Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-37546948

ABSTRACT

Most human pancreatic ductal adenocarcinoma (PDAC) are not infiltrated with cytotoxic T cells and are highly resistant to immunotherapy. Over 90% of PDAC have oncogenic KRAS mutations, and phosphoinositide 3-kinases (PI3Ks) are direct effectors of KRAS. Our previous study demonstrated that ablation of Pik3ca in KPC (KrasG12D; Trp53R172H; Pdx1-Cre) pancreatic cancer cells induced host T cells to infiltrate and completely eliminate the tumors in a syngeneic orthotopic implantation mouse model. Now, we show that implantation of Pik3ca-/- KPC (named αKO) cancer cells induces clonal expansion of cytotoxic T cells infiltrating the pancreatic tumors. To identify potential molecules that can regulate the activity of these anti-tumor T cells, we conducted an in vivo genome-wide gene-deletion screen using αKO cells implanted in the mouse pancreas. The result shows that deletion of propionyl-CoA carboxylase subunit B gene (Pccb) in αKO cells (named p-αKO) leads to immune evasion, tumor progression and death of host mice. Surprisingly, p-αKO tumors are still infiltrated with clonally expanded CD8+ T cells but they are inactive against tumor cells. However, blockade of PD-L1/PD1 interaction reactivated these clonally expanded T cells infiltrating p-αKO tumors, leading to slower tumor progression and improve survival of host mice. These results indicate that Pccb can modulate the activity of cytotoxic T cells infiltrating some pancreatic cancers and this understanding may lead to improvement in immunotherapy for this difficult-to-treat cancer.

2.
Infect Immun ; 90(4): e0007022, 2022 04 21.
Article in English | MEDLINE | ID: mdl-35311578

ABSTRACT

Persistent infections generally involve a complex balance between protective immunity and immunopathology. We used a murine model to investigate the role of inflammatory monocytes in immunity and host defense against persistent salmonellosis. Mice exhibit increased susceptibility to persistent infection when inflammatory monocytes cannot be recruited into tissues or when they are depleted at specific stages of persistent infection. Inflammatory monocytes contribute to the pathology of persistent salmonellosis and cluster with other cells in pathogen-containing granulomas. Depletion of inflammatory monocytes during the chronic phase of persistent salmonellosis causes regression of already established granulomas with resultant pathogen growth and spread in tissues. Thus, inflammatory monocytes promote granuloma-mediated control of persistent salmonellosis and may be key to uncovering new therapies for granulomatous diseases.


Subject(s)
Monocytes , Salmonella Infections , Animals , Granuloma , Mice , Receptors, CCR2
3.
Mucosal Immunol ; 15(1): 176-187, 2022 01.
Article in English | MEDLINE | ID: mdl-34462572

ABSTRACT

Although murine γδ T cells are largely considered innate immune cells, they have recently been reported to form long-lived memory populations. Much remains unknown about the biology and specificity of memory γδ T cells. Here, we interrogated intestinal memory Vγ4 Vδ1 T cells generated after foodborne Listeria monocytogenes (Lm) infection to uncover an unanticipated complexity in the specificity of these cells. Deep TCR sequencing revealed that a subset of non-canonical Vδ1 clones are selected by Lm infection, consistent with antigen-specific clonal expansion. Ex vivo stimulations and in vivo heterologous challenge infections with diverse pathogenic bacteria revealed that Lm-elicited memory Vγ4 Vδ1 T cells are broadly reactive. The Vγ4 Vδ1 T cell recall response to Lm, Salmonella enterica serovar Typhimurium (STm) and Citrobacter rodentium was largely mediated by the γδTCR as internalizing the γδTCR prevented T cell expansion. Both broadly-reactive canonical and pathogen-selected non-canonical Vδ1 clones contributed to memory responses to Lm and STm. Interestingly, some non-canonical γδ T cell clones selected by Lm infection also responded after STm infection, suggesting some level of cross-reactivity. These findings underscore the promiscuous nature of memory γδ T cells and suggest that pathogen-elicited memory γδ T cells are potential targets for broad-spectrum anti-infective vaccines.


