Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Inflamm Bowel Dis ; 21(12): 2758-65, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26222341

ABSTRACT

BACKGROUND: Intestinal inflammation is associated with systemic translocation of commensal antigens and the consequent activation of B and T lymphocytes. The long-term consequences of such immune activation are not completely understood. METHODS: C57BL/6 mice were subjected to 2 courses of treatment with dextran sulfate sodium (DSS) to induce colitis. Two to 7 weeks after the DSS treatment, the mice were infected intraperitoneally with Salmonella enterica serovar Typhimurium. The outcome of infection was evaluated based on survival and tissue pathogen burden. RESULTS: Mice that had recovered from DSS colitis displayed a significant increase in resistance to S. Typhimurium infection as indicated by improved survival and decreased tissue pathogen numbers. The colitis-induced increase in resistance to systemic salmonellosis lasted for as long as 7 weeks after discontinuing DSS and was dependent on T lymphocytes but not on B cells. Interestingly, depletion of CD4 and CD8 T cells just before the Salmonella infection did not alter the colitis-induced increase in resistance. Mice that had recovered from colitis had evidence of persistent activation of resident peritoneal macrophages and enhanced Salmonella-induced neutrophil recruitment to the peritoneum. Macrophage depletion with clodronate liposomes abrogated the colitis-induced increase in resistance to Salmonella. CONCLUSIONS: Taken together, our results indicate that DSS colitis leads to a long-lasting increase in resistance to Salmonella infection that is initiated in a T cell-dependent manner but is ultimately mediated independently of B and T cells as a result of persistent changes in innate immune cell function.


Subject(s)
B-Lymphocytes/immunology , Colitis/immunology , Macrophages/immunology , Salmonella Infections, Animal/immunology , T-Lymphocytes/immunology , Animals , Bacterial Load , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Colitis/chemically induced , Colitis/microbiology , Dextran Sulfate , Disease Models, Animal , Intestines/immunology , Male , Mice , Mice, Inbred C57BL , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/immunology
2.
Eur J Immunol ; 45(9): 2553-67, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26046550

ABSTRACT

Orally administrated iron is suspected to increase susceptibility to enteric infections among children in infection endemic regions. Here we investigated the effect of dietary iron on the pathology and local immune responses in intestinal infection models. Mice were held on iron-deficient, normal iron, or high iron diets and after 2 weeks they were orally challenged with the pathogen Citrobacter rodentium. Microbiome analysis by pyrosequencing revealed profound iron- and infection-induced shifts in microbiota composition. Fecal levels of the innate defensive molecules and markers of inflammation lipocalin-2 and calprotectin were not influenced by dietary iron intervention alone, but were markedly lower in mice on the iron-deficient diet after infection. Next, mice on the iron-deficient diet tended to gain more weight and to have a lower grade of colon pathology. Furthermore, survival of the nematode Caenorhabditis elegans infected with Salmonella enterica serovar Typhimurium was prolonged after iron deprivation. Together, these data show that iron limitation restricts disease pathology upon bacterial infection. However, our data also showed decreased intestinal inflammatory responses of mice fed on high iron diets. Thus additionally, our study indicates that the effects of iron on processes at the intestinal host-pathogen interface may highly depend on host iron status, immune status, and gut microbiota composition.


Subject(s)
Caenorhabditis elegans/drug effects , Enterobacteriaceae Infections/pathology , Intestinal Mucosa/pathology , Intestines/pathology , Iron, Dietary/administration & dosage , Salmonella Infections, Animal/metabolism , Acute-Phase Proteins/biosynthesis , Acute-Phase Proteins/immunology , Animals , Body Weight/immunology , Caenorhabditis elegans/immunology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/microbiology , Citrobacter rodentium/immunology , Diet/methods , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/metabolism , Enterobacteriaceae Infections/microbiology , Feces/microbiology , Female , Immunity, Innate , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestines/immunology , Intestines/microbiology , Iron, Dietary/adverse effects , Leukocyte L1 Antigen Complex/biosynthesis , Leukocyte L1 Antigen Complex/immunology , Lipocalin-2 , Lipocalins/biosynthesis , Lipocalins/immunology , Mice , Mice, Inbred C57BL , Oncogene Proteins/biosynthesis , Oncogene Proteins/immunology , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/mortality , Salmonella typhimurium/immunology , Survival Analysis
3.
Clin Vaccine Immunol ; 21(10): 1437-42, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25121777

