20 results on '"Jason W. Tam"'
Search Results
2. Inflammatory monocytes provide a niche for Salmonella expansion in the lumen of the inflamed intestine.
- Author
-
Patrick A McLaughlin, Julie A Bettke, Jason W Tam, Jesse Leeds, James B Bliska, Brian P Butler, and Adrianus W M van der Velden
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - 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.
- Published
- 2019
- Full Text
- View/download PDF
3. Inflammatory Monocytes Promote Granuloma-Mediated Control of Persistent Salmonella Infection
- Author
-
Julie A, Bettke, Jason W, Tam, Valeria, Montoya, Brian P, Butler, and Adrianus W M, van der Velden
- Subjects
Granuloma ,Receptors, CCR2 ,animal diseases ,Immunology ,chemical and pharmacologic phenomena ,Bacterial Infections ,biochemical phenomena, metabolism, and nutrition ,Microbiology ,Monocytes ,Mice ,Infectious Diseases ,Salmonella Infections ,Animals ,bacteria ,Parasitology - 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.
- Published
- 2022
4. TRAF3IP3 negatively regulates cytosolic RNA induced anti-viral signaling by promoting TBK1 K48 ubiquitination
- Author
-
W. June Brickey, Shao Cong Sun, Beckley K. Davis, Kaixin Liang, Jenny P.-Y. Ting, Lufei Wang, Jason W. Tam, Sirui Li, Yang Zhang, Alex Petrucelli, Haitao Guo, Brian J. Conti, Meng Deng, Ching-Chang Ko, Lu Zhang, Yu Lei, and Xiaobo Luo
- Subjects
0301 basic medicine ,TRAF3 ,THP-1 Cells ,General Physics and Astronomy ,Diseases ,Jurkat cells ,Jurkat Cells ,0302 clinical medicine ,Cytosol ,Interferon ,Chlorocebus aethiops ,Myeloid Cells ,lcsh:Science ,Cells, Cultured ,Innate immunity ,Mice, Knockout ,Multidisciplinary ,biology ,Chemistry ,Cell biology ,Vesicular stomatitis virus ,Interferon Type I ,Infectious diseases ,RNA, Viral ,medicine.drug ,Science ,Immunology ,Mice, Transgenic ,Protein Serine-Threonine Kinases ,General Biochemistry, Genetics and Molecular Biology ,Article ,Vesicular stomatitis Indiana virus ,Cell Line ,03 medical and health sciences ,Immune system ,medicine ,Animals ,Humans ,Vero Cells ,Mitochondrial antiviral-signaling protein ,Innate immune system ,Lysine ,Ubiquitination ,RNA ,Membrane Proteins ,General Chemistry ,biology.organism_classification ,Mice, Inbred C57BL ,030104 developmental biology ,HEK293 Cells ,Gene Expression Regulation ,lcsh:Q ,Carrier Proteins ,030217 neurology & neurosurgery ,HeLa Cells - Abstract
Innate immunity to nucleic acids forms the backbone for anti-viral immunity and several inflammatory diseases. Upon sensing cytosolic viral RNA, retinoic acid-inducible gene-I-like receptors (RLRs) interact with the mitochondrial antiviral signaling protein (MAVS) and activate TANK-binding kinase 1 (TBK1) to induce type I interferon (IFN-I). TRAF3-interacting protein 3 (TRAF3IP3, T3JAM) is essential for T and B cell development. It is also well-expressed by myeloid cells, where its role is unknown. Here we report that TRAF3IP3 suppresses cytosolic poly(I:C), 5’ppp-dsRNA, and vesicular stomatitis virus (VSV) triggers IFN-I expression in overexpression systems and Traf3ip3−/− primary myeloid cells. The mechanism of action is through the interaction of TRAF3IP3 with endogenous TRAF3 and TBK1. This leads to the degradative K48 ubiquitination of TBK1 via its K372 residue in a DTX4-dependent fashion. Mice with myeloid-specific gene deletion of Traf3ip3 have increased RNA virus-triggered IFN-I production and reduced susceptibility to virus. These results identify a function of TRAF3IP3 in the regulation of the host response to cytosolic viral RNA in myeloid cells., RNA viruses can be detected by immune cell pattern recognition receptors, such as RLRs, resulting in MAVS-TBK1-IRF3 signalling and production of antiviral type 1 interferons. Here the authors show that macrophage TRAF3-interacting protein 3 regulates this signalling pathway by interacting with TRAF3 and TBK1 to suppress interferon responses.