Subject(s)
Bacterial Infections/immunology , Bacterial Vaccines/immunology , Citrobacter rodentium/physiology , Listeria monocytogenes/physiology , Memory T Cells/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Salmonella typhi/physiology , Animals , Antigens, Bacterial/immunology , Cells, Cultured , Cross Reactions , High-Throughput Nucleotide Sequencing , Immunity, Heterologous , Memory T Cells/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell, gamma-delta/genetics , T-Cell Antigen Receptor Specificity
4.
Cancer Cell ; 39(4): 494-508.e5, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33545063

ABSTRACT

Mutant p53 (mtp53) proteins can exert cancer-promoting gain-of-function activities. We report a mechanism by which mtp53 suppresses both cell-autonomous and non-cell-autonomous signaling to promote cancer cell survival and evasion of tumor immune surveillance. Mtp53 interferes with the function of the cytoplasmic DNA sensing machinery, cGAS-STING-TBK1-IRF3, that activates the innate immune response. Mtp53, but not wild-type p53, binds to TANK-binding protein kinase 1 (TBK1) and prevents the formation of a trimeric complex between TBK1, STING, and IRF3, which is required for activation, nuclear translocation, and transcriptional activity of IRF3. Inactivation of innate immune signaling by mtp53 alters cytokine production, resulting in immune evasion. Restoring TBK1 signaling is sufficient to bypass mtp53 and lead to restored immune cell function and cancer cell eradication. This work is of translational interest because therapeutic approaches that restore TBK1 function could potentially reactivate immune surveillance and eliminate mtp53 tumors.


Subject(s)
Carcinogenesis/immunology , Immunity, Innate/immunology , Signal Transduction/immunology , Tumor Suppressor Protein p53/genetics , Animals , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/metabolism , Cytosol/metabolism , Gene Expression/genetics , Gene Expression/immunology , Membrane Proteins/genetics , Mice , Nucleotidyltransferases/genetics , Tumor Suppressor Protein p53/immunology
5.
Mucosal Immunol ; 14(2): 389-401, 2021 03.
Article in English | MEDLINE | ID: mdl-33060802

ABSTRACT

Interleukin-22 (IL-22) signaling in the intestines is critical for promoting tissue-protective functions. However, since a diverse array of cell types (absorptive and secretory epithelium as well as stem cells) express IL-22Ra1, a receptor for IL-22, it has been difficult to determine what cell type(s) specifically respond to IL-22 to mediate intestinal mucosal host defense. Here, we report that IL-22 signaling in the small intestine is positively correlated with Paneth cell differentiation programs. Our Il22Ra1fl/fl;Lgr5-EGFP-creERT2-specific knockout mice and, independently, our lineage-tracing findings rule out the involvement of Lgr5+ intestinal stem cell (ISC)-dependent IL-22Ra1 signaling in regulating the lineage commitment of epithelial cells, including Paneth cells. Using novel Paneth cell-specific IL-22Ra1 knockout mice (Il22Ra1fl/fl;Defa6-cre), we show that IL-22 signaling in Paneth cells is required for small intestinal host defense. We show that Paneth cell maturation, antimicrobial effector function, expression of specific WNTs, and organoid morphogenesis are dependent on cell-intrinsic IL-22Ra1 signaling. Furthermore, IL-22 signaling in Paneth cells regulates the intestinal commensal bacteria and microbiota-dependent IL-17A immune responses. Finally, we show ISC and, independently, Paneth cell-specific IL-22Ra1 signaling are critical for providing immunity against Salmonella enterica serovar Typhimurium. Collectively, our findings illustrate a previously unknown role of IL-22 in Paneth cell-mediated small intestinal host defense.


Subject(s)
Interleukins/metabolism , Microbiota/physiology , Paneth Cells/metabolism , Receptors, Interleukin/metabolism , Salmonella typhi/physiology , Th17 Cells/immunology , Typhoid Fever/immunology , Animals , Cell Differentiation , Immunity, Mucosal , Interleukins/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Paneth Cells/pathology , Receptors, Interleukin/genetics , Signal Transduction , Interleukin-22
6.
PLoS Pathog ; 15(7): e1007847, 2019 07.
Article in English | MEDLINE | ID: mdl-31306468

ABSTRACT

Salmonella exploit host-derived nitrate for growth in the lumen of the inflamed intestine. The generation of host-derived nitrate is dependent on Nos2, which encodes inducible nitric oxide synthase (iNOS), an enzyme that catalyzes nitric oxide (NO) production. However, the cellular sources of iNOS and, therefore, NO-derived nitrate used by Salmonella for growth in the lumen of the inflamed intestine remain unidentified. Here, we show that iNOS-producing inflammatory monocytes infiltrate ceca of mice infected with Salmonella. In addition, we show that inactivation of type-three secretion system (T3SS)-1 and T3SS-2 renders Salmonella unable to induce CC- chemokine receptor-2- and CC-chemokine ligand-2-dependent inflammatory monocyte recruitment. Furthermore, we show that the severity of the pathology of Salmonella- induced colitis as well as the nitrate-dependent growth of Salmonella in the lumen of the inflamed intestine are reduced in mice that lack Ccr2 and, therefore, inflammatory monocytes in the tissues. Thus, inflammatory monocytes provide a niche for Salmonella expansion in the lumen of the inflamed intestine.


