Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Nat Microbiol ; 4(5): 800-812, 2019 05.
Article in English | MEDLINE | ID: mdl-30858572

ABSTRACT

Host NOD-like receptor family pyrin domain-containing 6 (NLRP6) regulates innate immune responses and gastrointestinal homeostasis. Its protective role in intestinal colitis and tumorigenesis is dependent on the host microbiome. Host innate immunity and microbial diversity also play a role in the severity of allogeneic immune-mediated gastrointestinal graft-versus-host disease (GVHD), the principal toxicity after allogeneic haematopoietic cell transplantation. Here, we examined the role of host NLRP6 in multiple murine models of allogeneic bone marrow transplantation. In contrast to its role in intestinal colitis, host NLRP6 aggravated gastrointestinal GVHD. The impact of host NLRP6 deficiency in mitigating GVHD was observed regardless of co-housing, antibiotic treatment or colonizing littermate germ-free wild-type and NLRP6-deficient hosts with faecal microbial transplantation from specific pathogen-free wild-type and Nlrp6-/- animals. Chimaera studies were performed to assess the role of NLRP6 expression on host haematopoietic and non-haematopoietic cells. The allogeneic [B6Ly5.2 → Nlrp6-/-] animals demonstrated significantly improved survival compared to the allogeneic [B6Ly5.2 → B6] animals, but did not alter the therapeutic graft-versus-tumour effects after haematopoietic cell transplantation. Our results unveil an unexpected, pathogenic role for host NLRP6 in gastrointestinal GVHD that is independent of variations in the intestinal microbiome and in contrast to its well-appreciated microbiome-dependent protective role in intestinal colitis and tumorigenesis.


Subject(s)
Bacteria/isolation & purification , Gastrointestinal Microbiome , Graft vs Host Disease/microbiology , Receptors, Cell Surface/immunology , Animals , Bacteria/classification , Bacteria/genetics , Bone Marrow Transplantation , Feces/microbiology , Female , Graft vs Host Disease/immunology , Humans , Intestines/immunology , Intestines/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cell Surface/genetics , Specific Pathogen-Free Organisms , Transplantation, Homologous
2.
Cell ; 175(6): 1651-1664.e14, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30392956

ABSTRACT

The activator and composition of the NLRP6 inflammasome remain poorly understood. We find that lipoteichoic acid (LTA), a molecule produced by Gram-positive bacteria, binds and activates NLRP6. In response to cytosolic LTA or infection with Listeria monocytogenes, NLRP6 recruited caspase-11 and caspase-1 via the adaptor ASC. NLRP6 activation by LTA induced processing of caspase-11, which promoted caspase-1 activation and interleukin-1ß (IL-1ß)/IL-18 maturation in macrophages. Nlrp6-/- and Casp11-/- mice were less susceptible to L. monocytogenes infection, which was associated with reduced pathogen loads and impaired IL-18 production. Administration of IL-18 to Nlrp6-/- or Casp11-/- mice restored the susceptibility of mutant mice to L. monocytogenes infection. These results reveal a previously unrecognized innate immunity pathway triggered by cytosolic LTA that is sensed by NLRP6 and exacerbates systemic Gram-positive pathogen infection via the production of IL-18.


Subject(s)
Immunity, Innate , Inflammasomes/immunology , Lipopolysaccharides/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Receptors, Cell Surface/immunology , Teichoic Acids/immunology , Animals , Caspase 1/genetics , Caspase 1/immunology , Caspases/genetics , Caspases/immunology , Caspases, Initiator , Inflammasomes/genetics , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Listeriosis/genetics , Listeriosis/pathology , Mice , Mice, Knockout , Receptors, Cell Surface/genetics
3.
Diabetes ; 67(4): 769-781, 2018 04.
Article in English | MEDLINE | ID: mdl-29362226

ABSTRACT

Tight junctions (TJs) involve close apposition of transmembrane proteins between cells. Although TJ proteins have been studied in detail, the role of lipids is largely unknown. We addressed the role of very long-chain (VLC ≥26) ceramides in TJs using diabetes-induced loss of the blood-retinal barrier as a model. VLC fatty acids that incorporate into VLC ceramides are produced by elongase elongation of very long-chain fatty acids protein 4 (ELOVL4). ELOVL4 is significantly reduced in the diabetic retina. Overexpression of ELOVL4 significantly decreased basal permeability, inhibited vascular endothelial growth factor (VEGF)- and interleukin-1ß-induced permeability, and prevented VEGF-induced decrease in occludin expression and border staining of TJ proteins ZO-1 and claudin-5. Intravitreal delivery of AAV2-hELOVL4 reduced diabetes-induced increase in vascular permeability. Ultrastructure and lipidomic analysis revealed that ω-linked acyl-VLC ceramides colocalize with TJ complexes. Overall, normalization of retinal ELOVL4 expression could prevent blood-retinal barrier dysregulation in diabetic retinopathy through an increase in VLC ceramides and stabilization of TJs.