- Published
- 2020
5. Protective and aggressive bacterial subsets and metabolites modify hepatobiliary inflammation and fibrosis in PSC
- Author
-
Viswanathan A, Yury Popov, Jason W. Tam, Corinna Bang, Andre Franke, Jenny P.-Y. Ting, Muyiwa Awoniyi, Schnabl B, Farmer M, Yunjia Lai, Martin Kummen, Kun Lu, Hov, Louise B. Thingholm, Stephanie A. Montgomery, Francis H, Jin Wang, Christoph Schramm, Ngo B, Ryan Balfour Sartor, and Meadows
- Subjects
medicine.drug_class ,business.industry ,Antibiotics ,Hepatobiliary disease ,Lachnospiraceae ,Inflammation ,medicine.disease ,Primary sclerosing cholangitis ,Transplantation ,Fibrosis ,Immunology ,medicine ,Vancomycin ,medicine.symptom ,business ,medicine.drug - Abstract
ObjectiveConflicting microbiota data exist for primary sclerosing cholangitis (PSC) and experimental models. Goal: Define complex interactions between resident microbes and their association in PSC patients by studying antibiotic-treated specific pathogen-free (SPF) and germ-free (GF) multi-drug-resistant 2 deficient (mdr2-/-) mice.DesignWe measured weights, liver enzymes, RNA expression, histological, immunohistochemical and fibrotic biochemical parameters, fecal 16s rRNA gene profiling, and metabolomic endpoints in gnotobiotic and antibiotic-treated SPF mdr2-/- mice and targeted metagenomic analysis in PSC patients.ResultsGF mdr2-/- mice had exaggerated hepatic inflammation and fibrosis with 100% mortality by 8 weeks; early SPF autologous stool transplantation rescued liver-related mortality. Broad-spectrum antibiotics and vancomycin alone accelerated disease in weanling SPF mdr2-/- mice, indicating that vancomycin-sensitive resident microbiota protect against hepatobiliary disease. Vancomycin treatment selectively decreased Lachnospiraceae and short-chain fatty acids (SCFAs) but expanded Enterococcus and Enterobacteriaceae. Antibiotics increased cytolysin-expressing E. faecalis and E. coli liver translocation; colonization of gnotobiotic mdr2-/- mice with translocated E. faecalis and E. coli strains accelerated liver inflammation and mortality. Lachnospiraceae colonization of antibiotic pre-treated mdr2-/- mice reduced liver fibrosis, inflammation and translocation of pathobionts, while Lachnospiraceae-produced SCFA decreased fibrosis. Fecal E. faecalis/ Enterobacteriaceae was positively and Lachnospiraceae was negatively associated with PSC patients’ clinical severity Mayo risk scores.ConclusionsWe identified specific functionally protective and detrimental resident bacterial species in mdr2-/- mice and PSC patients with associated clinical outcomes. These insights may guide personalized targeted therapeutic interventions in PSC patients.
- Published
- 2021
6. CCR2+ Inflammatory Dendritic Cells and Translocation of Antigen by Type III Secretion Are Required for the Exceptionally Large CD8+ T Cell Response to the Protective YopE69-77 Epitope during Yersinia Infection.
- Author
-
Yue Zhang, Jason W Tam, Patricio Mena, Adrianus W M van der Velden, and James B Bliska
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - 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.