Subject(s)
Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Monocytes/metabolism , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Animals , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Female , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Humans , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Monocytes/pathology , Nitric Oxide Synthase Type II/metabolism , Receptors, CCR2/deficiency , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Salmonella Infections, Animal/metabolism , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/pathology , Salmonella typhimurium/genetics , Type III Secretion Systems/metabolism
7.
J Clin Invest ; 129(8): 3264-3276, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31112530

ABSTRACT

The presence of tumor-infiltrating T cells is associated with favorable patient outcomes, yet most pancreatic cancers are immunologically silent and resistant to currently available immunotherapies. Here we show using a syngeneic orthotopic implantation model of pancreatic cancer that Pik3ca regulates tumor immunogenicity. Genetic silencing of Pik3ca in KrasG12D/Trp53R172H-driven pancreatic tumors resulted in infiltration of T cells, complete tumor regression, and 100% survival of immunocompetent host mice. By contrast, Pik3ca-null tumors implanted in T cell-deficient mice progressed and killed all of the animals. Adoptive transfer of tumor antigen-experienced T cells eliminated Pik3ca-null tumors in immunodeficient mice. Loss of PIK3CA or inhibition of its effector, AKT, increased the expression of MHC Class I and CD80 on tumor cells. These changes contributed to the increased susceptibility of Pik3ca-null tumors to T cell surveillance. Our results indicate that tumor cell PIK3CA-AKT signaling limits T cell recognition and clearance of pancreatic cancer cells. Strategies that target this pathway may yield an effective immunotherapy for this cancer.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms, Experimental/immunology , Pancreatic Neoplasms/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , B7-1 Antigen/genetics , B7-1 Antigen/immunology , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Knockout , Mice, SCID , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction/genetics , T-Lymphocytes/pathology , Xenograft Model Antitumor Assays
8.
Infect Immun ; 86(3)2018 03.
Article in English | MEDLINE | ID: mdl-29263104

ABSTRACT

Murine Ly6Chi inflammatory monocytes (IMs) require CCR2 to leave the bone marrow and enter mesenteric lymph nodes (MLNs) and other organs in response to Yersinia pseudotuberculosis infection. We are investigating how IMs, which can differentiate into CD11c+ dendritic cells (DCs), contribute to innate and adaptive immunity to Y. pseudotuberculosis Previously, we obtained evidence that IMs are important for a dominant CD8+ T cell response to the epitope YopE69-77 and host survival using intravenous infections with attenuated Y. pseudotuberculosis Here we challenged CCR2+/+ or CCR2-/- mice orally with wild-type Y. pseudotuberculosis to investigate how IMs contribute to immune responses during intestinal infection. Unexpectedly, CCR2-/- mice did not have reduced survival but retained body weight better and their MLNs cleared Y. pseudotuberculosis faster and with reduced lymphadenopathy compared to controls. Enhanced bacterial clearance in CCR2-/- mice correlated with reduced numbers of IMs in spleens and increased numbers of neutrophils in livers. In situ imaging of MLNs and spleens from CCR2-GFP mice showed that green fluorescent protein-positive (GFP+) IMs accumulated at the periphery of neutrophil-rich Yersinia-containing pyogranulomas. GFP+ IMs colocalized with CD11c+ cells and YopE69-77-specific CD8+ T cells in MLNs, suggesting that IM-derived DCs prime adaptive responses in Yersinia pyogranulomas. Consistently, CCR2-/- mice had reduced numbers of splenic DCs, YopE69-77-specific CD8+ T cells, CD4+ T cells, and B cells in organs and lower levels of serum antibodies to Y. pseudotuberculosis antigens. Our data suggest that IMs differentiate into DCs in MLN pyogranulomas and direct adaptive responses in T cells at the expense of innate immunity during oral Y. pseudotuberculosis infection.