Subject(s)
Blood-Retinal Barrier/metabolism , Capillary Permeability/genetics , Ceramides/metabolism , Endothelial Cells/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Retinal Vessels/metabolism , Tight Junctions/metabolism , Animals , Cattle , Claudin-5/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/etiology , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Endothelial Cells/ultrastructure , Humans , Interleukin-1beta/metabolism , Mice , Occludin/metabolism , Retina/metabolism , Retinal Vessels/ultrastructure , Tight Junctions/ultrastructure , Vascular Endothelial Growth Factor A/metabolism , Zonula Occludens-1 Protein/metabolism
5.
Cell Rep ; 19(4): 733-745, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28445725

ABSTRACT

Dysfunction in host immune responses and pathologic alterations in the gut microbiota, referred to as dysbiosis, can both contribute to the development of inflammatory bowel disease (IBD). However, it remains unclear how specific changes in host immunity or the microbiota cause disease. We previously demonstrated that the loss of the innate immune receptor NLRP6 in mice resulted in impaired production of interleukin-18 (IL-18) and increased susceptibility to epithelial-induced injury. Here, we show that NLRP6 is important for suppressing the development of spontaneous colitis in the Il10-/- mice model of IBD and that NLRP6 deficiency results in the enrichment of Akkermansia muciniphila. A. muciniphila was sufficient for promoting intestinal inflammation in both specific-pathogen-free and germ-free Il10-/- mice. Our results demonstrate that A. muciniphila can act as a pathobiont to promote colitis in a genetically susceptible host and that NLRP6 is a key regulator of its abundance.


Subject(s)
Colitis/etiology , Interleukin-10/genetics , Receptors, Cell Surface/metabolism , Verrucomicrobia/physiology , Animals , Bacteroides/genetics , Bacteroides/physiology , Colitis/microbiology , Colitis/pathology , Colon/microbiology , Colon/pathology , DNA, Bacterial/chemistry , DNA, Bacterial/isolation & purification , DNA, Bacterial/metabolism , Disease Susceptibility , Female , Hyperplasia/etiology , Hyperplasia/pathology , Inflammation/etiology , Inflammation/pathology , Interleukin-10/deficiency , Interleukin-18/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Sequence Analysis, DNA , Verrucomicrobia/genetics
6.
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
7.
J Immunol ; 196(12): 5121-9, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27183588

ABSTRACT

Chronic intestinal inflammation is a major risk factor for the development of colorectal cancer. Nod1, a member of the Nod-like receptor (NLR) family of pattern recognition receptors, is a bacterial sensor that has been previously demonstrated to reduce susceptibility of mice to chemically induced colitis and subsequent tumorigenesis, but the mechanism by which it mediates its protection has not been elucidated. In this study, we show that Nod1 expression in the hematopoietic cell compartment is critical for limiting inflammation-induced intestinal tumorigenesis. Specifically, Nod1-deficient T cells exhibit impaired IFN-γ production during dextran sulfate sodium (DSS)-induced acute inflammation in vivo, and administration of the Nod1 ligand KF1B enhances IFN-γ responses by anti-CD3-activated T cells in vitro. Absence of IFN-γ signaling results in increased inflammation-associated tumors in mice, and adoptive transfer of Nod1(-/-) or IFNγ(-/-) T cells into T cell-deficient mice results in increased tumorigenesis as compared with T cell-deficient mice that were adoptively transferred with wild-type T cells. Collectively, these results suggest a previously unappreciated role for the innate immune receptor Nod1 in suppressing colitis-associated tumorigenesis through a T cell-mediated mechanism.