ABSTRACT

Individuals with human immunodeficiency virus (HIV) infection have increased susceptibility to invasive disease caused by Salmonella enterica serovar Typhimurium. Studies from Africa have suggested that this susceptibility is related in part to the development of a high level of lipopolysaccharide (LPS)-specific IgG that is able to inhibit the killing of S. Typhimurium by bactericidal antibodies in healthy individuals. To explore this issue further, we examined the bactericidal activity against S. Typhimurium using serum and plasma samples from healthy controls and various clinical subgroups of HIV-infected adults in the United States. We found that the bactericidal activity in the samples from HIV-positive elite controllers was comparable to that from healthy individuals, whereas it was significantly reduced in HIV-positive viremic controllers and untreated chronic progressors. As demonstrated previously for healthy controls, the bactericidal activity of the plasma from the elite controllers was inhibited by preincubation with S. Typhimurium LPS, suggesting that it was mediated by anti-LPS antibodies. S. Typhimurium LPS-specific IgG was significantly reduced in all subgroups of HIV-infected individuals. Interestingly, and in contrast to the healthy controls, plasma from all HIV-positive subgroups inhibited in vitro killing of S. Typhimurium by plasma from a healthy individual. Our results, together with the findings from Africa, suggest that multiple mechanisms may be involved in the HIV-induced dysregulation of humoral immunity to S. Typhimurium.


Subject(s)
Antibodies, Bacterial/blood , Blood Bactericidal Activity , HIV Infections/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Adult , Africa , Humans , Immunoglobulin G/blood , Lipopolysaccharides/immunology , Plasma/immunology , United States
4.
J Immunol ; 193(3): 1398-407, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24973448

ABSTRACT

States of chronic inflammation such as inflammatory bowel disease are often associated with dysregulated iron metabolism and the consequent development of an anemia that is caused by maldistribution of iron. Abnormally elevated expression of the hormone hepcidin, the central regulator of systemic iron homeostasis, has been implicated in these abnormalities. However, the mechanisms that regulate hepcidin expression in conditions such as inflammatory bowel disease are not completely understood. To clarify this issue, we studied hepcidin expression in mouse models of colitis. We found that dextran sulfate sodium-induced colitis inhibited hepcidin expression in wild-type mice but upregulated it in IL-10-deficient animals. We identified two mechanisms contributing to this difference. Firstly, erythropoietic activity, as indicated by serum erythropoietin concentrations and splenic erythropoiesis, was higher in the wild-type mice, and pharmacologic inhibition of erythropoiesis prevented colitis-associated hepcidin downregulation in these animals. Secondly, the IL-10 knockout mice had higher expression of multiple inflammatory genes in the liver, including several controlled by STAT3, a key regulator of hepcidin. The results of cohousing and fecal transplantation experiments indicated that the microbiota was involved in modulating the expression of hepcidin and other STAT3-dependent hepatic genes in the context of intestinal inflammation. Our observations thus demonstrate the importance of erythropoietic activity and the microbiota in influencing hepcidin expression during colitis and provide insight into the dysregulated iron homeostasis seen in inflammatory diseases.


Subject(s)
Erythropoiesis/immunology , Erythropoietin/metabolism , Hepcidins/genetics , Inflammation Mediators/blood , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/immunology , Iron/physiology , Microbiota/immunology , Animals , Bacteroides fragilis/immunology , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Dextran Sulfate/administration & dosage , Disease Models, Animal , Erythropoiesis/genetics , Erythropoietin/blood , Female , Hepcidins/biosynthesis , Homeostasis/immunology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Interleukin-10/deficiency , Interleukin-10/genetics , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , STAT3 Transcription Factor/physiology , Streptococcaceae/immunology
5.
Clin Vaccine Immunol ; 20(10): 1491-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23803904