Subject(s)
Adaptive Immunity , Immunity, Innate , Monocytes/immunology , Mouth/microbiology , Receptors, CCR2/immunology , Yersinia pseudotuberculosis Infections/immunology , Yersinia pseudotuberculosis/immunology , Animals , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Female , Humans , Mice , Mice, Inbred C57BL , Receptors, CCR2/genetics , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/physiology , Yersinia pseudotuberculosis Infections/genetics , Yersinia pseudotuberculosis Infections/microbiology
9.
Infect Immun ; 85(2)2017 02.
Article in English | MEDLINE | ID: mdl-27849183

ABSTRACT

Salmonellae are pathogenic bacteria that cause significant morbidity and mortality in humans worldwide. Salmonellae establish infection and avoid clearance by the immune system by mechanisms that are not well understood. We previously showed that l-asparaginase II produced by Salmonella enterica serovar Typhimurium (S Typhimurium) inhibits T cell responses and mediates virulence. In addition, we previously showed that asparagine deprivation such as that mediated by l-asparaginase II of S Typhimurium causes suppression of activation-induced T cell metabolic reprogramming. Here, we report that STM3997, which encodes a homolog of disulfide bond protein A (dsbA) of Escherichia coli, is required for l-asparaginase II stability and function. Furthermore, we report that l-asparaginase II localizes primarily to the periplasm and acts together with l-asparaginase I to provide S Typhimurium the ability to catabolize asparagine and assimilate nitrogen. Importantly, we determined that, in a murine model of infection, S Typhimurium lacking both l-asparaginase I and II genes competes poorly with wild-type S Typhimurium for colonization of target tissues. Collectively, these results indicate that asparagine catabolism contributes to S Typhimurium virulence, providing new insights into the competition for nutrients at the host-pathogen interface.


Subject(s)
Asparagine/metabolism , Salmonella Infections/microbiology , Salmonella/metabolism , Salmonella/pathogenicity , Animals , Asparaginase/metabolism , Catalysis , Cysteine/metabolism , Disease Models, Animal , Enzyme Stability , Female , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mutation , Nitrogen/metabolism , Salmonella/genetics , Salmonella/immunology , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Virulence , Virulence Factors/genetics
10.
Cell Host Microbe ; 20(5): 551-552, 2016 11 09.
Article in English | MEDLINE | ID: mdl-27832583

ABSTRACT

How bacterial pathogens evade adaptive immunity is not well understood. In this issue of Cell Host & Microbe, Bayer-Santos et al. (2016) show that the Salmonella effector protein SteD mediates MARCH8-dependent ubiquitination of class II MHC molecules, thereby inhibiting antigen presentation and limiting T cell responses.


Subject(s)
Histocompatibility Antigens Class II/immunology , Salmonella , Antigen Presentation , T-Lymphocytes/immunology , Ubiquitination
11.
J Leukoc Biol ; 99(2): 387-98, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26497246

ABSTRACT

Salmonellae are pathogenic bacteria that induce immunosuppression by mechanisms that remain largely unknown. Previously, we showed that a putative type II l-asparaginase produced by Salmonella Typhimurium inhibits T cell responses and mediates virulence in a murine model of infection. Here, we report that this putative L-asparaginase exhibits L-asparagine hydrolase activity required for Salmonella Typhimurium to inhibit T cells. We show that L-asparagine is a nutrient important for T cell activation and that L-asparagine deprivation, such as that mediated by the Salmonella Typhimurium L-asparaginase, causes suppression of activation-induced mammalian target of rapamycin signaling, autophagy, Myc expression, and L-lactate secretion. We also show that L-asparagine deprivation mediated by the Salmonella Typhimurium L-asparaginase causes suppression of cellular processes and pathways involved in protein synthesis, metabolism, and immune response. Our results advance knowledge of a mechanism used by Salmonella Typhimurium to inhibit T cell responses and mediate virulence, and provide new insights into the prerequisites of T cell activation. We propose a model in which l-asparagine deprivation inhibits T cell exit from quiescence by causing suppression of activation-induced metabolic reprogramming.


Subject(s)
Asparaginase/physiology , Asparagine/physiology , Bacterial Proteins/physiology , Immune Evasion/physiology , Salmonella typhimurium/enzymology , T-Lymphocyte Subsets/metabolism , Amino Acid Sequence , Animals , Asparaginase/genetics , Asparaginase/pharmacology , Asparagine/deficiency , Asparagine/pharmacology , Autophagy/drug effects , Bacterial Proteins/genetics , Cells, Cultured , Female , Genes, myc , Immune Evasion/genetics , Interleukin-2/biosynthesis , Interleukin-2/genetics , Lactic Acid/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Proto-Oncogene Proteins c-myc/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/genetics , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , TOR Serine-Threonine Kinases/metabolism , Virulence
12.
PLoS Pathog ; 11(10): e1005167, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26468944