- Published
- 2015
- Full Text
- View/download PDF
7. STING Agonist Mitigates Experimental Autoimmune Encephalomyelitis by Stimulating Type I IFN–Dependent and –Independent Immune-Regulatory Pathways
- Author
-
Matthew D. Gallovic, Sara A. Gibson, Brandon M. Johnson, Kristy M. Ainslie, Jonathan W. Williams, Wei-Chun Chou, Cole J. Batty, Jenny P.-Y. Ting, Glenn K. Matsushima, Eric M. Bachelder, Meng Deng, Jason W. Tam, Toru Uchimura, Silva Markovic-Plese, and Madhan Thamilarasan
- Subjects
business.industry ,medicine.medical_treatment ,T cell ,Encephalomyelitis ,Immunology ,Experimental autoimmune encephalomyelitis ,medicine.disease ,medicine.disease_cause ,Article ,Autoimmunity ,03 medical and health sciences ,0302 clinical medicine ,Cytokine ,medicine.anatomical_structure ,Immune system ,Cancer immunotherapy ,Immunity ,medicine ,Immunology and Allergy ,business ,030215 immunology - Abstract
The cGAS–cyclic GMP–AMP (cGAMP)–stimulator of IFN genes (STING) pathway induces a powerful type I IFN (IFN-I) response and is a prime candidate for augmenting immunity in cancer immunotherapy and vaccines. IFN-I also has immune-regulatory functions manifested in several autoimmune diseases and is a first-line therapy for relapsing–remitting multiple sclerosis. However, it is only moderately effective and can induce adverse effects and neutralizing Abs in recipients. Targeting cGAMP in autoimmunity is unexplored and represents a challenge because of the intracellular location of its receptor, STING. We used microparticle (MP)–encapsulated cGAMP to increase cellular delivery, achieve dose sparing, and reduce potential toxicity. In the C57BL/6 experimental allergic encephalomyelitis (EAE) model, cGAMP encapsulated in MPs (cGAMP MPs) administered therapeutically protected mice from EAE in a STING-dependent fashion, whereas soluble cGAMP was ineffective. Protection was also observed in a relapsing–remitting model. Importantly, cGAMP MPs protected against EAE at the peak of disease and were more effective than rIFN-β. Mechanistically, cGAMP MPs showed both IFN-I–dependent and –independent immunosuppressive effects. Furthermore, it induced the immunosuppressive cytokine IL-27 without requiring IFN-I. This augmented IL-10 expression through activated ERK and CREB. IL-27 and subsequent IL-10 were the most important cytokines to mitigate autoreactivity. Critically, cGAMP MPs promoted IFN-I as well as the immunoregulatory cytokines IL-27 and IL-10 in PBMCs from relapsing–remitting multiple sclerosis patients. Collectively, this study reveals a previously unappreciated immune-regulatory effect of cGAMP that can be harnessed to restrain T cell autoreactivity.
- Published
- 2021
8. Multi-omics analyses of radiation survivors identify radioprotective microbes and metabolites
- Author
-
Jonathan U. Peled, Kun Lu, Yunjia Lai, Andrew N. Macintyre, Jenny P.-Y. Ting, W. June Brickey, R. Balfour Sartor, Matthew J. French, Jason W. Tam, Kaixin Liang, Xin Li, Hao Guo, Antonio L.C. Gomes, Nathan D. Montgomery, Xianming Tan, Anthony D. Sung, Lauren Bohannon, Marcel R.M. van den Brink, Gregory D. Sempowski, Wei-Chun Chou, Liang Chen, and Nelson J. Chao
- Subjects
0301 basic medicine ,Immunology ,Population ,Enterococcaceae ,Gut flora ,Article ,Proinflammatory cytokine ,Microbiology ,Mice ,03 medical and health sciences ,Radiation Protection ,0302 clinical medicine ,Immunology and Allergy ,Animals ,Humans ,Metabolomics ,Medicine ,Survivors ,education ,Clostridiales ,education.field_of_study ,Radiation ,Multidisciplinary ,biology ,business.industry ,Research ,Lachnospiraceae ,Gastrointestinal Microbiome ,Tryptophan ,Acute Radiation Syndrome ,Fatty Acids, Volatile ,biology.organism_classification ,medicine.disease ,Mice, Inbred C57BL ,Leukemia ,030104 developmental biology ,030220 oncology & carcinogenesis ,business - Abstract
Ionizing-radiation causes acute radiation syndrome leading to hematopoietic, gastrointestinal and cerebrovascular injuries. Substantial federal efforts have been made to develop therapeutic radioprotectants for routine clinical use, however it remains a long-standing and unresolved problem. We investigated a population of mice that recovered from high-dose radiation to live normal lifespans. These elite-survivors harbored distinct gut microbiota that developed post-radiation and protected against radiation-induced damage and death in germ-free and conventionally housed recipients. Elevated abundances of members of the bacterial taxa Lachnospiraceae and Enterococcaceae were associated with post-radiation restoration of hematopoiesis and gastrointestinal repair. These bacteria were also found to be more abundant in leukemia patients undergoing radiotherapy who also displayed milder gastrointestinal dysfunction. Metabolomics revealed increased fecal concentrations of microbially derived propionate and tryptophan metabolites in elite-survivors. The concentrations of these metabolites correlated with long-term radioprotection, mitigation of hematopoietic and gastrointestinal syndromes, and a reduction in pro-inflammatory responses. The novelty of our work lies in the first identification of gut microbiota and downstream metabolites in providing protection against lethal radiation. Furthermore, our work provides a comprehensive omics dataset at the bacteria and metabolite levels that is of broad interest and serves as a powerful resource and example for the identification of actionable therapeutic targets derived from microbiome studies.
