Animals , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/mortality , Cells, Cultured , Cellular Reprogramming , Female , Gene Expression Regulation, Neoplastic , Humans , Immune Tolerance , Kidney Neoplasms/immunology , Kidney Neoplasms/mortality , Male , Melanoma/immunology , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental , Signal Transduction , Signaling Lymphocytic Activation Molecule Family/genetics , Skin Neoplasms/immunology , Survival Analysis , Tumor Microenvironment
6.
J Immunol ; 202(1): 228-238, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30530590

ABSTRACT

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.


Subject(s)
HIV Infections/immunology , HIV-1/physiology , Interferon-alpha/metabolism , Neutrophils/immunology , Signaling Lymphocytic Activation Molecule Family/metabolism , Cells, Cultured , Chemokine CCL3/metabolism , Chemokine CXCL10/metabolism , Disease Progression , Disease Susceptibility , Humans , Phenotype , Receptors, CCR5/metabolism , Signal Transduction , Up-Regulation
7.
Clin Vaccine Immunol ; 24(6)2017 Jun.
Article in English | MEDLINE | ID: mdl-28381403

ABSTRACT

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.


Subject(s)
Adenovirus Vaccines/immunology , Adenoviruses, Human/immunology , Drug Carriers , Gene Expression , Genetic Vectors , Adenovirus Vaccines/genetics , Adenoviruses, Human/genetics , Cells, Cultured , Cytokines/metabolism , Humans , Leukocytes, Mononuclear/immunology , Serogroup , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
8.
Vaccine ; 34(27): 3109-3118, 2016 06 08.
Article in English | MEDLINE | ID: mdl-27151882

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Killer Cells, Natural/immunology , Protein Transport , Signaling Lymphocytic Activation Molecule Family/immunology , AIDS Vaccines/immunology , Animals , Cytokines/immunology , Immunity, Innate , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RAW 264.7 Cells , Recombinant Fusion Proteins/immunology , Transcription Factors/immunology
9.
J Immunol ; 196(4): 1741-52, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26792800

ABSTRACT

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.


Subject(s)
Adaptive Immunity/immunology , Adjuvants, Immunologic/pharmacology , Antigens/immunology , Cyclic GMP/analogs & derivatives , Immunotherapy/methods , Adenoviridae/immunology , Animals , Blotting, Western , Cyclic GMP/biosynthesis , Cyclic GMP/immunology , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Flow Cytometry , Genetic Vectors , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Transduction, Genetic
10.
Cancer Immunol Immunother ; 64(4): 479-91, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25655760

ABSTRACT

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.


Subject(s)
Adenoviridae/genetics , Antigens, Neoplasm/immunology , Cancer Vaccines/pharmacology , Carcinoembryonic Antigen/immunology , Colonic Neoplasms/immunology , Eimeria/immunology , T-Lymphocytes, Cytotoxic/immunology , Adenoviridae/immunology , Adjuvants, Immunologic , Animals , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Carcinoembryonic Antigen/genetics , Colonic Neoplasms/prevention & control , Eimeria/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Genetic Vectors/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Prohibitins , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Vaccination
11.
J Innate Immun ; 7(3): 275-89, 2015.
Article in English | MEDLINE | ID: mdl-25591727

ABSTRACT

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.


Subject(s)
Aminopeptidases/immunology , Autoimmune Diseases/immunology , Immunity, Innate , Leukocytes, Mononuclear/immunology , Adenoviridae , Aminopeptidases/genetics , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Carrier Proteins/genetics , Carrier Proteins/immunology , Cell Line , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Leukocytes, Mononuclear/pathology , Mice , Minor Histocompatibility Antigens , NLR Family, Pyrin Domain-Containing 3 Protein , Transduction, Genetic
12.
Clin Vaccine Immunol ; 21(11): 1550-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25230938

ABSTRACT

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.


Subject(s)
Adenoviridae/enzymology , Adjuvants, Immunologic/metabolism , Cyclic GMP/analogs & derivatives , Escherichia coli Proteins/metabolism , Immunity, Innate/drug effects , Phosphorus-Oxygen Lyases/metabolism , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Clostridioides difficile/genetics , Clostridioides difficile/immunology , Cyclic GMP/metabolism , Escherichia coli Proteins/genetics , Male , Mice, Inbred BALB C , Phosphorus-Oxygen Lyases/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transduction, Genetic
13.
Int Immunol ; 26(12): 685-95, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25087231

ABSTRACT

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.


Subject(s)
Aminopeptidases/metabolism , Antigens/immunology , Cytotoxicity, Immunologic , Immunodominant Epitopes/immunology , Immunologic Memory , Peptides/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adaptive Immunity , Aminopeptidases/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Clonal Selection, Antigen-Mediated , Cytokines/biosynthesis , Cytotoxicity, Immunologic/genetics , Immunodominant Epitopes/chemistry , Immunodominant Epitopes/genetics , Immunologic Memory/genetics , Mice , Mice, Knockout , Minor Histocompatibility Antigens
14.
Int Immunol ; 26(5): 291-303, 2014 May.
Article in English | MEDLINE | ID: mdl-24374770

ABSTRACT

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.