ABSTRACT

During Yersinia pseudotuberculosis infection of C57BL/6 mice, an exceptionally large CD8+ T cell response to a protective epitope in the type III secretion system effector YopE is produced. At the peak of the response, up to 50% of splenic CD8+ T cells recognize the epitope YopE69-77. The features of the interaction between pathogen and host that result in this large CD8+ T cell response are unknown. Here, we used Y. pseudotuberculosis strains defective for production, secretion and/or translocation of YopE to infect wild-type or mutant mice deficient in specific dendritic cells (DCs). Bacterial colonization of organs and translocation of YopE into spleen cells was measured, and flow cytometry and tetramer staining were used to characterize the cellular immune response. We show that the splenic YopE69-77-specific CD8+ T cells generated during the large response are polyclonal and are produced by a "translocation-dependent" pathway that requires injection of YopE into host cell cytosol. Additionally, a smaller YopE69-77-specific CD8+ T cell response (~10% of the large expansion) can be generated in a "translocation-independent" pathway in which CD8α+ DCs cross present secreted YopE. CCR2-expressing inflammatory DCs were required for the large YopE69-77-specific CD8+ T cell expansion because this response was significantly reduced in Ccr2-/- mice, YopE was translocated into inflammatory DCs in vivo, inflammatory DCs purified from infected spleens activated YopE69-77-specific CD8+ T cells ex vivo and promoted the expansion of YopE69-77-specific CD8+ T cells in infected Ccr2-/- mice after adoptive transfer. A requirement for inflammatory DCs in producing a protective CD8+ T cell response to a bacterial antigen has not previously been demonstrated. Therefore, the production of YopE69-77-specific CD8+ T cells by inflammatory DCs that are injected with YopE during Y. pseudotuberculosis infection represents a novel mechanism for generating a massive and protective adaptive immune response.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Type III Secretion Systems/immunology , Yersinia pseudotuberculosis Infections/immunology , Adoptive Transfer , Animals , Antigen Presentation/immunology , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Female , Flow Cytometry , Immunoblotting , Inflammation/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Protein Transport/immunology , Receptors, CCR2/immunology , Virulence Factors/immunology , Yersinia pseudotuberculosis/immunology , Yersinia pseudotuberculosis/pathogenicity
13.
J Virol ; 89(13): 6562-74, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25855746

ABSTRACT

UNLABELLED: Gammaherpesviruses establish lifelong infections that are associated with the development of cancer. These viruses subvert many aspects of the innate and adaptive immune response of the host. The inflammasome, a macromolecular protein complex that controls inflammatory responses to intracellular danger signals generated by pathogens, is both activated and subverted during human gammaherpesvirus infection in culture. The impact of the inflammasome response on gammaherpesvirus replication and latency in vivo is not known. Caspase-1 is the inflammasome effector protease that cleaves the proinflammatory cytokines interleukin-1ß (IL-1ß) and IL-18. We infected caspase-1-deficient mice with murine gammaherpesvirus 68 (MHV68) and observed no impact on acute replication in the lung or latency and reactivation from latency in the spleen. This led us to examine the effect of viral infection on inflammasome responses in bone marrow-derived macrophages. We determined that infection of macrophages with MHV68 led to a robust interferon response but failed to activate caspase-1 or induce the secretion of IL-1ß. In addition, MHV68 infection led to a reduction in IL-1ß production after extrinsic lipopolysaccharide stimulation or upon coinfection with Salmonella enterica serovar Typhimurium. Interestingly, this impairment occurred at the proIL-1ß transcript level and was independent of the RTA, the viral lytic replication and transcription activator. Taken together, MHV68 impairs the inflammasome response by inhibiting IL-1ß production during the initial stages of infection. IMPORTANCE: Gammaherpesviruses persist for the lifetime of the host. To accomplish this, they must evade recognition and clearance by the immune system. The inflammasome consists of proteins that detect foreign molecules in the cell and respond by secreting proinflammatory signaling proteins that recruit immune cells to clear the infection. Unexpectedly, we found that murine gammaherpesvirus pathogenesis was not enhanced in mice lacking caspase-1, a critical inflammasome component. This led us to investigate whether the virus actively impairs the inflammasome response. We found that the inflammasome was not activated upon macrophage cell infection with murine gammaherpesvirus 68. Infection also prevented the host cell inflammasome response to other pathogen-associated molecular patterns, indicated by reduced production of the proinflammatory cytokine IL-1ß upon bacterial coinfection. Taken together, murine gammaherpesvirus impairment of the inflammatory cytokine IL-1ß in macrophages identifies one mechanism by which the virus may inhibit caspase-1-dependent immune responses in the infected animal.