Subject(s)
Carcinogenesis , Colitis/complications , Gene Expression Regulation , Interferon-gamma/biosynthesis , Nod1 Signaling Adaptor Protein/metabolism , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Colitis/chemically induced , Colitis/immunology , Dextran Sulfate , Disease Models, Animal , Immunity, Innate , Inflammation/chemically induced , Inflammation/immunology , Interferon-gamma/immunology , Intestines/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod1 Signaling Adaptor Protein/deficiency , Nod1 Signaling Adaptor Protein/genetics
8.
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
9.
J Immunol ; 195(3): 1112-20, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26116500

ABSTRACT

A small pool of NK1.1(+) CD8(+) T cells is harbored among the conventional CD8(+) T cell compartment. Conclusions drawn from the analysis of immune responses mediated by cytotoxic CD8(+) T cells are often based on the total population, which includes these contaminating NK1.1(+) CD8(+) T cells. An unresolved question is whether NK1.1(+) CD8(+) cells are conventional T cells that acquire NK1.1 expression upon activation or delineation into memory phenotype or whether they are a distinct cell population that induces immune responses in a different manner than conventional T cells. To address this question, we used the Listeria monocytogenes model of infection and followed CD8(+) NK1.1(+) T cells and NK1.1(-) CD8(+) T cells during each phase of the immune response: innate, effector, and memory. Our central finding is that CD8(+) NK1.1(+) cells and conventional NK1.1(-) CD8(+) T cells both contribute to the adaptive immune response to Listeria, but only CD8(+) NK1.1(+) cells were equipped with the ability to provide a rapid innate immune response, as demonstrated by early and Ag-independent IFN-γ production, granzyme B expression, and degranulation. More importantly, purified conventional CD8(+) T cells alone, in the absence of any contaminating CD8(+) NK1.1(+) cells, were not sufficient to provide early protection to lethally infected mice. These results highlight the role of CD8(+) NK1.1(+) T cells in mounting early innate responses that are important for host defense and support the therapeutic potential of this subset to improve the effectiveness of protective immunity.


Subject(s)
Immunity, Innate/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Natural Killer T-Cells/immunology , T-Lymphocytes, Cytotoxic/immunology , Adoptive Transfer , Animals , Antigens, Ly/genetics , Granzymes/biosynthesis , Interferon-gamma/biosynthesis , Listeriosis/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily B/genetics , Natural Killer T-Cells/cytology , Natural Killer T-Cells/transplantation , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/transplantation
10.
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
11.
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
12.
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
13.
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
14.
PLoS One ; 8(11): e79745, 2013.
Article in English | MEDLINE | ID: mdl-24244554

ABSTRACT

Although we stimulate enterocytic proliferation to ameliorate short gut syndrome or mucosal atrophy, less effort has been directed at enterocytic differentiation. Schlafen 3 (Slfn3) is a poorly understood protein induced during IEC-6 enterocytic differentiation. We hypothesized that exogenous manipulation of Slfn3 would regulate enterocytic differentiation in vivo. Adenoviral vector coding for Slfn3 cDNA (Ad-GFP-Slfn3) or silencing RNA for Slfn3 (siSlfn3) was introduced intraluminally into rat intestine. We assessed Slfn3, villin, sucrase-isomaltase (SI), Dpp4, and Glut2 by qRT-PCR, Western blot, and immunohistochemistry. We also studied Slfn3 and these differentiation markers in atrophic defunctionalized jejunal mucosa and the crypt-villus axis of normal jejunum. Ad-GFP-Slfn3 but not Ad-GFP increased Slfn3, villin and Dpp4 expression in human Caco-2 intestinal epithelial cells. Injecting Ad-GFP-Slfn3 into rat jejunum in vivo increased mucosal Slfn3 mRNA three days later vs. intraluminal Ad-GFP. This Slfn3 overexpression was associated with increases in all four differentiation markers. Injecting siSlfn3 into rat jejunum in vivo substantially reduced Slfn3 and all four intestinal mucosal differentiation markers three days later, as well as Dpp4 specific activity. Endogenous Slfn3 was reduced in atrophic mucosa from a blind-end Roux-en-Y anastomosis in parallel with differentiation marker expression together with AKT and p38 signaling. Slfn3 was more highly expressed in the villi than the crypts, paralleling Glut2, SI and Dpp4. Slfn3 is a key intracellular regulator of rat enterocytic differentiation. Understanding how Slfn3 works may identify targets to promote enterocytic differentiation and maintain mucosal function in vivo, facilitating enteral nutrition and improving survival in patients with mucosal atrophy or short gut syndrome.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/genetics , Intestinal Mucosa/metabolism , Proteins/genetics , RNA, Small Interfering/genetics , Animals , Apoptosis , Atrophy , Biomarkers , Caco-2 Cells , Cell Differentiation/genetics , Cell Proliferation , Gene Expression , Gene Transfer Techniques , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Jejunum/metabolism , Male , Microfilament Proteins/genetics , RNA, Messenger/genetics , Rats , Transduction, Genetic , Transfection , Transgenes
15.
Proc Natl Acad Sci U S A ; 110(49): 19890-5, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24248368