- Published
- 2020
- Full Text
- View/download PDF
9. NLRP12 regulates anti-viral RIG-I activation via interaction with TRIM25
- Author
-
Chih Wei Chao, Jenny P.-Y. Ting, Yen Po Tsao, Ivan Dzhagalov, Liang Chen, Moon Jung Song, Hye Ri Kang, Diego Shih Chieh Lin, Szu Ting Chen, W. June Brickey, Fei Ju Li, Haitao Guo, Sei Yi Chen, Jae U. Jung, Jason W. Tam, and Wei Ting Tseng
- Subjects
Male ,TRIM25 ,viruses ,chemical and pharmacologic phenomena ,Microbiology ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,RNA Virus Infections ,Ubiquitin ,Interferon ,Virology ,medicine ,Animals ,Humans ,RNA Viruses ,030304 developmental biology ,0303 health sciences ,Innate immune system ,biology ,RIG-I ,Intracellular Signaling Peptides and Proteins ,Ubiquitination ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,Type I interferon production ,biology.organism_classification ,Ubiquitin ligase ,Cell biology ,DNA-Binding Proteins ,Mice, Inbred C57BL ,Vesicular stomatitis virus ,biology.protein ,DEAD Box Protein 58 ,Parasitology ,Female ,Interferons ,biological phenomena, cell phenomena, and immunity ,030217 neurology & neurosurgery ,medicine.drug ,Protein Binding ,Transcription Factors - Abstract
Establishing the balance between positive and negative innate immune mechanisms is crucial for maintaining homeostasis. Here we uncover the regulatory crosstalk between two previously unlinked innate immune receptor families: RIG-I, an anti-viral cytosolic receptor activated type I interferon production, and NLR (nucleotide-binding domain, leucine repeat domain-containing protein). We show that NLRP12 dampens RIG-I-mediated immune signaling against RNA viruses by controlling RIG-I's association with its adaptor MAVS. The nucleotide-binding domain of NLRP12 interacts with the ubiquitin ligase TRIM25 to prevent TRIM25-mediated, Lys63-linked ubiquitination and activation of RIG-I. NLRP12 also enhances RNF125-mediated, Lys48-linked degradative ubiquitination of RIG-I. Vesicular stomatitis virus (VSV) infection downregulates NLRP12 expression to allow RIG-I activation. Myeloid-cell-specific Nlrp12-deficient mice display a heightened interferon and TNF response and are more resistant to VSV infection. These results indicate that NLRP12 functions as a checkpoint for anti-viral RIG-I activation.
- Published
- 2019
10. The Innate Immune Sensor NLRC3 Acts as a Rheostat that Fine-Tunes T Cell Responses in Infection and Autoimmunity
- Author
-
W. June Brickey, Meng Deng, Xianming Tan, Kevin D. Cook, Yoshitaka Oyama, Haitao Guo, Alex Petrucelli, Elena Rampanelli, Ichiro Misumi, Jason W. Tam, Jenny P.-Y. Ting, Justin E. Wilson, Jason K. Whitmire, Brandon M. Johnson, Toru Uchimura, Wei-Chun Chou, and Liang Chen
- Subjects
0301 basic medicine ,Encephalomyelitis, Autoimmune, Experimental ,T-Lymphocytes ,T cell ,Immunology ,Autoimmunity ,Cell Cycle Proteins ,Inflammation ,Lymphocytic Choriomeningitis ,Biology ,medicine.disease_cause ,Article ,03 medical and health sciences ,Immune system ,NLRC3 ,medicine ,Animals ,Humans ,Lymphocytic choriomeningitis virus ,Immunology and Allergy ,Eukaryotic Initiation Factors ,Adaptor Proteins, Signal Transducing ,Mice, Knockout ,TNF Receptor-Associated Factor 6 ,Innate immune system ,Experimental autoimmune encephalomyelitis ,NF-kappa B ,Th1 Cells ,Phosphoproteins ,medicine.disease ,Immunity, Innate ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Intercellular Signaling Peptides and Proteins ,Th17 Cells ,Tumor necrosis factor alpha ,medicine.symptom ,Carrier Proteins - Abstract
NLRC3 limits inflammatory signaling in myeloid cells, but its role in T cells has been unclear. Uchimura et al. reveal that T cell expression of Nlrc3 restricts autoimmune and virus-specific CD4 + T cell responses by attenuating T cell signaling and metabolic pathways in CD4 + T cells. �� 2018 Elsevier Inc.Appropriate immune responses require a fine balance between immune activation and attenuation. NLRC3, a non-inflammasome-forming member of the NLR innate immune receptor family, attenuates inflammation in myeloid cells and proliferation in epithelial cells. T lymphocytes express the highest amounts of Nlrc3 transcript where its physiologic relevance is unknown. We show that NLRC3 attenuated interferon-�� and TNF expression by CD4 + T cells and reduced T helper 1 (Th1) and Th17 cell proliferation. Nlrc3 ���/��� mice exhibited increased and prolonged CD4 + T cell responses to lymphocytic choriomeningitis virus infection and worsened experimental autoimmune encephalomyelitis (EAE). These functions of NLRC3 were executed in a T-cell-intrinsic fashion: NLRC3 reduced K63-linked ubiquitination of TNF-receptor-associated factor 6 (TRAF6) to limit NF-��B activation, lowered phosphorylation of eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1), and diminished glycolysis and oxidative phosphorylation. This study reveals an unappreciated role for NLRC3 in attenuating CD4 + T cell signaling and metabolism.