ABSTRACT

Recent observations from Africa have rekindled interest in the role of serum bactericidal antibodies in protecting against systemic infection with Salmonella enterica serovar Typhimurium. To determine whether the findings are applicable to other populations, we analyzed serum samples collected from healthy individuals in the United States. We found that all but 1 of the 49 adult samples tested had robust bactericidal activity against S. Typhimurium in a standard in vitro assay. The activity was dependent on complement and could be reproduced by immunoglobulin G (IgG) purified from the sera. The bactericidal activity was inhibited by competition with soluble lipopolysaccharide (LPS) from S. Typhimurium but not from Escherichia coli, consistent with recognition of a determinant in the O-antigen polysaccharide. Sera from healthy children aged 10 to 48 months also had bactericidal activity, although it was significantly less than in the adults, correlating with lower levels of LPS-specific IgM and IgG. The lone sample in our collection that lacked bactericidal activity was able to inhibit killing of S. Typhimurium by the other sera. The inhibition correlated with the presence of an LPS-specific IgM and was associated with decreased complement deposition on the bacterial surface. Our results indicate that healthy individuals can have circulating antibodies to LPS that either mediate or inhibit killing of S. Typhimurium. The findings contrast with the observations from Africa, which linked bactericidal activity to antibodies against an S. Typhimurium outer membrane protein and correlated the presence of inhibitory anti-LPS antibodies with human immunodeficiency virus infection.


Subject(s)
Blood Bactericidal Activity , Immunoglobulins/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Serum/immunology , Adolescent , Adult , Aged , Child, Preschool , Complement System Proteins/immunology , Female , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Infant , Male , Middle Aged , United States , Young Adult
6.
Infect Immun ; 81(1): 166-72, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23115043

ABSTRACT

Melanin-concentrating hormone (MCH) was initially identified in mammals as a hypothalamic neuropeptide regulating appetite and energy balance. However, the wide distribution of MCH receptors in peripheral tissues suggests additional functions for MCH which remain largely unknown. We have previously reported that mice lacking MCH develop attenuated intestinal inflammation when exposed to Clostridium difficile toxin A. To further characterize the role of MCH in host defense mechanisms against intestinal pathogens, Salmonella enterocolitis (using Salmonella enterica serovar Typhimurium) was induced in MCH-deficient mice and their wild-type littermates. In the absence of MCH, infected mice had increased mortality associated with higher bacterial loads in blood, liver, and spleen. Moreover, the knockout mice developed more-severe intestinal inflammation, based on epithelial damage, immune cell infiltrates, and local and systemic cytokine levels. Paradoxically, these enhanced inflammatory responses in the MCH knockout mice were associated with disproportionally lower levels of macrophages infiltrating the intestine. Hence, we investigated potential direct effects of MCH on monocyte/macrophage functions critical for defense against intestinal pathogens. Using RAW 264.7 mouse monocytic cells, which express endogenous MCH receptor, we found that treatment with MCH enhanced the phagocytic capacity of these cells. Taken together, these findings reveal a previously unappreciated role for MCH in host-bacterial interactions.


Subject(s)
Hypothalamic Hormones/immunology , Hypothalamic Hormones/metabolism , Melanins/immunology , Melanins/metabolism , Pituitary Hormones/immunology , Pituitary Hormones/metabolism , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/metabolism , Salmonella typhimurium/immunology , Animals , Cell Movement/immunology , Cytokines/immunology , Cytokines/metabolism , Disease Susceptibility/immunology , Inflammation/immunology , Inflammation/metabolism , Inflammation/microbiology , Intestinal Mucosa/metabolism , Intestines/immunology , Intestines/microbiology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Monocytes/microbiology , Phagocytosis/immunology , Receptors, Somatostatin/immunology , Receptors, Somatostatin/metabolism , Salmonella Infections, Animal/microbiology
7.
PLoS One ; 7(5): e38136, 2012.
Article in English | MEDLINE | ID: mdl-22675442

ABSTRACT

BACKGROUND: Abnormal expression of the liver peptide hormone hepcidin, a key regulator of iron homeostasis, contributes to the pathogenesis of anemia in conditions such as inflammatory bowel disease (IBD). Since little is known about the mechanisms that control hepcidin expression during states of intestinal inflammation, we sought to shed light on this issue using mouse models. METHODOLOGY/PRINCIPAL FINDINGS: Hepcidin expression was evaluated in two types of intestinal inflammation caused by innate immune activation-dextran sulfate sodium (DSS)-induced colitis in wild-type mice and the spontaneous colitis occurring in T-bet/Rag2-deficient (TRUC) mice. The role of tumor necrosis factor (TNF) α was investigated by in vivo neutralization, and by treatment of a hepatocyte cell line, as well as mice, with the recombinant cytokine. Expression and activation of Smad1, a positive regulator of hepcidin transcription, were assessed during colitis and following administration or neutralization of TNFα. Hepcidin expression progressively decreased with time during DSS colitis, correlating with changes in systemic iron distribution. TNFα inhibited hepcidin expression in cultured hepatocytes and non-colitic mice, while TNFα neutralization during DSS colitis increased it. Similar results were obtained in TRUC mice. These effects involved a TNFα-dependent decrease in Smad1 protein but not mRNA. CONCLUSIONS/SIGNIFICANCE: TNFα inhibits hepcidin expression in two distinct types of innate colitis, with down-regulation of Smad1 protein playing an important role in this process. This inhibitory effect of TNFα may be superseded by other factors in the context of T cell-mediated colitis given that in the latter form of intestinal inflammation hepcidin is usually up-regulated.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Colitis/genetics , Gene Expression Regulation/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antimicrobial Cationic Peptides/metabolism , Bone Morphogenetic Proteins/metabolism , Cell Line , Colitis/chemically induced , Colitis/metabolism , Dextran Sulfate/adverse effects , Disease Models, Animal , Female , Hepcidins , Humans , Iron/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/drug effects , Smad Proteins/metabolism
8.
Inflamm Bowel Dis ; 18(1): 112-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21351217