Subject(s)
Caspase 1/metabolism , Caspases/metabolism , Herpesviridae Infections/pathology , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Rhadinovirus/immunology , Tumor Virus Infections/pathology , Animals , Caspase 1/deficiency , Caspases, Initiator , Herpesviridae Infections/immunology , Interferons/metabolism , Lipopolysaccharides/immunology , Lung/virology , Macrophages , Mice, Inbred C57BL , Mice, Knockout , Rhadinovirus/physiology , Salmonella typhimurium/immunology , Spleen/virology , Tumor Virus Infections/immunology , Virus Activation , Virus Latency , Virus Replication
14.
Infect Immun ; 82(6): 2606-14, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24711563

ABSTRACT

Immature myeloid cells in bone marrow are a heterogeneous population of cells that, under normal conditions, provide tissues with protective cell types such as granulocytes and macrophages. Under certain pathological conditions, myeloid cell homeostasis is altered and immature forms of these cells appear in tissues. Murine immature myeloid cells that express CD11b and Ly6C or Ly6G (two isoforms of Gr-1) have been associated with immunosuppression in cancer (in the form of myeloid-derived suppressor cells) and, more recently, infection. Here, we found that CD11b(+) Ly6C(hi) Ly6G(-) and CD11b(+) Ly6C(int) Ly6G(+) cells accumulated and persisted in tissues of mice infected with Salmonella enterica serovar Typhimurium (S. Typhimurium). Recruitment of CD11b(+) Ly6C(hi) Ly6G(-) but not CD11b(+) Ly6C(int) Ly6G(+) cells from bone marrow into infected tissues depended on chemokine receptor CCR2. The CD11b(+) Ly6C(hi) Ly6G(-) cells exhibited a mononuclear morphology, whereas the CD11b(+) Ly6C(int) Ly6G(+) cells exhibited a polymorphonuclear or band-shaped nuclear morphology. The CD11b(+) Ly6C(hi) Ly6G(-) cells differentiated into macrophage-like cells following ex vivo culture and could present antigen to T cells in vitro. However, significant proliferation of T cells was observed only when the ability of the CD11b(+) Ly6C(hi) Ly6G(-) cells to produce nitric oxide was blocked. CD11b(+) Ly6C(hi) Ly6G(-) cells recruited in response to S. Typhimurium infection could also present antigen to T cells in vivo, but increasing their numbers by adoptive transfer did not cause a corresponding increase in T cell response. Thus, CD11b(+) Ly6C(hi) Ly6G(-) immature myeloid cells recruited in response to S. Typhimurium infection exhibit protective and immunosuppressive properties that may influence the outcome of infection.


Subject(s)
Antigens, Ly/immunology , CD11b Antigen/immunology , Myeloid Cells/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Analysis of Variance , Animals , Cell Differentiation/physiology , Cell Proliferation , Immunity, Innate/physiology , Immunosuppression Therapy , Liver/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, CCR2/physiology , Spleen/cytology , T-Lymphocytes/immunology
15.
PLoS One ; 9(4): e92807, 2014.
Article in English | MEDLINE | ID: mdl-24717768

ABSTRACT

Pancreatitis, a known risk factor for the development of pancreatic ductal adenocarcinoma, is a serious, widespread medical condition usually caused by alcohol abuse or gallstone-mediated ductal obstruction. However, many cases of pancreatitis are of an unknown etiology. Pancreatitis has been linked to bacterial infection, but causality has yet to be established. Here, we found that persistent infection of mice with the bacterial pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium) was sufficient to induce pancreatitis reminiscent of the human disease. Specifically, we found that pancreatitis induced by persistent S. Typhimurium infection was characterized by a loss of pancreatic acinar cells, acinar-to-ductal metaplasia, fibrosis and accumulation of inflammatory cells, including CD11b+ F4/80+, CD11b+ Ly6Cint Ly6G+ and CD11b+ Ly6Chi Ly6G- cells. Furthermore, we found that S. Typhimurium colonized and persisted in the pancreas, associated with pancreatic acinar cells in vivo, and could invade cultured pancreatic acinar cells in vitro. Thus, persistent infection of mice with S. Typhimurium may serve as a useful model for the study of pancreatitis as it relates to bacterial infection. Increased knowledge of how pathogenic bacteria can cause pancreatitis will provide a more integrated picture of the etiology of the disease and could lead to the development of new therapeutic approaches for treatment and prevention of pancreatitis and pancreatic ductal adenocarcinoma.