- Published
- 2018
- Full Text
- View/download PDF
11. CD11b + Ly6C hi Ly6G − Immature Myeloid Cells Recruited in Response to Salmonella enterica Serovar Typhimurium Infection Exhibit Protective and Immunosuppressive Properties
- Author
-
Adrianus W. M. van der Velden, James B. Bliska, Amy L. Kullas, Patricio Mena, and Jason W. Tam
- Subjects
CD40 ,Immunology ,Biology ,Microbiology ,Molecular biology ,Interleukin 21 ,Infectious Diseases ,Antigen ,TheoryofComputation_ANALYSISOFALGORITHMSANDPROBLEMCOMPLEXITY ,biology.protein ,Interleukin 12 ,Cytotoxic T cell ,Parasitology ,Myeloid cell homeostasis ,Antigen-presenting cell ,Interleukin 3 - 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.
- Published
- 2014
12. Inflammatory monocytes provide a niche for Salmonella expansion in the lumen of the inflamed intestine
- Author
-
Jesse Leeds, Jason W. Tam, James B. Bliska, Brian P. Butler, Julie A. Bettke, Adrianus W. M. van der Velden, and Patrick A. McLaughlin
- Subjects
Bacterial Diseases ,Male ,Salmonella typhimurium ,Salmonella ,CCR2 ,Chemokine ,Nitric Oxide Synthase Type II ,Secretion Systems ,Pathology and Laboratory Medicine ,medicine.disease_cause ,Monocytes ,Mice ,chemistry.chemical_compound ,Mathematical and Statistical Techniques ,Microbial Physiology ,Medicine and Health Sciences ,Type III Secretion Systems ,Bacterial Physiology ,Intestinal Mucosa ,Biology (General) ,Immune Response ,Chemokine CCL2 ,Mice, Knockout ,0303 health sciences ,biology ,Statistics ,030302 biochemistry & molecular biology ,Animal Models ,Colitis ,Bacterial Pathogens ,3. Good health ,Nitric oxide synthase ,Chemistry ,Infectious Diseases ,Experimental Organism Systems ,Medical Microbiology ,Physical Sciences ,Female ,Anatomy ,Pathogens ,medicine.symptom ,Research Article ,Virulence Factors ,Receptors, CCR2 ,QH301-705.5 ,Immunology ,Lumen (anatomy) ,Mouse Models ,Inflammation ,Gastroenterology and Hepatology ,Research and Analysis Methods ,Models, Biological ,Microbiology ,Nitric oxide ,03 medical and health sciences ,Signs and Symptoms ,Model Organisms ,Enterobacteriaceae ,Diagnostic Medicine ,Virology ,Genetics ,medicine ,Animals ,Humans ,Secretion ,Statistical Methods ,Microbial Pathogens ,Molecular Biology ,030304 developmental biology ,Analysis of Variance ,Salmonella Infections, Animal ,Nitrates ,Bacteria ,Host Microbial Interactions ,Inflammatory Bowel Disease ,Chemical Compounds ,Organisms ,Biology and Life Sciences ,Bacteriology ,RC581-607 ,Gastrointestinal Tract ,Mice, Inbred C57BL ,chemistry ,Animal Studies ,biology.protein ,Parasitology ,Immunologic diseases. Allergy ,Digestive System ,Mathematics - 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., Author summary Salmonella exploit gut inflammation to edge out competing microbes in the intestinal lumen and establish infection. Notably, Salmonella use inflammation-derived nitrate for growth in the intestinal lumen. The generation of inflammation-derived nitrate is dependent on Nos2, which encodes inducible nitric oxide synthase (iNOS), an enzyme that catalyzes nitric oxide production. However, the cellular sources of iNOS and, therefore, the nitric oxide-derived nitrate used by Salmonella for growth in the inflamed intestine remain unidentified, presenting an important, long-standing open question. Here, we show that iNOS-producing inflammatory monocytes, which are phagocyte precursors that play a critical role in immunity and host defense, promote nitrate-dependent Salmonella expansion in the inflamed intestine, providing new insights into how Salmonella exploit gut inflammation to establish infection.