ABSTRACT

Intracellular aminopeptidases endoplasmic reticulum aminopeptidases 1 and 2 (ERAP1 and ERAP2), and as well as insulin-regulated aminopeptidase (IRAP) process antigenic epitope precursors for loading onto MHC class I molecules and regulate the adaptive immune response. Their activity greatly affects the antigenic peptide repertoire presented to cytotoxic T lymphocytes and as a result can regulate cytotoxic cellular responses contributing to autoimmunity or immune evasion by viruses and cancer cells. Therefore, pharmacological regulation of their activity is a promising avenue for modulating the adaptive immune response with possible applications in controlling autoimmunity, in boosting immune responses to pathogens, and in cancer immunotherapy. In this study we exploited recent structural and biochemical analysis of ERAP1 and ERAP2 to design and develop phosphinic pseudopeptide transition state analogs that can inhibit this family of enzymes with nM affinity. X-ray crystallographic analysis of one such inhibitor in complex with ERAP2 validated our design, revealing a canonical mode of binding in the active site of the enzyme, and highlighted the importance of the S2' pocket for achieving inhibitor potency. Antigen processing and presentation assays in HeLa and murine colon carcinoma (CT26) cells showed that these inhibitors induce increased cell-surface antigen presentation of transfected and endogenous antigens and enhance cytotoxic T-cell responses, indicating that these enzymes primarily destroy epitopes in those systems. This class of inhibitors constitutes a promising tool for controlling the cellular adaptive immune response in humans by modulating the antigen processing and presentation pathway.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Antigen Presentation/immunology , Models, Molecular , T-Lymphocytes, Cytotoxic/immunology , Aminopeptidases/chemistry , Aminopeptidases/metabolism , Animals , Antigen Presentation/drug effects , Binding Sites/immunology , Cell Line, Tumor , Crystallography, X-Ray , Cystinyl Aminopeptidase/metabolism , HeLa Cells , Humans , Mice , Minor Histocompatibility Antigens , Molecular Structure , Phosphinic Acids , Protein Engineering , T-Lymphocytes, Cytotoxic/drug effects
16.
Autoimmunity ; 46(8): 497-508, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24028501

ABSTRACT

Ankylosing spondylitis (AS) is a chronic systemic arthritic disease that leads to significant disability and loss of quality of life in the ∼0.5% of the worldwide human population it affects. There is currently no cure for AS and mechanisms underlying its pathogenesis remain unclear. AS is highly genetic, with over 70% of the genetic risk being associated with the presence of HLA-B27 and endoplasmic reticulum aminopeptidase-1 (ERAP1) alleles. Furthermore, gene-gene interactions between HLA-B27 and ERAP1 AS risk alleles have recently been confirmed. Here, we demonstrate that various ERAP1 alleles can differentially mediate surface expression of antigens presented by HLA-B27 on human cells. Specifically, for all peptides tested, we found that an ERAP1 variant containing high AS risk SNPs reduced the amount of the peptide presented by HLA-B27, relative to low AS risk ERAP1 variants. These results were further validated using peptide catalysis assays in vitro, suggesting that high AS risk alleles have an enhanced catalytic activity that more rapidly destroys many HLA-B27-destined peptides, a result that correlated with decreased HLA-B27 presentation of the same peptides. These findings suggest that one mechanism underlying AS pathogenesis may involve an altered ability for AS patients harboring both HLA-B27 and high AS risk ERAP1 alleles to correctly display a variety of peptides to the adaptive arm of the immune system, potentially exposing such individuals to higher AS risk due to abnormal display of pathogen or self-derived peptides by the adaptive immune system.