Subject(s)
Pancreatitis/etiology , Salmonella Infections, Animal/complications , Acinar Cells/microbiology , Acinar Cells/pathology , Animals , Disease Models, Animal , Humans , Lipopolysaccharides , Mice, Inbred C57BL , Pancreas/microbiology , Pancreas/pathology , Pancreatitis/pathology , Salmonella Infections, Animal/pathology , Salmonella typhimurium/physiology
16.
Cell Host Microbe ; 12(6): 791-8, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-23245323

ABSTRACT

Salmonella enterica serovar Typhimurium avoids clearance by the host immune system by suppressing T cell responses; however, the mechanisms that mediate this immunosuppression remain unknown. We show that S. Typhimurium inhibit T cell responses by producing L-Asparaginase II, which catalyzes the hydrolysis of L-asparagine to aspartic acid and ammonia. L-Asparaginase II is necessary and sufficient to suppress T cell blastogenesis, cytokine production, and proliferation and to downmodulate expression of the T cell receptor. Furthermore, S. Typhimurium-induced inhibition of T cells in vitro is prevented upon addition of L-asparagine. S. Typhimurium lacking the L-Asparaginase II gene (STM3106) are unable to inhibit T cell responses and exhibit attenuated virulence in vivo. L-Asparaginases are used to treat acute lymphoblastic leukemia through mechanisms that likely involve amino acid starvation of leukemic cells, and these findings indicate that pathogens similarly use L-asparagine deprivation to limit T cell responses.


Subject(s)
Asparaginase/metabolism , Immune Evasion , Salmonella typhimurium/enzymology , Salmonella typhimurium/pathogenicity , T-Lymphocytes/immunology , Virulence Factors/metabolism , Ammonia/metabolism , Animals , Asparaginase/genetics , Asparagine/metabolism , Aspartic Acid/metabolism , Cell Proliferation , Cytokines/metabolism , Gene Deletion , Mice , Mice, Inbred C57BL , Salmonella typhimurium/immunology , Virulence , Virulence Factors/genetics
17.
Sci Signal ; 5(249): ra79, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23131846

ABSTRACT

Angiogenesis inhibitors, which target tumor cells, confer only short-term benefits on tumor growth. We report that ablation of the lipid signaling enzyme phospholipase D1 (PLD1) in the tumor environment compromised the neovascularization and growth of tumors. PLD1 deficiency suppressed the activation of Akt and mitogen-activated protein kinase signaling pathways by vascular endothelial growth factor in vascular endothelial cells, resulting in decreased integrin-dependent cell adhesion to, and migration on, extracellular matrices, as well as reduced tumor angiogenesis in a xenograft model. In addition, mice lacking PLD1 incurred fewer lung metastases than did wild-type mice. Bone marrow transplantation and binding studies identified a platelet-derived mechanism involving decreased tumor cell-platelet interactions, in part because of impaired activation of αIIbß3 integrin in platelets, which decreased the seeding of tumor cells into the lung parenchyma. Treatment with a small-molecule inhibitor of PLD1 phenocopied PLD1 deficiency, efficiently suppressing both tumor growth and metastasis in mice. These findings reveal that PLD1 in the tumor environment promotes tumor growth and metastasis and, taken together with previous reports on the roles of PLD in tumor cell-intrinsic adaptations to stress, suggest the potential use of PLD inhibitors as cancer therapeutics.


Subject(s)
Breast Neoplasms/enzymology , Endothelial Cells/enzymology , Lung Neoplasms/enzymology , Neovascularization, Pathologic/enzymology , Phospholipase D/metabolism , Signal Transduction , Tumor Microenvironment , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Endothelial Cells/pathology , Female , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice , Mice, Knockout , Neoplasm Metastasis , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Phospholipase D/genetics , Transplantation, Heterologous
18.
mBio ; 3(4): e00226-12, 2012.
Article in English | MEDLINE | ID: mdl-22893385