- Published
- 2019
13. Viral DNA Binding to NLRC3, an Inhibitory Nucleic Acid Sensor, Unleashes STING, a Cyclic Dinucleotide Receptor that Activates Type I Interferon
- Author
-
Pingwei Li, Cheng Zhu, Song Zhang, Hengming Ke, Jenny P.-Y. Ting, Joseph A. Duncan, Nikolay V. Dokholyan, Baoyu Zhao, Lu Zhang, Alex Petrucelli, Pablo Ariel, Jinyao Mo, Xin Li, Meng Deng, and Jason W. Tam
- Subjects
0301 basic medicine ,Immunology ,Biology ,Article ,Cell Line ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Interferon ,Cell Line, Tumor ,Nucleic Acids ,medicine ,NLRC3 ,Animals ,Humans ,Immunology and Allergy ,Innate immune system ,Membrane Proteins ,NOD-like receptor ,Immunity, Innate ,Cell biology ,Mice, Inbred C57BL ,HEK293 Cells ,030104 developmental biology ,Infectious Diseases ,chemistry ,030220 oncology & carcinogenesis ,Stimulator of interferon genes ,DNA, Viral ,Interferon Type I ,Nucleic acid ,Intercellular Signaling Peptides and Proteins ,Interferon type I ,DNA ,HeLa Cells ,Protein Binding ,medicine.drug - Abstract
Immune suppression is a crucial component of immunoregulation and a subgroup of nucleotide-binding domain (NBD), leucine-rich repeat (LRR)-containing proteins (NLRs) attenuate innate immunity. How this inhibitory function is controlled is unknown. A key question is whether microbial ligands can regulate this inhibition. NLRC3 is a negative regulator that attenuates type I interferon (IFN-I) response by sequestering and attenuating stimulator of interferon genes (STING) activation. Here, we report that NLRC3 binds viral DNA and other nucleic acids through its LRR domain. DNA binding to NLRC3 increases its ATPase activity, and ATP-binding by NLRC3 diminishes its interaction with STING, thus licensing an IFN-I response. This work uncovers a mechanism wherein viral nucleic acid binding releases an inhibitory innate receptor from its target.
- Published
- 2019
14. Phenotypic, Morphological, and Functional Heterogeneity of Splenic Immature Myeloid Cells in the Host Response to Tularemia
- Author
-
Nihal A. Okan, Patricio Mena, Adrianus W. M. van der Velden, Jorge L. Benach, Jason W. Tam, David G. Thanassi, Martha B. Furie, and John W. Rasmussen
- Subjects
T-Lymphocytes ,Immunology ,Population ,Spleen ,Microbiology ,Immunophenotyping ,Tularemia ,Mice ,medicine ,Animals ,Myeloid Cells ,Francisella tularensis ,education ,B-Lymphocytes ,Mice, Inbred BALB C ,Mice, Inbred C3H ,education.field_of_study ,CD11b Antigen ,biology ,Bacterial Infections ,biology.organism_classification ,medicine.disease ,Virology ,Phenotype ,Mice, Inbred C57BL ,Disease Models, Animal ,Infectious Diseases ,medicine.anatomical_structure ,Integrin alpha M ,biology.protein ,Female ,Receptors, Chemokine ,Parasitology ,Ex vivo - 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.