Subject(s)
Alleles , Aminopeptidases/genetics , Antigen Presentation/immunology , HLA-B27 Antigen/immunology , Spondylitis, Ankylosing/genetics , Spondylitis, Ankylosing/immunology , Amino Acid Substitution , Epitopes/chemistry , Epitopes/immunology , Gene Expression , Genetic Predisposition to Disease , HLA-B27 Antigen/genetics , Humans , Minor Histocompatibility Antigens , Peptides/chemistry , Peptides/immunology , Polymorphism, Single Nucleotide , Reproducibility of Results , Transfection
17.
PLoS One ; 8(7): e69539, 2013.
Article in English | MEDLINE | ID: mdl-23894499

ABSTRACT

Endoplasmic reticulum aminopeptidase-1 (ERAP1) is a multifunctional, ubiquitously expressed enzyme whose peptide-trimming role during antigen processing for presentation by MHC I molecules is well established, however, a role for ERAP1 in modulating global innate immune responses has not been described to date. Here we demonstrate that, relative to wild type mice, mice lacking ERAP1 exhibit exaggerated innate immune responses early during pathogen recognition, as characterized by increased activation of splenic and hepatic NK and NKT cells and enhanced production of pro-inflammatory cytokines such as IL12 and MCP1. Our data also revealed that ERAP1 is playing a critical role in NK cell development and function. We observed higher frequencies of terminally matured NK cells, as well as higher frequencies of licensed NK cells (expressing the Ly49C and Ly49I receptors) in ERAP1-KO mice, results that positively correlated with an enhanced NK activation and IFNγ production by ERAP1-KO mice challenged with pro-inflammatory stimuli. Furthermore, during pathogen recognition, ERAP1 regulates IL12 production by CD11c(+) DCs specifically, with increases in IL12 production positively correlated with an increased phagocytic activity of splenic DCs and macrophages. Collectively, our results demonstrate a previously unrecognized, more central role for the ERAP1 protein in modulating several aspects of both the development of the innate immune system, and its responses during the initial stages of pathogen recognition. Such a role may explain why ERAP1 has been implicated by GWAS in the pathogenesis of autoimmune diseases that may be precipitated by aberrant responses to pathogen encounters.


Subject(s)
Aminopeptidases/metabolism , Immunity, Innate/physiology , Aminopeptidases/genetics , Animals , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunity, Innate/genetics , Interleukin-12/metabolism , Killer Cells, Natural/metabolism , Liver/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , Reverse Transcriptase Polymerase Chain Reaction , Spleen/cytology
18.
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
19.
J Immunol ; 189(5): 2383-92, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22837489

ABSTRACT

Endoplasmic reticulum aminopeptidases 1 and 2 (ERAP1 and ERAP2) cooperate to trim antigenic peptide precursors for loading onto MHC class I molecules and help regulate the adaptive immune response. Common coding single nucleotide polymorphisms in ERAP1 and ERAP2 have been linked with predisposition to human diseases ranging from viral and bacterial infections to autoimmunity and cancer. It has been hypothesized that altered Ag processing by these enzymes is a causal link to disease etiology, but the molecular mechanisms are obscure. We report in this article that the common ERAP2 single nucleotide polymorphism rs2549782 that codes for amino acid variation N392K leads to alterations in both the activity and the specificity of the enzyme. Specifically, the 392N allele excises hydrophobic N-terminal residues from epitope precursors up to 165-fold faster compared with the 392K allele, although both alleles are very similar in excising positively charged N-terminal amino acids. These effects are primarily due to changes in the catalytic turnover rate (k(cat)) and not in the affinity for the substrate. X-ray crystallographic analysis of the ERAP2 392K allele suggests that the polymorphism interferes with the stabilization of the N terminus of the peptide both directly and indirectly through interactions with key residues participating in catalysis. This specificity switch allows the 392N allele of ERAP2 to supplement ERAP1 activity for the removal of hydrophobic N-terminal residues. Our results provide mechanistic insight to the association of this ERAP2 polymorphism with disease and support the idea that polymorphic variation in Ag processing enzymes constitutes a component of immune response variability in humans.


Subject(s)
Aminopeptidases/genetics , Antigen Presentation/immunology , Endoplasmic Reticulum/enzymology , Immunoglobulin Class Switching/immunology , Polymorphism, Single Nucleotide/immunology , Amino Acid Sequence , Antigen Presentation/genetics , Crystallography, X-Ray , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/immunology , Enzyme Activation/genetics , Enzyme Activation/immunology , Genetic Variation/immunology , HeLa Cells , Humans , Immunoglobulin Class Switching/genetics , Molecular Sequence Data , Substrate Specificity/genetics , Substrate Specificity/immunology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...