ABSTRACT

The microbiota of the mammalian intestinal tract represents a formidable barrier to colonization by pathogens. To overcome this resistance to colonization, bacterial pathogens use virulence factors to induce intestinal inflammation, which liberates nutrients for selective use by the infecting microbe. Studies of Salmonella enterica serovar Typhimurium (S. Typhimurium) infection in a streptomycin-treated mouse colitis model show how virulence factor-induced inflammation can produce nutrients used selectively by the pathogen. Type III secreted effectors of invading S. Typhimurium induce inflammation in the intestine (epithelial cells and lamina propria macrophages) that causes changes in the composition of the lumen. For example, neutrophils entering the intestine produce superoxide, resulting in production of tetrathionate, which S. Typhimurium in the lumen uses as an electron acceptor for anaerobic respiration. In their recent study, Lopez et al. demonstrate that S. Typhimurium strains that are lysogenized with a phage encoding type III effector SopE induce the host to produce nitric oxide synthetase (iNOS) in the intestine (C. A. Lopez et al., mBio 3:e00143-12, 2012). Nitric oxide is converted to a highly favorable electron acceptor, nitrate. As a result, growth of sopE(+) S. Typhimurium in the intestine lumen is boosted by nitrate respiration. This is a striking example of how acquisition of a virulence factor by horizontal gene transfer can increase the metabolic fitness of a pathogen. Interestingly, survival of the invading bacteria is probably decreased as a result of the SopE-induced immune response, and yet the S. Typhimurium bacteria that multiply in the lumen of the intestine can efficiently disseminate to another host, ensuring success for the pathogen.


Subject(s)
Colitis/microbiology , Colon/microbiology , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Animals , Bacterial Proteins/metabolism , Colitis/pathology , Disease Models, Animal , Host-Pathogen Interactions , Mice , Nitrates/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Salmonella Infections, Animal/pathology , Salmonella typhimurium/growth & development , Virulence Factors/metabolism
19.
Infect Immun ; 80(7): 2371-81, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22526678

ABSTRACT

Recent studies have linked accumulation of the Gr-1⁺ CD11b⁺ cell phenotype with functional immunosuppression in diverse pathological conditions, including bacterial and parasitic infections and cancer. Gr-1⁺ CD11b⁺ cells were the largest population of cells present in the spleens of mice infected with sublethal doses of the Francisella tularensis live vaccine strain (LVS). In contrast, the number of T cells present in the spleens of these mice did not increase during early infection. There was a significant delay in the kinetics of accumulation of Gr-1⁺ CD11b⁺ cells in the spleens of B-cell-deficient mice, indicating that B cells play a role in recruitment and maintenance of this population in the spleens of mice infected with F. tularensis. The splenic Gr-1⁺ CD11b⁺ cells in tularemia were a heterogeneous population that could be further subdivided into monocytic (mononuclear) and granulocytic (polymorphonuclear) cells using the Ly6C and Ly6G markers and differentiated into antigen-presenting cells following ex vivo culture. Monocytic, CD11b⁺ Ly6C(hi) Ly6G⁻ cells but not granulocytic, CD11b⁺ Ly6C(int) Ly6G⁺ cells purified from the spleens of mice infected with F. tularensis suppressed polyclonal T-cell proliferation via a nitric oxide-dependent pathway. Although the monocytic, CD11b⁺ Ly6C(hi) Ly6G⁻ cells were able to suppress the proliferation of T cells, the large presence of Gr-1⁺ CD11b⁺ cells in mice that survived F. tularensis infection also suggests a potential role for these cells in the protective host response to tularemia.


Subject(s)
Francisella tularensis/pathogenicity , Myeloid Cells/cytology , Myeloid Cells/physiology , Spleen/immunology , Spleen/pathology , Tularemia/immunology , Tularemia/pathology , Animals , B-Lymphocytes/immunology , CD11b Antigen/analysis , Disease Models, Animal , Female , Immunophenotyping , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Receptors, Chemokine/analysis , T-Lymphocytes/immunology
20.
J Am Chem Soc ; 132(24): 8244-5, 2010 Jun 23.
Article in English | MEDLINE | ID: mdl-20504019

ABSTRACT

Type III protein secretion is essential for many gram-negative bacterial infections of host cells and an attractive target for new antibacterial drugs. Here, we describe a bacterial protein effector-carboxypeptidase G2 (CPG2) reporter system for fluorescence and visible detection of type III protein secretion in Salmonella typhimurium. This system provides a general method for measuring protein expression and secretion as well as a high-throughput and quantitative assay for analyzing type III protein secretion inhibitors.


Subject(s)
Bacterial Proteins/metabolism , Salmonella enterica , Spectrometry, Fluorescence/methods , Bacterial Proteins/antagonists & inhibitors , Color , HeLa Cells , Humans , gamma-Glutamyl Hydrolase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...