- Published
- 2012
15. The Inhibitory Innate Immune Sensor NLRP12 Maintains a Threshold against Obesity by Regulating Gut Microbiota Homeostasis
- Author
-
Ning Cheng, Alex Petrucelli, Hao Guo, Daniel E. Cooper, Mihai G. Netea, Mark J. Koenigsknecht, Wei-Chun Chou, Vincent B. Young, Liang Chen, Rinke Stienstra, Stephanie A. Montgomery, Justin E. Wilson, Rosalind A. Coleman, Jenny P.-Y. Ting, R. Balfour Sartor, Rongrong Liu, Agnieszka D. Truax, Jason W. Tam, W. June Brickey, and A. Alicia Koblansky
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Adipose tissue ,Inflammation ,Biology ,Gut flora ,Bio-Organic Chemistry ,Microbiology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Virology ,Internal medicine ,medicine ,Animals ,Homeostasis ,Humans ,Obesity ,Aged ,Mice, Knockout ,Innate immune system ,Bacteria ,Lachnospiraceae ,Intracellular Signaling Peptides and Proteins ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Inflammasome ,Middle Aged ,biology.organism_classification ,medicine.disease ,Immunity, Innate ,Gastrointestinal Microbiome ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,Adipose Tissue ,Female ,Parasitology ,medicine.symptom ,Dysbiosis ,030217 neurology & neurosurgery ,medicine.drug - Abstract
In addition to high-fat diet (HFD) and inactivity, inflammation and microbiota composition contribute to obesity. Inhibitory immune receptors, such as NLRP12, dampen inflammation and are important for resolving inflammation, but their role in obesity is unknown. We show that obesity in humans correlates with reduced expression of adipose tissue NLRP12. Similarly, Nlrp12 ���/��� mice show increased weight gain, adipose deposition, blood glucose, NF-��B/MAPK activation, and M1-macrophage polarization. Additionally, NLRP12 is required to mitigate HFD-induced inflammasome activation. Co-housing with wild-type animals, antibiotic treatment, or germ-free condition was sufficient to restrain inflammation, obesity, and insulin tolerance in Nlrp12 ���/��� mice, implicating the microbiota. HFD-fed Nlrp12 ���/��� mice display dysbiosis marked by increased obesity-associated Erysipelotrichaceae, but reduced Lachnospiraceae family and the associated enzymes required for short-chain fatty acid (SCFA) synthesis. Lachnospiraceae or SCFA administration attenuates obesity, inflammation, and dysbiosis. These findings reveal that Nlrp12 reduces HFD-induced obesity by maintaining beneficial microbiota. Truax et al. show that myeloid-expressed NLRP12 restrains high-fat-diet-induced obesity and type 2 diabetes by attenuating TNF, IL-6, NF-kB, MAPK, M1-macrophage polarization, and inflammasome activation in adipose tissue. This protective function of NLRP12 is microbiota dependent, and is associated with Lachnospiraceae and their metabolites, which mitigate obesity.
- Published
- 2018
16. CCR2+ Inflammatory Dendritic Cells and Translocation of Antigen by Type III Secretion Are Required for the Exceptionally Large CD8+ T Cell Response to the Protective YopE69-77 Epitope during Yersinia Infection
- Author
-
Patricio Mena, Jason W. Tam, Yue Zhang, Adrianus W. M. van der Velden, and James B. Bliska
- Subjects
lcsh:Immunologic diseases. Allergy ,Receptors, CCR2 ,Virulence Factors ,T cell ,Immunology ,Immunoblotting ,Epitopes, T-Lymphocyte ,Yersinia pseudotuberculosis Infections ,Biology ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Microbiology ,Epitope ,Mice ,Immune system ,Antigen ,Virology ,Genetics ,medicine ,Type III Secretion Systems ,Cytotoxic T cell ,Yersinia pseudotuberculosis ,Animals ,Molecular Biology ,lcsh:QH301-705.5 ,Inflammation ,Antigen Presentation ,Dendritic Cells ,Acquired immune system ,biology.organism_classification ,Flow Cytometry ,Molecular biology ,Adoptive Transfer ,Mice, Mutant Strains ,3. Good health ,Mice, Inbred C57BL ,Disease Models, Animal ,Protein Transport ,medicine.anatomical_structure ,lcsh:Biology (General) ,Parasitology ,Female ,lcsh:RC581-607 ,CD8 ,Bacterial Outer Membrane Proteins ,Research Article - 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., Author Summary Dendritic cells (DCs) direct host protective adaptive immune responses during infection. How different subpopulations of DCs contribute to the formation of antigen-specific CD8+ T cells is incompletely understood. Infection of C57BL/6 mice with the bacterial pathogen Yersinia pseudotuberculosis results in the production of an exceptionally large CD8+ T cell response to an epitope in the type III secretion system effector YopE. Here, we show that this large CD8+ T cell response requires translocation of YopE into inflammatory DCs, which express CCR2 and accumulate in infected tissues. In contrast, when mice are infected with a Y. pseudotuberculosis strain that can secrete but not translocate YopE, a smaller response is seen, and under these conditions the generation of YopE-specific CD8+ T cell requires CD8α+ DCs. Our results indicate that distinct DC subsets participate in constructing the CD8+ T cell response to secreted, versus translocated, YopE. Furthermore our data indicate that inflammatory DCs are a driving force behind the massive CD8+ T cell response to a protective epitope in a bacterial virulence factor that is translocated into host cells.