ABSTRACT

BACKGROUND: A debilitating anemia associated with low serum iron often accompanies inflammatory bowel disease (IBD). Increased production of the iron regulatory hormone hepcidin is implicated in its pathogenesis and may also contribute to the inflammatory process itself. Hepcidin expression is dependent on bone morphogenetic proteins (BMPs) like BMP6, but the mechanisms that increase hepcidin levels during intestinal inflammation are not clear. Here we test the hypothesis that inhibiting hepcidin expression may have beneficial effects in IBD, and also shed light on the mechanism of colitis-induced hepcidin upregulation. METHODS: Mice with T cell transfer colitis were treated with vehicle or one of three anti-BMP reagents: HJV.Fc, a recombinant protein that prevents binding of BMPs to their receptor, LDN-193189, a small molecule inhibitor of BMP signal transduction, and an anti-BMP6 antibody. The effects of these reagents on colitis severity, liver hepcidin mRNA, and serum iron were determined. The mechanism of hepcidin upregulation was investigated by examining BMP6 expression and activity and the effects of IL-6 deficiency. RESULTS: All the anti-BMP reagents inhibited hepcidin expression and increased serum iron levels in the colitic mice. They also produced modest reductions in colon inflammatory cytokine expression. Although hepcidin upregulation during colitis was dependent on BMP6, it was not associated with increased BMP6 expression or activity. IL-6 was required for increased hepcidin expression during colitis. CONCLUSIONS: Inhibiting hepcidin expression may help to correct the anemia of IBD and may also attenuate intestinal inflammation. The mechanism of colitis-induced hepcidin upregulation involves both BMP6 and IL-6.


Subject(s)
Anti-Bacterial Agents/metabolism , Antimicrobial Cationic Peptides/metabolism , Bone Morphogenetic Protein 6/metabolism , Colitis/pathology , Inflammatory Bowel Diseases/pathology , Animals , Anti-Bacterial Agents/antagonists & inhibitors , Antimicrobial Cationic Peptides/antagonists & inhibitors , Antimicrobial Cationic Peptides/genetics , Blotting, Western , Bone Morphogenetic Protein 6/antagonists & inhibitors , Bone Morphogenetic Protein 6/genetics , Colitis/etiology , Colitis/metabolism , Hepcidins , Inflammatory Bowel Diseases/metabolism , Interleukin-6/metabolism , Iron/blood , Male , Mice , Mice, Inbred C57BL , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction , T-Lymphocytes/transplantation
9.
J Innate Immun ; 3(5): 437-46, 2011.
Article in English | MEDLINE | ID: mdl-21494017