Subject(s)
Immune System/immunology , Immunity, Innate/immunology , Immunomodulation/immunology , Transcription Factors/immunology , Antigen Presentation/genetics , Antigen Presentation/immunology , Cells, Cultured , Coculture Techniques , Cytokines/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Immune System/cytology , Immune System/metabolism , Immunity, Innate/genetics , Immunomodulation/genetics , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , K562 Cells , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Microscopy, Fluorescence , Monocytes/immunology , Monocytes/metabolism , Mutation , Natural Cytotoxicity Triggering Receptor 2/immunology , Natural Cytotoxicity Triggering Receptor 2/metabolism , Phagocytosis/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , src Homology Domains/genetics , src Homology Domains/immunology
15.
Hum Immunol ; 74(8): 916-26, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23639554

ABSTRACT

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.


Subject(s)
Antigens, Protozoan/immunology , Eimeria/immunology , Immunity, Innate/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Recombinant Proteins/immunology , Antigens, CD/metabolism , Antigens, Protozoan/pharmacology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation , Humans , Immunity, Innate/drug effects , Immunophenotyping , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/drug effects , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Phenotype , Prohibitins , Recombinant Proteins/pharmacology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists
16.
J Immunol ; 189(3): 1349-59, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22745373

ABSTRACT

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.


Subject(s)
AIDS Vaccines/pharmacology , Cancer Vaccines/pharmacology , Immunity, Innate , Immunologic Memory , T-Lymphocyte Subsets/immunology , Transcription Factors/physiology , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Adaptive Immunity/genetics , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line , Cells, Cultured , Genetic Vectors , Immunity, Innate/genetics , Immunologic Memory/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/pharmacology
17.
Vaccine ; 30(8): 1492-501, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22200503

ABSTRACT

Clostridium difficile associated diarrhea (CDAD) is a critical public health problem worldwide with over 300,000 cases every year in the United States alone. Clearly, a potent vaccine preventing the morbidity and mortality caused by this detrimental pathogen is urgently required. However, vaccine efforts to combat C. difficile infections have been limited both in scope as well as to efficacy, as such there is not a vaccine approved for use against C. difficile to date. In this study, we have used a highly potent Adenovirus (Ad) based platform to create a vaccine against C. difficile. The Ad-based vaccine was able to generate rapid and robust humoral as well as cellular (T-cell) immune responses in mice that correlated with provision of 100% protection from lethal challenge with C. difficile toxin A. Most relevant to the clinical utility of this vaccine formulation was our result that toxin A specific IgGs were readily detected in plasma of Ad immunized mice as early as 3 days post vaccination. In addition, we found that several major immuno-dominant T cell epitopes were identified in toxin A, suggesting that the role of the cellular arm in protection from C. difficile infections may be more significant than previously appreciated. Therefore, our studies confirm that an Adenovirus based-C. difficile vaccine could be a promising candidate for prophylactic vaccination both for use in high risk patients and in high-risk environments.


Subject(s)
Adenoviridae/genetics , Antitoxins/blood , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/immunology , Bacterial Vaccines/immunology , Drug Carriers/administration & dosage , Enterotoxins/antagonists & inhibitors , Enterotoxins/immunology , Vaccination/methods , Adenoviridae/immunology , Animals , Bacterial Toxins/genetics , Bacterial Vaccines/administration & dosage , Clostridium Infections/prevention & control , Disease Models, Animal , Enterotoxins/genetics , Epitopes, T-Lymphocyte/immunology , Immunity, Cellular , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Poisoning/prevention & control , Survival Analysis
18.
PLoS One ; 6(8): e24147, 2011.
Article in English | MEDLINE | ID: mdl-21912619

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Immunity, Cellular/immunology , Immunity, Innate/immunology , Immunologic Factors/immunology , Protozoan Proteins/immunology , Vaccines/genetics , Vaccines/immunology , Adaptor Proteins, Signal Transducing/genetics , Adenoviridae/genetics , Animals , Antibody Specificity , Antigen Presentation/immunology , B-Lymphocytes/immunology , Immune Tolerance/immunology , Immunologic Factors/genetics , Male , Mice , Mice, Inbred BALB C , Protozoan Proteins/genetics , T-Lymphocytes/immunology , Toll-Like Receptors/agonists
19.
PLoS One ; 6(7): e22064, 2011.
Article in English | MEDLINE | ID: mdl-21760953

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Dendritic Cells/immunology , Toll-Like Receptors/agonists , Animals , Antigens, Protozoan/immunology , B-Lymphocytes/immunology , Chemokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/parasitology , Eimeria tenella/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Inflammation Mediators , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Models, Immunological , Myeloid Differentiation Factor 88/metabolism , Natural Killer T-Cells/immunology , Phosphorylation , Prohibitins , Toll-Like Receptors/immunology
20.
Hum Gene Ther ; 22(9): 1083-94, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21388344

ABSTRACT

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.


Subject(s)
Adaptive Immunity , Adenoviridae/genetics , Adenoviridae/immunology , CD55 Antigens/genetics , Genetic Vectors/immunology , Transgenes , Animals , Epitopes/immunology , Gene Transfer Techniques , Genetic Vectors/administration & dosage , HEK293 Cells , Humans , Immunity, Cellular , Immunity, Humoral , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...