- Published
- 2015
17. CD11b+ Ly6Chi Ly6G- immature myeloid cells recruited in response to Salmonella enterica serovar Typhimurium infection exhibit protective and immunosuppressive properties
- Author
-
Jason W, Tam, Amy L, Kullas, Patricio, Mena, James B, Bliska, and Adrianus W M, van der Velden
- Subjects
Immunosuppression Therapy ,Salmonella typhimurium ,Analysis of Variance ,Host Response and Inflammation ,CD11b Antigen ,Receptors, CCR2 ,T-Lymphocytes ,Cell Differentiation ,Mice, Transgenic ,Immunity, Innate ,Mice, Inbred C57BL ,Mice ,Liver ,Salmonella Infections ,Animals ,Antigens, Ly ,Myeloid Cells ,Spleen ,Cell Proliferation - 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.
- Published
- 2014
18. Persistent salmonellosis causes pancreatitis in a murine model of infection
- Author
-
Jason W. Tam, Gangadaar Thotakura, Jason C. Hall, Howard C. Crawford, Adrianus W. M. van der Velden, and Kathleen E. DelGiorno
- Subjects
Lipopolysaccharides ,Salmonella typhimurium ,Bacterial Diseases ,Pathology ,Salmonellosis ,Population Modeling ,lcsh:Medicine ,Acinar Cells ,Pathogenesis ,medicine.disease_cause ,Pathology and Laboratory Medicine ,Fibrosis ,Salmonella ,Metaplasia ,Gastrointestinal Cancers ,Medicine and Health Sciences ,Gastrointestinal Infections ,lcsh:Science ,Pathogen ,Multidisciplinary ,3. Good health ,Bacterial Pathogens ,medicine.anatomical_structure ,Infectious Diseases ,Medical Microbiology ,Host-Pathogen Interactions ,medicine.symptom ,Anatomy ,Pancreas ,Bacterial and Foodborne Illness ,Biliary Disorders ,Research Article ,medicine.medical_specialty ,Inflammation ,Endocrine System ,Gastroenterology and Hepatology ,Biology ,Microbiology ,In vivo ,medicine ,Animals ,Humans ,Microbial Pathogens ,Salmonella Infections, Animal ,lcsh:R ,Biology and Life Sciences ,Computational Biology ,Pathogenic bacteria ,Bacteriology ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,Pancreatitis ,Immunology ,lcsh:Q ,Infectious Disease Modeling - 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.
- Published
- 2014
19. L-Asparaginase II Produced by Salmonella Typhimurium Inhibits T Cell Responses and Mediates Virulence
- Author
-
Michael McClelland, Steffen Porwollik, Adrianus W. M. van der Velden, Patricio Mena, Lydia M. Bogomolnaya, Jason W. Tam, AnnMarie Torres, Hee-Jeong Yang, Joseph B. McPhee, Helene Andrews-Polymenis, and Amy L. Kullas
- Subjects
Salmonella typhimurium ,Cancer Research ,Virulence Factors ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,Virulence ,Biology ,Microbiology ,Article ,Mice ,03 medical and health sciences ,Immune system ,Ammonia ,Immunology and Microbiology(all) ,Virology ,medicine ,Animals ,Asparaginase ,Molecular Biology ,Cell Proliferation ,Immune Evasion ,030304 developmental biology ,Aspartic Acid ,0303 health sciences ,030306 microbiology ,Cell growth ,T-cell receptor ,biology.organism_classification ,In vitro ,3. Good health ,Mice, Inbred C57BL ,Cytokine ,medicine.anatomical_structure ,Salmonella enterica ,Cytokines ,Parasitology ,Asparagine ,Gene Deletion - Abstract
SummarySalmonella 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.
- Full Text
- View/download PDF
20. Persistent salmonellosis causes pancreatitis in a murine model of infection.
- Author
-
Kathleen E DelGiorno, Jason W Tam, Jason C Hall, Gangadaar Thotakura, Howard C Crawford, and Adrianus W M van der Velden
- Subjects
Medicine ,Science - 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.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.