ABSTRACT

Lipopolysaccharide (LPS)-induced production of tumor necrosis factor (TNF)-α requires the recruitment of two pairs of adaptors to the Toll-like receptor 4 cytoplasmic domain. The contribution of one pair - Toll-interleukin-1 receptor domain-containing adaptor inducing interferon-ß (TRIF) and TRIF-related adaptor molecule (TRAM) - to TNF-α expression is not well understood. To clarify this issue, we studied TRAM knockout bone marrow-derived macrophages (BMDM). LPS-stimulated TRAM-deficient BMDM had decreased TNF-α protein expression even at times when TNF-α mRNA levels were normal, suggesting impaired translation. Consistent with this idea, knockdown of TRAM in RAW264.7 macrophages decreased translation of a reporter controlled by the TNF-α 3' untranslated region, while transfection of TRAM in HEK293T cells increased translation of this reporter. Also consistent with a role for TRAM in TNF-α translation, LPS-induced activation of MK2, a kinase involved in this process, was impaired in TRAM-deficient BMDM. TRIF did not increase translation of the TNF-α 3' untranslated region reporter when expressed in HEK293T cells. However, BMDM that lacked functional TRIF produced reduced levels of TNF-α protein in response to LPS despite normal amounts of the mRNA. Unlike BMDM, LPS-stimulated TRAM-deficient peritoneal macrophages displayed equivalent reductions in TNF-α protein and mRNA. Our results indicate that TRAM- and TRIF-dependent signals have a previously unappreciated, cell type-specific role in regulating TNF-α translation.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Lipopolysaccharides/immunology , Macrophages/metabolism , Receptors, Interleukin/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/metabolism , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Biosynthesis , Protein Serine-Threonine Kinases/metabolism , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Transgenes/genetics , Tumor Necrosis Factor-alpha/immunology
10.
FEMS Immunol Med Microbiol ; 58(1): 138-45, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19863663

ABSTRACT

Siderocalin is a secreted protein that binds to siderophores to prevent bacterial iron acquisition. While it has been shown to inhibit the growth of Mycobacterium tuberculosis (M.tb) in extracellular cultures, its effect on this pathogen within macrophages is not clear. Here, we show that siderocalin expression is upregulated following M.tb infection of mouse macrophage cell lines and primary murine alveolar macrophages. Furthermore, siderocalin added exogenously as a recombinant protein or overexpressed in the RAW264.7 macrophage cell line inhibited the intracellular growth of the pathogen. A variant form of siderocalin, which is expressed only in the macrophage cytosol, inhibited intracellular M.tb growth as effectively as the normal, secreted form, an observation that provides mechanistic insight into how siderocalin might influence iron acquisition by the bacteria in the phagosome. Our findings are consistent with an important role for siderocalin in protection against M.tb infection and suggest that exogenously administered siderocalin may have therapeutic applications in tuberculosis.


Subject(s)
Carrier Proteins/physiology , Macrophages, Alveolar/microbiology , Mycobacterium tuberculosis/growth & development , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Cell Line , Cell Proliferation , Female , Humans , Lipocalin-2 , Lipocalins/metabolism , Lipocalins/pharmacology , Lung/metabolism , Mice , Mice, Inbred C57BL , Mutation , Mycobacterium tuberculosis/drug effects , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology
11.
J Clin Invest ; 119(11): 3322-8, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19809161

ABSTRACT

Mice deficient in the hemochromatosis gene, Hfe, have attenuated inflammatory responses to Salmonella infection associated with decreased macrophage TNF-alpha and IL-6 biosynthesis after exposure to LPS. In this study, we show that the abnormal cytokine production is related to impaired TLR4 signaling. Despite their abnormal response to LPS, Hfe KO macrophages produced amounts of TNF-alpha similar to those in WT cells after TLR2 stimulation. Consistent with this finding, LPS-induced activation of Mal/MyD88-dependent events was normal in the mutant macrophages. However, LPS-induced IFN-beta expression, a TRAM/TRIF-dependent response activated by TLR4, was reduced by Hfe deficiency. This reduction could be replicated in WT macrophages with the use of iron chelators. In contrast, TLR3-activated expression of IFN-beta, a TRIF-dependent response, was normal in Hfe KO macrophages and was unaffected by iron chelation. Our data suggest that low intracellular iron selectively impairs signaling via the TLR4/TRAM/TRIF pathway proximal to TRIF and results in reduced LPS-induced cytokine expression. Furthermore, by mimicking the altered iron metabolism associated with Hfe deficiency, we found that 3 different inhibitors of hepcidin attenuated Salmonella-induced and noninfectious enterocolitis. Thus, manipulation of iron homeostasis could represent a new therapeutic approach to controlling inflammation.


Subject(s)
Gene Expression Regulation , Homeostasis/immunology , Iron/immunology , Toll-Like Receptor 4/immunology , Animals , Enterocolitis/genetics , Enterocolitis/immunology , Gene Expression Regulation/drug effects , Hemochromatosis/genetics , Immunity, Innate/genetics , Inflammation/immunology , Lipopolysaccharides/pharmacology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Salmonella Infections/genetics , Salmonella Infections/immunology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...